1
|
Lu Y, Shen M, Yang Z, Zhang X, Li D, Wei Z, Li B, Zhao X, Wu N, Wu B, Yu W, Zhong Y. Optic disc changes in Chinese patients with NLRP3-associated autoinflammatory disease. Ann Med 2025; 57:2438842. [PMID: 39673196 DOI: 10.1080/07853890.2024.2438842] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2023] [Revised: 08/23/2024] [Accepted: 11/25/2024] [Indexed: 12/16/2024] Open
Abstract
OBJECTIVE To investigate the optic disc changes (ODC) in Chinese patients with NLRP3-associated autoinflammatory disease (NLRP3-AID). METHODS Patients who were diagnosed with NLRP3-AID at the Department of Rheumatology, Peking Union Medical College Hospital between April 2015 and December 2022 were retrospectively reviewed and analyzed. RESULTS A total of 20 patients were enrolled in this retrospective study. All 20 patients had a moderate MWS NLRP3-AID phenotype. Thirteen patients (65%) had ocular involvements. The interval between symptoms onset and diagnosis was significantly longer in patients with ocular involvement than in patients without (p = 0.044). The incidence of hearing loss was significantly higher in patients with ocular involvement (p = 0.017), while the incidence of abdominal pain was significantly lower when compared to patients without ocular involvement (p = 0.007). Optic disc swelling (ODS) (50%) was the most common ODC. All of the four T348M mutation carriers within our cohort exhibited ODS with visual-field defects. There was a significant difference between patients with/without ODS regarding the number of patients carrying T348M mutation (p = 0.014). The occurrence of hearing loss and CNS involvement was significantly higher in the group with ODS compared to the group without (p = 0.0014, p = 0.0198). Of the eight patients who underwent lumbar puncture, five presented with intracranial hypertension (IH). ODS was observed in all patients with IH. The serum inflammatory markers were significantly higher in patients with ODS than in those without. Two patients receiving regular subcutaneous IL-1 inhibitor treatment showed improvements in ODC. CONCLUSIONS ODC is common among Chinese patients with NLRP3-AID, with ODS being the most common manifestation. Hearing loss and CNS involvement often accompany the occurrence of ODS. The serum inflammatory markers are associated with ODS. The T348M mutation is more likely to lead to ODC with visual-field defects.
Collapse
Affiliation(s)
- Yuezhu Lu
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Min Shen
- Department of Rare Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, PUMCH, Beijing, China
| | - Zhikun Yang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xiao Zhang
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Donghui Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Zhangwanyu Wei
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Bing Li
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Xufeng Zhao
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Na Wu
- Department of Rare Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, PUMCH, Beijing, China
| | - Bingxuan Wu
- Department of Rare Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences, Peking Union Medical College, Beijing, China
- Department of Rheumatology and Clinical Immunology, National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology, Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, PUMCH, Beijing, China
| | - Weihong Yu
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| | - Yong Zhong
- Department of Ophthalmology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences, Beijing, China
- Key Laboratory of Ocular Fundus Diseases, State Key Laboratory of Complex Severe and Rare Diseases, Chinese Academy of Medical Sciences, Beijing, China
| |
Collapse
|
2
|
Su K, Tang M, Wu J, Ye N, Jiang X, Zhao M, Zhang R, Cai X, Zhang X, Li N, Peng J, Lin L, Wu W, Ye H. Mechanisms and therapeutic strategies for NLRP3 degradation via post-translational modifications in ubiquitin-proteasome and autophagy lysosomal pathway. Eur J Med Chem 2025; 289:117476. [PMID: 40056798 DOI: 10.1016/j.ejmech.2025.117476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 02/20/2025] [Accepted: 03/03/2025] [Indexed: 03/10/2025]
Abstract
The NLRP3 inflammasome is crucial for immune responses. However, its overactivation can lead to severe inflammatory diseases, underscoring its importance as a target for therapeutic intervention. Although numerous inhibitors targeting NLRP3 exist, regulating its degradation offers an alternative and promising strategy to suppress its activation. The degradation of NLRP3 is primarily mediated by the proteasomal and autophagic pathways. The review not only elaborates on the traditional concepts of ubiquitination and NLRP3 degradation but also investigates the important roles of indirect regulatory modifications, such as phosphorylation, acetylation, ubiquitin-like modifications, and palmitoylation-key post-translational modifications (PTMs) that influence NLRP3 degradation. Additionally, we also discuss the potential targets that may affect NLRP3 degradation during the proteasomal and autophagic pathways. By unraveling these complex regulatory mechanisms, the review aims to enhance the understanding of NLRP3 regulation and its implications for developing therapeutic strategies to combat inflammatory diseases.
Collapse
Affiliation(s)
- Kaiyue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Minghai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jie Wu
- Key Laboratory of Hydrodynamics (Ministry of Education), School of Ocean and Civil Engineering, Shanghai Jiao Tong University, Shanghai, 200240, China
| | - Neng Ye
- Scaled Manufacturing Center of Biological Products, Management Office of National Facility for Translational Medicine, West China Hospital, Sichuan University Chengdu 610041, China
| | - Xueqin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Min Zhao
- Laboratory of Metabolomics and Drug-induced Liver Injury, Department of Gastroenterology & Hepatology, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ruijia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiaoying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xinlu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Lei Lin
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Wenshuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China.
| | - Haoyu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China.
| |
Collapse
|
3
|
Chen Y, Chen S, Liu Z, Wang Y, An N, Chen Y, Peng Y, Liu Z, Liu Q, Hu X. Red blood cells undergo lytic programmed cell death involving NLRP3. Cell 2025:S0092-8674(25)00389-7. [PMID: 40252640 DOI: 10.1016/j.cell.2025.03.039] [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: 08/23/2023] [Revised: 08/27/2024] [Accepted: 03/24/2025] [Indexed: 04/21/2025]
Abstract
The canonical complement-mediated lysis of mature red blood cells (RBCs) leads to severe pathogenesis. However, inhibition strategies targeting complement are not always as efficient as expected, indicating that unknown mechanisms are awaiting elucidation. In this study, we investigate the intracellular events in mature RBCs following complement activation. The collected evidence demonstrates that complement-induced hemolysis is a caspase-8-dependent programmed RBC death. Furthermore, short NLRP3 (miniNLRP3) fragments in RBCs are identified to engage in the assembly of NLRP3-apoptosis-associated speck-like protein containing a CARD (ASC)-caspase-8 complex. Activated caspase-8 directly induces the proteolysis of β-spectrin, thereby disrupting the skeletal network of the RBC membrane, a process we refer to as spectosis. Spectosis signaling is also activated in autoimmune hemolytic anemia or paroxysmal nocturnal hemoglobinuria, and the inhibition of spectosis significantly reduced complement-induced hemolysis. These findings reveal a programmed death cascade in mature RBCs, which may have important implications for the treatment of hemolytic disorders.
Collapse
Affiliation(s)
- Yaozhen Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Shouwen Chen
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China.
| | - Zhixin Liu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yafen Wang
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Ning An
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yutong Chen
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China
| | - Yihao Peng
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen 518115, Guangdong, China
| | - Zheng Liu
- Kobilka Institute of Innovative Drug Discovery, School of Medicine, Chinese University of Hong Kong, Shenzhen 518115, Guangdong, China
| | - Qin Liu
- State Key Laboratory of Bioreactor Engineering, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, East China University of Science and Technology, Shanghai 200237, China.
| | - Xingbin Hu
- Department of Transfusion Medicine, Xijing Hospital, Fourth Military Medical University, Xi'an 710032, Shaanxi, China.
| |
Collapse
|
4
|
Jing J, Yang F, Wang K, Cui M, Kong N, Wang S, Qiao X, Kong F, Zhao D, Ji J, Tang L, Gao J, Cong Y, Ding D, Chen K. UFMylation of NLRP3 Prevents Its Autophagic Degradation and Facilitates Inflammasome Activation. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2406786. [PMID: 39985286 PMCID: PMC12005806 DOI: 10.1002/advs.202406786] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 01/29/2025] [Indexed: 02/24/2025]
Abstract
NLRP3 (NOD, LRR and pyrin domain-containing protein 3) inflammasome is important for host defense against infections and maintaining homeostasis. Aberrant activation of NLRP3 inflammasome is closely related to various inflammatory diseases. Post-translational modifications are critical for NLRP3 inflammasome regulation. However, the mechanism of NLRP3 inflammasome activation remains incompletely understood. Here, it is demonstrated that the Ufm1 E3 ligase Ufl1 mediated UFMylation is essential for NLRP3 inflammasome activation. Mechanistically, Ufl1 binds and UFMylates NLRP3 in the priming stage of NLRP3 activation, thereby sustaining the stability of NLRP3 by preventing NLRP3 K63-linked ubiquitination and the subsequent autophagic degradation. It is further demonstrated that myeloid cell-specific Ufl1 or Ufm1 deficiency in mice significantly alleviated inflammatory responses and tissue damage following lipopolysaccharide (LPS)-induced endotoxemia and alum-induced peritonitis. Thus, the findings offer new insights into potential therapeutic targets for NLRP3 inflammasome-related diseases by targeting the UFMylation system.
Collapse
Affiliation(s)
- Jiongjie Jing
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Fan Yang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Ke Wang
- Shanghai Key Laboratory of Maternal Fetal MedicineClinical and Translational Research Center of Shanghai First Maternity and Infant HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Mintian Cui
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Ni Kong
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Shixi Wang
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Xiaoyue Qiao
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
| | - Fanyu Kong
- Department of Internal Emergency MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
| | - Dongyang Zhao
- Department of Internal Emergency MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
| | - Jinlu Ji
- Department of Internal Emergency MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
| | - Lunxian Tang
- Department of Internal Emergency MedicineShanghai East HospitalSchool of MedicineTongji UniversityShanghai200120China
| | - Jiaxin Gao
- State Key Laboratory of MycologyInstitute of MicrobiologyChinese Academy of SciencesBeijing100101China
| | - Yu‐Sheng Cong
- Key Laboratory of Aging and Cancer Biology of Zhejiang ProvinceInstitute of Aging ResearchSchool of MedicineHangzhou Normal UniversityHangzhou311121China
| | - Deqiang Ding
- Shanghai Key Laboratory of Maternal Fetal MedicineClinical and Translational Research Center of Shanghai First Maternity and Infant HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
- Shanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| | - Kun Chen
- State Key Laboratory of Cardiovascular Diseases and Medical Innovation CenterShanghai East HospitalSchool of Life Sciences and TechnologyTongji UniversityShanghai200127China
- Shanghai Key Laboratory of Signaling and Disease ResearchFrontier Science Center for Stem Cell ResearchSchool of Life Sciences and TechnologyTongji UniversityShanghai200092China
| |
Collapse
|
5
|
Öksel A, Şahin N, Günlemez A. When should a neonatologist consult a rheumatologist? Eur J Pediatr 2025; 184:256. [PMID: 40100445 PMCID: PMC11920313 DOI: 10.1007/s00431-025-06086-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/12/2024] [Revised: 02/28/2025] [Accepted: 03/06/2025] [Indexed: 03/20/2025]
Abstract
Pediatric rheumatologic diseases are complex conditions that can present with various clinical manifestations, including fever, rash, joint involvement, and diarrhea, impacting more than one organ system and affecting all pediatric age groups from 0 to 18 years. This review focuses on rheumatologic diseases in neonates, encompassing both primary neonatal-onset conditions and those influenced by maternal autoimmune diseases and treatments during pregnancy. Diagnosing rheumatologic diseases in neonates is challenging due to their nonspecific symptoms, which can overlap with other conditions. While primary neonatal-onset diseases such as cryopyrin-associated periodic syndromes (CAPS), deficiency of IL-1 receptor antagonist (DIRA), and neonatal-onset juvenile idiopathic arthritis (JIA) are rare, maternal autoimmune diseases and their treatments can also impact neonatal health. Conditions like systemic lupus erythematosus (SLE) and antiphospholipid syndrome (APS) may increase neonatal risks, leading to complications such as thrombosis or pregnancy loss. Identifying these conditions early and providing the proper care is crucial to reduce morbidity and mortality in this vulnerable group. CONCLUSION Persistent fever, rash, or unexplained joint involvement warrants early referral to a pediatric rheumatologist. A multidisciplinary approach involving obstetricians, rheumatologists, and neonatologists is essential for timely diagnosis and optimal neonatal outcomes. WHAT IS KNOWN • Diagnosis of neonatal rheumatologic diseases is difficult because their symptoms are nonspecific and may overlap with other neonatal diseases. • Maternal autoantibodies transmitted through the placenta may lead to neonatal complications (e.g. congenital heart block, thrombosis). WHAT IS NEW • Long-term follow-up of autoinflammatory diseases is essential, as the absence of neonatal-specific damage indices limits the ability to assess disease progression and treatment outcomes, underscoring the need for validated scoring systems tailored to neonates. • Novel biomarkers, such as elevated levels of cord C-reactive protein, NT-proBNP, MMP-2, uPA, uPAR, and plasminogen, have been identified, offering new insights into potential diagnostic tools for cardiac neonatal lupus.
Collapse
Affiliation(s)
- Ali Öksel
- Medicalpark Hospital, Pediatrics, Kocaeli, Turkey.
| | - Nihal Şahin
- Department of Pediatric Rheumatology, Kocaeli University, Kocaeli, Turkey
| | - Ayla Günlemez
- Department of Neoanatology, Kocaeli University, Kocaeli, Turkey
| |
Collapse
|
6
|
Zhang J, Shen M. The Role of IL-17 in Systemic Autoinflammatory Diseases: Mechanisms and Therapeutic Perspectives. Clin Rev Allergy Immunol 2025; 68:27. [PMID: 40074883 DOI: 10.1007/s12016-025-09042-5] [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] [Accepted: 03/03/2025] [Indexed: 03/14/2025]
Abstract
Interleukin (IL)-17, a pro-inflammatory cytokine, plays a pivotal role in immune regulation by bridging innate and adaptive responses. Beyond its canonical involvement in T helper-17 cells-mediated immunity, IL-17 contributes significantly to the pathogenesis of systemic autoinflammatory diseases (SAIDs) including Familial Mediterranean Fever (FMF), nucleotide-binding oligomerization domain-like receptor protein 3 (NLRP3)-associated autoinflammatory diseases, and synovitis, acne, pustulosis, hyperostosis, and osteitis (SAPHO) syndrome. Dysregulated IL-17 signaling drives inflammasome activation, neutrophil recruitment, and chronic tissue inflammation. IL-17 inhibitors have demonstrated efficacy in refractory SAIDs, though challenges such as increased infection risks, paradoxical inflammatory reactions, and uncertainties regarding long-term safety persist. Currently, there is insufficient data to support the use of IL-17 inhibitors as first-line treatments, and their role in managing SAIDs is yet to be fully defined. This review highlights the mechanistic role of IL-17 in SAIDs and emerging therapeutic strategies, including IL-17-targeted monotherapies and combination approaches with IL-1 or tumor necrosis factor (TNF) inhibitors. Future research should focus on biomarker development, combination therapies, and long-term studies to optimize the safety and efficacy of IL-17-targeted therapies in SAIDs.
