1
|
Deng X, Zhou J, Fang W, Sun R, Yan G, Sun Y. pH-triggered small molecule nanodrugs self-assembled from tryptamine-cinnamaldehyde and fisetin for targeted sepsis-associated encephalopathy therapy. J Biomater Appl 2025; 39:1165-1176. [PMID: 39894462 DOI: 10.1177/08853282251318052] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Sepsis-associated encephalopathy (SAE) is an acute diffuse brain dysfunction, but its clinical treatment just focuses on antibiotics and supportive therapy, which fail to directly limit the development of SAE. Herein, this work highlights the development of pH-triggered small molecule nanodrugs self-assembled from tryptamine (Try)-cinnamaldehyde (CA) and fisetin for targeted SAE therapy. The imine linkage in Try-CA and acid-dependent protonation of Try and fisetin endow the nanodrugs with pH-triggered dynamic changes of particle sizes, surficial charges, and drug release. Moreover, the combined use of Try-CA and fisetin also endows the nanodrugs with superior antioxidative, anti-inflammatory and antibacterial capabilities compared to their individual use. These characteristics of the nanodrugs facilitate long-term circulation stability, effective penetration through BBB, selective accumulation in the brain, and target to central and peripheral focal areas, thereby achieving comprehensive treatment or relief of SAE. Thus, these attractive experimental results illuminate the enormous potential of such pH-triggered small molecule nanodrugs for targeted SAE therapy, advancing their use in clinics.
Collapse
Affiliation(s)
- Ximing Deng
- Department of Critical Care Medicine, The Second Hospital of Anhui Medical University, Hefei, China
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Jinyao Zhou
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Wei Fang
- School of Pharmacy, Anhui University of Chinese Medicine, Hefei, China
| | - Rao Sun
- Department of Critical Care Medicine, First Affiliated Hospital of Bengbu Medical University, Bengbu, China
| | - Guoqing Yan
- Engineering Research Center for Biomedical Materials, Anhui Key Laboratory of Modern Biomanufacturing, School of Life Sciences, Anhui University, Hefei, China
| | - Yun Sun
- Department of Critical Care Medicine, The Second Hospital of Anhui Medical University, Hefei, China
| |
Collapse
|
2
|
Chen X, Zhu Y, Xia L, Su S, Fan S, Lu Y, Chen Q, Wei Y, Huang Q, Liu X, Peng X. Glutamine limits NLRP3 inflammasome activation and pyroptosis in macrophages by sustaining the IRG1/itaconate axis. FEBS J 2025. [PMID: 40296302 DOI: 10.1111/febs.70119] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2024] [Revised: 01/18/2025] [Accepted: 04/17/2025] [Indexed: 04/30/2025]
Abstract
Aberrant activation of NACHT, LRR, and PYD domains-containing protein 3 (NLRP3) inflammasome increases the release of mature pro-inflammatory cytokines interleukin (IL)-1β and IL-18, and enhances pyroptosis; thereby necessitating tight regulation of the NLRP3 inflammasome. Dysfunctional glutamine metabolism contributes to the pathogenesis of multiple inflammatory disorders, and the precise mechanism remains to be elucidated. Here, we provide evidence that glutamine deprivation enhances NLRP3 inflammasome activation in macrophages. Indeed, the absence of exogenous glutamine specifically enhanced NLRP3 inflammasome assembly, thereby accelerating pyroptosis and promoting the maturation of IL-1β and IL-18. Inhibition of glutaminolysis exhibited a similar effect to glutamine deprivation, whereas this effect was reversed by α-ketoglutarate (α-KG), a tricarboxylic acid (TCA)-cycle intermediate that can be replenished by glutamine supply. We further observed reduced generation of endogenous itaconate by glutamine deprivation and verified that both exogenous supplementation of itaconate derivative and increased endogenous itaconate production by overexpressing immune-responsive gene 1 [IRG1; also known as aconitate decarboxylase 1 (ACOD1)] could replace glutamine to inhibit the NLRP3 inflammasome. Mechanistically, glutamine deprivation decreased the source of substrate and inhibited transcription factor EB (TFEB)-dependent transcriptional upregulation of IRG1, thereby impairing the IRG1/itaconate axis that suppresses the NLRP3 inflammasome. Furthermore, glutamine deficiency was detected in a murine sepsis model, whereas extrinsic glutamine supplementation conferred protection against intestinal inflammation and tissue damage in septic mice. Taken together, our findings provide a novel insight into the link between glutamine metabolism and NLRP3 inflammasome activation, highlighting the target of glutamine metabolism, which holds as a potential therapeutic strategy for inflammatory diseases.
