1
|
You X, Li H, Li Q, Zhang Q, Cao Y, Fu W, Wang B. Astragaloside IV-PESV facilitates pyroptosis by enhancing palmitoylation of GSDMD protein mediated by ZDHHC1. NAUNYN-SCHMIEDEBERG'S ARCHIVES OF PHARMACOLOGY 2025:10.1007/s00210-025-04122-x. [PMID: 40237800 DOI: 10.1007/s00210-025-04122-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2024] [Accepted: 03/28/2025] [Indexed: 04/18/2025]
Abstract
Prostate cancer (PCa) is an epithelial malignancy affecting the prostate gland. Astragaloside IV combined with polypeptide extract from scorpion venom (PESV) has been reported to inhibit the growth of PCa. This study aimed to investigate the mechanisms by which this combination mitigates the progression of PCa. Bioinformatic analysis was utilized to investigate the correlation between zinc finger DHHC-type containing 1 (ZDHHC1) expression and PCa progression. The extent of pyroptosis in PCa cells was assessed by measuring cell viability, IL-1β and IL-18 secretion, LDH release, and HMGB1 content. PCa mouse models were constructed by subcutaneous injection of DU145 or PC-3 cells into nude mice, with subsequent monitoring of tumor weight and volume. ZDHHC1 expression was significantly lower in PCa patient tissues, which correlated with a poor prognosis. ZDHHC1 overexpression inhibited PC-3 and DU145 cell viability and increased IL-1β, IL-18, LDH, and HMGB1 levels in cell supernatants. Notably, the pyroptosis inhibitor LDC7559 partially reversed these effects. Co-IP assay demonstrated an interaction between ZDHHC1 and GSDMD. ZDHHC1 overexpression significantly enhanced GSDMD palmitoylation-mediated membrane translocation and pyroptosis; however, this effect was partially reversed by the palmitoylation inhibitor 2-BP. The combination of Astragaloside IV and PESV promoted GSDMD membrane translocation and pyroptosis in PCa cells, with ZDHHC1 knockdown partially reversing the effects of Astragaloside IV-PESV. Furthermore, treatment with Astragaloside IV-PESV significantly inhibited tumor tissue growth in tumor-bearing nude mouse models. Astragaloside IV-PESV enhances palmitoylation-mediated membrane translocation of GSDMD-N by upregulating ZDHHC1 expression, thereby facilitating pyroptosis in PCa cells and attenuating PCa progression.
Collapse
Affiliation(s)
- Xujun You
- Department of Andrology, Dongcheng District, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hai Yun Cang on the 5 th, Beijing, 100700, China
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Honghan Li
- The Seventh Clinical Medical College of Guangzhou, University of Traditional Chinese Medicine, Shenzhen, 518133, China
| | - Qixin Li
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Qing Zhang
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Yiguo Cao
- Department of Urology Surgery, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China
| | - Wei Fu
- Department of Andrology, Shenzhen Bao'an Traditional Chinese Medicine Hospital, Guangzhou University of Chinese Medicine, Shenzhen, 518101, China.
| | - Bin Wang
- Department of Andrology, Dongcheng District, Dongzhimen Hospital, Beijing University of Chinese Medicine, Hai Yun Cang on the 5 th, Beijing, 100700, China.
| |
Collapse
|
2
|
Quach A, Lao Y, Troelnikov A, Hissaria P, Chan D, Wells A, Kulkarni A, Rawlings L, Poplawski N, Ferrante A. Rapid validation of NLRP3 gain-of-function variants in cryopyrin-associated periodic syndromes. J Allergy Clin Immunol 2025; 155:1392-1395. [PMID: 39891630 DOI: 10.1016/j.jaci.2025.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/05/2024] [Accepted: 01/10/2025] [Indexed: 02/03/2025]
Affiliation(s)
- Alex Quach
- Department of Immunopathology at SA Pathology, Women's and Children's Hospital, North Adelaide, Australia; School of Biomedicine and the Robinson Research Institute, University of Adelaide, Adelaide, Australia.
| | - Yunyu Lao
- Department of Immunopathology at SA Pathology, Women's and Children's Hospital, North Adelaide, Australia; School of Biomedicine and the Robinson Research Institute, University of Adelaide, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia
| | | | - Pravin Hissaria
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Division of Human Immunology, SA Pathology, Adelaide, Australia; Clinical Immunology and Allergy, Royal Adelaide Hospital, Australia
| | - Damien Chan
- Department of Paediatric Allergy and Clinical Immunology, Women's and Children's Hospital, North Adelaide, Australia
| | - Amanda Wells
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - Abhijit Kulkarni
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - Lesley Rawlings
- Genetics and Molecular Pathology, SA Pathology, Adelaide, Australia
| | - Nicola Poplawski
- Adelaide Medical School, University of Adelaide, Adelaide, Australia; Adult Genetics Unit, Royal Adelaide Hospital, Australia
| | - Antonio Ferrante
- Department of Immunopathology at SA Pathology, Women's and Children's Hospital, North Adelaide, Australia; School of Biomedicine and the Robinson Research Institute, University of Adelaide, Adelaide, Australia; Adelaide Medical School, University of Adelaide, Adelaide, Australia; School of Biological Sciences, University of Adelaide, Adelaide, Australia
| |
Collapse
|
3
|
McCutcheon CR, Gaddy JA, Aronoff DM, Manning SD, Petroff MG. Group B Streptococcal Membrane Vesicles Induce Proinflammatory Cytokine Production and Are Sensed in an NLRP3 Inflammasome-Dependent Mechanism in a Human Macrophage-like Cell Line. ACS Infect Dis 2025; 11:453-462. [PMID: 39761308 PMCID: PMC11833861 DOI: 10.1021/acsinfecdis.4c00641] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 02/20/2025]
Abstract
Group B Streptococcus (GBS) is a major cause of fetal and neonatal mortality worldwide. Many of the adverse effects of invasive GBS are associated with inflammation; therefore, understanding bacterial factors that promote inflammation is of critical importance. Membrane vesicles (MVs), which are produced by many bacteria, may modulate host inflammatory responses. While it is known that mice injected intra-amniotically with GBS MVs exhibit large-scale leukocyte infiltration, preterm birth, and subsequent fetal death, the immune effectors driving this response remain unclear. Here, we hypothesized that THP-1 macrophage-like cells respond to GBS-derived MVs by producing proinflammatory cytokines and are recognized through one or more pattern recognition receptors. We show that THP-1s produce high levels of neutrophil- and monocyte-specific chemokines in response to MVs derived from different clinical isolates of GBS. Using antibody microarrays and multiplex Luminex assays, we found that GBS MVs elicit significantly (p < 0.05) higher levels of CCL1, CCL2, CCL20, CXCL1, CXCL10, and IL-1β relative to untreated THP-1s. Using chemical inhibitors in combination with caspase-1 activity assays and Luminex assays, we further demonstrate that GBS MVs upregulated IL-1β production in a caspase-1 and NLRP3-dependent manner, ultimately identifying NLRP3 as a sensor of GBS MVs. These data indicate that MVs contain one or more pathogen-associated molecular patterns that can be sensed by the immune system and show that the NLRP3 inflammasome is a novel sensor of GBS MVs. Our data additionally indicate that MVs may serve as immune effectors that can be targeted for immunotherapeutics.