Collapse
Affiliation(s)
- Jingyuan Zhang
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China
| | - Min Shen
- Department of Rare Diseases, Peking Union Medical College Hospital (PUMCH), Chinese Academy of Medical Sciences & Peking Union Medical College; State Key Laboratory of Complex Severe and Rare Diseases, PUMCH; Department of Rheumatology and Clinical Immunology, PUMCH; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, 100730, China.
| |
Collapse
|
7
|
Cabral JE, Lin S, Zhou H, Wu A, Lackner A, Pham MA, Chi F, McNulty R. NLRP10 Cleaves Oxidized DNA inhibited by OGG1 inhibitors: A Newly Identified Role in DNA Damage Processing and Senescence Regulation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.07.642110. [PMID: 40161730 PMCID: PMC11952316 DOI: 10.1101/2025.03.07.642110] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/02/2025]
Abstract
Mitochondrial DNA (mtDNA) release into the cytosol is a critical event in innate immune activation, often acting as a damage-associated molecular pattern (DAMP) that triggers inflammasome assembly. Here, we demonstrate that NLRP3 plays a direct role in cleaving and facilitating the release of D-loop mtDNA into the cytosol. We further show that NLRP3 interacts with NLRP10. NLRP10-mediated ox-DNA cleavage involves a Schiff base intermediate and is inhibited by small molecules known to inhibit glycosylases. These findings support a model where NLRP10 interaction with oxidized DNA may contribute to long-term senescence secretory phenotype and modulate inflammasome activation. Our study highlights a novel mechanism by which NLRP10 can respond to mitochondrial stress signals to influence innate immunity and suggests therapeutic potential for targeting these interactions in inflammatory diseases.
Collapse
|
8
|
Feng S, Wierzbowski MC, Hrovat-Schaale K, Dumortier A, Zhang Y, Zyulina M, Baker PJ, Reygaerts T, Steiner A, De Nardo D, Narayanan DL, Milhavet F, Pinzon-Charry A, Arostegui JI, Khubchandani RP, Geyer M, Boursier G, Masters SL. Mechanisms of NLRP3 activation and inhibition elucidated by functional analysis of disease-associated variants. Nat Immunol 2025; 26:511-523. [PMID: 39930093 PMCID: PMC11876074 DOI: 10.1038/s41590-025-02088-9] [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: 02/27/2024] [Accepted: 01/13/2025] [Indexed: 02/16/2025]
Abstract
The NLRP3 inflammasome is a multiprotein complex that mediates caspase-1 activation and the release of proinflammatory cytokines, including interleukin (IL)-1β and IL-18. Gain-of-function variants in the gene encoding NLRP3 (also called cryopyrin) lead to constitutive inflammasome activation and excessive IL-1β production in cryopyrin-associated periodic syndromes (CAPS). Here we present functional screening and automated analysis of 534 NLRP3 variants from the international INFEVERS registry and the ClinVar database. This resource captures the effect of NLRP3 variants on ASC speck formation spontaneously, at low temperature, after inflammasome stimulation and with the specific NLRP3 inhibitor MCC950. Most notably, our analysis facilitated the updated classification of NLRP3 variants in INFEVERS. Structural analysis suggested multiple mechanisms by which CAPS variants activate NLRP3, including enhanced ATP binding, stabilizing the active NLRP3 conformation, destabilizing the inactive NLRP3 complex and promoting oligomerization of the pyrin domain. Furthermore, we identified pathogenic variants that can hypersensitize the activation of NLRP3 in response to nigericin and cold temperature exposure. We also found that most CAPS-related NLRP3 variants can be inhibited by MCC950; however, NLRP3 variants with changes to proline affecting helices near the inhibitor binding site are resistant to MCC950, as are variants in the pyrin domain, which likely trigger activation directly with the pyrin domain of ASC. Our findings could help stratify the CAPS population for NLRP3 inhibitor clinical trials and our automated methodologies can be implemented for molecules with a different mechanism of activation and in laboratories worldwide that are interested in adding new functionally validated NLRP3 variants to the resource. Overall, our study provides improved diagnosis for patients with CAPS, mechanistic insight into the activation of NLRP3 and stratification of patients for the future application of targeted therapeutics.
Collapse
Affiliation(s)
- Shouya Feng
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Matthew C Wierzbowski
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Immunology Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
| | - Katja Hrovat-Schaale
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Andreas Dumortier
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Yaoyuan Zhang
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Maria Zyulina
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Paul J Baker
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
| | - Thomas Reygaerts
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Annemarie Steiner
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Dominic De Nardo
- Department of Biochemistry and Molecular Biology, Immunity Program, Biomedicine Discovery Institute, Monash University, Clayton, Victoria, Australia
| | - Dhanya Lakshmi Narayanan
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia
| | - Florian Milhavet
- Department of Molecular genetics and Cytogenomics, CHU Montpellier, Rare and Autoinflammatory Diseases Unit, University of Montpellier, CEREMAIA, Institute for Regenerative Medicine and Biotherapy, INSERMU1183, Montpellier, France
| | - Alberto Pinzon-Charry
- Queensland Paediatric Immunology and Allergy Service, Children's Health Queensland, Brisbane, Queensland, Australia
| | - Juan Ignacio Arostegui
- Department of Immunology, Hospital Clínic, Barcelona, Spain
- Institut d'Investigacions Biomèdiques August Pi i Sunyer, Barcelona, Spain
- School of Medicine, University of Barcelona, Barcelona, Spain
| | - Raju P Khubchandani
- Sectional Head Pediatric Rheumatology, SRCC Children's Hospital, Mumbai, India
| | - Matthias Geyer
- Institute of Structural Biology, Medical Faculty, University of Bonn, Bonn, Germany
| | - Guilaine Boursier
- Department of Molecular genetics and Cytogenomics, CHU Montpellier, Rare and Autoinflammatory Diseases Unit, University of Montpellier, CEREMAIA, Institute for Regenerative Medicine and Biotherapy, INSERMU1183, Montpellier, France
| | - Seth L Masters
- Centre for Innate Immunity and Infectious Diseases, Hudson Institute of Medical Research, Clayton, Victoria, Australia.
- Department of Molecular and Translational Science, Monash University, Clayton, Victoria, Australia.
- Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria, Australia.
- Department of Medical Biology, University of Melbourne, Parkville, Victoria, Australia.
| |
Collapse
|
9
|
Deng J, Guo H, Kong R, Gao J. Case Report: PFAPA (Periodic Fever, Aphthous Stomatitis, Pharyngitis, and Cervical Adenitis) Syndrome With a Novel TNFAIP3 Mutation. Immun Inflamm Dis 2025; 13:e70178. [PMID: 40071725 PMCID: PMC11898003 DOI: 10.1002/iid3.70178] [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: 06/22/2024] [Revised: 02/18/2025] [Accepted: 03/04/2025] [Indexed: 03/15/2025] Open
Abstract
BACKGROUND Periodic fever, aphthous stomatitis, pharyngitis, and cervical adenitis (PFAPA) syndrome has been considered as a childhood syndrome. Its etiopathogeny is unknown however, currently considered as auto-immune inflammatory disease. Recently, a few cases of adult-onset of PFAPA syndrome have been reported. However, there is no report about the adult-onset of PFAPA case with a novel TNFAIP3 Mutation. OBJECTIVE AND METHOD Followed by detailed clinical inquiry, related laboratory tests, genetic sequencing and treatment, we reported a case with the adult-onset of PFAPA syndrome with a novel TNFAIP3 mutation. RESULTS We have found a novel mutation in the gene TNFAIP3 in an adult patient with periodic fever, aphthous stomatitis, pharyngitis, and adenitis-the PFAPA syndrome, under the environmental factor-COVID-19 vaccination. CONCLUSION This case demonstrated adult-onset of PFAPA symptoms, including periodic fever of unknown origin, which can occur in adult patients with the familial hereditary TNFAIP3 mutation and environmental factors. And the therapeutic measures provide some reference and practical significance for the treatment of PFAPA syndrome.
Collapse
Affiliation(s)
- Jiewen Deng
- Department of Cardiovascular DiseasesThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Hui Guo
- Department of Pharmacy, Shanghai Tenth People's HospitalTongji University School of MedicineShanghaiChina
| | - Ruina Kong
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| | - Jie Gao
- Department of Rheumatology and ImmunologyThe First Affiliated Hospital of Naval Medical UniversityShanghaiChina
| |
Collapse
|
10
|
Inagaki Y, Kamikubo T, Kuriwaki I, Watanabe J, Yamaki S, Iida M, Tomita K, Kakefuda K, Kurokawa J, Kiso T, Saba K, Koike T. Discovery of 2-(6-{[(1R,2R)-2-hydroxycyclohexyl]amino}-4,5-dimethylpyridazin-3-yl)-5-(trifluoromethyl)phenol (ASP0965): A potent, orally active and brain-penetrable NLRP3 inflammasome inhibitor with a novel scaffold for the treatment of α-synucleinopathy. Bioorg Med Chem 2025; 118:118042. [PMID: 39742857 DOI: 10.1016/j.bmc.2024.118042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2024] [Revised: 12/03/2024] [Accepted: 12/07/2024] [Indexed: 01/04/2025]
Abstract
NLRP3 inflammasome inhibitor is a highly attractive drug target for the treatment of various inflammatory diseases. Here, we report the discovery of pyridazine derivatives as a new class of scaffold for NLRP3 inflammasome inhibitors. We optimized HTS hit 2a to improve both in vitro IL-1β inhibitory activity and the mean photo effect (MPE) value in the in vitro 3T3 neutral red uptake (NRU) phototoxicity test. As a result, we identified compound 5e (ASP0965) with brain penetrability and showing efficacy in the brain on oral administration in the rat pharmacodynamics (PD) model and the mouse α-synuclein injection model. These findings suggest that compound 5e is a promising clinical candidate for α-synucleinopathy therapeutics.
Collapse
Affiliation(s)
- Yusuke Inagaki
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan.
| | - Takashi Kamikubo
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Ikumi Kuriwaki
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Junko Watanabe
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Susumu Yamaki
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Maiko Iida
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Kyoko Tomita
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Kenichi Kakefuda
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Jun Kurokawa
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Tetsuo Kiso
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Kengo Saba
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| | - Takanori Koike
- Tsukuba Research Center, Astellas Pharma Inc., 21, Miyukigaoka, Tsukuba, Ibaraki 305-8585, Japan
| |
Collapse
|
11
|
Baker MG, Ford LS, Campbell DE, Sampson HA. Just scratching the surface: A review of pediatric skin allergies. Pediatr Allergy Immunol 2025; 36:e70038. [PMID: 39953855 DOI: 10.1111/pai.70038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2024] [Revised: 11/16/2024] [Accepted: 01/23/2025] [Indexed: 02/17/2025]
Abstract
The skin is a large and sophisticated organ populated by innate and adaptive immune effector cells. These immune cells provide a critical first line of defense against pathogens, but genetic and environmental factors can lead to inappropriate signaling that may manifest as hypersensitivity. The most common cutaneous allergic disorders in children include atopic dermatitis, urticaria/angioedema, and contact dermatitis. In this review, we will briefly review these conditions, with a focus on recent developments in our understanding of the diagnosis and management of these disorders.
Collapse
Affiliation(s)
- Mary Grace Baker
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| | - Lara S Ford
- Department of Allergy & Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
| | - Dianne E Campbell
- Department of Allergy & Immunology, The Children's Hospital at Westmead, Sydney, New South Wales, Australia
- Faculty of Medicine and Health, Sydney Medical School, The University of Sydney, Sydney, New South Wales, Australia
- DBV Technologies, Montrouge, France
| | - Hugh A Sampson
- Division of Pediatric Allergy & Immunology, Department of Pediatrics, Elliot and Roslyn Jaffe Food Allergy Institute, Icahn School of Medicine at Mount Sinai, Kravis Children's Hospital, New York, New York, USA
| |
Collapse
|
12
|
Wei W, Wang F, Wang S, Wu H, Liu S, Wang G. Renal transplantation in patients with cryopyrin-associated periodic syndrome: A case report and literature review. Int Immunopharmacol 2025; 146:113879. [PMID: 39709908 DOI: 10.1016/j.intimp.2024.113879] [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: 04/25/2024] [Revised: 12/05/2024] [Accepted: 12/15/2024] [Indexed: 12/24/2024]
Abstract
BACKGROUND The cryopyrin-associated periodic syndrome (CAPS) is a rare autosomal dominant hereditary inflammatory disease clinically characterized by three overlapping types and associated with interleukin (IL)-1β.We reported a rare case of CAPS in a patient with accompanying symptoms such as growth retardation and urticaria-like rash. These clinical manifestations were caused by mutations in the NALP3 gene. CASE PRESENTATION A 28-year-old male patient, with a height of 1.3 m and a weight of 33.4 kg. He began to experience fever, rash, joint swelling and pain, as well as recurrent cerebral infarction, delayed growth and development, and decreased hearing, among other neuroendocrine manifestations six months after birth. After being examined at Peking Union Medical College Hospital in September 2014, a mutation in the NALP3 gene related to the encoding of Cryopyrin protein was detected,leading to a diagnosis of CAPS with a high probability of CINCA. CONCLUSION This is a rare case of CAPS, a disease inherited in an autosomal dominant manner and associated with mutations in the NALP3 gene. Based on its clinical manifestations and severity, it can be divided into three subtypes. The clinical presentation involves autoimmune inflammatory disease, with inflammation affecting multiple organs throughout the body, including the joints, skin, face, and kidneys. Involvement of the kidneys can lead to kidney failure, which is one of its most severe complications.