Collapse
Affiliation(s)
- Xiaoli Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yuanfeng Zhu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lin Xia
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sen Su
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Shijun Fan
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yongling Lu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qian Chen
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Yan Wei
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Qianying Huang
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xin Liu
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xi Peng
- Clinical Medical Research Center, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
- State Key Laboratory of Trauma and Chemical Poisoning, Southwest Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| |
Collapse
|
3
|
Fan Y, Meng S, Song Y, Zhang Y, Song Y, Chen Z, Xie K. Interaction, diagnosis, and treatment of lung microbiota-NLRP3 inflammasome-target organ axis in sepsis. Int Immunopharmacol 2025; 149:114222. [PMID: 39923579 DOI: 10.1016/j.intimp.2025.114222] [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: 11/01/2024] [Revised: 01/14/2025] [Accepted: 01/31/2025] [Indexed: 02/11/2025]
Abstract
Sepsis is defined as a life-threatening condition caused by a dysregulated host response to infection, leading to multi-organ dysfunction, and representing a significant global health burden. The progression of sepsis is closely linked to disruptions in lung microbiota, including bacterial translocation, impaired barrier function, and local microenvironmental disturbances. Conversely, the worsening of sepsis exacerbates lung microbiota imbalances, contributing to multi-organ dysfunction. Recent culture-independent microbiological techniques have unveiled the complexity of the respiratory tract microbiome, necessitating a reassessment of the interactions between the host, microbes, and pathogenesis in sepsis. This review synthesizes current insights into the causes of microbiota dysbiosis and the regulatory mechanisms of the NOD-like receptor pyrin domain containing 3 (NLRP3) inflammasome, as well as their interactions during sepsis and sepsis-induced organ dysfunction. In addition, we summarize novel diagnostic and therapeutic approaches from the current study that may offer promising prospects for the management of sepsis.
Collapse
Affiliation(s)
- Yan Fan
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Shuqi Meng
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Yu Song
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Ying Zhang
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China
| | - Yan Song
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin China
| | - Zhe Chen
- Department of Geriatrics, Tianjin Geriatrics Institute, Tianjin Medical University General Hospital, Tianjin China.
| | - Keliang Xie
- Department of Critical Care Medicine, Tianjin Medical University General Hospital, No. 154, Anshan Road, Heping District, Tianjin 300052, China; Laboratory of Anesthesia and Critical Care Medicine in Colleges and Universities of Shandong Province, School of Anesthesiology, Shandong Second Medical University, China.
| |
Collapse
|
4
|
Yu X, Gao J, Zhang C. Sepsis-induced cardiac dysfunction: mitochondria and energy metabolism. Intensive Care Med Exp 2025; 13:20. [PMID: 39966268 PMCID: PMC11836259 DOI: 10.1186/s40635-025-00728-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2024] [Accepted: 02/04/2025] [Indexed: 02/20/2025] Open
Abstract
Sepsis is a life-threatening multi-organ dysfunction syndrome caused by dysregulated host response to infection, posing a significant global healthcare challenge. Sepsis-induced myocardial dysfunction (SIMD) is a common complication of sepsis, significantly increasing mortality due to its high energy demands and low compensatory reserves. The substantial mitochondrial damage rather than cell apoptosis in SIMD suggests disrupted cardiac energy metabolism as a crucial pathophysiological mechanism. Therefore, we systematically reviewed the mechanisms underlying energy metabolism dysfunction in SIMD, including alterations in myocardial cell energy metabolism substrates, excitation-contraction coupling processes, mitochondrial dysfunction, and mitochondrial autophagy and biogenesis, summarizing potential therapeutic targets within them.
Collapse
Affiliation(s)
- Xueting Yu
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Jie Gao
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China
| | - Chunxiang Zhang
- Key Laboratory of Medical Electrophysiology, Ministry of Education, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China.
- Department of Cardiology, Institute of Cardiovascular Research, the Affiliated Hospital of Southwest Medical University, Southwest Medical University, Luzhou, Sichuan, China.
- FACC, Southwest Medical University, Luzhou, Sichuan, China.
| |
Collapse
|
5
|
Zhu W, Bao X, Yang Y, Xing M, Xiong S, Chen S, Zhong Y, Hu X, Lu Q, Wang K, Ling Q, Cui S. Peripheral Evolution of Tanshinone IIA and Cryptotanshinone for Discovery of a Potent and Specific NLRP3 Inflammasome Inhibitor. J Med Chem 2025; 68:3460-3479. [PMID: 39847657 DOI: 10.1021/acs.jmedchem.4c02648] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2025]
Abstract
Natural products (NPs) continue to serve as an invaluable source in drug discovery, and peripheral evolution of NPs is a highly efficient evolution strategy. Herein, we describe a unified "methyl to amide" peripheral evolution of Tanshinone IIA and Cryptotanshinone for discovery of NLRP3 inflammasome inhibitors. There were 54 compounds designed and prepared, while the chemoinformatic analysis revealed that these evolved NP analogues occupy a unique chemical space. Biological evaluation identified 5m as an NLRP3 inflammasome inhibitor, and 5m could directly bind to the NACHT domain of the NLRP3 protein and block the interaction of NLRP3 and ASC, thus suppressing ASC oligomerization and NLRP3 inflammasome assembly. Molecular dynamic stimulations revealed that the amide moiety played a vital role in the binding mode. Moreover, 5m exhibited therapeutical efficacy in sepsis and the NASH mouse model. In conclusion, this protocol provides a new vision of NPs' peripheral evolution and a novel NLRP3 inflammasome inhibitor.