Collapse
Affiliation(s)
- Cole R. McCutcheon
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Jennifer A. Gaddy
- Department
of Medicine, Division of Infectious Disease, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Department
of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, United States
- Tennessee
Valley Healthcare System, Department of
Veterans Affairs, Nashville, Tennessee 37212, United States
| | - David M. Aronoff
- Department
of Medicine, Indiana University School of
Medicine, Indianapolis, Indiana 46202, United States
| | - Shannon D. Manning
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
| | - Margaret G. Petroff
- Department
of Microbiology, Genetics, and Immunology, Michigan State University, East Lansing, Michigan 48824, United States
- Department
of Pathobiology and Diagnostic Investigation, Michigan State University, East
Lansing, Michigan 48824, United States
| |
Collapse
|
4
|
La Mensa A, Buscetta M, Woldhuis RR, Cimino M, Giuffrè MR, Cristaldi M, Dino P, Fiore L, Fucarino A, Lo Iacono G, Bertani A, Brandsma CA, Bucchieri F, Cipollina C. Caspase inhibition restores collagen Iα1 and fibronectin release in cigarette smoke extract-exposed human lung fibroblasts. Am J Physiol Lung Cell Mol Physiol 2025; 328:L239-L252. [PMID: 39772929 DOI: 10.1152/ajplung.00214.2024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 12/13/2024] [Accepted: 12/26/2024] [Indexed: 01/11/2025] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is a progressive lung disease characterized by obstructed airflow, airway remodeling, and inflammation, with cigarette smoke (CS) exposure being the main risk factor. Although CS extract (CSE) has been shown to activate caspases in various cell types, the role of caspases in human lung fibroblasts (hLFs) in COPD remains poorly understood. Recent studies have linked caspases to extracellular matrix (ECM) remodeling in skin and kidney fibrosis. Caspase activation can be triggered by oxidative stress, with active caspase-3 executing the pore-forming protein gasdermin E (GSDME) in the cleaved N-terminal form GSDME-NT. We investigated whether CSE activates caspases and GSDME in hLFs and their role in ECM remodeling. MRC-5 lung fibroblasts were treated with CSE with or without the antioxidant N-acetyl-cysteine (NAC) and the caspase-8 inhibitor z-IETD-fmk. We measured the effects on caspase-1-8-3/7 activation, GSDME cleavage, ECM remodeling (procollagen Iα1, COLIα1, and fibronectin, FN), and mitochondrial superoxide (mSOX) generation. Key findings were validated in patient-derived hLFs (phLFs). Our results showed that CSE induced caspase-1-8-3/7 activation, mSOX generation, and decreased COLIα1 and FN levels in MRC-5. CSE caused caspase-8-dependent activation of caspase-3, leading to GSDME cleavage. Treatment with NAC inhibited mSOX and caspase activation. Inhibition of caspase-8 and mSOX restored FN and COLIα1 levels. In phLFs, we confirmed caspase-1 and -8 activation, mSOX increase, COLIα1/FN decrease, and the effects of NAC. Our findings highlight the role of the axis caspase-8-3/7-GSDME and mSOX in regulating ECM protein, suggesting that these pathways may contribute to remodeling in COPD.NEW & NOTEWORTHY This research investigates the connection between caspases, gasdermins, and extracellular matrix (ECM) remodeling in the context of cigarette smoke-associated lung diseases. The study found that cigarette smoke extract (CSE) activates caspases and gasdermin E in human lung fibroblasts, leading to decreased ECM protein expression and release. Findings herein reported suggest that targeting the caspase-8-3/7-gasdermin axis and mitochondrial reactive oxygen species may help restore ECM remodeling in chronic lung diseases associated with cigarette smoke exposure.
Collapse
Affiliation(s)
- Agnese La Mensa
- Ri.MED Foundation, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | - Roy R Woldhuis
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Maura Cimino
- Ri.MED Foundation, Palermo, Italy
- IRCCS ISMETT-UPMC Italy, Palermo, Italy
| | | | | | - Paola Dino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Luigi Fiore
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università di Messina, Messina, Italy
| | - Alberto Fucarino
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | | | | | - Corry-Anke Brandsma
- University Medical Center Groningen, University of Groningen, Groningen, The Netherlands
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics, University of Palermo, Palermo, Italy
| | - Chiara Cipollina
- Ri.MED Foundation, Palermo, Italy
- Istituto di Farmacologia Traslazionale-CNR, Palermo, Italy
| |
Collapse
|
5
|
Niharika, Roy A, Sadhukhan R, Patra SK. Screening and identification of gene expression in large cohorts of clinical tissue samples unveils the major involvement of EZH2 and SOX2 in lung cancer. Cancer Genet 2025; 290-291:16-35. [PMID: 39647236 DOI: 10.1016/j.cancergen.2024.11.006] [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: 09/19/2024] [Revised: 11/13/2024] [Accepted: 11/29/2024] [Indexed: 12/10/2024]
Abstract
Lung adenocarcinoma (LUAD), the primary subtype of Non-Small Cell Lung Cancer (NSCLC), accounts for 80 % to 85 % of cases. Due to suboptimal screening method, LUAD is often detected in late stage, leading to aggressive progression and poor outcomes. Therefore, early disease prognosis for the LUAD is high priority. In order to identify early detection biomarkers, we conducted a meta-analysis of mRNA expression TCGA and GTEx datasets from LUAD patients. A total of 795 differentially expressed genes (DEGs) were identified by exploring the Network-Analyst tool and utilizing combined effect size methods. DEGs refer to genes whose expression levels are significantly different (either higher or lower) compared to their normal baseline expression levels. KEGG pathway enrichment analysis highlighted the TNF signaling pathway as being prominently associated with these DEGs. Subsequently, using the MCODE and CytoHubba plugins in Cytoscape software, we filtered out the top 10 genes. Among these, SOX2 was the only gene exhibiting higher expression, while the others were downregulated. Consequently, our subsequent research focused on SOX2. Further transcription factor-gene network analysis revealed that enhancer of zeste homolog 2 (EZH2) is a significant partner of SOX2, potentially playing a crucial role in euchromatin-heterochromatin dynamics. Structure of SOX2 protein suggest that it is a non-druggable transcription factor, literature survey suggests the same. SOX2 is considered challenging to target directly, or "non-druggable," because of several intrinsic properties that make it difficult to design effective therapeutic agents against it. The primary function of SOX2 is to bind DNA and regulates gene expression. Unlike enzymes or receptors with defined active sites or binding pockets, transcription factors typically have relatively flat or diffuse surfaces that do not offer obvious "pockets" for small molecules to bind effectively. Hence, we drove our focus to investigate on potential drug(s) targeting EZH2. Molecular docking analyses predicted most probable inhibitors of EZH2. We employed several predictive analysis tools and identified GSK343, as a promising inhibitor of EZH2.