Collapse
Affiliation(s)
- Wei Wei
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Fulin Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Shaokun Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Hao Wu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Si Liu
- Department of Urology, The First Hospital of Jilin University, Changchun, China
| | - Gang Wang
- Department of Urology, The First Hospital of Jilin University, Changchun, China.
| |
Collapse
|
13
|
Gao H, Sun M, Gao H, Sun Y, Chen W, Dong N. Genome-wide screen based on 2DG activated NLRP3 inflammasome reveals the priming signal of TLR2/4 to IKKβ but not IKKα. Int Immunopharmacol 2025; 145:113781. [PMID: 39657538 DOI: 10.1016/j.intimp.2024.113781] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2024] [Revised: 11/27/2024] [Accepted: 12/01/2024] [Indexed: 12/12/2024]
Abstract
NLRP3 inflammasome activation is a pivotal area of research in innate immunity, yet the precise priming and activation signal remain unclear. In this study, we demonstrate that glycolysis inhibitor 2-Deoxy-D-glucose (2DG) triggers NLRP3-driven pyroptosis in human leukemia monocyte THP-1 cells by interfering glycosylation rather than glycolysis, which occurs independent of potassium efflux but requires the involvement of glycolysis rate-limiting enzyme PFKP. Using a CRISPR-Cas9 mediated large-scale screen, with 2DG as a new tool for probing NLRP3 activation, we identified that TLR2, rather than TLR4, initiates a rapid and robust priming signal for NLRP3 inflammasome activation. Importantly, both TLR2 and TLR4 depend entirely on MyD88, but not TRIF, for signal transduction. Furthermore, we discovered that TAK1, IKKβ and NEMO, but not IKKα, are essential for the priming signal. Additionally, we observed that deficiency in the linear ubiquitin assembly complex (LUBAC) subunits HOIP and HOIL-1, but not SHARPIN, is sufficient to inhibit 2DG-induced pyroptotic cell death. Collectively, our study reveals some common mechanism in the NLRP3 priming signals, as well as specific mechanisms upstream of NLRP3 triggered by 2DG, and underscores the potential of 2DG as a trigger to facilitate further detailed analysis of the underlying mechanisms of NLRP3 inflammasome activation. One Sentence Summary: Priming signal by IKKβ is essential for NLRP3 activation.
Collapse
Affiliation(s)
- Hui Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Mengning Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Hang Gao
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Yi Sun
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Wenjuan Chen
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China
| | - Na Dong
- State Key Laboratory of Animal Nutrition and Feeding, College of Animal Science and Technology, China Agricultural University, Yuanmingyuan West Road 2#, Haidian District, Beijing 100193, China.
| |
Collapse
|
14
|
Gupta S, Cassel SL, Sutterwala FS, Dagvadorj J. Regulation of the NLRP3 inflammasome by autophagy and mitophagy. Immunol Rev 2025; 329:e13410. [PMID: 39417249 DOI: 10.1111/imr.13410] [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: 10/19/2024]
Abstract
The NLRP3 inflammasome is a multiprotein complex that upon activation by the innate immune system drives a broad inflammatory response. The primary initial mediators of this response are pro-IL-1β and pro-IL-18, both of which are in an inactive form. Formation and activation of the NLRP3 inflammasome activates caspase-1, which cleaves pro-IL-1β and pro-IL-18 and triggers the formation of gasdermin D pores. Gasdermin D pores allow for the secretion of active IL-1β and IL-18 initiating the organism-wide inflammatory response. The NLRP3 inflammasome response can be beneficial to the host; however, if the NLRP3 inflammasome is inappropriately activated it can lead to significant pathology. While the primary components of the NLRP3 inflammasome are known, the precise details of assembly and activation are less well defined and conflicting. Here, we discuss several of the proposed pathways of activation of the NLRP3 inflammasome. We examine the role of subcellular localization and the reciprocal regulation of the NLRP3 inflammasome by autophagy. We focus on the roles of mitochondria and mitophagy in activating and regulating the NLRP3 inflammasome. Finally, we detail the impact of pathologic NLRP3 responses in the development and manifestations of pulmonary disease.
Collapse
Affiliation(s)
- Suman Gupta
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Suzanne L Cassel
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Fayyaz S Sutterwala
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| | - Jargalsaikhan Dagvadorj
- Department of Medicine, Cedars-Sinai Medical Center, Los Angeles, California, USA
- Women's Guild Lung Institute, Cedars-Sinai Medical Center, Los Angeles, California, USA
| |
Collapse
|
15
|
Meier DT, de Paula Souza J, Donath MY. Targeting the NLRP3 inflammasome-IL-1β pathway in type 2 diabetes and obesity. Diabetologia 2025; 68:3-16. [PMID: 39496966 DOI: 10.1007/s00125-024-06306-1] [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: 03/28/2024] [Accepted: 09/30/2024] [Indexed: 11/06/2024]
Abstract
Increased activity of the NACHT, LRR and PYD domains-containing protein 3 (NLRP3) inflammasome-IL-1β pathway is observed in obesity and contributes to the development of type 2 diabetes and its complications. In this review, we describe the pathological activation of IL-1β by metabolic stress, ageing and the microbiome and present data on the role of IL-1β in metabolism. We explore the physiological role of the IL-1β pathway in insulin secretion and the relationship between circulating levels of IL-1β and the development of diabetes and associated diseases. We highlight the paradoxical nature of IL-1β as both a friend and a foe in glucose regulation and provide details on clinical translation, including the glucose-lowering effects of IL-1 antagonism and its impact on disease modification. We also discuss the potential role of IL-1β in obesity, Alzheimer's disease, fatigue, gonadal dysfunction and related disorders such as rheumatoid arthritis and gout. Finally, we address the safety of NLRP3 inhibition and IL-1 antagonists and the prospect of using this therapeutic approach for the treatment of type 2 diabetes and its comorbidities.
Collapse
Affiliation(s)
- Daniel T Meier
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| | - Joyce de Paula Souza
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland
- Department of Biomedicine, University of Basel, Basel, Switzerland
| | - Marc Y Donath
- Clinic of Endocrinology, Diabetes and Metabolism, University Hospital Basel, Basel, Switzerland.
- Department of Biomedicine, University of Basel, Basel, Switzerland.
| |
Collapse
|
16
|
Shippy DC, Evered AH, Ulland TK. Ketone body metabolism and the NLRP3 inflammasome in Alzheimer's disease. Immunol Rev 2025; 329:e13365. [PMID: 38989642 PMCID: PMC11724017 DOI: 10.1111/imr.13365] [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: 07/12/2024]
Abstract
Alzheimer's disease (AD) is a degenerative brain disorder and the most common form of dementia. AD pathology is characterized by senile plaques and neurofibrillary tangles (NFTs) composed of amyloid-β (Aβ) and hyperphosphorylated tau, respectively. Neuroinflammation has been shown to drive Aβ and tau pathology, with evidence suggesting the nod-like receptor family pyrin domain containing 3 (NLRP3) inflammasome as a key pathway in AD pathogenesis. NLRP3 inflammasome activation in microglia, the primary immune effector cells of the brain, results in caspase-1 activation and secretion of IL-1β and IL-18. Recent studies have demonstrated a dramatic interplay between the metabolic state and effector functions of immune cells. Microglial metabolism in AD is of particular interest, as ketone bodies (acetone, acetoacetate (AcAc), and β-hydroxybutyrate (BHB)) serve as an alternative energy source when glucose utilization is compromised in the brain of patients with AD. Furthermore, reduced cerebral glucose metabolism concomitant with increased BHB levels has been demonstrated to inhibit NLRP3 inflammasome activation. Here, we review the role of the NLRP3 inflammasome and microglial ketone body metabolism in AD pathogenesis. We also highlight NLRP3 inflammasome inhibition by several ketone body therapies as a promising new treatment strategy for AD.
Collapse
Affiliation(s)
- Daniel C. Shippy
- Department of Pathology and Laboratory Medicine, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Abigail H. Evered
- Department of Pathology and Laboratory Medicine, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
- Cellular and Molecular Pathology Graduate Program, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| | - Tyler K. Ulland
- Department of Pathology and Laboratory Medicine, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
- Wisconsin Alzheimer's Disease Research Center, School of Medicine and Public HealthUniversity of WisconsinMadisonWisconsinUSA
| |
Collapse
|
17
|
Mackay A, Velcicky J, Gommermann N, Mattes H, Janser P, Wright M, Dubois C, Brenneisen S, Ilic S, Vangrevelinghe E, Stiefl N, Boettcher A, Schoenboerner M, Vogelsanger M, Muller-Bentz S, Kamke M, Rubert J, Kauffmann M, Desrayaud S, Trunzer M, Srinivas H, Hinniger A, von Burg N, Beltz K, Dekker C, Farady CJ. Discovery of NP3-253, a Potent Brain Penetrant Inhibitor of the NLRP3 Inflammasome. J Med Chem 2024; 67:20780-20798. [PMID: 39574318 DOI: 10.1021/acs.jmedchem.4c02350] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2024]
Abstract
Activation of the NLRP3 inflammasome in response to danger signals is a key innate immune mechanism and results in the production of the pro-inflammatory cytokines interleukin-1β (IL-1β) and interleukin-18 (IL-18) as well as pyroptotic cell death. Aberrant NLRP3 activation has been linked to many acute and chronic conditions ranging from atherosclerosis to Alzheimer's disease and cancer, and based on the clinical success of IL-1-targeting therapies, NLRP3 has emerged as an attractive therapeutic target. Herein we describe our discovery, characterization, and structure-based optimization of a pyridazine-based series of NLRP3 inhibitors initiating from an high-throughput screening campaign. The scaffold, exemplified by lead molecule NP3-253, has excellent potency and physicochemical and pharmacokinetic properties, including good brain penetration. The establishment of pharmacokinetic/pharmacodynamic relationships in the periphery and central nervous system in mechanistic models facilitates the use of NP3-253 as a tool to further interrogate the biology of NLRP3 in peripheral and neuroinflammatory models.
Collapse
Affiliation(s)
- Angela Mackay
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Juraj Velcicky
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Henri Mattes
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Philipp Janser
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Michael Wright
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Celine Dubois
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | - Slavica Ilic
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | | | | | | | | | - Marion Kamke
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Joelle Rubert
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | - Markus Trunzer
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | | | | | | - Karen Beltz
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | - Carien Dekker
- Novartis Biomedical Research, CH-4002 Basel, Switzerland
| | | |
Collapse
|
18
|
Rajkhowa S, Jha S. The role of NLRP3 and NLRP12 inflammasomes in glioblastoma. Genes Immun 2024; 25:541-551. [PMID: 39604503 DOI: 10.1038/s41435-024-00309-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Revised: 11/06/2024] [Accepted: 11/12/2024] [Indexed: 11/29/2024]
Abstract
Glioblastoma (GBM) is the deadliest malignant brain tumor, with a survival of less than 14 months after diagnosis. The highly invasive nature of GBM makes total surgical resection challenging, leading to tumor recurrence and declined survival. The heterocellular composition of the GBM reprograms its microenvironment, favoring tumor growth, proliferation, and migration. The innate immune cells in the GBM tumor microenvironment, including microglia, astrocytes, and macrophages, express pattern recognition receptors such as NLRs (Nucleotide-binding domain and leucine-rich repeat-containing) that sense pathogen- and damage-associated molecular patterns initiating inflammation. Upon activation, NLRP3 promotes inflammation by NLRP3 inflammasome formation. Auto-proteolytic cleavage and activation of Caspase-1 within the inflammasome leads to caspase-1-mediated cleavage, activation, and conversion of pro-IL-1ß and pro-IL-18 to IL-1ß and IL-18, leading to pyroptosis. In contrast, NLRP12 downregulates inflammatory responses in microglia and macrophages by regulating the NF-κB pathway. NLRP3 and NLRP12 have been implicated in the disease pathophysiology of several cancers with cell-context-dependent, pro- or anti-tumorigenic roles. In this review, we discuss the current literature on the mechanistic roles of NLRP3 and NLRP12 in GBM and the gaps in the scientific literature in the context of GBM pathophysiology with potential for targeted therapeutics.
Collapse
Affiliation(s)
- Sushmita Rajkhowa
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India
| | - Sushmita Jha
- Department of Bioscience and Bioengineering, Indian Institute of Technology Jodhpur, Jodhpur, Rajasthan, India.
| |
Collapse
|
19
|
Zhang C, Qiao P, Zhang J, Luo Y, Xiao C, Shen S, Hasegawa A, Qiao H, Wang G, Abe R, Fu M. A carbamazepine metabolite activates NLRP3 and controls skin homing of CD8 + T-cells in SJS/TEN. J Dermatol Sci 2024; 116:80-89. [PMID: 39500682 DOI: 10.1016/j.jdermsci.2024.10.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2022] [Revised: 10/05/2024] [Accepted: 10/18/2024] [Indexed: 12/09/2024]
Abstract
BACKGROUND Stevens-Johnson syndrome (SJS) and toxic epidermal necrolysis (TEN) are severe adverse drug reactions with extensive keratinocyte death. Carbamazepine (CBZ), the most commonly implicated drug in SJS/TEN, is metabolized by the cytochrome P450 enzyme 3A4 (CYP3A4) into carbamazepine-10,11-epoxide (CBZE) in the liver. While CD8+ cytotoxic T cells play an important role in SJS/TEN, the underlying mechanism of exuberant immune response by CD8+ T cells in these conditions remains incompletely understood. OBJECTIVES To examine the expression of NLRP3 inflammasome and their skin migration in CBZE-induced SJS/TEN. METHODS The expression of the NLRP3 inflammasome complex in skin lesions, sera, and blister fluids of SJS/TEN patients were analyzed by immunohistochemistry and enzyme-linked immunosorbent assay. NLRP3 formation and CD8+ T cell activation status and their functions were examined by immunoblotting, immunofluorescence, and chemotaxis assays. RESULTS The expression of the NLRP3 inflammasome complex was greatly increased in skin lesions of SJS/TEN patients. Moreover, IL-1β and IL-18 levels in sera and blister fluids of SJS/TEN patients were approximately 3-fold higher than those in healthy individuals, with a linear correlation between IL-1β levels and disease activity. CBZE induced NLRP3 inflammasome formation, upregulated CXCL9/CXCL10 levels, and activated CD8+ cytotoxic T cell functions via IL-1β/IL-1R or IL-18/IL-18R signaling in SJS/TEN keratinocytes, which promoted CD8+ cytotoxic T cell migration in SJS/TEN patients. CONCLUSION This study showed that CBZE promoted NLRP3 inflammasome formation and strengthened the activation and function of CD8+ cytotoxic T cells in the skin, which contributed to the initiation and progression of SJS/TEN.