Collapse
Affiliation(s)
- Wenqi Zhu
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xiaodong Bao
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yuyan Yang
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Muqiong Xing
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Sijie Xiong
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Siyu Chen
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Yongxin Zhong
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
| | - Xueping Hu
- Institute of Frontier Chemistry, School of Chemistry and Chemical Engineering, Shandong University, Qingdao 266237, China
| | - Qianrang Lu
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Kairong Wang
- School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
| | - Qi Ling
- Division of Hepatobiliary and Pancreatic Surgery, Department of Surgery, The First Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
| | - Sunliang Cui
- College of Pharmaceutical Sciences, State Key Laboratory of Advanced Drug Delivery and Release Systems, Zhejiang University, Hangzhou 310058, China
- Department of Burns and Wound Care, The Second Affiliated Hospital, School of Medicine, Zhejiang University, Hangzhou 310009, China
- Jinhua Institute of Zhejiang University, Jinhua 321299, China
| |
Collapse
|
6
|
Xu K, Tan J, Lin D, Jiang H, Chu Y, Zhou L, Zhang J, Lu Y. Gut microbes of the cecum versus the colon drive more severe lethality and multi-organ damage. Int Immunopharmacol 2025; 147:114029. [PMID: 39793233 DOI: 10.1016/j.intimp.2025.114029] [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: 01/02/2025] [Accepted: 01/03/2025] [Indexed: 01/13/2025]
Abstract
Intestinal perforations lead to a high risk of sepsis-associated morbidity and multi-organ dysfunctions. A perforation allows intestinal contents (IC) to enter the peritoneal cavity, causing abdominal infections. Right- and left-sided perforations have different prognoses in humans, but the mechanisms associated with different cecum and colon perforations remain unclear. This study investigates how gut flora influences outcomes from perforations at different sites in mice. Using fecal-induced peritonitis mouse model, isolated IC from the cecum or colon was injected peritoneally at 2 mg/kg. Bacterial burden was quantified with quantitative PCR, and microbial communities were analyzed using 16S rRNA gene sequencing. Survival rates were monitored, and blood biochemical indices, histological changes, cytokines expression, immunological signaling and multiple-organ damage were assessed at 16 h post-injections. The results showed cecum IC developed more severe sepsis than colon IC, with shorter median survival time and greater multi-organ damage. Mice treated with cecum IC displayed elevated tissue damage markers in the liver, heart, and kidneys, contributing to worsened pathology. This was likely driven by systematic inflammatory cytokines production and lung inflammation. Mechanistically, cecum IC triggered stronger cGAS-STING and TBK1-NF-κB signaling, promoting systemic inflammation compared to the colon IC. Moreover, bacterial analysis demonstrated that cecum IC carry a higher bacterial burden than colon IC and exhibit a different microbial community. A detailed microbiome comparison revealed an increased abundance of potentially pathogenic bacteria in the cecum IC. These findings suggest that the site of intestinal perforation influences sepsis severity, with the cecum being associated with a higher bacterial burden and a relatively increased abundance of potentially pathogenic bacteria compared to the colon. Our findings first compared the lethality associated with the microbial composition of the cecum and colon, indicating the perforation site could help providers predict the severity of sepsis, thereby introducing a novel perspective to microbiology and sepsis research.
Collapse
Affiliation(s)
- Kejia Xu
- Department of Anesthesiology and Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Juan Tan
- Department of Pathology, The Third Xiangya Hospital of Central South University, No. 138 Tongzipo Road, Yuelu District, Changsha Hunan 410013, China
| | - Dongyang Lin
- Department of Anesthesiology and Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Haoran Jiang
- Department of Anesthesiology and Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Yimin Chu
- Digestive Endoscopy Center, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China
| | - Luting Zhou
- Department of Pathology, Shanghai Jiao Tong University Medical School Affiliated Ruijin Hospital, Shanghai 200025, China
| | - Junjie Zhang
- Department of Anesthesiology and Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| | - Yinzhong Lu
- Department of Anesthesiology and Hongqiao International Institute of Medicine, Tongren Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai 200336, China.