Collapse
Affiliation(s)
- Niharika
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Ankan Roy
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India
| | - Ratan Sadhukhan
- Department of Radiation Oncology, University of Arkansas for Medical Sciences, Little Rock, Arkansas, USA
| | - Samir Kumar Patra
- Epigenetics and Cancer Research Laboratory, Biochemistry and Molecular Biology Group, Department of Life Science, National Institute of Technology, Rourkela, India.
| |
Collapse
|
6
|
Vervaeke A, Lamkanfi M. MAP Kinase Signaling at the Crossroads of Inflammasome Activation. Immunol Rev 2025; 329:e13436. [PMID: 39754394 DOI: 10.1111/imr.13436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2024] [Accepted: 12/14/2024] [Indexed: 01/06/2025]
Abstract
Inflammasomes are crucial mediators of both antimicrobial host defense and inflammatory pathology, requiring stringent regulation at multiple levels. This review explores the pivotal role of mitogen-activated protein kinase (MAPK) signaling in modulating inflammasome activation through various regulatory mechanisms. We detail recent advances in understanding MAPK-mediated regulation of NLRP3 inflammasome priming, licensing and activation, with emphasis on MAPK-induced activator protein-1 (AP-1) signaling in NLRP3 priming, ERK1 and JNK in NLRP3 licensing, and TAK1 in connecting death receptor signaling to NLRP3 inflammasome activation. Furthermore, we discuss novel insights into MAPK signaling in human NLRP1 inflammasome activation, focusing on the MAP3K member ZAKα as a key kinase linking ribosomal stress to inflammasome activation. Lastly, we review recent work elucidating how Bacillus anthracis lethal toxin (LeTx) manipulates host MAPK signaling to induce macrophage apoptosis as an immune evasion strategy, and the counteraction of this effect through genotype-specific Nlrp1b inflammasome activation in certain rodent strains.
Collapse
Affiliation(s)
- Alex Vervaeke
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Mohamed Lamkanfi
- Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| |
Collapse
|
7
|
Rosarda JD, Stanton CR, Chen EB, Bollong MJ, Wiseman RL. Pharmacologic Targeting of PDIA1 Inhibits NLRP3 Inflammasome Assembly and Activation. Isr J Chem 2024; 64:e202300125. [PMID: 40370770 PMCID: PMC12077611 DOI: 10.1002/ijch.202300125] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Indexed: 05/16/2025]
Abstract
The NLRP3 inflammasome is a cytosolic protein complex that regulates innate immune signaling in response to diverse pathogenic insults through the proteolytic processing and secretion of pro-inflammatory cytokines such as IL-1β. Hyperactivation of NLRP3 inflammasome signaling is implicated in the onset and pathogenesis of numerous diseases, motivating the discovery of new strategies to suppress NLRP3 inflammasome activity. We sought to define the potential for the proteostasis regulator AA147 to inhibit the assembly and activation of the NLRP3 inflammasome. AA147 is a pro-drug that is metabolically converted to a reactive metabolite at the endoplasmic reticulum (ER) membrane to covalently modify ER-localized proteins such as protein disulfide isomerases (PDIs). We show that AA147 inhibits NLRP3 inflammasome activity in monocytes and monocyte-derived macrophages through a mechanism involving impaired assembly of the active inflammasome complex. This inhibition is mediated through AA147-dependent covalent modification of PDIA1. Genetic depletion or treatment with other highly selective PDIA1 inhibitors similarly blocks NLRP3 inflammasome assembly and activation. Our results identify PDIA1 as a potential therapeutic target to mitigate NLRP3 inflammasome-mediated pro-inflammatory signaling implicated in etiologically diverse diseases.
Collapse
Affiliation(s)
- Jessica D Rosarda
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, CA 92037
| | - Caroline R Stanton
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, CA 92037
- Department of Chemistry, Scripps Research, La Jolla, CA 92037
| | - Emily B Chen
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, CA 92037
| | | | - R Luke Wiseman
- Department of Molecular and Cellular Biology, Scripps Research, La Jolla, CA 92037
| |
Collapse
|
8
|
Hosseinkhani S, Amandadi M, Ghanavatian P, Zarein F, Ataei F, Nikkhah M, Vandenabeele P. Harnessing luciferase chemistry in regulated cell death modalities and autophagy: overview and perspectives. Chem Soc Rev 2024; 53:11557-11589. [PMID: 39417351 DOI: 10.1039/d3cs00743j] [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: 10/19/2024]
Abstract
Regulated cell death is a fate of cells in (patho)physiological conditions during which extrinsic or intrinsic signals or redox equilibrium pathways following infection, cellular stress or injury are coupled to cell death modalities like apoptosis, necroptosis, pyroptosis or ferroptosis. An immediate survival response to cellular stress is often induction of autophagy, a process that deals with removal of aggregated proteins and damaged organelles by a lysosomal recycling process. These cellular processes and their regulation are crucial in several human diseases. Exploiting high-throughput assays which discriminate distinct cell death modalities and autophagy are critical to identify potential therapeutic agents that modulate these cellular responses. In the past few years, luciferase-based assays have been widely developed for assessing regulated cell death and autophagy pathways due to their simplicity, sensitivity, known chemistry, different spectral properties and high-throughput potential. Here, we review basic principles of bioluminescent reactions from a mechanistic perspective, along with their implication in vitro and in vivo for probing cell death and autophagy pathways. These include applying luciferase-, luciferin-, and ATP-based biosensors for investigating regulated cell death modalities. We discuss multiplex bioluminescence platforms which simultaneously distinguish between the various cell death phenomena and cellular stress recovery processes such as autophagy. We also highlight the recent technological achievements of bioluminescent tools for the prediction of drug effectiveness in pathways associated with regulated cell death.
Collapse
Affiliation(s)
- Saman Hosseinkhani
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Mojdeh Amandadi
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Parisa Ghanavatian
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Fateme Zarein
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Farangis Ataei
- Department of Biochemistry, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran.
| | - Maryam Nikkhah
- Department of Nanobiotechnology, Faculty of Biological Sciences, Tarbiat Modares University, Tehran, Iran
| | - Peter Vandenabeele
- Cell Death and Inflammation Unit, VIB-UGent Center for Inflammation Research (IRC), Ghent, Belgium
- Department of Biomedical Molecular Biology (DBMB), Ghent University, Ghent, Belgium
- Cancer Research Institute Ghent (CRIG), Ghent, Belgium
| |
Collapse
|
9
|
Behrendt I, Becker K, Steingass CB, Schweiggert R, Michel G, Friedrich E, Grote D, Martin Z, Dötzer HP, Fasshauer M, Speckmann M, Kuntz S. Acylated Anthocyanins From Black Carrots and Their Related Phenolic Acids Diminish Priming and Activation of the NLRP3 Inflammasome in THP-1 Monocytes. Mol Nutr Food Res 2024; 68:e2400356. [PMID: 39425563 PMCID: PMC11605781 DOI: 10.1002/mnfr.202400356] [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: 05/15/2024] [Revised: 09/05/2024] [Indexed: 10/21/2024]
Abstract
SCOPE Excessive activation of the nucleotide-binding oligomerization domain-like receptor pyrin domain-containing protein 3 (NLRP3) inflammasome contributes to chronic inflammation. Thus, targeting NLRP3 inflammasome activation by anthocyanins may prevent inflammatory diseases. Therefore, the present study determines the influence of a black carrot extract (BCE) with high amounts of acylated anthocyanins and their related phenolic acids on the NLRP3 inflammasome. METHODS AND RESULTS THP-1 monocytes are pretreated with a BCE, cyanidin-3-glucoside (C3G), or hydroxycinnamic acids. NLRP3 inflammasome assembly is initiated by priming THP-1 monocytes with lipopolysaccharide and/or activating the NLRP3 inflammasome with nigericin. Flow cytometry is used to assess apoptosis-associated speck-like protein containing a caspase recruitment domain (ASC) speck formation, as well as ASC and NLRP3 protein expression. Caspase-1 activity is measured using a bioluminescent assay, and cytokine concentrations are determined by enzyme-linked immunosorbent assays (ELISA). C3G and phenolic acids diminish ASC and NLRP3 protein expression. In addition, C3G and phenolic acids attenuate ASC speck formation. Furthermore, the BCE and C3G decline caspase-1 activity. Consistently, IL-1β and IL-18 secretion are reduced upon NLRP3 inflammasome activation. CONCLUSION The present study shows that a BCE with high amounts of acylated anthocyanins and their related phenolic acids diminish priming and activation of the NLRP3 inflammasome in THP-1 monocytes.