Collapse
Affiliation(s)
- Chen Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Pei Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - JieYu Zhang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - YiXin Luo
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - ChunYing Xiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - ShengXian Shen
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Akio Hasegawa
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan
| | - HongJiang Qiao
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Gang Wang
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China
| | - Riichiro Abe
- Division of Dermatology, Niigata University Graduate School of Medical and Dental Sciences, Niigata, Japan.
| | - Meng Fu
- Department of Dermatology, Xijing Hospital, Fourth Military Medical University, Xi'an, China.
| |
Collapse
|
20
|
Xu S, Wang D, Tan L, Lu J. The role of NLRP3 inflammasome in type 2 inflammation related diseases. Autoimmunity 2024; 57:2310269. [PMID: 38332696 DOI: 10.1080/08916934.2024.2310269] [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: 08/30/2023] [Accepted: 01/21/2024] [Indexed: 02/10/2024]
Abstract
Type 2 inflammation related diseases, such as atopic dermatitis, asthma, and allergic rhinitis, are diverse and affect multiple systems in the human body. It is common for individuals to have multiple co-existing type 2 inflammation related diseases, which can impose a significant financial and living burden on patients. However, the exact pathogenesis of these diseases is still unclear. The NLRP3 inflammasome is a protein complex composed of the NLRP3 protein, ASC, and Caspase-1, and is activated through various mechanisms, including the NF-κB pathway, ion channels, and lysosomal damage. The NLRP3 inflammasome plays a role in the immune response to pathogens and cellular damage. Recent studies have indicated a strong correlation between the abnormal activation of NLRP3 inflammasome and the onset of type 2 inflammation. Additionally, it has been demonstrated that suppressing NLRP3 expression effectively diminishes the inflammatory response, highlighting its promising therapeutic applications. Therefore, this article reviews the role of NLRP3 inflammasome in the development and therapy of multiple type 2 inflammation related diseases.
Collapse
Affiliation(s)
- Shenming Xu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Dan Wang
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Lina Tan
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| | - Jianyun Lu
- Department of Dermatology, Third Xiangya Hospital, Central South University, Changsha, Hunan, People's Republic of China
- Medical Ozone Research Center of Central South University, Changsha, Hunan, People's Republic of China
| |
Collapse
|
21
|
Li Y, Chen W, Koo S, Liu H, Saiding Q, Xie A, Kong N, Cao Y, Abdi R, Serhan CN, Tao W. Innate immunity-modulating nanobiomaterials for controlling inflammation resolution. MATTER 2024; 7:3811-3844. [PMID: 40123651 PMCID: PMC11925551 DOI: 10.1016/j.matt.2024.09.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/25/2025]
Abstract
The acute inflammatory response is an inherent protective mechanism, its unsuccessful resolution can contribute to disease pathogenesis and potentially lead to death. Innate immune cells are the first line of host defenders and play a substantial role in inflammation initiation, amplification, resolution, or subsequent disease progression. As the resolution of inflammation is an active and highly regulated process, modulating innate immune cells, including neutrophils, monocytes and macrophages, and endothelial cells, and their interactions offer opportunities to control excessive inflammation. Nanobiomaterials have shown superior therapeutic potential in inflammation-related diseases by manipulating inflammatory responses because nanobiomaterials can target and interact with innate immune cells. Versatile nanobiomaterials can be designed for targeted modulation of specific innate immune responses. Nanopro-resolving medicines have been prepared both with pro-resolving lipid mediators and peptides each demonstrated to active resolution of inflammation in animal disease models. Here, we review innovative nanobiomaterials for modulating innate immunity and alleviating inflammation. We summarise the strategies converging the design of nanobiomaterials and the nano-bio interaction in modulating innate immune profiles and propelling the advancement of nanobiomaterials for inflammatory disease treatments. We also propose the future perspectives and translational challenges of nanobiomaterials that need to be overcome in this swiftly rising field.
Collapse
Affiliation(s)
- Yongjiang Li
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Wei Chen
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
- These authors contributed equally: Yongjiang Li, Wei Chen
| | - Seyoung Koo
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Haijun Liu
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Qimanguli Saiding
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Angel Xie
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Na Kong
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Yihai Cao
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institute, Stockholm 17177, Sweden
| | - Reza Abdi
- Transplantation Research Center, Renal Division, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Charles N. Serhan
- Center for Experimental Therapeutics and Reperfusion Injury, Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| | - Wei Tao
- Center for Nanomedicine and Department of Anesthesiology, Perioperative and Pain Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA 02115, USA
| |
Collapse
|
22
|
Ahmed HS. The Impact of Prevotella on Neurobiology in Aging: Deciphering Dendritic Cell Activity and Inflammatory Dynamics. Mol Neurobiol 2024; 61:9240-9251. [PMID: 38613648 DOI: 10.1007/s12035-024-04156-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2023] [Accepted: 03/29/2024] [Indexed: 04/15/2024]
Abstract
Prevotella species, notably Prevotella copri, significantly populate the human gut. In particular, P. copri is prevalent among non-Western populations with diets high in fiber. These species show complex relationships with diverse health aspects, associating with beneficial outcomes, including reduced visceral fat and improved glucose tolerance. Studies implicate various Prevotella species in specific diseases. Prevotella nigrescens and Porphyromonas gingivalis were linked to periodontal disease, promoting immune responses and influencing T helper type 17 (Th17) cells. Prevotella bivia was associated with bacterial vaginosis and a specific increase in activated cells in the vaginal mucosa. In contrast, they have shown substantial potential for inducing connective tissue degradation and alveolar bone resorption. Prevotella's role in neuroinflammatory disorders and autoinflammatory conditions such as Alzheimer's disease and Parkinson's disease has also been noted. The complex relationship between Prevotella and age-related conditions further extends to neurobiological changes in aging, with varying associations with Alzheimer's, Parkinson's, and other inflammatory conditions. Studies have also identified Prevotella to be implicated in cognitive decline in middle aged and the elderly. Future directions in this research area are anticipated to explore Prevotella-associated inflammatory mechanisms and therapeutic interventions. Investigating specific drug targets and immunomodulatory measures could lead to novel therapeutic strategies. Understanding how Prevotella-induced inflammation interacts with aging diseases would offer promising insights for treatments and interventions. This review urges ongoing research to discover therapeutic targets and mechanisms for moderating Prevotella-associated inflammation to further enhance our understanding and improve health outcomes.
Collapse
Affiliation(s)
- H Shafeeq Ahmed
- Bangalore Medical College and Research Institute, K.R Road, Bangalore, 560002, Karnataka, India.
| |
Collapse
|
23
|
Deng J, Han M, Gong J, Ma H, Hao Y, Fang C, Zhang H, Li J, Jiang W. Transcriptomic analysis of spleen-derived macrophages in response to lipopolysaccharide shows dependency on the MyD88-independent pathway in Chinese giant salamanders (Andrias davidianus). BMC Genomics 2024; 25:1005. [PMID: 39465384 PMCID: PMC11514755 DOI: 10.1186/s12864-024-10888-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2024] [Accepted: 10/09/2024] [Indexed: 10/29/2024] Open
Abstract
BACKGROUND Gram-negative bacteria are the main bacterial pathogens infecting Chinese giant salamanders (Andrias davidianus; CGS) in captivity and the wild, causing substantial economic losses in the CGS industry. However, the molecular mechanisms underlying pathogenesis following infection remain unclear. RESULTS Spleen-derived macrophages from healthy CGS were isolated, cultured, and identified using density gradient centrifugation and immunofluorescence. A macrophage transcriptome database was established 0, 6, and 12 h post lipopolysaccharide stimulation using RNA-sequencing. In the final database 76,743 unigenes and 4,698 differentially expressed genes (DEGs) were functionally annotated. Gene Ontology and Kyoto Encyclopedia of Genes and Genomes enrichment results showed that DEGs were concentrated in toll-like receptor-nuclear factor kappa B-related immune pathways. Ten DEGs were validated 12 h after lipopolysaccharide (LPS) stimulation. Although the common LPS recognition receptor toll-like receptor 4 was not activated and the key adaptor protein MyD88 showed no significant response, we observed significant up-regulation of the following adaptors: toll/interleukin-1 receptor domain-containing adaptor inducing interferon-β, tumour necrosis factor receptor-associated factor 6, and transforming growth factor-β activated kinase 1, which are located downstream of the non-classical MyD88 pathway. CONCLUSIONS In contrast to that in other species, macrophage activation in CGS could depend on the non-classical MyD88 pathway in response to bacterial infection. Our study provides insights into the molecular mechanisms regulating CGS antibacterial responses, with implications for disease prevention and understanding immune evolution in amphibians.
Collapse
Affiliation(s)
- Jie Deng
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an, 710032, China
| | - Mengdi Han
- College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, China
| | - Jingyu Gong
- College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, China
| | - Hongying Ma
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an, 710032, China
| | - Yinting Hao
- College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, China
| | - Cheng Fang
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an, 710032, China
| | - Han Zhang
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an, 710032, China
| | - Jia Li
- College of Life Sciences, Shaanxi Normal University, No. 620, West Chang'an Avenue, Chang'an District, Xi'an, 710119, China
| | - Wei Jiang
- Shaanxi Key Laboratory of Qinling Ecological Security, Shaanxi Institute of Zoology, Xi'an, 710032, China.
| |
Collapse
|
24
|
Hu H, Wang S, Chen C. Pathophysiological role and potential drug target of NLRP3 inflammasome in the metabolic disorders. Cell Signal 2024; 122:111320. [PMID: 39067838 DOI: 10.1016/j.cellsig.2024.111320] [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: 04/07/2024] [Revised: 07/17/2024] [Accepted: 07/23/2024] [Indexed: 07/30/2024]
Abstract
NLRP3 plays a role in the development of autoinflammatory diseases. NLRP3, ASC, and Caspases 1 or 8 make up the NLRP3 inflammasome, which is an important part of innate immune system. The NLRP3 inflammasome-mediated inflammatory cytokines may also participate in metabolic disorders, such as diabetes, hyperlipidemia, atherosclerosis, non-alcoholic fatty liver disease, and gout. Hence, an overview of the NLRP3 regulation in these metabolic diseases and the potential drugs targeting NLRP3 is the focus of this review.
Collapse
Affiliation(s)
- Huiming Hu
- School of pharmacy, Nanchang Medical College, Nanchang, Jiangxi, China; School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia; Key Laboratory of Pharmacodynamics and Safety Evaluation, Health Commission of Jiangxi Province, Jiangxi, China
| | - Shuwen Wang
- Jiangxi University of Chinese Medicine, Nanchang, Jiangxi, China
| | - Chen Chen
- School of Biomedical Sciences, University of Queensland, St Lucia, Brisbane, Queensland, Australia.
| |
Collapse
|
25
|
Verlinden SF. The genetic advantage of healthy centenarians: unraveling the central role of NLRP3 in exceptional healthspan. FRONTIERS IN AGING 2024; 5:1452453. [PMID: 39301197 PMCID: PMC11410711 DOI: 10.3389/fragi.2024.1452453] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/20/2024] [Accepted: 08/19/2024] [Indexed: 09/22/2024]
Abstract
Despite extensive research into extending human healthspan (HS) and compressing morbidity, the mechanisms underlying aging remain elusive. However, a better understanding of the genetic advantages responsible for the exceptional HS of healthy centenarians (HC), who live in good physical and mental health for one hundred or more years, could lead to innovative health-extending strategies. This review explores the role of NLRP3, a critical component of innate immunity that significantly impacts aging. It is activated by pathogen-associated signals and self-derived signals that increase with age, leading to low-grade inflammation implicated in age-related diseases. Furthermore, NLRP3 functions upstream in several molecular aging pathways, regulates cellular senescence, and may underlie the robust health observed in HC. By targeting NLRP3, mice exhibit a phenotype akin to that of HC, the HS of monkeys is extended, and aging symptoms are reversed in humans. Thus, targeting NLRP3 could offer a promising approach to extend HS. Additionally, a paradigm shift is proposed. Given that the HS of the broader population is 30 years shorter than that of HC, it is postulated that they suffer from a form of accelerated aging. The term 'auto-aging' is suggested to describe accelerated aging driven by NLRP3.
Collapse
|
26
|
Payne FM, Dabb AR, Harrison JC, Sammut IA. Inhibitors of NLRP3 Inflammasome Formation: A Cardioprotective Role for the Gasotransmitters Carbon Monoxide, Nitric Oxide, and Hydrogen Sulphide in Acute Myocardial Infarction. Int J Mol Sci 2024; 25:9247. [PMID: 39273196 PMCID: PMC11395567 DOI: 10.3390/ijms25179247] [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: 07/26/2024] [Revised: 08/21/2024] [Accepted: 08/21/2024] [Indexed: 09/15/2024] Open
Abstract
Myocardial ischaemia reperfusion injury (IRI) occurring from acute coronary artery disease or cardiac surgical interventions such as bypass surgery can result in myocardial dysfunction, presenting as, myocardial "stunning", arrhythmias, infarction, and adverse cardiac remodelling, and may lead to both a systemic and a localised inflammatory response. This localised cardiac inflammatory response is regulated through the nucleotide-binding oligomerisation domain (NACHT), leucine-rich repeat (LRR)-containing protein family pyrin domain (PYD)-3 (NLRP3) inflammasome, a multimeric structure whose components are present within both cardiomyocytes and in cardiac fibroblasts. The NLRP3 inflammasome is activated via numerous danger signals produced by IRI and is central to the resultant innate immune response. Inhibition of this inherent inflammatory response has been shown to protect the myocardium and stop the occurrence of the systemic inflammatory response syndrome following the re-establishment of cardiac circulation. Therapies to prevent NLRP3 inflammasome formation in the clinic are currently lacking, and therefore, new pharmacotherapies are required. This review will highlight the role of the NLRP3 inflammasome within the myocardium during IRI and will examine the therapeutic value of inflammasome inhibition with particular attention to carbon monoxide, nitric oxide, and hydrogen sulphide as potential pharmacological inhibitors of NLRP3 inflammasome activation.