| |
Collapse
|
7
|
Li D, Ma H, Dong X, Fang Z, Zhang C, Song Q, Wang J, Wan X. ATF3 deficiency promotes alveolar macrophage pyroptosis in sepsis-induced acute lung injury. Mol Immunol 2024; 176:84-95. [PMID: 39591838 DOI: 10.1016/j.molimm.2024.11.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Revised: 10/21/2024] [Accepted: 11/13/2024] [Indexed: 11/28/2024]
Abstract
BACKGROUND Acute lung injury (ALI) ranks among the leading reasons for death in septic patients. As an essential transcription factor associated with stress, activating transcription factor 3 (ATF3) participates in a variety of pathophysiological processes, including immunology and inflammation. However, the specific mechanism of ATF3 in pyroptosis of sepsis-induced ALI remains elusive. METHODS A mouse model of ALI was established by administering lipopolysaccharide (LPS) in vivo and LPS combined with adenosine triphosphate (ATP) in vitro to compare differences in ATF3 expression level. The role of ATF3 in pyroptosis was then assessed by knocking down ATF3 using small interfering RNA. The levels of interleukin-6 (IL-6), tumor necrosis factor-α, IL-1β, and IL-18 in mouse serum and cell culture supernatants were measured using enzyme-linked immunosorbent assay. Moreover, immunohistochemistry, immunofluorescence, Western blotting, co-immunoprecipitation, and quantitative reverse transcription polymerase chain reaction were employed for examining pyroptosis and pyroptotic pathways. RESULTS Both in vitro and in vivo ALI models were successfully established. LPS could activate the pyroptotic signaling pathway, and the expression level of ATF3 peaked 6 h after LPS and ATP stimulation in vitro. ATF3 could interact with NLRP3 and potentially influence the assembly of the inflammasome. This mechanism could involve the inhibition of the classical pyroptotic pathway, including Caspase-1 and Gasdermin D transcription and cleavage, as well as the inhibition of the non-classical pyroptotic pathway, including transcription and cleavage of Caspase-11. CONCLUSION The results indicated that inhibition of ATF3 could exacerbate sepsis-induced ALI by regulating pyroptotic pathways. The potential of targeting ATF3 as a future treatment strategy for ALI is noteworthy.
Collapse
Affiliation(s)
- Dandan Li
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Huanyu Ma
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Xue Dong
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Zhiyao Fang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Chenyang Zhang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Qing Song
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China
| | - Jia Wang
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China.
| | - Xianyao Wan
- Department of Critical Care Medicine, The First Affiliated Hospital of Dalian Medical University, Dalian 116021, China.
| |
Collapse
|
8
|
Kong R, Peng L, Bao H, Sun L, Feng Y, Li H, Wang D. The role of Gαq in regulating NLRP3 inflammasome activation. Inflamm Res 2024; 73:2249-2261. [PMID: 39455437 DOI: 10.1007/s00011-024-01961-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: 12/22/2023] [Revised: 09/20/2024] [Accepted: 10/11/2024] [Indexed: 10/28/2024] Open
Abstract
BACKGROUND G proteins are a class of important signal transducers in mammalians. G proteins can corpoarated with G proteincoupled receptors (GPCRs) and transmit signals from extracellular stimuli into intracellular response, which will regulate a series of biological functions. G-proteins are heterotrimeric proteins composed of Gα, Gβ, and Gγ subunits. Based on structural and functional similarity of their α-subunits, G proteins are typically grouped into four classes (Gi, Gs, Gq/11, and G12/13). The Gq/11 subfamily consists of Gq, G11, G14, and G15/16 proteins. Gαq is the α-subunit of Gq protein and encoded by GNAQ. Our previous studies revealed that Gαq play an important role in regulating T cell survival and T cell differentiation. Inflammasomes are multiprotein complexes that play a critical role in modulating innate inflammatory response. NLRP3 inflammasome is currently the most extensively studied inflammasome. METHODS We found that Gαq suppressed NLRP3 inflammasome activation in macrophage, Gαq also suppressed NLRP3 inflammasome activation in a LPS-induced sepsis mouse model. Gαq can locate to mitochondria and Gαq was required for the maintenance of mitochondrial homeostasis. Gαq regulated NLRP3 inflammasome activation by modulating mitochondrial reactive oxygen species (mtROS). RESULTS We found that Gαq suppressed NLRP3 inflammasome activation in macrophage, Gαq also suppressed NLRP3 inflammasome activation in a LPS-induced sepsis mouse model. Gαq can locate to mitochondria and Gαq was required for the maintenance of mitochondrial homeostasis. Gαq regulated NLRP3 inflammasome activation by modulating mitochondrial reactive oxygen species (mtROS). CONCLUSION Our results indicate that Gαq regulates NLRP3 inflammasome activation by modulating mitochondrial ROS production. Our research provides new mechanistic insight into the activation of NLRP3 inflammasome. As it has been proved that NLRP3 inflammasome plays an important role in the pathogenesis many diseases such as Alzheimer's disease, cancer, and inflammatory bowel disease, Gαq might become a novel drug target for these diseases in future.
Collapse
Affiliation(s)
- Ruixue Kong
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Lijun Peng
- Department of Gastroenterology, Linyi People's Hospital, Linyi, 276000, Shandong, China
| | - Honggang Bao
- Department of Laboratory Medicine, Linyi Cancer Hospital, Linyi, 276000, Shandong, China
| | - Lulu Sun
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Yan Feng
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China
| | - Hua Li
- College of Life Sciences, Shandong Normal University, Jinan, 250014, Shandong, China.
| | - Dashan Wang
- Research Center, Shandong Medical College, Linyi, 276000, Shandong, China.