Collapse
Affiliation(s)
- Inken Behrendt
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Katharina Becker
- Chair of Analysis and Technology of Plant‐based Foods – Focus on BeveragesDepartment of Beverage ResearchGeisenheim UniversityGeisenheimGermany
| | - Christof Björn Steingass
- Chair of Analysis and Technology of Plant‐based Foods – Focus on BeveragesDepartment of Beverage ResearchGeisenheim UniversityGeisenheimGermany
| | - Ralf Schweiggert
- Chair of Analysis and Technology of Plant‐based Foods – Focus on BeveragesDepartment of Beverage ResearchGeisenheim UniversityGeisenheimGermany
| | - Gabriela Michel
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Department of MedicineJustus‐Liebig‐University GiessenGiessenGermany
- Flow Cytometry Core FacilityDepartment of MedicineJustus‐Liebig‐University GiessenGiessenGermany
| | - Elvira Friedrich
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Daniela Grote
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Zoe Martin
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Hanna Pauline Dötzer
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Mathias Fasshauer
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| | - Martin Speckmann
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Department of MedicineJustus‐Liebig‐University GiessenGiessenGermany
- Flow Cytometry Core FacilityDepartment of MedicineJustus‐Liebig‐University GiessenGiessenGermany
| | - Sabine Kuntz
- Institute of Nutritional Science, Department of Nutritional ScienceJustus‐Liebig‐University GiessenGiessenGermany
| |
Collapse
|
10
|
Dino P, Giuffrè MR, Buscetta M, Di Vincenzo S, La Mensa A, Cristaldi M, Bucchieri F, Lo Iacono G, Bertani A, Pace E, Cipollina C. Release of IL-1β and IL-18 in human primary bronchial epithelial cells exposed to cigarette smoke is independent of NLRP3. Eur J Immunol 2024; 54:e2451053. [PMID: 39072707 DOI: 10.1002/eji.202451053] [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: 02/06/2024] [Revised: 07/16/2024] [Accepted: 07/18/2024] [Indexed: 07/30/2024]
Abstract
Cigarette smoke (CS) is a major risk factor for chronic lung diseases and promotes activation of pattern recognition receptors in the bronchial epithelium. NOD-like receptor family, pyrin domain-containing 3 (NLRP3) is a pattern recognition receptor whose activation leads to caspase-1 cleavage, maturation/release of IL-1β and IL-18, and eventually pyroptosis. Whether the NLRP3 inflammasome participates in CS-induced inflammation in bronchial epithelial cells is still unclear. Herein, we evaluated the involvement of NLRP3 in CS-induced inflammatory responses in human primary bronchial epithelial cells. To this purpose, human primary bronchial epithelial cells were stimulated with CS extracts (CSE) and lytic cell death, caspase activation (-1, -8, -3/7), cytokine release (IL-1β, IL-18, and IL-8), NLRP3, pro-IL-1β/pro-IL-18 mRNA, and protein expression were measured. The impact of inhibitors of NLRP3 (MCC950), caspases, and the effect of the antioxidant N-acetyl cysteine were evaluated. We found that CSE increased pro-IL-1β expression and induced activation of caspase-1 and release of IL-1β and IL-18. These events were independent of NLRP3 and we found that NLRP3 was not expressed. N-acetyl cysteine reverted CSE-induced caspase-1 activation. Overall, our findings support that the bronchial epithelium may play a central role in the release of IL-1 family cytokines independently of NLRP3 in the lungs of smokers.
Collapse
Affiliation(s)
- Paola Dino
- Ri.MED Foundation, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
- Ospedale Civile di Venezia SS. Giovanni e Paolo, Venezia, Italy
| | | | | | | | - Agnese La Mensa
- Ri.MED Foundation, Palermo, Italy
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | | | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | | | | | - Elisabetta Pace
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| | - Chiara Cipollina
- Ri.MED Foundation, Palermo, Italy
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| |
Collapse
|
11
|
Tengesdal IW, Banks M, Dinarello CA, Marchetti C. Screening NLRP3 drug candidates in clinical development: lessons from existing and emerging technologies. Front Immunol 2024; 15:1422249. [PMID: 39188718 PMCID: PMC11345644 DOI: 10.3389/fimmu.2024.1422249] [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: 04/23/2024] [Accepted: 07/11/2024] [Indexed: 08/28/2024] Open
Abstract
Decades of evidence positioned IL-1β as a master regulatory cytokine in acute and chronic inflammatory diseases. Approved biologics aimed at inhibiting IL-1 signaling have shown efficacy but variable safety. More recently, targeting NLRP3 activation, an upstream mediator of IL-1β, has garnered the most attention. Aberrant NLRP3 activation has been demonstrated to participate in the progression of several pathological conditions from neurogenerative diseases to cardio-metabolic syndromes and cancer. Pharmacological and genetic strategies aimed to limit NLRP3 function have proven effective in many preclinical models of diseases. These evidences have lead to a significant effort in the generation and clinical testing of small orally active molecules that can target NLRP3. In this report, we discuss different properties of these molecules with translational potential and describe the technologies currently available to screen NLRP3 targeting molecules highlighting advantages and limitations of each method.