Collapse
Affiliation(s)
- Fergus M Payne
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Alisha R Dabb
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Joanne C Harrison
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| | - Ivan A Sammut
- Department of Pharmacology and Toxicology and HeartOtago, School of Biomedical Sciences, University of Otago, Dunedin 9054, New Zealand
| |
Collapse
|
27
|
Dirks J, Wölfl M, Speer CP, Härtel C, Morbach H. Inborn Errors of Immunity in Early Childhood: Essential Insights for the Neonatologist. Neonatology 2024; 121:646-655. [PMID: 39182489 DOI: 10.1159/000540436] [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: 05/05/2024] [Accepted: 07/16/2024] [Indexed: 08/27/2024]
Abstract
BACKGROUND Inborn errors of immunity (IEI), formerly referred to as primary immunodeficiencies, manifest with a wide range of symptoms such as increased susceptibility to infections, immune dysregulation, and autoinflammation. Although most cases manifest in childhood, onset during the neonatal period is rare but potentially critical. SUMMARY In this review, we discuss the diverse clinical presentations of IEI and the specific challenges they pose to neonatologists. Rather than detailing every molecular defect, we focus on common clinical scenarios in neonates and young infants, providing practical diagnostic strategies to ensure timely and effective therapeutic interventions. KEY MESSAGES Clinical presentations of IEI in neonates may include delayed separation of the umbilical cord, skin rashes such as eczema and erythroderma, and recurrent episodes of inflammation. We also highlight immunological emergencies that require urgent medical attention, such as hyperinflammatory activity mimicking acute neonatal liver failure, sometimes seen in hemophagocytic lymphohistiocytosis. We also discuss appropriate medical action in the case of a positive newborn screening for severe T-cell defects. Early medical intervention in such circumstances may significantly improve outcomes.
Collapse
Affiliation(s)
- Johannes Dirks
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Matthias Wölfl
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Christian P Speer
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, Germany
| | - Henner Morbach
- Department of Pediatrics, University Hospital Würzburg, Würzburg, Germany
- Center for Primary Immunodeficiencies and Autoinflammatory Diseases, Centre for Rare Diseases - Reference Centre Northern Bavaria (ZESE), University Hospital, Würzburg, Germany
| |
Collapse
|
28
|
Cescato M, Zhu YYJ, Le Corre L, Py BF, Georgin-Lavialle S, Rodero MP. Implication of the LRR Domain in the Regulation and Activation of the NLRP3 Inflammasome. Cells 2024; 13:1365. [PMID: 39195255 PMCID: PMC11352923 DOI: 10.3390/cells13161365] [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: 07/09/2024] [Revised: 08/02/2024] [Accepted: 08/04/2024] [Indexed: 08/29/2024] Open
Abstract
The NLRP3 inflammasome is a critical component of the innate immune response. NLRP3 activation is a tightly controlled process involving an initial priming to express NLRP3, pro-IL-1 β, and pro-IL-18, followed by an activation signal. The precise mechanism of activation is not fully understood due to the diverse range of activators, yet it effectively orchestrates the activation of caspase-1, which subsequently triggers the release of proinflammatory cytokines IL-1β and IL-18. NLRP3 dysregulation can lead to a variety of inflammatory diseases, highlighting its significant role in immune response and disease pathogenesis. NLRP3 is divided into three domains: the PYD, the NACHT, and the LRR domains. This review focuses on the LRR domain of NLRP3, detailing its structural characteristics, its function in pathogen sensing, its role in the degradation process, and its involvement in inflammasome auto-inhibition and activation. Additionally, we discuss the impact of mutations within the LRR domain found in atypical Cryopyrin-Associated Periodic Syndromes (CAPS), highlighting the clinical relevance of this domain.
Collapse
Affiliation(s)
- Margaux Cescato
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| | - Yixiang Y J Zhu
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
- National Reference Center for Autoinflammatory Diseases and AA Amyloidosis, Department of Internal Medicine, Tenon Hospital, Sorbonne University, Assistance Publique—Hôpitaux de Paris (APHP), 75020 Paris, France;
| | - Laurent Le Corre
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| | - Bénédicte F Py
- CIRI, International Center for Research in Infectiology, Inserm, University Claude Bernard Lyon 1, 69007 Lyon, France;
| | - Sophie Georgin-Lavialle
- National Reference Center for Autoinflammatory Diseases and AA Amyloidosis, Department of Internal Medicine, Tenon Hospital, Sorbonne University, Assistance Publique—Hôpitaux de Paris (APHP), 75020 Paris, France;
| | - Mathieu P Rodero
- Laboratory of Pharmacological and Toxicological Chemistry and Biochemistry, CNRS, Paris Cité University, 75006 Paris, France; (M.C.); (Y.Y.J.Z.); (L.L.C.)
| |
Collapse
|
29
|
Lackner A, Cabral JE, Qiu Y, Zhou H, Leonidas L, Pham MA, Macapagal A, Lin S, Armanus E, McNulty R. Small molecule inhibitor binds to NOD-like receptor family pyrin domain containing 3 and prevents inflammasome activation. iScience 2024; 27:110459. [PMID: 39104412 PMCID: PMC11298654 DOI: 10.1016/j.isci.2024.110459] [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: 02/02/2024] [Revised: 05/10/2024] [Accepted: 07/02/2024] [Indexed: 08/07/2024] Open
Abstract
Despite recent advances in the mechanism of oxidized DNA activating NLRP3, the molecular mechanism and consequence of oxidized DNA associating with NLRP3 remains unknown. Cytosolic NLRP3 binds oxidized DNA which has been released from the mitochondria, which subsequently triggers inflammasome activation. Human glycosylase (hOGG1) repairs oxidized DNA damage which inhibits inflammasome activation. The fold of NLRP3 pyrin domain contains amino acids and a protein fold similar to hOGG1. Amino acids that enable hOGG1 to bind and cleave oxidized DNA are conserved in NLRP3. We found NLRP3 could bind and cleave oxidized guanine within mitochondrial DNA. The binding of oxidized DNA to NLRP3 was prevented by small molecule drugs which also inhibit hOGG1. These same drugs also inhibited inflammasome activation. Elucidating this mechanism will enable the design of drug memetics that treat inflammasome pathologies, illustrated herein by NLRP3 pyrin domain inhibitors which suppressed interleukin-1β (IL-1β) production in macrophages.
Collapse
Affiliation(s)
- Angela Lackner
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Julia Elise Cabral
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Yanfei Qiu
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Haitian Zhou
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Lemuel Leonidas
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Minh Anh Pham
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Alijah Macapagal
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Sophia Lin
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Emy Armanus
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| | - Reginald McNulty
- Laboratory of Macromolecular Structure, Department of Molecular Biology and Biochemistry, Charlie Dunlop School of Biological Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
- Department of Pharmaceutical Sciences, University of California, Irvine, Steinhaus Hall, Irvine, CA 92694-3900, USA
| |
Collapse
|
30
|
Tweedell RE, Kanneganti TD. Inflammasomes at the Foundation of Inflammatory Cell Death Complexes. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2024; 213:247-249. [PMID: 39008793 PMCID: PMC11302449 DOI: 10.4049/jimmunol.2400288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/21/2024] [Accepted: 05/21/2024] [Indexed: 07/17/2024]
Abstract
This Pillars of Immunology article is a commentary on “The Inflammasome: A Molecular Platform Triggering Activation of Inflammatory Caspases and Processing of proIL-β,” a pivotal article written by F. Martinon, K. Burns, and J. Tschopp, published in Molecular Cell in 2002. https://doi.org/10.1016/S1097-2765(02)00599-3.
Collapse
Affiliation(s)
- Rebecca E Tweedell
- Department of Immunology, St. Jude Children's Research Hospital, Memphis, TN, 38105, USA
| | | |
Collapse
|
31
|
Lecomte K, Toniolo A, Hoste E. Cell death as an architect of adult skin stem cell niches. Cell Death Differ 2024; 31:957-969. [PMID: 38649745 PMCID: PMC11303411 DOI: 10.1038/s41418-024-01297-3] [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: 12/16/2023] [Revised: 04/09/2024] [Accepted: 04/11/2024] [Indexed: 04/25/2024] Open
Abstract
Our skin provides a physical and immunological barrier against dehydration and environmental insults ranging from microbial attacks, toxins and UV irradiation to wounding. Proper functioning of the skin barrier largely depends on the interplay between keratinocytes- the epithelial cells of the skin- and immune cells. Two spatially distinct populations of keratinocyte stem cells (SCs) maintain the epidermal barrier function and the hair follicle. These SCs are inherently long-lived, but cell death can occur within their niches and impacts their functionality. The default cell death programme in skin is apoptosis, an orderly and non-inflammatory suicide programme. However, recent findings are shedding light on the significance of various modes of regulated necrotic cell death, which are lytic and can provoke inflammation within the local skin environment. While the presence of dying cells was generally regarded as a mere consequence of inflammation, findings in various human dermatological conditions and experimental mouse models of aberrant cell death control demonstrated that cell death programmes in keratinocytes (KCs) can drive skin inflammation and even tumour initiation. When cells die, they need to be removed by phagocytosis and KCs can function as non-professional phagocytes of apoptotic cells with important implications for their SC capacities. It is becoming apparent that in conditions of heightened SC activity, distinct cell death modalities differentially impact the different skin SC populations in their local niches. Here, we describe how regulated cell death modalities functionally affect epidermal SC niches along with their relevance to injury repair, inflammatory skin disorders and cancer.
Collapse
Affiliation(s)
- Kim Lecomte
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Annagiada Toniolo
- VIB Center for Inflammation Research, 9052, Ghent, Belgium
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium
| | - Esther Hoste
- VIB Center for Inflammation Research, 9052, Ghent, Belgium.
- Department of Biomedical Molecular Biology, Ghent University, 9052, Ghent, Belgium.
| |
Collapse
|
32
|
Ando T, Abe Y, Yamaji K, Nishikomori R, Tamura N. A case of cryopyrin-associated periodic syndrome due to somatic mosaic mutation complicated with recurrent circinate erythematous psoriasis. Mod Rheumatol Case Rep 2024; 8:368-372. [PMID: 38036300 DOI: 10.1093/mrcr/rxad067] [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: 10/03/2023] [Revised: 11/13/2023] [Accepted: 11/22/2023] [Indexed: 12/02/2023]
Abstract
Cryopyrin-associated periotic syndrome (CAPS) is a rare autoinflammatory disease caused by genetic variants in innate immunity genes. Autoinflammatory diseases, including CAPS, mediate proinflammatory cytokines such as interleukin (IL)-1 and IL-18 and result in severe systemic inflammation. A gain-of-function mutation in the NLR family pyrin domain-containing 3 (NLRP3) gene, which encodes the protein cryopyrin, was identified to be responsible for CAPS in 2001, and since then several additional pathogenic mutations have been found. Moreover, other phenotypes have been identified based on severity and symptomatology, including familial cold autoinflammatory syndrome, Muckle-Wells syndrome, and neonatal-onset multisystem inflammatory disease/chronic neurologic cutaneous articular syndrome. Prompt diagnosis of CAPS remains challenging, however, due to unspecific, extensive clinical signs, and delayed diagnosis and treatment targeting IL-1 lead to multiorgan damage. Another factor complicating diagnosis is the existence of somatic mosaic mutations in the NLRP3 gene in some cases, resulting in symptoms and clinical courses that are atypical. The frequency of somatic mosaic mutations in CAPS was estimated to be 19% in a systematic review. Psoriasis is a chronic inflammatory skin disease that affects ∼3% of the global population. Although no reports have shown complication between CAPS and psoriasis, these diseases have several similarities and potential relationships, for instance activation of T helper 17 cells in the dermis and increased NLRP3 gene expression in psoriatic skin compared with normal skin. Here, we report a case of CAPS due to a somatic mosaic mutation with recurrent circinate erythematous psoriasis.
Collapse
Affiliation(s)
- Taiki Ando
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Yoshiyuki Abe
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ken Yamaji
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| | - Ryuta Nishikomori
- The Department of Pediatrics and Child Health, Kurume University School of Medicine, Fukuoka, Japan
| | - Naoto Tamura
- Department of Internal Medicine and Rheumatology, Juntendo University School of Medicine, Tokyo, Japan
| |
Collapse
|
33
|
Fu J, Schroder K, Wu H. Mechanistic insights from inflammasome structures. Nat Rev Immunol 2024; 24:518-535. [PMID: 38374299 PMCID: PMC11216901 DOI: 10.1038/s41577-024-00995-w] [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] [Accepted: 01/17/2024] [Indexed: 02/21/2024]
Abstract
Inflammasomes are supramolecular complexes that form in the cytosol in response to pathogen-associated and damage-associated stimuli, as well as other danger signals that perturb cellular homoeostasis, resulting in host defence responses in the form of cytokine release and programmed cell death (pyroptosis). Inflammasome activity is closely associated with numerous human disorders, including rare genetic syndromes of autoinflammation, cardiovascular diseases, neurodegeneration and cancer. In recent years, a range of inflammasome components and their functions have been discovered, contributing to our knowledge of the overall machinery. Here, we review the latest advances in inflammasome biology from the perspective of structural and mechanistic studies. We focus on the most well-studied components of the canonical inflammasome - NAIP-NLRC4, NLRP3, NLRP1, CARD8 and caspase-1 - as well as caspase-4, caspase-5 and caspase-11 of the noncanonical inflammasome, and the inflammasome effectors GSDMD and NINJ1. These structural studies reveal important insights into how inflammasomes are assembled and regulated, and how they elicit the release of IL-1 family cytokines and induce membrane rupture in pyroptosis.
Collapse
Affiliation(s)
- Jianing Fu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA
| | - Kate Schroder
- Institute for Molecular Bioscience, The University of Queensland, St. Lucia, Queensland, Australia
| | - Hao Wu
- Department of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, Boston, MA, USA.