| |
Collapse
|
9
|
Chen PR, Li CY, Yazal T, Chen IC, Liu PL, Chen YT, Liu CC, Lo J, Lin TC, Chang CT, Wu HE, Chen YR, Cheng WC, Chiu CC, Chen CS, Wang SC. Protective effects of nordalbergin against LPS-induced endotoxemia through inhibiting MAPK/NF-κB signaling pathway, NLRP3 inflammasome activation, and ROS production. Inflamm Res 2024; 73:1657-1670. [PMID: 39052062 DOI: 10.1007/s00011-024-01922-4] [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: 02/05/2024] [Revised: 06/17/2024] [Accepted: 07/15/2024] [Indexed: 07/27/2024] Open
Abstract
OBJECTIVE Nordalbergin is a coumarin extracted from Dalbergia sissoo DC. To date, the biological effects of nordalbergin have not been well investigated. To investigate the anti-inflammatory responses and the anti-oxidant abilities of nordalbergin using lipopolysaccharide (LPS)-activated macrophages and LPS-induced sepsis mouse model. MATERIALS AND METHODS Production of nitrite oxide (NO), prostaglandin E2 (PGE2), pro-inflammatory cytokines (tumor necrosis factor (TNF)-α, interleukin (IL)-6 and IL-1β), reactive oxygen species (ROS), tissue damage and serum inflammatory markers, and the activation of the NLRP3 inflammasome were examined. RESULTS Our results indicated that nordalbergin reduced the production of NO and pro-inflammatory cytokines in vitro and ex vivo. Nordalbergin also suppressed iNOS and cyclooxygenase-2 expressions, decreased NF-κB activity, and attenuated MAPKs signaling pathway activation by decreasing JNK and p38 phosphorylation by LPS-activated J774A.1 macrophages. Notably, nordalbergin diminished NLRP3 inflammasome activation via repressing the maturation of IL-1β and caspase-1 and suppressing ROS production by LPS/ATP- and LPS/nigericin-activated J774A.1 macrophages. Furthermore, nordalbergin exhibited protective effects against the infiltration of inflammatory cells and also inhibited the levels of organ damage markers (AST, ALT, BUN) by LPS-challenged mice. CONCLUSION Nordalbergin possesses anti-inflammatory effects in macrophage-mediated innate immune responses, alleviates ROS production, decreases NLRP3 activation, and exhibits protective effects against LPS-induced tissue damage in mice.
Collapse
Grants
- NSTC 111-2218-E-037-001, NSTC 111-2314-B-037-071-MY3, NSTC 112-2314-B-037-127, NSTC 112-2314-B-037-128, NSTC 112-2926-I-037-501-G, NSTC 112-2314-B-037-089, NSTC 112-2311-B-039 -001, NSTC 112-2622-E-039-001, NSTC 111-2622-E-039-004 and NSTC 112-2218-E-037-001 National Science and Technology Council, Taiwan, R.O.C.
- KT113P010 NTHU-KMU Joint Research Project
- CMU111-IP-04 and CMU112-MF-25 China Medical University, Taiwan
- DMR-112-056, DMR-113-190, and DMR-113-191 China Medical University Hospital
Collapse
Affiliation(s)
- Pin-Rong Chen
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chia-Yang Li
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Biological Science and Technology, National Pingtung University of Science and Technology, Pingtung, 91201, Taiwan
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
| | - Taha Yazal
- School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - I-Chen Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Pediatrics, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan
- Department of Pediatrics, School of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Po-Len Liu
- Department of Respiratory Therapy, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yi-Ting Chen
- Department of Pathology, Taichung Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, Taichung, 427213, Taiwan
| | - Ching-Chih Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Chi Mei Medical Center, Tainan, 71004, Taiwan
| | - Jung Lo
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Ophthalmology, Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, 83301, Taiwan
| | - Tzu-Chieh Lin
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Ching-Tang Chang
- Graduate Institute of Clinical Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Division of Cardiology, Department of Internal Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
- Department of Internal Medicine, Kaohsiung Municipal Ta-Tung Hospital, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Hsin-En Wu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Yuan-Ru Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Wei-Chung Cheng
- Cancer Biology and Precision Therapeutics Center, China Medical University, Taichung, 40403, Taiwan
- Ph.D. Program for Cancer Biology and Drug Discovery, China Medical University, Academia Sinica, Taipei, 40403, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan
| | - Chi-Shuo Chen
- Department of Biomedical Engineering and Environmental Sciences, National Tsing Hua University, Hsinchu, 30013, Taiwan
| | - Shu-Chi Wang
- Department of Medical Research, Kaohsiung Medical University Hospital, Kaohsiung, 80756, Taiwan.
- Department of Medical Laboratory Science and Biotechnology, Kaohsiung Medical University, Kaohsiung, 80708, Taiwan.