Collapse
Affiliation(s)
- Isak W. Tengesdal
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Migachelle Banks
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Charles A. Dinarello
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
| | - Carlo Marchetti
- Department of Medicine, University of Colorado Denver, Aurora, CO, United States
- Department of Research, Rocky Mountain Regional Veteran Affairs (VA) Medical Center, Aurora, CO, United States
| |
Collapse
|
12
|
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
|
13
|
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
|
14
|
Caproni A, Nordi C, Fontana R, Facchini M, Melija S, Pappadà M, Buratto M, Marconi P. Herpes Simplex Virus ICP27 Protein Inhibits AIM 2-Dependent Inflammasome Influencing Pro-Inflammatory Cytokines Release in Human Pigment Epithelial Cells (hTert-RPE 1). Int J Mol Sci 2024; 25:4608. [PMID: 38731826 PMCID: PMC11083950 DOI: 10.3390/ijms25094608] [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: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/19/2024] [Indexed: 05/13/2024] Open
Abstract
Although Herpes simplex virus type 1 (HSV-1) has been deeply studied, significant gaps remain in the fundamental understanding of HSV-host interactions: our work focused on studying the Infected Cell Protein 27 (ICP27) as an inhibitor of the Absent-in-melanoma-2 (AIM 2) inflammasome pathway, leading to reduced pro-inflammatory cytokines that influence the activation of a protective innate immune response to infection. To assess the inhibition of the inflammasome by the ICP27, hTert-immortalized Retinal Pigment Epithelial cells (hTert-RPE 1) infected with HSV-1 wild type were compared to HSV-1 lacking functional ICP27 (HSV-1∆ICP27) infected cells. The activation of the inflammasome by HSV-1∆ICP27 was demonstrated by quantifying the gene and protein expression of the inflammasome constituents using real-time PCR and Western blot. The detection of the cleavage of the pro-caspase-1 into the active form was performed by using a bioluminescent assay, while the quantification of interleukins 1β (IL-1β) and 18 (IL-18)released in the supernatant was quantified using an ELISA assay. The data showed that the presence of the ICP27 expressed by HSV-1 induces, in contrast to HSV-1∆ICP27 vector, a significant downregulation of AIM 2 inflammasome constituent proteins and, consequently, the release of pro-inflammatory interleukins into the extracellular environment reducing an effective response in counteracting infection.
Collapse
Affiliation(s)
- Anna Caproni
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Chiara Nordi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Riccardo Fontana
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Martina Facchini
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Sara Melija
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Mariangela Pappadà
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Mattia Buratto
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
| | - Peggy Marconi
- Department of Chemical, Pharmaceutical and Agricultural Sciences, University of Ferrara, 44121 Ferrara, Italy; (A.C.); (C.N.); (R.F.); (M.F.); (S.M.); (M.P.); (M.B.)
- LTTA Laboratory for Advanced Therapies, Technopole of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
15
|
Behrendt I, Röder I, Will F, Michel G, Friedrich E, Grote D, Martin Z, Dötzer HP, Fasshauer M, Speckmann M, Kuntz S. Grape/Blueberry Anthocyanins and Their Gut-Derived Metabolites Attenuate LPS/Nigericin-Induced Inflammasome Activation by Inhibiting ASC Speck Formation in THP-1 Monocytes. Metabolites 2024; 14:203. [PMID: 38668331 PMCID: PMC11051782 DOI: 10.3390/metabo14040203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2024] [Revised: 03/28/2024] [Accepted: 03/28/2024] [Indexed: 04/28/2024] Open
Abstract
Inflammasomes are multi-protein complexes, which are formed in response to tissue injury, infections, and metabolic stress. However, aberrant inflammasome activation has been linked to several inflammatory diseases. Anthocyanins have been reported to attenuate NLR family pyrin domain-containing 3 (NLRP3) inflammasome activation, but the influence of grape/blueberry anthocyanins and especially their gut-derived metabolites on NLRP3 inflammasome activation in human monocytes remains unclear. Therefore, human leukemic monocytes (THP-1 cells, Tohoku Hospital Pediatrics-1 cells) were preincubated with different concentrations of grape/blueberry anthocyanins, homovanillyl alcohol, or 2,4,6-trihydroxybenzaldehyde (THBA) before the NLRP3 inflammasome was activated by lipopolysaccharide and/or nigericin. Apoptosis-associated speck-like protein containing a CARD (ASC) speck formation, as well as ASC and NLRP3 protein expression, were determined using flow cytometry. Caspase-1 activity was measured in cultured cells, and pro-inflammatory cytokine secretion was determined using enzyme-linked immunosorbent assays. Anthocyanins and their metabolites had no effect on ASC or NLRP3 protein expression. However, THBA significantly inhibited ASC speck formation in primed and unprimed THP-1 monocytes, while caspase-1 activity was significantly declined by grape/blueberry anthocyanins. Furthermore, reduced inflammasome activation resulted in lower pro-inflammatory cytokine secretion. In conclusion, our results show for the first time that grape/blueberry anthocyanins and their gut-derived metabolites exert anti-inflammatory effects by attenuating NLRP3 inflammasome activation in THP-1 monocytes.
Collapse
Affiliation(s)
- Inken Behrendt
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Isabella Röder
- Department of Beverage Research, Hochschule Geisenheim University, 65366 Geisenheim, Germany; (I.R.); (F.W.)
| | - Frank Will
- Department of Beverage Research, Hochschule Geisenheim University, 65366 Geisenheim, Germany; (I.R.); (F.W.)
| | - Gabriela Michel
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (G.M.); (M.S.)
- Flow Cytometry Core Facility, Department of Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Elvira Friedrich
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Daniela Grote
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Zoe Martin
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Hanna Pauline Dötzer
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Mathias Fasshauer
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| | - Martin Speckmann
- Institute for Clinical Immunology, Transfusion Medicine and Hemostaseology, Justus-Liebig-University Giessen, 35392 Giessen, Germany; (G.M.); (M.S.)
- Flow Cytometry Core Facility, Department of Medicine, Justus-Liebig-University Giessen, 35392 Giessen, Germany
| | - Sabine Kuntz
- Institute of Nutritional Science, Justus-Liebig-University Giessen, 35390 Giessen, Germany; (E.F.); (D.G.); (Z.M.); (H.P.D.); (M.F.); (S.K.)
| |
Collapse
|
16
|
Teske KA, Corona C, Wilkinson J, Mamott D, Good DA, Zambrano D, Lazar DF, Cali JJ, Robers MB, O'Brien MA. Interrogating direct NLRP3 engagement and functional inflammasome inhibition using cellular assays. Cell Chem Biol 2024; 31:349-360.e6. [PMID: 37858335 DOI: 10.1016/j.chembiol.2023.09.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2023] [Revised: 07/20/2023] [Accepted: 09/25/2023] [Indexed: 10/21/2023]
Abstract
As a key regulator of the innate immune system, the NLRP3 inflammasome responds to a variety of environmental insults through activation of caspase-1 and release of the proinflammatory cytokines IL-1β and IL-18. Aberrant NLRP3 inflammasome function is implicated in numerous inflammatory diseases, spurring drug discovery efforts at NLRP3 as a therapeutic target. A diverse array of small molecules is undergoing preclinical/clinical evaluation with a reported mode of action involving direct modulation of the NLRP3 pathway. However, for a subset of these ligands the functional link between live-cell target engagement and pathway inhibition has yet to be fully established. Herein we present a cohort of mechanistic assays to both query direct NLRP3 engagement in cells, and functionally interrogate different nodes of NLRP3 pathway activity. This system enabled the stratification of potency for five confirmed NLRP3 inhibitors, and identification of two reported NLRP3 inhibitors that failed to demonstrate direct pathway antagonism.