- Program in Cellular and Molecular Medicine, Boston Children's Hospital, Boston, MA, USA.
| |
Collapse
|
34
|
Mulazzani E, Böhm L, Christmann T, Krumbholz M, Kümpfel T, Havla J. Optical coherence tomography assessment of disease activity in cryopyrin-associated periodic syndrome. Eur J Neurol 2024; 31:e16301. [PMID: 38628041 PMCID: PMC11235936 DOI: 10.1111/ene.16301] [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: 01/26/2024] [Revised: 03/18/2024] [Accepted: 03/28/2024] [Indexed: 07/28/2024]
Abstract
BACKGROUND AND PURPOSE Cryopyrin-associated periodic syndrome is a rare autoinflammatory disease caused by gain-of-function mutations or variants in the NLRP3 gene. Clinically, patients suffer from a broad spectrum of both systemic and neurological symptoms. The aim of this study was to determine whether systemic inflammation demonstrated by serum amyloid A (SAA) elevation is associated with neuroinflammation assessed by optical coherence tomography (OCT). METHODS Thirty eyes of 15 patients with NLRP3 low penetrance mutations (PwNLRP3) and 20 eyes of 10 age- and sex-matched healthy controls were examined by spectral-domain OCT as part of routine clinical care. All retinal layers and clinical features were evaluated. RESULTS At baseline no significant retinal neuroaxonal inflammation or degeneration was observed in all measured retinal layers amongst PwNLRP3 compared with healthy controls. In a pooled analysis of all individual OCT time points a significant difference regarding the macular retinal nerve fibre layer was detected. Increased levels of SAA showed a positive association with averaged combined outer plexiform layer and outer nuclear layer volumes (ρ < 0.0001, r2 = 0.35). CONCLUSION In cryopyrin-associated periodic syndrome increased combined outer plexiform layer and outer nuclear layer volumes are mirrored by SAA increase, an acute phase reactant indicating systemic inflammation. Our findings identify OCT as a candidate biomarker to monitor subclinical neuroinflammation and to assess disease activity in PwNLRP3.
Collapse
Affiliation(s)
- E. Mulazzani
- Institute of Clinical NeuroimmunologyLMU University Hospital, LMU MunichMunichGermany
| | - L. Böhm
- Institute of Clinical NeuroimmunologyLMU University Hospital, LMU MunichMunichGermany
| | - T. Christmann
- Institute of Clinical NeuroimmunologyLMU University Hospital, LMU MunichMunichGermany
| | - M. Krumbholz
- Department of Neurology and Pain Treatment, Immanuel Klinik RüdersdorfUniversity Hospital of the Brandenburg Medical School Theodor FontaneRüdersdorf bei BerlinGermany
- Faculty of Health Sciences BrandenburgBrandenburg Medical School Theodor FontaneRüdersdorf bei BerlinGermany
- Department of Neurology and StrokeUniversity Hospital of TübingenTübingenGermany
| | - T. Kümpfel
- Institute of Clinical NeuroimmunologyLMU University Hospital, LMU MunichMunichGermany
| | - J. Havla
- Institute of Clinical NeuroimmunologyLMU University Hospital, LMU MunichMunichGermany
| |
Collapse
|
35
|
Healy LD, Fernández JA, Aiolfi R, Mosnier LO, Griffin JH. An orthosteric/allosteric bivalent peptide agonist comprising covalently linked protease-activated receptor-derived peptides mimics in vitro and in vivo activities of activated protein C. J Thromb Haemost 2024; 22:2039-2051. [PMID: 38670314 PMCID: PMC11610403 DOI: 10.1016/j.jtha.2024.04.007] [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: 10/12/2023] [Revised: 03/22/2024] [Accepted: 04/09/2024] [Indexed: 04/28/2024]
Abstract
BACKGROUND Activated protein C (APC) has anticoagulant and cytoprotective cell-signaling activities, which often require protease-activated receptor (PAR) 1 and PAR3 and PAR cleavages at noncanonical sites (R46-N47 and R41-G42, respectively). Some PAR1-derived (P1) peptides and PAR3-derived (P3) peptides, eg, P1-47-66 and P3-42-65, mimic APC's cell signaling. In anti-inflammatory assays, these 2 peptides at low concentrations synergistically attenuate cellular inflammation. OBJECTIVES To determine whether a P1 peptide covalently linked to a P3 peptide mimics APC's anti-inflammatory and endothelial barrier stabilization activities. METHODS Anti-inflammatory assays employed stimulated THP-1 cells and caspase-1 measurements. Cultured human EA.hy926 or murine aortic endothelial cells (ECs) exposed to thrombin were monitored for transendothelial electrical resistance. Bivalent covalently linked P1:P3 peptides were studied for APC-like activities. RESULTS In anti-inflammatory assays, P1-47-55 was as active as P1-47-66 and some P3 peptides (eg, P3-44-54 and P3-51-65) were as active as P3-42-65. The bivalent P1:P3 peptide comprising P1-47-55-(Gly[10 residues])-P3-51-65 (designated "G10 peptide") was more potently anti-inflammatory than the P1 or P3 peptide alone. In transendothelial electrical resistance studies of thrombin-challenged ECs, P1-47-55 and the G10 peptide mimicked APC's protective actions. In dose-response studies, the G10 peptide was more potent than the P1-47-55 peptide. In murine EC studies, the murine PAR-sequence-derived G10 peptide mimicked murine APC's activity. Anti-PAR1 and anti-PAR3 antibodies, but not anti-endothelial protein C receptor antibodies, abated G10's cytoprotection, showing that G10's actions involve PAR1:PAR3. G10 significantly increased survival in murine endotoxemia. CONCLUSION The PAR-sequence-derived G10 peptide is a bivalent agonist that mimics APC's cytoprotective, anti-inflammatory, and endothelial barrier-stabilizing actions and APC's protection against endotoxemic mortality.
Collapse
Affiliation(s)
- Laura D Healy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - José A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Roberto Aiolfi
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - Laurent O Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, California, USA.
| |
Collapse
|
36
|
Cordone V. Biochemical and molecular determinants of the subclinical inflammatory mechanisms in Rett syndrome. Arch Biochem Biophys 2024; 757:110046. [PMID: 38815782 DOI: 10.1016/j.abb.2024.110046] [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: 04/16/2024] [Revised: 05/24/2024] [Accepted: 05/25/2024] [Indexed: 06/01/2024]
Abstract
To date, Rett syndrome (RTT), a genetic disorder mainly caused by mutations in the X-linked MECP2 gene, is increasingly considered a broad-spectrum pathology, instead of just a neurodevelopmental disease, due to the multitude of peripheral co-morbidities and the compromised metabolic pathways, affecting the patients. The altered molecular processes include an impaired mitochondrial function, a perturbed redox homeostasis, a chronic subclinical inflammation and an improper cholesterol metabolism. The persistent subclinical inflammatory condition was first defined ten years ago, as a previously unrecognized feature of RTT, playing a role in the pathology progress and modulation of phenotypical severity. In light of this, the present work aims at reviewing the current knowledge on the chronic inflammatory status and the altered immune/inflammatory functions in RTT, as well as investigating the emerging mechanisms underlying this condition with a special focus on the latest findings about inflammasome system, autoimmunity responses and intestinal micro- and mycobiota. On these bases, although further research is needed, future therapeutic strategies able to re-establish an adequate immune/inflammatory response could represent potential approaches for RTT patients.
Collapse
Affiliation(s)
- Valeria Cordone
- Department of Environmental and Prevention Sciences, University of Ferrara, Ferrara, Italy.
| |
Collapse
|
37
|
Ahmed M, Kurungottu P, Swetha K, Atla S, Ashok N, Nagamalleswari E, Bonam SR, Sahu BD, Kurapati R. Role of NLRP3 inflammasome in nanoparticle adjuvant-mediated immune response. Biomater Sci 2024. [PMID: 38867716 DOI: 10.1039/d4bm00439f] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/14/2024]
Abstract
The nucleotide-binding oligomerization domain (NOD)-like receptor (NLR) family pyrin domain-containing 3 (NLRP3) inflammasome is pivotal in orchestrating the immune response induced by nanoparticle adjuvants. Understanding the intricate mechanisms underlying the activation of NLRP3 inflammasome by these adjuvants is crucial for deciphering their immunomodulatory properties. This review explores the involvement of the NLRP3 inflammasome in mediating immune responses triggered by nanoparticle adjuvants. It delves into the signaling pathways and cellular mechanisms involved in NLRP3 activation, highlighting its significance in modulating the efficacy and safety of nanoparticle-based adjuvants. A comprehensive grasp of the interplay between NLRP3 inflammasome and nanoparticle adjuvants holds promise for optimizing vaccine design and advancing immunotherapeutic strategies.
Collapse
Affiliation(s)
- Momitul Ahmed
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Pavithra Kurungottu
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - K Swetha
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Sandeep Atla
- Texas A&M Drug Discovery Center, Department of Chemistry, Texas A&M University, College Station, Texas 77843, USA
| | - Nivethitha Ashok
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| | - Easa Nagamalleswari
- MTCC and Gene Bank, CSIR-Institute of Microbial Technology, Sector 39A, Chandigarh, 160036, India
| | - Srinivasa Reddy Bonam
- Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX 77555, USA.
| | - Bidya Dhar Sahu
- Department of Pharmacology & Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Guwahati 781101, India.
| | - Rajendra Kurapati
- School of Chemistry, Indian Institute of Science Education and Research, Thiruvananthapuram 695551, India.
| |
Collapse
|
38
|
Kurdi H, Lavalle L, Moon JCC, Hughes D. Inflammation in Fabry disease: stages, molecular pathways, and therapeutic implications. Front Cardiovasc Med 2024; 11:1420067. [PMID: 38932991 PMCID: PMC11199868 DOI: 10.3389/fcvm.2024.1420067] [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: 04/19/2024] [Accepted: 05/28/2024] [Indexed: 06/28/2024] Open
Abstract
Fabry disease, a multisystem X-linked disorder caused by mutations in the alpha-galactosidase gene. This leads to the accumulation of globotriaosylceramide (Gb3) and globotriaosylsphingosine (Lyso-Gb3), culminating in various clinical signs and symptoms that significantly impact quality of life. Although treatments such as enzyme replacement, oral chaperone, and emerging therapies like gene therapy exist; delayed diagnosis often curtails their effectiveness. Our review highlights the importance of delineating the stages of inflammation in Fabry disease to enhance the timing and efficacy of diagnosis and interventions, particularly before the progression to fibrosis, where treatment options are less effective. Inflammation is emerging as an important aspect of the pathogenesis of Fabry disease. This is thought to be predominantly mediated by the innate immune response, with growing evidence pointing towards the potential involvement of adaptive immune mechanisms that remain poorly understood. Highlighted by the fact that Fabry disease shares immune profiles with systemic autoinflammatory diseases, blurring the distinctions between these disorders and highlighting the need for a nuanced understanding of immune dynamics. This insight is crucial for developing targeted therapies and improving the administration of current treatments like enzyme replacement. Moreover, our review discusses the complex interplay between these inflammatory processes and current treatments, such as the challenges posed by anti-drug antibodies. These antibodies can attenuate the effectiveness of therapies, necessitating more refined approaches to mitigate their impact. By advancing our understanding of the molecular changes, inflammatory mediators and causative factors that drive inflammation in Fabry disease, we aim to clarify their role in the disease's progression. This improved understanding will help us see how these processes fit into the current landscape of Fabry disease. Additionally, it will guide the development of more effective diagnostic and therapeutic approaches, ultimately improving patient care.
Collapse
Affiliation(s)
- Hibba Kurdi
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Lucia Lavalle
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| | - James C. C. Moon
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Cardiovascular Imaging Department, Barts Heart Centre, London, United Kingdom
| | - Derralynn Hughes
- Institute of Cardiovascular Science, University College London, London, United Kingdom
- Lysosomal Storage Disorders Unit, The Royal Free Hospital, London, United Kingdom
| |
Collapse
|
39
|
Vande Walle L, Said M, Paerewijck O, Bertoni A, Gattorno M, Linclau B, Lamkanfi M. Novel chemotype NLRP3 inhibitors that target the CRID3-binding pocket with high potency. Life Sci Alliance 2024; 7:e202402644. [PMID: 38519142 PMCID: PMC10961714 DOI: 10.26508/lsa.202402644] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2024] [Revised: 03/06/2024] [Accepted: 03/14/2024] [Indexed: 03/24/2024] Open
Abstract
The NLRP3 inflammasome plays a central role in various human diseases. Despite significant interest, most clinical-grade NLRP3 inhibitors are derived from sulfonylurea inhibitor CRID3 (also called MCC950). Here, we describe a novel chemical class of NLRP3-inhibiting compounds (NIC) that exhibit potent and selective NLRP3 inflammasome inhibition in human monocytes and mouse macrophages. BRET assays demonstrate that they physically interact with NLRP3. Structural modeling further reveals they occupy the same binding site of CRID3 but in a critically different conformation. Furthermore, we show that NIC-11 and NIC-12 lack the off-target activity of CRID3 against the enzymatic activity of carbonic anhydrases I and II. NIC-12 selectively reduces circulating IL-1ß levels in the LPS-endotoxemia model in mice and inhibits NLRP3 inflammasome activation in CAPS patient monocytes and mouse macrophages with about tenfold increased potency compared with CRID3. Altogether, this study unveils a new chemical class of highly potent and selective NLRP3-targeted inhibitors with a well-defined molecular mechanism to complement existing CRID3-based NLRP3 inhibitors in pharmacological studies and serve as novel chemical leads for the development of NLRP3-targeted therapies.