| |
Collapse
|
10
|
Thom RE, D’Elia RV. Future applications of host direct therapies for infectious disease treatment. Front Immunol 2024; 15:1436557. [PMID: 39411713 PMCID: PMC11473292 DOI: 10.3389/fimmu.2024.1436557] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2024] [Accepted: 09/06/2024] [Indexed: 10/19/2024] Open
Abstract
New and emerging pathogens, such as SARS-CoV2 have highlighted the requirement for threat agnostic therapies. Some antibiotics or antivirals can demonstrate broad-spectrum activity against pathogens in the same family or genus but efficacy can quickly reduce due to their specific mechanism of action and for the ability of the disease causing agent to evolve. This has led to the generation of antimicrobial resistant strains, making infectious diseases more difficult to treat. Alternative approaches therefore need to be considered, which include exploring the utility of Host-Directed Therapies (HDTs). This is a growing area with huge potential but difficulties arise due to the complexity of disease profiles. For example, a HDT given early during infection may not be appropriate or as effective when the disease has become chronic or when a patient is in intensive care. With the growing understanding of immune function, a new generation of HDT for the treatment of disease could allow targeting specific pathways to augment or diminish the host response, dependent upon disease profile, and allow for bespoke therapeutic management plans. This review highlights promising and approved HDTs that can manipulate the immune system throughout the spectrum of disease, in particular to viral and bacterial pathogens, and demonstrates how the advantages of HDT will soon outweigh the potential side effects.
Collapse
Affiliation(s)
- Ruth E. Thom
- Chemical, Biological and Radiological Division, Defence Science and Technology Laboratory, Porton Down, Salisbury, United Kingdom
| | - R V. D’Elia
- Strathclyde Institute of Pharmacy & Biomedical Sciences, University of Strathclyde, Glasgow, United Kingdom
| |
Collapse
|
11
|
Liu ML, Wong WT, Weng YM, Ho CL, Hsu HT, Hua KF, Wu CH, Li LH. Cinnamaldehyde, A Bioactive Compound from the Leaves of Cinnamomum osmophloeum Kaneh, Ameliorates Dextran Sulfate Sodium-Induced Colitis in Mice by Inhibiting the NLRP3 Inflammasome. JOURNAL OF PHYSIOLOGICAL INVESTIGATION 2024; 67:139-152. [PMID: 38902958 DOI: 10.4103/ejpi.ejpi-d-24-00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Accepted: 05/07/2024] [Indexed: 06/22/2024]
Abstract
Inflammatory bowel disease (IBD) comprises a group of idiopathic intestinal disorders, including ulcerative colitis and Crohn's disease, significantly impacting the quality of life for affected individuals. The effective management of these conditions remains a persistent challenge. The NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome, a complex molecular structure, regulates the production of pro-inflammatory cytokines such as interleukin-1β. Abnormal activation of the NLRP3 inflammasome plays a pivotal role in the development of IBD, making it a compelling target for therapeutic intervention. Our research revealed that cinnamaldehyde (CA), a major bioactive compound found in the leaves of Cinnamomum osmophloeum kaneh, demonstrated a remarkable ability to alleviate colitis induced by dextran sulfate sodium (DSS) in a mouse model. This effect was attributed to CA's ability to downregulate the activation of the NLRP3 inflammasome and reduce the expression of pro-inflammatory mediators in the colon. In the mechanism study, we observed that CA inhibited the NLRP3 inflammasome in macrophages, at least partially, by enhancing the autophagic response, without reducing mitochondrial damage. These findings collectively suggest that CA holds significant potential as a therapeutic agent for enhancing the management of IBD, offering a promising avenue for further research and development.
Collapse
Affiliation(s)
- May-Lan Liu
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
- Department of Early Childhood Educare, WuFeng University, Chiayi, Taiwan
| | - Wei-Ting Wong
- Taiwan Autoantibody Biobank Initiative, Hualien Tzu Chi Hospital, Hualien, Taiwan
| | - Yih-Ming Weng
- Department of Food Science, National Chiayi University, Chiayi, Taiwan
| | - Chen-Lung Ho
- Division of Wood Cellulose, Taiwan Forestry Research Institute, Taipei, Taiwan
| | - Hsien-Ta Hsu
- Division of Neurosurgery, Taipei Tzu Chi Hospital, Buddhist Tzu Chi Medical Foundation, New Taipei City, Taiwan
- School of Medicine, Buddhist Tzu Chi University, Hualien, Taiwan
| | - Kuo-Feng Hua
- Department of Biotechnology and Animal Science, National Ilan University, Ilan, Taiwan
- Department of Medical Research, China Medical University Hospital, China Medical University, Taichung, Taiwan
| | - Chun-Hsien Wu
- Department of Internal Medicine, Division of Cardiology, Tri-Service General Hospital, National Defense Medical Center, Taipei, Taiwan
| | - Lan-Hui Li
- Department of Laboratory Medicine, Linsen, Chinese Medicine and Kunming Branch, Taipei City Hospital, Taipei, Taiwan
| |
Collapse
|
12
|
Ziehe D, Marko B, Thon P, Rahmel T, Palmowski L, Nowak H, von Busch A, Wolf A, Witowski A, Vonheder J, Ellger B, Wappler F, Schwier E, Henzler D, Köhler T, Zarbock A, Ehrentraut SF, Putensen C, Frey UH, Anft M, Babel N, Adamzik M, Koos B, Bergmann L, Unterberg M, Rump K. The Aquaporin 3 Polymorphism (rs17553719) Is Associated with Sepsis Survival and Correlated with IL-33 Secretion. Int J Mol Sci 2024; 25:1400. [PMID: 38338680 PMCID: PMC10855683 DOI: 10.3390/ijms25031400] [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: 12/11/2023] [Revised: 01/16/2024] [Accepted: 01/22/2024] [Indexed: 02/12/2024] Open
Abstract
Sepsis is a common life-threatening disease caused by dysregulated immune response and metabolic acidosis which lead to organ failure. An abnormal expression of aquaporins plays an important role in organ failure. Additionally, genetic variants in aquaporins impact on the outcome in sepsis. Thus, we investigated the polymorphism (rs17553719) and expression of aquaporin-3 (AQP3) and correlated these measurements with the survival of sepsis patients. Accordingly, we collected blood samples on several days (plus clinical data) from 265 sepsis patients who stayed in different ICUs in Germany. Serum plasma, DNA, and RNA were then separated to detect the promotor genotypes of AQP3 mRNA expression of AQP3 and several cytokines. The results showed that the homozygote CC genotype exhibited a significant decrease in 30-day survival (38.9%) compared to the CT (66.15%) and TT genotypes (76.3%) (p = 0.003). Moreover, AQP3 mRNA expression was significantly higher and nearly doubled in the CC compared to the CT (p = 0.0044) and TT genotypes (p = 0.018) on the day of study inclusion. This was accompanied by an increased IL-33 concentration in the CC genotype (day 0: p = 0.0026 and day 3: p = 0.008). In summary, the C allele of the AQP3 polymorphism (rs17553719) shows an association with increased AQP3 expression and IL-33 concentration accompanied by decreased survival in patients with sepsis.