Collapse
Affiliation(s)
- Kelly A Teske
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Cesear Corona
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | | | - Daniel Mamott
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - David A Good
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Delia Zambrano
- Promega Corporation, Research & Development, San Luis Obispo, CA 93401, USA
| | - Dan F Lazar
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - James J Cali
- Promega Corporation, Research & Development, Madison, WI 53711, USA
| | - Matthew B Robers
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| | - Martha A O'Brien
- Promega Corporation, Research & Development, Madison, WI 53711, USA.
| |
Collapse
|
17
|
Vande Walle L, Lamkanfi M. Drugging the NLRP3 inflammasome: from signalling mechanisms to therapeutic targets. Nat Rev Drug Discov 2024; 23:43-66. [PMID: 38030687 DOI: 10.1038/s41573-023-00822-2] [Citation(s) in RCA: 100] [Impact Index Per Article: 100.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 10/06/2023] [Indexed: 12/01/2023]
Abstract
Diseases associated with chronic inflammation constitute a major health burden across the world. As central instigators of the inflammatory response to infection and tissue damage, inflammasomes - and the NACHT, LRR and PYD domain-containing protein 3 (NLRP3) inflammasome in particular - have emerged as key regulators in diverse rheumatic, metabolic and neurodegenerative diseases. Similarly to other inflammasome sensors, NLRP3 assembles a cytosolic innate immune complex that activates the cysteine protease caspase-1, which in turn cleaves gasdermin D (GSDMD) to induce pyroptosis, a regulated mode of lytic cell death. Pyroptosis is highly inflammatory, partly because of the concomitant extracellular release of the inflammasome-dependent cytokines IL-1β and IL-18 along with a myriad of additional danger signals and intracellular antigens. Here, we discuss how NLRP3 and downstream inflammasome effectors such as GSDMD, apoptosis-associated speck-like protein containing a CARD (ASC) and nerve injury-induced protein 1 (NINJ1) have gained significant traction as therapeutic targets. We highlight the recent progress in developing small-molecule and biologic inhibitors that are advancing into the clinic and serving to harness the broad therapeutic potential of modulating the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Lieselotte Vande Walle
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium
| | - Mohamed Lamkanfi
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, Belgium.
| |
Collapse
|
18
|
Nandi D, Forster J, Ramesh A, Nguyen A, Bharadwaj H, Kulkarni A. Caspase-1 Responsive Nanoreporter for In Vivo Monitoring of Inflammasome Immunotherapy. ACS APPLIED MATERIALS & INTERFACES 2023; 15:55545-55558. [PMID: 37990965 PMCID: PMC11056827 DOI: 10.1021/acsami.3c15733] [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] [Indexed: 11/23/2023]
Abstract
Inflammasomes are multimeric protein signaling complexes that are assembled in innate immune cells in response to a multitude of pathogen and damage-associated signals. They are essential for generating robust inflammatory responses to prevent pathogenic insults. However, inflammasome dysregulation can induce cascading immune responses, resulting in systemic toxicities and inflammatory disease. In this sense, there is a strong need to develop potent inflammasome inhibiting therapies as well as technologies to monitor their efficacy, yet current systems lack the ability to effectively image inflammasome activation and track therapy response early. To overcome these limitations, we report a novel nanoparticle system delivering both a caspase-1 cleavable inflammasome detecting probe and the NLRP3 inhibitor drug MCC-950, providing dual capabilities of monitoring and regulation of inflammasome activation in a biocompatible, tissue penetrating, and sustained release liposomal formulation. We observed this liposomal nanoreporter's ability to reduce and detect inflammasome activation both in vitro in immortalized bone marrow-derived macrophages and in vivo in a DSS-induced ulcerative colitis mouse model. Our results exhibited the nanoreporter's ability to penetrate inflammatory tissues and detect inflammasome activation early and in real-time for multiple days while alleviating inflammation in the groups coencapsulating imaging reporter and inflammasome inhibitor. Overall, the developed liposomal nanoreporter platform enables spatiotemporal delivery of imaging probe and inhibitor, captures early and sustained inflammasome detection, and induces inflammasome amelioration, thus establishing a novel tool for the real-time monitoring and treatment of inflammasome-mediated disease with high potential for clinical application.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering and Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - James Forster
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Anujan Ramesh
- Department of Chemical Engineering and Department of Biomedical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Anh Nguyen
- Department of Chemical Engineering, University of Massachusetts, Amherst, Massachusetts 01003, United States
| | - Hariharan Bharadwaj
- Department of Pathology, UMass Chan, Medical School-Baystate, Springfield, Massachusetts 01107, United States
| | - Ashish Kulkarni
- Department of Chemical Engineering, Department of Veterinary and Animal Sciences, Department of Biomedical Engineering, and Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, Massachusetts 01003, United States
| |
Collapse
|
19
|
Cristaldi M, Buscetta M, Cimino M, La Mensa A, Giuffrè MR, Fiore L, Carcione C, Bucchieri F, Rappa F, Coronnello C, Sciaraffa N, Amato S, Aronica TS, Lo Iacono G, Bertani A, Pace E, Cipollina C. Caspase-8 activation by cigarette smoke induces pro-inflammatory cell death of human macrophages exposed to lipopolysaccharide. Cell Death Dis 2023; 14:773. [PMID: 38007509 PMCID: PMC10676397 DOI: 10.1038/s41419-023-06318-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2023] [Revised: 11/03/2023] [Accepted: 11/15/2023] [Indexed: 11/27/2023]
Abstract
Cigarette smoking impairs the lung innate immune response making smokers more susceptible to infections and severe symptoms. Dysregulation of cell death is emerging as a key player in chronic inflammatory conditions. We have recently reported that short exposure of human monocyte-derived macrophages (hMDMs) to cigarette smoke extract (CSE) altered the TLR4-dependent response to lipopolysaccharide (LPS). CSE caused inhibition of the MyD88-dependent inflammatory response and activation of TRIF/caspase-8/caspase-1 pathway leading to Gasdermin D (GSDMD) cleavage and increased cell permeability. Herein, we tested the hypothesis that activation of caspase-8 by CSE increased pro-inflammatory cell death of LPS-stimulated macrophages. To this purpose, we measured apoptotic and pyroptotic markers as well as the expression/release of pro-inflammatory mediators in hMDMs exposed to LPS and CSE, alone or in combination, for 6 and 24 h. We show that LPS/CSE-treated hMDMs, but not cells treated with CSE or LPS alone, underwent lytic cell death (LDH release) and displayed apoptotic features (activation of caspase-8 and -3/7, nuclear condensation, and mitochondrial membrane depolarization). Moreover, the negative regulator of caspase-8, coded by CFLAR gene, was downregulated by CSE. Activation of caspase-3 led to Gasdermin E (GSDME) cleavage. Notably, lytic cell death caused the release of the damage-associated molecular patterns (DAMPs) heat shock protein-60 (HSP60) and S100A8/A9. This was accompanied by an impaired inflammatory response resulting in inhibited and delayed release of IL6 and TNF. Of note, increased cleaved caspase-3, higher levels of GSDME and altered expression of cell death-associated genes were found in alveolar macrophages of smoker subjects compared to non-smoking controls. Overall, our findings show that CSE sensitizes human macrophages to cell death by promoting pyroptotic and apoptotic pathways upon encountering LPS. We propose that while the delayed inflammatory response may result in ineffective defenses against infections, the observed cell death associated with DAMP release may contribute to establish chronic inflammation. CS exposure sensitizes human macrophages to pro-inflammatory cell death. Upon exposure to LPS, CS inhibits the TLR4/MyD88 inflammatory response, downregulating the pro-inflammatory genes TNF and IL6 and the anti-apoptotic gene CFLAR, known to counteract caspase-8 activity. CS enhances caspase-8 activation through TLR4/TRIF, with a partial involvement of RIPK1, resulting on the activation of caspase-1/GSDMD axis leading to increased cell permeability and DAMP release through gasdermin pores [19]. At later timepoints caspase-3 becomes strongly activated by caspase-8 triggering apoptotic events which are associated with mitochondrial membrane depolarization, gasdermin E cleavage and secondary necrosis with consequent massive DAMP release.