Collapse
Affiliation(s)
- Lieselotte Vande Walle
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Madhukar Said
- Research Group Organic and Medicinal Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Oonagh Paerewijck
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Arinna Bertoni
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Marco Gattorno
- UOC Reumatologia e Malattie Autoinfiammatorie, IRCCS Istituto G. Gaslini, Genova, Italy
| | - Bruno Linclau
- Research Group Organic and Medicinal Chemistry, Department of Organic and Macromolecular Chemistry, Ghent University, Ghent, Belgium
| | - Mohamed Lamkanfi
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
40
|
Miyamoto T, Izawa K, Masui S, Yamazaki A, Yamasaki Y, Matsubayashi T, Shiraki M, Ohnishi H, Yasumura J, Kawabe T, Miyamae T, Matsubara T, Arakawa N, Ishige T, Takizawa T, Shimbo A, Shimizu M, Kimura N, Maeda Y, Maruyama Y, Shigemura T, Furuta J, Sato S, Tanaka H, Izumikawa M, Yamamura M, Hasegawa T, Kaneko H, Nakagishi Y, Nakano N, Iida Y, Nakamura T, Wakiguchi H, Hoshina T, Kawai T, Murakami K, Akizuki S, Morinobu A, Ohmura K, Eguchi K, Sonoda M, Ishimura M, Furuno K, Kashiwado M, Mori M, Kawahata K, Hayama K, Shimoyama K, Sasaki N, Ito T, Umebayashi H, Omori T, Nakamichi S, Dohmoto T, Hasegawa Y, Kawashima H, Watanabe S, Taguchi Y, Nakaseko H, Iwata N, Kohno H, Ando T, Ito Y, Kataoka Y, Saeki T, Kaneko U, Murase A, Hattori S, Nozawa T, Nishimura K, Nakano R, Watanabe M, Yashiro M, Nakamura T, Komai T, Kato K, Honda Y, Hiejima E, Yonezawa A, Bessho K, Okada S, Ohara O, Takita J, Yasumi T, Nishikomori R. Clinical Characteristics of Cryopyrin-Associated Periodic Syndrome and Long-Term Real-World Efficacy and Tolerability of Canakinumab in Japan: Results of a Nationwide Survey. Arthritis Rheumatol 2024; 76:949-962. [PMID: 38268504 DOI: 10.1002/art.42808] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2023] [Revised: 01/09/2024] [Accepted: 01/22/2024] [Indexed: 01/26/2024]
Abstract
OBJECTIVE We assess the clinical characteristics of patients with cryopyrin-associated periodic syndrome (CAPS) in Japan and evaluate the real-world efficacy and safety of interleukin-1 (IL-1) inhibitors, primarily canakinumab. METHODS Clinical information was collected retrospectively, and serum concentrations of canakinumab and cytokines were analyzed. RESULTS A total of 101 patients were included, with 86 and 15 carrying heterozygous germline and somatic mosaic mutations, respectively. We identified 39 mutation types, and the common CAPS-associated symptoms corresponded with those in previous reports. Six patients (5.9% of all patients) died, with four of the deaths caused by CAPS-associated symptoms. Notably, 73.7% of patients (100%, 79.6%, and 44.4% of familial cold autoinflammatory syndrome, Muckle-Wells syndrome, and chronic infantile neurological cutaneous articular syndrome/neonatal onset multisystem inflammatory disease, respectively) achieved complete remission with canakinumab, and early therapeutic intervention was associated with better auditory outcomes. In some patients, canakinumab treatment stabilized the progression of epiphysial overgrowth and improved height gain, visual acuity, and renal function. However, 23.7% of patients did not achieve inflammatory remission with crucial deterioration of organ damage, with two dying while receiving high-dose canakinumab treatment. Serological analysis of canakinumab and cytokine concentrations revealed that the poor response was not related to canakinumab shortage. Four inflammatory nonremitters developed inflammatory bowel disease (IBD)-unclassified during canakinumab treatment. Dual biologic therapy with canakinumab and anti-tumor necrosis factor-α agents was effective for IBD- and CAPS-associated symptoms not resolved by canakinumab monotherapy. CONCLUSION This study provides one of the largest epidemiologic data sets for CAPS. Although early initiation of anti-IL-1 treatment with canakinumab is beneficial for improving disease prognosis, some patients do not achieve remission despite a high serum concentration of canakinumab. Moreover, IBD may develop in CAPS after canakinumab treatment.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Tomoyo Matsubara
- Dokkyo Medical University Saitama Medical Center, Saitama, Japan
| | | | | | | | - Asami Shimbo
- Tokyo Medical and Dental University, Tokyo, Japan
| | | | - Naoki Kimura
- Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | - Satoshi Sato
- Saitama Children's Medical Center, Saitama, Japan
| | | | | | | | | | - Hiroshi Kaneko
- National Center for Global Health and Medicine, Tokyo, Japan
| | | | - Naoko Nakano
- Ehime Prefectural Central Hospital, Matsuyama, Japan
| | | | | | | | - Takayuki Hoshina
- University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Toshinao Kawai
- National Center for Child Health and Development, Tokyo, Japan
| | | | | | | | | | | | | | | | | | | | - Masaaki Mori
- Tokyo Medical and Dental University, Tokyo, Japan, and St. Marianna University School of Medicine, Kawasaki, Japan
| | | | | | | | - Natsuko Sasaki
- University of Occupational and Environmental Health, Japan, Kitakyushu, Japan
| | - Taisuke Ito
- Hamamatsu University School of Medicine, Hamamatsu, Japan
| | | | - Tae Omori
- Tokyo Metropolitan Bokutoh Hospital, Tokyo, Japan
| | | | | | | | | | | | - Yuichiro Taguchi
- Department of Rheumatology, Nagoya Ekisaikai Hospital, Nagoya, Japan
| | | | - Naomi Iwata
- Aichi Children's Health and Medical Center, Obu, Japan
| | - Hiroki Kohno
- Tokyo Women's Medical University Hospital, Tokyo, Japan
| | | | - Yasuhiko Ito
- Nagoya City University West Medical Center, Nagoya, Japan
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | - Osamu Ohara
- Kazusa DNA Research Institute, Kisarazu, Japan
| | | | | | | |
Collapse
|
41
|
Hayashida Y, Hatano M, Ito K, Sugie M, Kunieda J, Shimizu M, Morio T, Morioka C. Case Report: A neonatal case of cryopyrin-associated periodic syndrome with severe funisitis and neonatal asphyxia. Front Pediatr 2024; 12:1397412. [PMID: 38808101 PMCID: PMC11130403 DOI: 10.3389/fped.2024.1397412] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/07/2024] [Accepted: 04/25/2024] [Indexed: 05/30/2024] Open
Abstract
Cryopyrin-associated periodic syndrome (CAPS) is a genetic disorder and autoinflammatory disease characterized by chronic inflammation throughout the body. The most severe form of CAPS, Chronic Infantile Neurologic Cutaneous, and Articular (CINCA) syndrome, also known as Neonatal Onset Multisystem Inflammatory Disease (NOMID), has three main features: skin rash, CNS involvement, and joint symptoms. Although these symptoms are typically reported shortly after birth, there have been a few reports of prenatal inflammation. Here, we report our experience managing a case of a CAPS infant born in severe neonatal asphyxia due to a ruptured cord associated with severe funisitis. The baby was born at 38 weeks and 6 days of gestation, weighing 2,898 g, through an ultra-emergency Caesarian section prompted by variable deceleration. The Apgar score was 1 point at 1 min and 4 points at 5 min, necessitating intensive care due to hypoxic-ischemic encephalopathy. Upon delivery, it was observed that the umbilical cord had partially ruptured at the site of attachment to the baby, accompanied by arterial hemorrhage. Umbilical cord rupture was considered to be the cause of the sudden decrease in fetal heart rate. Pathological examination also showed that the inflammation of the cord was more severe on the side attached to the fetus and on the arterial side, suggesting that the inflammation had extended from the fetus. The father carried a genetic mutation associated with CINCA syndrome/NOMID (NLRP3 c.2068G>A p.Glu690Lys Hetero), which was also found in the child. Histopathologic examination of the placenta and umbilical cord can provide crucial insights into the intrauterine onset of inflammation, which is the first manifestation of CINCA syndrome/NOMID in newborns. It should be noted that births with a genetic predisposition to CAPS may have complications related to the placenta and umbilical cord.
Collapse
Affiliation(s)
- Yuri Hayashida
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Maho Hatano
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Kazuyuki Ito
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Manabu Sugie
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Junko Kunieda
- Department of Comprehensive Pathology, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Masaki Shimizu
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Tomohiro Morio
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| | - Chikako Morioka
- Department of Pediatrics, Tokyo Medical and Dental University Hospital, Tokyo, Japan
| |
Collapse
|
42
|
Doedens JR, Smolak P, Nguyen M, Wescott H, Diamond C, Schooley K, Billinton A, Harrison D, Koller BH, Watt AP, Gabel CA. Pharmacological Analysis of NLRP3 Inflammasome Inhibitor Sodium [(1,2,3,5,6,7-Hexahydro-s-indacen-4-yl)carbamoyl][(1-methyl-1 H-pyrazol-4-yl)({[(2 S)-oxolan-2-yl]methyl})sulfamoyl]azanide in Cellular and Mouse Models of Inflammation Provides a Translational Framework. ACS Pharmacol Transl Sci 2024; 7:1438-1456. [PMID: 38751618 PMCID: PMC11091978 DOI: 10.1021/acsptsci.4c00061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2024] [Revised: 03/19/2024] [Accepted: 04/09/2024] [Indexed: 05/18/2024]
Abstract
Interleukin (IL)-1β is an apex proinflammatory cytokine produced in response to tissue injury and infection. The output of IL-1β from monocytes and macrophages is regulated not only by transcription and translation but also post-translationally. Release of the active cytokine requires activation of inflammasomes, which couple IL-1β post-translational proteolysis with pyroptosis. Among inflammasome platforms, NOD-like receptor pyrin domain-containing protein 3 (NLRP3) is implicated in the pathogenesis of numerous human disorders in which disease-specific danger-associated molecular patterns (DAMPS) are positioned to drive its activation. As a promising therapeutic target, numerous candidate NLRP3-targeting therapeutics have been described and demonstrated to provide benefits in the context of animal disease models. While showing benefits, published preclinical studies have not explored dose-response relationships within the context of the models. Here, the preclinical pharmacology of a new chemical entity, [(1,2,3,5,6,7-hexahydro-s-indacen-4-yl)carbamoyl][(1-methyl-1H-pyrazol-4-yl)({[(2S)-oxolan-2-yl]methyl})sulfamoyl]azanide (NT-0249), is detailed, establishing its potency and selectivity as an NLRP3 inhibitor. NT-0249 also is evaluated in two acute in vivo mouse challenge models where pharmacodynamic/pharmacokinetic relationships align well with in vitro blood potency assessments. The therapeutic utility of NT-0249 is established in a mouse model of cryopyrin-associated periodic syndrome (CAPS). In this model, mice express a human gain-of-function NLRP3 allele and develop chronic and progressive IL-1β-dependent autoinflammatory disease. NT-0249 dose-dependently reduced multiple inflammatory biomarkers in this model. Significantly, NT-0249 decreased mature IL-1β levels in tissue homogenates, confirming in vivo target engagement. Our findings highlight not only the pharmacological attributes of NT-0249 but also provide insight into the extent of target suppression that will be required to achieve clinical benefit.
Collapse
Affiliation(s)
| | - Pamela Smolak
- NodThera,
Inc., Seattle, Washington 98103, United States
| | - MyTrang Nguyen
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | | | | | - Ken Schooley
- NodThera,
Inc., Seattle, Washington 98103, United States
| | - Andy Billinton
- NodThera
Ltd, Little Chesterford,
Saffron Walden, Essex CB10
1XL, U.K.
| | - David Harrison
- NodThera
Ltd, Little Chesterford,
Saffron Walden, Essex CB10
1XL, U.K.
| | - Beverly H. Koller
- Department
of Genetics, University of North Carolina
at Chapel Hill, Chapel
Hill, North Carolina 27599, United States
| | - Alan P. Watt
- NodThera
Ltd, Little Chesterford,
Saffron Walden, Essex CB10
1XL, U.K.
| | | |
Collapse
|
43
|
Wu B, Luo Y, Wu D, Wang Y, Shen M. Phenotypic and genotypic characterization of Chinese adult patients with NLRP3-associated autoinflammatory disease with hearing loss. Rheumatology (Oxford) 2024; 63:1690-1698. [PMID: 37656934 DOI: 10.1093/rheumatology/kead451] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2023] [Revised: 07/22/2023] [Accepted: 08/02/2023] [Indexed: 09/03/2023] Open
Abstract
OBJECTIVES Nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3-associated autoinflammatory disease (NLRP3-AID) is a rare autosomal dominant systemic autoinflammatory disease. We aimed to summarize the phenotypic and genotypic features of Chinese adult NLRP3-AID patients with hearing loss. METHODS A retrospective cohort study of twenty-one adult patients with NLRP3-AID was conducted in Peking Union Medical College Hospital between July 2015 and March 2023. All patients underwent whole exome sequencing and otorhinolaryngologic assessments. Clinical features and therapeutic data were collected and analysed. RESULTS We found that 13/21 (61.90%) of patients had hearing loss with high-frequency impairment in the majority, and most patients presented with vestibular dysfunction as a new finding. The NLRP3-AID patients with early-onset, cold or stress triggered episodes, red eyes, fatigue, hypopsia and mutations located in the NACHT domain of the NLRP3 protein were more likely to suffer from hearing loss, especially sensorineural hearing loss, perhaps as a result of pathogenic variants of high penetrance. By a series of audiological evaluations, TNF-α inhibitors were confirmed to improve or reverse hearing loss. CONCLUSIONS We reported the first cohort of Chinese adult NLRP3-AID patients with hearing loss and characterized vestibular dysfunction, highlighted the necessity for attention to high-frequency hearing and provided potential alternative treatment.
Collapse
Affiliation(s)
- Bingxuan Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yi Luo
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Di Wu
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| | - Yi Wang
- Department of Otolaryngological, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College, Beijing, China
| | - Min Shen
- Department of Rheumatology and Clinical Immunology, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences & Peking Union Medical College; National Clinical Research Center for Dermatologic and Immunologic Diseases (NCRC-DID), Ministry of Science & Technology; State Key Laboratory of Complex Severe and Rare Diseases, Peking Union Medical College Hospital; Key Laboratory of Rheumatology and Clinical Immunology, Ministry of Education, Beijing, China
| |
Collapse
|
44
|
Daman AW, Cheong JG, Berneking L, Josefowicz SZ. The potency of hematopoietic stem cell reprogramming for changing immune tone. Immunol Rev 2024; 323:197-208. [PMID: 38632868 DOI: 10.1111/imr.13335] [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: 04/19/2024]
Abstract
Innate immune memory endows innate immune cells with antigen independent heightened responsiveness to subsequent challenges. The durability of this response can be mediated by inflammation induced epigenetic and metabolic reprogramming in hematopoietic stem and progenitor cells (HSPCs) that are maintained through differentiation to mature immune progeny. Understanding the mechanisms and extent of trained immunity induction by pathogens and vaccines, such as BCG, in HSPC remains a critical area of exploration with important implications for health and disease. Here we review these concepts and present new analysis to highlight how inflammatory reprogramming of HSPC can potently alter immune tone, including to enhance specific anti-tumor responses. New findings in the field pave the way for novel HSPC targeting therapeutic strategies in cancer and other contexts of immune modulation. Future studies are expected to unravel diverse and extensive effects of infections, vaccines, microbiota, and sterile inflammation on hematopoietic progenitor cells and begin to illuminate the broad spectrum of immunologic tuning that can be established through altering HSPC phenotypes. The purpose of this review is to draw attention to emerging and speculative topics in this field where we posit that focused study of HSPC in the framework of trained immunity holds significant promise.