Collapse
Affiliation(s)
- Dominik Ziehe
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Britta Marko
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Patrick Thon
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Tim Rahmel
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Lars Palmowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Hartmuth Nowak
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
- Center for Artificial Intelligence, Medical Informatics and Data Science, University Hospital Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany
| | - Alexander von Busch
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Alexander Wolf
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Andrea Witowski
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Jolene Vonheder
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Björn Ellger
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Klinikum Westfalen, 44309 Dortmund, Germany;
| | - Frank Wappler
- Department of Anesthesiology and Operative Intensive Care Medicine, University of Witten/Herdecke, Cologne Merheim Medical School, 51109 Cologne, Germany;
| | - Elke Schwier
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Dietrich Henzler
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Thomas Köhler
- Department of Anesthesiology, Surgical Intensive Care, Emergency and Pain Medicine, Ruhr-University Bochum, Klinikum Herford, 32049 Herford, Germany; (E.S.); (D.H.); (T.K.)
| | - Alexander Zarbock
- Klinik für Anästhesiologie, Operative Intensivmedizin und Schmerztherapie, Universitätsklinikum Münster, 48149 Münster, Germany;
| | - Stefan Felix Ehrentraut
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, 53127 Bonn, Germany; (S.F.E.); (C.P.)
| | - Christian Putensen
- Klinik für Anästhesiologie und Operative Intensivmedizin, Universitätsklinikum Bonn, 53127 Bonn, Germany; (S.F.E.); (C.P.)
| | - Ulrich Hermann Frey
- Marien Hospital Herne, Universitätsklinikum der Ruhr-Universität Bochum, 44625 Herne, Germany;
| | - Moritz Anft
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Herne, Germany; (M.A.); (N.B.)
| | - Nina Babel
- Center for Translational Medicine, Medical Clinic I, Marien Hospital Herne, University Hospital of the Ruhr-University Bochum, 44625 Herne, Germany; (M.A.); (N.B.)
| | - Michael Adamzik
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Björn Koos
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Lars Bergmann
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Matthias Unterberg
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| | - Katharina Rump
- Klinik für Anästhesiologie, Intensivmedizin und Schmerztherapie, Universitätsklinikum Knappschaftskrankenhaus Bochum, 44892 Bochum, Germany; (D.Z.); (B.M.); (P.T.); (T.R.); (L.P.); (H.N.); (A.v.B.); (A.W.); (A.W.); (J.V.); (M.A.); (B.K.); (L.B.); (M.U.)