Collapse
Affiliation(s)
| | - Marco Buscetta
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
| | - Maura Cimino
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
| | - Agnese La Mensa
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | | | - Luigi Fiore
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy
- Dipartimento di Scienze Biomediche, Odontoiatriche e delle Immagini Morfologiche e Funzionali, Università di Messina, Piazza Pugliatti, 1, 98122, Messina, Italy
| | | | - Fabio Bucchieri
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Via del Vespro 129, 90127, Palermo, Italy
| | - Francesca Rappa
- Dipartimento di Biomedicina, Neuroscienze e Diagnostica Avanzata, Università di Palermo, Via del Vespro 129, 90127, Palermo, Italy
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Via Ugo la Malfa 153, 90146, Palermo, Italy
| | | | | | - Santina Amato
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Piazza Nicola Leotta 4, 90127, Palermo, Italy
| | - Tommaso Silvano Aronica
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Piazza Nicola Leotta 4, 90127, Palermo, Italy
| | | | | | - Elisabetta Pace
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Via Ugo la Malfa 153, 90146, Palermo, Italy
| | - Chiara Cipollina
- Fondazione Ri.MED, Via Bandiera 11, 90133, Palermo, Italy.
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Via Ugo la Malfa 153, 90146, Palermo, Italy.
| |
Collapse
|
20
|
Goncalves BG, Heise RM, Banerjee IA. Development of Self-Assembled Biomimetic Nanoscale Collagen-like Peptide-Based Scaffolds for Tissue Engineering: An In Silico and Laboratory Study. Biomimetics (Basel) 2023; 8:548. [PMID: 37999189 PMCID: PMC10669358 DOI: 10.3390/biomimetics8070548] [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: 09/22/2023] [Revised: 11/01/2023] [Accepted: 11/02/2023] [Indexed: 11/25/2023] Open
Abstract
Development of biocomposite scaffolds has gained tremendous attention due to their potential for tissue regeneration. However, most scaffolds often contain animal-derived collagen that may elicit an immunological response, necessitating the development of new biomaterials. Herein, we developed a new collagen-like peptide,(Pro-Ala-His)10 (PAH)10, and explored its ability to be utilized as a functional biomaterial by incorporating it with a newly synthesized peptide-based self-assembled gel. The gel was prepared by conjugating a pectin derivative, galataric acid, with a pro-angiogenic peptide (LHYQDLLQLQY) and further functionalized with a cortistatin-derived peptide, (Phe-Trp-Lys-Thr)4 (FWKT)4, and the bio-ionic liquid choline acetate. The self-assembly of (PAH)10 and its interactions with the galactarate-peptide conjugates were examined using replica exchange molecular dynamics (REMD) simulations. Results revealed the formation of a multi-layered scaffold, with enhanced stability at higher temperatures. We then synthesized the scaffold and examined its physicochemical properties and its ability to integrate with aortic smooth muscle cells. The scaffold was further utilized as a bioink for bioprinting to form three-dimensional cell-scaffold matrices. Furthermore, the formation of actin filaments and elongated cell morphology was observed. These results indicate that the (PAH)10 hybrid scaffold provides a suitable environment for cell adhesion, proliferation and growth, making it a potentially valuable biomaterial for tissue engineering.
Collapse
Affiliation(s)
| | | | - Ipsita A. Banerjee
- Department of Chemistry, Fordham University, 441 East Fordham Road, Bronx, New York, NY 10458, USA; (B.G.G.); (R.M.H.)
| |
Collapse
|
21
|
Choi YH. Reduction of high glucose-induced oxidative injury in human retinal pigment epithelial cells by sarsasapogenin through inhibition of ROS generation and inactivation of NF-κB/NLRP3 inflammasome pathway. Genes Genomics 2023; 45:1153-1163. [PMID: 37354257 DOI: 10.1007/s13258-023-01417-2] [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: 05/30/2023] [Accepted: 06/19/2023] [Indexed: 06/26/2023]
Abstract
BACKGROUND Hyperglycemia-induced accumulation of reactive oxygen species (ROS) is a major risk factor for diabetic retinopathy (DR). Sarsasapogenin is a natural steroidal saponin that is known to have excellent antidiabetic effects and improve diabetic complications, but its potential efficacy and mechanism for DR are unknown. OBJECTIVES The current study was designed to explore whether sarsasapogenin inhibits hyperglycemia-induced oxidative stress in human retinal pigment epithelial (RPE) ARPE-19 cells and to elucidate the molecular mechanisms. METHODS To mimic hyperglycemic conditions, ARPE-19 cells were cultured in medium containing high glucose (HG). The suppressive effects of sarsasapogenin on HG-induced cell viability reduction, apoptosis and ROS production were investigated. In addition, the relevance of the nuclear factor-kappa B (NF-κB)/NOD-like receptor family pyrin domain-containing 3 (NLRP3) inflammasome signaling pathway was explored to investigate the mechanism of antioxidant and anti-inflammatory activity of sarsasapogenin. RESULTS Sarsasapogenin significantly alleviated cytotoxicity and apoptosis in HG-treated ARPE-19 cells through inhibition of intracellular ROS generation. Sarsasapogenin also effectively attenuated HG-induced excess accumulation of mitochondrial superoxide, reduction of glutathione content, and inactivation of manganese superoxide dismutase and glutathione peroxidase. The HG condition markedly increased the expression and maturation of interleukin (IL)-1β and IL-18 through the activation of the NF-kB signaling pathway, whereas sarsasapogenin reversed these effects. Moreover, although the expression of NLRP3 inflammasome multiprotein complex molecules was increased in ARPE-19 cells cultured under HG conditions, their levels remained similar to the control group in the presence of sarsasapogenin. CONCLUSION Sarsasapogenin could protect RPE cells from HG-induced injury by inhibiting ROS generation and NF-κB/NLRP3 inflammasome pathway, suggesting its potential as a therapeutic agent to improve the symptoms of DR.
Collapse
Affiliation(s)
- Yung Hyun Choi
- Anti-Aging Research Center, Dong-eui University, Busan, 47340, Republic of Korea.