Collapse
Affiliation(s)
- Andrew W Daman
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Jin Gyu Cheong
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| | - Laura Berneking
- Human Oncology & Pathogenesis Program, Memorial Sloan Kettering Cancer Center, New York, New York, USA
| | - Steven Z Josefowicz
- Immunology and Microbial Pathogenesis Program, Weill Cornell Medical College, New York, New York, USA
- Department of Pathology and Laboratory Medicine, Weill Cornell Medical College, New York, New York, USA
| |
Collapse
|
45
|
Zhao M, Ye N, Liu L, Zhang RJ, Li N, Peng J, Cai XY, Jiang XQ, Su KY, Zhang XL, Rao QR, Liu KJ, Deng DX, Peng AH, Tang MH, Chen LJ, Wu WS, Ye HY. Novel Isoalantolactone-Based Derivatives as Potent NLRP3 Inflammasome Inhibitors: Design, Synthesis, and Biological Characterization. J Med Chem 2024. [PMID: 38686671 DOI: 10.1021/acs.jmedchem.4c00357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2024]
Abstract
The NLRP3 inflammasome has been recognized as a promising therapeutic target in drug discovery for inflammatory diseases. Our initial research identified a natural sesquiterpene isoalantolactone (IAL) as the active scaffold targeting NLRP3 inflammasome. To improve its activity and metabolic stability, a total of 64 IAL derivatives were designed and synthesized. Among them, compound 49 emerged as the optimal lead, displaying the most potent inhibitory efficacy on nigericin-induced IL-1β release in THP-1 cells, with an IC50 value of 0.29 μM, approximately 27-fold more potent than that of IAL (IC50: 7.86 μM), and exhibiting higher metabolic stability. Importantly, 49 remarkably improved DSS-induced ulcerative colitis in vivo. Mechanistically, we demonstrated that 49 covalently bound to cysteine 279 in the NACHT domain of NLRP3, thereby inhibiting the assembly and activation of NLRP3 inflammasome. These results provided compelling evidence to further advance the development of more potent NLRP3 inhibitors based on this scaffold.
Collapse
Affiliation(s)
- Min Zhao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Neng Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ling Liu
- School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu 611137, China
| | - Rui-Jia Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Na Li
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Jing Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xiao-Ying Cai
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xue-Qin Jiang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kai-Yue Su
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Xin-Lu Zhang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Qian-Ru Rao
- Department of Urology, Institute of Urology (Laboratory of Reconstructive Urology), West China Hospital, Sichuan University, Chengdu 610041, China
| | - Kong-Jun Liu
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - De-Xin Deng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ai-Hua Peng
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Ming-Hai Tang
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Li-Juan Chen
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
- Chengdu Zenitar Biomedical Technology Co., Ltd., Chengdu 610041, China
| | - Wen-Shuang Wu
- Division of Thyroid Surgery, Department of General Surgery and Laboratory of Thyroid and Parathyroid Disease, Frontiers Science Center for Disease-Related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, China
| | - Hao-Yu Ye
- Department of Biotherapy, Cancer Center and State Key Laboratory of Biotherapy, West China Hospital, Sichuan University, Chengdu 610041, China
| |
Collapse
|
46
|
Carpenter S, O'Neill LAJ. From periphery to center stage: 50 years of advancements in innate immunity. Cell 2024; 187:2030-2051. [PMID: 38670064 PMCID: PMC11060700 DOI: 10.1016/j.cell.2024.03.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 02/24/2024] [Accepted: 03/25/2024] [Indexed: 04/28/2024]
Abstract
Over the past 50 years in the field of immunology, something of a Copernican revolution has happened. For a long time, immunologists were mainly concerned with what is termed adaptive immunity, which involves the exquisitely specific activities of lymphocytes. But the other arm of immunity, so-called "innate immunity," had been neglected. To celebrate Cell's 50th anniversary, we have put together a review of the processes and components of innate immunity and trace the seminal contributions leading to the modern state of this field. Innate immunity has joined adaptive immunity in the center of interest for all those who study the body's defenses, as well as homeostasis and pathology. We are now entering the era where therapeutic targeting of innate immune receptors and downstream signals hold substantial promise for infectious and inflammatory diseases and cancer.
Collapse
Affiliation(s)
- Susan Carpenter
- University of California Santa Cruz, 1156 High St., Santa Cruz, CA 95064, USA.
| | - Luke A J O'Neill
- School of Biochemistry and Immunology, Trinity Biomedical Sciences Institute, Trinity College Dublin, Dublin, Ireland.
| |
Collapse
|
47
|
Yu T, Hou D, Zhao J, Lu X, Greentree WK, Zhao Q, Yang M, Conde DG, Linder ME, Lin H. NLRP3 Cys126 palmitoylation by ZDHHC7 promotes inflammasome activation. Cell Rep 2024; 43:114070. [PMID: 38583156 PMCID: PMC11130711 DOI: 10.1016/j.celrep.2024.114070] [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: 01/21/2023] [Revised: 02/14/2024] [Accepted: 03/20/2024] [Indexed: 04/09/2024] Open
Abstract
Nucleotide oligomerization domain (NOD)-like receptor protein 3 (NLRP3) inflammasome hyperactivation contributes to many human chronic inflammatory diseases, and understanding how NLRP3 inflammasome is regulated can provide strategies to treat inflammatory diseases. Here, we demonstrate that NLRP3 Cys126 is palmitoylated by zinc finger DHHC-type palmitoyl transferase 7 (ZDHHC7), which is critical for NLRP3-mediated inflammasome activation. Perturbing NLRP3 Cys126 palmitoylation by ZDHHC7 knockout, pharmacological inhibition, or modification site mutation diminishes NLRP3 activation in macrophages. Furthermore, Cys126 palmitoylation is vital for inflammasome activation in vivo. Mechanistically, ZDHHC7-mediated NLRP3 Cys126 palmitoylation promotes resting NLRP3 localizing on the trans-Golgi network (TGN) and activated NLRP3 on the dispersed TGN, which is indispensable for recruitment and oligomerization of the adaptor ASC (apoptosis-associated speck-like protein containing a CARD). The activation of NLRP3 by ZDHHC7 is different from the termination effect mediated by ZDHHC12, highlighting versatile regulatory roles of S-palmitoylation. Our study identifies an important regulatory mechanism of NLRP3 activation that suggests targeting ZDHHC7 or the NLRP3 Cys126 residue as a potential therapeutic strategy to treat NLRP3-related human disorders.
Collapse
Affiliation(s)
- Tao Yu
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Dan Hou
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Jiaqi Zhao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Xuan Lu
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Wendy K Greentree
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Qian Zhao
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Min Yang
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Don-Gerard Conde
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14853, USA
| | - Maurine E Linder
- Department of Molecular Medicine, Cornell University, Ithaca, NY 14853, USA
| | - Hening Lin
- Howard Hughes Medical Institute, Department of Chemistry and Chemical Biology, Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY 14853, USA.
| |
Collapse
|
48
|
Xu Z, Kombe Kombe AJ, Deng S, Zhang H, Wu S, Ruan J, Zhou Y, Jin T. NLRP inflammasomes in health and disease. MOLECULAR BIOMEDICINE 2024; 5:14. [PMID: 38644450 PMCID: PMC11033252 DOI: 10.1186/s43556-024-00179-x] [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: 01/01/2024] [Accepted: 03/20/2024] [Indexed: 04/23/2024] Open
Abstract
NLRP inflammasomes are a group of cytosolic multiprotein oligomer pattern recognition receptors (PRRs) involved in the recognition of pathogen-associated molecular patterns (PAMPs) and danger-associated molecular patterns (DAMPs) produced by infected cells. They regulate innate immunity by triggering a protective inflammatory response. However, despite their protective role, aberrant NLPR inflammasome activation and gain-of-function mutations in NLRP sensor proteins are involved in occurrence and enhancement of non-communicating autoimmune, auto-inflammatory, and neurodegenerative diseases. In the last few years, significant advances have been achieved in the understanding of the NLRP inflammasome physiological functions and their molecular mechanisms of activation, as well as therapeutics that target NLRP inflammasome activity in inflammatory diseases. Here, we provide the latest research progress on NLRP inflammasomes, including NLRP1, CARD8, NLRP3, NLRP6, NLRP7, NLRP2, NLRP9, NLRP10, and NLRP12 regarding their structural and assembling features, signaling transduction and molecular activation mechanisms. Importantly, we highlight the mechanisms associated with NLRP inflammasome dysregulation involved in numerous human auto-inflammatory, autoimmune, and neurodegenerative diseases. Overall, we summarize the latest discoveries in NLRP biology, their forming inflammasomes, and their role in health and diseases, and provide therapeutic strategies and perspectives for future studies about NLRP inflammasomes.
Collapse
Affiliation(s)
- Zhihao Xu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Arnaud John Kombe Kombe
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Shasha Deng
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China
| | - Hongliang Zhang
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Songquan Wu
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China
| | - Jianbin Ruan
- Department of Immunology, University of Connecticut Health Center, Farmington, 06030, USA.
| | - Ying Zhou
- Department of Obstetrics and Gynecology, Core Facility Center, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
| | - Tengchuan Jin
- Center of Disease Immunity and Intervention, College of Medicine, Lishui University, Lishui, 323000, China.
- Laboratory of Structural Immunology, the CAS Key Laboratory of Innate Immunity and Chronic Disease, School of Basic Medical Sciences, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, 230027, China.
- Department of Obstetrics and Gynecology, Core Facility Center, Division of Life Sciences and Medicine, The First Affiliated Hospital of USTC, University of Science and Technology of China, Hefei, 230001, Anhui, China.
- Institute of Health and Medicine, Hefei Comprehensive National Science Center, Hefei, Anhui, China.
- Biomedical Sciences and Health Laboratory of Anhui Province, University of Science & Technology of China, Hefei, 230027, China.
- Clinical Research Hospital of Chinese Academy of Sciences (Hefei), University of Science and Technology of China, Hefei, 230001, China.
| |
Collapse
|
49
|
Hollingsworth LR, Veeraraghavan P, Paulo JA, Harper JW. Spatiotemporal proteomic profiling of cellular responses to NLRP3 agonists. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.04.19.590338. [PMID: 38659763 PMCID: PMC11042255 DOI: 10.1101/2024.04.19.590338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2024]
Abstract
Nucleotide-binding domain and leucine-rich repeat pyrin-domain containing protein 3 (NLRP3) is an innate immune sensor that forms an inflammasome in response to various cellular stressors. Gain-of-function mutations in NLRP3 cause autoinflammatory diseases and NLRP3 signalling itself exacerbates the pathogenesis of many other human diseases. Despite considerable therapeutic interest, the primary drivers of NLRP3 activation remain controversial due to the diverse array of signals that are integrated through NLRP3. Here, we mapped subcellular proteome changes to lysosomes, mitochondrion, EEA1-positive endosomes, and Golgi caused by the NLRP3 inflammasome agonists nigericin and CL097. We identified several common disruptions to retrograde trafficking pathways, including COPI and Shiga toxin-related transport, in line with recent studies. We further characterized mouse NLRP3 trafficking throughout its activation using temporal proximity proteomics, which supports a recent model of NLRP3 recruitment to endosomes during inflammasome activation. Collectively, these findings provide additional granularity to our understanding of the molecular events driving NLRP3 activation and serve as a valuable resource for cell biological research. We have made our proteomics data accessible through an open-access Shiny browser to facilitate future research within the community, available at: https://harperlab.connect.hms.harvard.edu/inflame/. We will display anonymous peer review for this manuscript on pubpub.org (https://harperlab.pubpub.org/pub/nlrp3/) rather than a traditional journal. Moreover, we invite community feedback on the pubpub version of this manuscript, and we will address criticisms accordingly.
Collapse
Affiliation(s)
- L. Robert Hollingsworth
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | | | - Joao A. Paulo
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| | - J. Wade Harper
- Department of Cell Biology, Harvard Medical School, Harvard
University, Boston, MA 02115, USA
| |
Collapse
|
50
|
Meyer M, Schwärzler J, Jukic A, Tilg H. Innate Immunity and MASLD. Biomolecules 2024; 14:476. [PMID: 38672492 PMCID: PMC11048298 DOI: 10.3390/biom14040476] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2024] [Revised: 04/08/2024] [Accepted: 04/11/2024] [Indexed: 04/28/2024] Open
Abstract
Metabolic dysfunction-associated steatotic liver disease (MASLD) has emerged as the most common liver disease worldwide in recent years. MASLD commonly presents as simple hepatic steatosis, but ~25% of patients develop liver inflammation, progressive fibrosis, liver cirrhosis and related hepatocellular carcinoma. Liver inflammation and the degree of fibrosis are key determinants of the prognosis. The pathophysiology of liver inflammation is incompletely understood and involves diverse factors and specifically innate and adaptive immune responses. More specifically, diverse mediators of innate immunity such as proinflammatory cytokines, adipokines, inflammasomes and various cell types like mononuclear cells, macrophages and natural killer cells are involved in directing the inflammatory process in MASLD. The activation of innate immunity is driven by various factors including excess lipids and lipotoxicity, insulin resistance and molecular patterns derived from gut commensals. Targeting pathways of innate immunity might therefore appear as an attractive therapeutic strategy in the future management of MASLD and possibly its complications.
Collapse
Affiliation(s)
| | | | | | - Herbert Tilg
- Department of Internal Medicine I, Gastroenterology, Hepatology, Endocrinology & Metabolism, Medical University Innsbruck, 6020 Innsbruck, Austria; (M.M.); (A.J.)
| |
Collapse
|