| |
Collapse
|
13
|
Sabnis RW. Pyridazine Derivatives as NLRP3 Inhibitors for Treating Asthma, COPD, Parkinson's Disease, and Alzheimer's Disease. ACS Med Chem Lett 2023; 14:1047-1048. [PMID: 37583813 PMCID: PMC10424320 DOI: 10.1021/acsmedchemlett.3c00311] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2023] [Indexed: 08/17/2023] Open
Abstract
Provided herein are novel pyridazine derivatives as NLRP3 inhibitors, pharmaceutical compositions, use of such compounds in treating asthma, COPD, Parkinson's disease, and Alzheimer's disease, and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell LLP, 1105 W. Peachtree Street NE, Suite
1000, Atlanta, Georgia 30309, United States
| |
Collapse
|
14
|
Sabnis RW. Fused Bicyclic Heteroaryl Compounds as NLRP3 Inhibitors for Treating Asthma or COPD. ACS Med Chem Lett 2023; 14:895-896. [PMID: 37465294 PMCID: PMC10351047 DOI: 10.1021/acsmedchemlett.3c00236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2023] [Indexed: 07/20/2023] Open
Abstract
Provided herein are novel fused bicyclic heteroaryl compounds as NLRP3 inhibitors, pharmaceutical compositions, use of such compounds in treating asthma or COPD and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell LLP, 1105 W. Peachtree Street NE, Suite
1000, Atlanta, Georgia 30309, United States
| |
Collapse
|
15
|
Awni AA, Hamed ZO, Abdul-Hassan Abbas A, Abdulamir AS. Effect of NLRP3 inflammasome genes polymorphism on disease susceptibility and response to TNF-α inhibitors in Iraqi patients with rheumatoid arthritis. Heliyon 2023; 9:e16814. [PMID: 37332933 PMCID: PMC10275785 DOI: 10.1016/j.heliyon.2023.e16814] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Revised: 04/19/2023] [Accepted: 05/29/2023] [Indexed: 06/20/2023] Open
Abstract
Background Rheumatoid arthritis (RA) is a genetically predisposed, systemic, chronic, inflammatory disease. Immune system dysregulation and inherited susceptibility polymorphisms suggest that this type of variation is functional and may help predict disease susceptibility and develop new therapeutic strategies. Anti-TNF-alpha (TNF-α) drugs are highly effective RA treatments, but not all patients respond the same way. It's important to figure out whether RA risk alleles can identify and predict anti-TNF-α-responsiveness in RA patients. Aims of the study Examine the function of the NLR family pyrin domain containing 3 (NLRP3) and caspase recruitment domain family member 8 (CARD8) genes polymorphisms and their morbid genotypes and alleles in RA patients and apparently healthy controls. In addition, their role in disease susceptibility, severity, and response to anti-TNF-α therapy. Also, examine how single nucleotide polymorphisms (SNPs) affect serum levels of pro-inflammatory cytokines like TNF-α and interleukin (IL)-1β. Materials and methods 100 RA patients (88 females, 12 males) and 100 apparently healthy people (86 females, 14 males) were examined. To measure serum TNF-α and IL-1β, Elabscience sandwich ELISA kits were used. Iraq Biotech, Turkey DNA extraction kit was used to extract genomic DNA from whole blood. CARD8 (rs2043211) and NLRP3 (rs4612666) were genotyped using Agilent, AriaMx, USA, through Tri-Plex SYBR Green-based real-time PCR allelic discrimination assays. Geneious software, version 2019.2.2, used to design primers from published sequences (GenBank accession no. GCA 009914755.1). Primer specificity was determined by NCBI's BLAST. Results Study found that there is association between cytokines serum level and 28-joints disease activity score (DAS-28). The level of TNF-α increases with the higher DAS-28 (r2 = 0.45, P < 0.0001). Also, IL- 1β level increases with higher DAS-28 (r2 = 0.51, P < 0.0001). There were no statistically significant variations between patients with RA and the control group in the distribution of CARD8 SNP rs2043211 and NLRP3 SNP rs4612666 genotypes (P = 0.17 and 0.08 respectively) as well their alleles (P = 0.059 and 0.879 respectively). CARD8 (rs2043211) TT genotype was more frequent in patients with higher DAS-28 (P < 0.0001) and higher TNF-α and IL-1β serum levels (P < 0.0001 for both). Also, NLRP3 (rs4612666) TT genotype was more frequent in patients with higher DAS-28 (P < 0.0001) and higher TNF-α and IL- 1β serum levels (P < 0.0001 for both). Interestingly, this study revealed that CARD8 (rs2043211) and NLRP3 (rs4612666) variant genotypes are associated with lower response to anti-TNF-α drugs. Conclusions Serum TNF-α and IL-1β correlate with DAS-28 and disease activity. Non-responders have elevated TNF-α and IL-1β. CARD8 rs2043211 and NLRP3 rs4612666 variant polymorphisms are associated with high serum TNF-α and IL-1β, active disease course, poor disease outcomes, and low response to anti-TNF-α therapy.
Collapse
Affiliation(s)
- Abdullah Abbas Awni
- College of Medical Sciences Techniques, The University of Mashreq, Baghdad, Iraq
| | - Zainab Oday Hamed
- Therapeutics and Clinical Pharmacy Department, Baghdad College of Medical Sciences, Baghdad, Iraq
| | | | | |
Collapse
|
16
|
Sabnis RW. 1,2,4-Triazine Derivatives as NLRP3 Inhibitors for Treating Diseases. ACS Med Chem Lett 2023; 14:547-548. [PMID: 37197471 PMCID: PMC10184145 DOI: 10.1021/acsmedchemlett.3c00122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2023] [Indexed: 05/19/2023] Open
Abstract
Provided herein are 1,2,4-triazine derivatives used as NLRP3 inhibitors, their pharmaceutical compositions, the use of such compounds in treating diseases and processes for preparing such compounds.
Collapse
Affiliation(s)
- Ram W. Sabnis
- Smith, Gambrell & Russell
LLP, 1105 W. Peachtree Street NE, Suite 1000, Atlanta, Georgia 30309, United States
| |
Collapse
|