- Department of Biochemistry, Dong-eui University College of Korean Medicine, 52-57 Yangjeong-ro, Busan, 47227, Republic of Korea.
| |
Collapse
|
22
|
Khalil BA, Sharif-Askari NS, Halwani R. Role of inflammasome in severe, steroid-resistant asthma. CURRENT RESEARCH IN IMMUNOLOGY 2023; 4:100061. [PMID: 37304814 PMCID: PMC10250931 DOI: 10.1016/j.crimmu.2023.100061] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/09/2023] [Accepted: 05/14/2023] [Indexed: 06/13/2023] Open
Abstract
Purpose of review Asthma is a common heterogeneous group of chronic inflammatory diseases with different pathological phenotypes classified based on the various clinical, physiological and immunobiological profiles of patients. Despite similar clinical symptoms, asthmatic patients may respond differently to treatment. Hence, asthma research is becoming more focused on deciphering the molecular and cellular pathways driving the different asthma endotypes. This review focuses on the role of inflammasome activation as one important mechanism reported in the pathogenesis of severe steroid resistant asthma (SSRA), a Th2-low asthma endotype. Although SSRA represents around 5-10% of asthmatic patients, it is responsible for the majority of asthma morbidity and more than 50% of asthma associated healthcare costs with clear unmet need. Therefore, deciphering the role of the inflammasome in SSRA pathogenesis, particularly in relation to neutrophil chemotaxis to the lungs, provides a novel target for therapy. Recent findings The literature highlighted several activators of inflammasomes that are elevated during SSRA and result in the release of proinflammatory mediators, mainly IL-1β and IL-18, through different signaling pathways. Consequently, the expression of NLRP3 and IL-1β is shown to be positively correlated with neutrophil recruitment and negatively correlated with airflow obstruction. Furthermore, exaggerated NLRP3 inflammasome/IL-1β activation is reported to be associated with glucocorticoid resistance. Summary In this review, we summarized the reported literature on the activators of the inflammasome during SSRA, the role of IL-1β and IL-18 in SSRA pathogenesis, and the pathways by which inflammasome activation contributes to steroid resistance. Finally, our review shed light on the different levels to target inflammasome involvement in an attempt to ameliorate the serious outcomes of SSRA.
Collapse
Affiliation(s)
- Bariaa A. Khalil
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
| | | | - Rabih Halwani
- Sharjah Institute of Medical Research, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Department of Clinical Sciences, College of Medicine, University of Sharjah, Sharjah, United Arab Emirates
- Prince Abdullah Ben Khaled Celiac Disease Research Chair, Department of Pediatrics, Faculty of Medicine, King Saud University, Saudi Arabia
| |
Collapse
|
23
|
Yadav AK, Chan J. Activity-based bioluminescence probes for in vivo sensing applications. Curr Opin Chem Biol 2023; 74:102310. [PMID: 37119771 PMCID: PMC10225331 DOI: 10.1016/j.cbpa.2023.102310] [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: 02/22/2023] [Revised: 03/17/2023] [Accepted: 03/28/2023] [Indexed: 05/01/2023]
Abstract
Bioluminescence imaging is a highly sensitive technique commonly used for various in vivo applications. Recent efforts to expand the utility of this modality have led to the development of a suite of activity-based sensing (ABS) probes for bioluminescence imaging by 'caging' of luciferin and its structural analogs. The ability to selectively detect a given biomarker has presented researchers with many exciting opportunities to study both health and disease states in animal models. Here, we highlight recent (2021-2023) bioluminescence-based ABS probes with an emphasis on probe design and in vivo validation experiments.
Collapse
Affiliation(s)
- Anuj K Yadav
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States
| | - Jefferson Chan
- Department of Chemistry, Beckman Institute for Advanced Science and Technology, and Cancer Center at Illinois, University of Illinois at Urbana-Champaign, Urbana, Illinois 61801, United States.
| |
Collapse
|
24
|
Mu W, Xu G, Wang Z, Li Q, Sun S, Qin Q, Li Z, Shi W, Dai W, Zhan X, Wang J, Bai Z, Xiao X. Tricyclic antidepressants induce liver inflammation by targeting NLRP3 inflammasome activation. Cell Commun Signal 2023; 21:123. [PMID: 37231437 DOI: 10.1186/s12964-023-01128-x] [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: 09/04/2022] [Accepted: 04/15/2023] [Indexed: 05/27/2023] Open
Abstract
BACKGROUND Idiosyncratic drug-induced liver injury (IDILI) is common in hepatology practices and, in some cases, lethal. Increasing evidence show that tricyclic antidepressants (TCAs) can induce IDILI in clinical applications but the underlying mechanisms are still poorly understood. METHODS We assessed the specificity of several TCAs for NLRP3 inflammasome via MCC950 (a selective NLRP3 inhibitor) pretreatment and Nlrp3 knockout (Nlrp3-/-) BMDMs. Meanwhile, the role of NLRP3 inflammasome in the TCA nortriptyline-induced hepatotoxicity was demonstrated in Nlrp3-/- mice. RESULTS We reported here that nortriptyline, a common TCA, induced idiosyncratic hepatotoxicity in a NLRP3 inflammasome-dependent manner in mildly inflammatory states. In parallel in vitro studies, nortriptyline triggered the inflammasome activation, which was completely blocked by Nlrp3 deficiency or MCC950 pretreatment. Furthermore, nortriptyline treatment led to mitochondrial damage and subsequent mitochondrial reactive oxygen species (mtROS) production resulting in aberrant activation of the NLRP3 inflammasome; a selective mitochondrial ROS inhibitor pretreatment dramatically abrogated nortriptyline-triggered the NLRP3 inflammasome activation. Notably, exposure to other TCAs also induced aberrant activation of the NLRP3 inflammasome by triggering upstream signaling events. CONCLUSION Collectively, our findings revealed that the NLRP3 inflammasome may act as a crucial target for TCA agents and suggested that the core structures of TCAs may contribute to the aberrant activation of NLRP3 inflammasome induced by them, an important factor involved in the pathogenesis of TCA-induced liver injury. Video Abstract.
Collapse
Affiliation(s)
- Wenqing Mu
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- State Key Laboratory of Radiation Medicine and Protection, Institutes for Translational Medicine, Soochow University, Suzhou, 215123, Jiangsu, China
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China
| | - Guang Xu
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- School of Traditional Chinese Medicine, Capital Medical University, Beijing, 100069, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Zhilei Wang
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- TCM Regulating Metabolic Diseases Key Laboratory of Sichuan Province, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Qiang Li
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Siqiao Sun
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Qin Qin
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhiyong Li
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wei Shi
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Wenzhang Dai
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Xiaoyan Zhan
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Jiabo Wang
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China
| | - Zhaofang Bai
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| | - Xiaohe Xiao
- Department of Hepatology, the Fifth Medical Center of PLA General Hospital, Beijing, 100039, China.
- Military Institute of Chinese Materia, Fifth Medical Center of Chinese PLA General Hospital, Beijing, 100039, China.
| |
Collapse
|
25
|
TXNIP shuttling - a key molecular link in regulating inflammation and mitochondrial dysfunction in freeze tolerant wood frogs. Gene 2023; 857:147184. [PMID: 36627089 DOI: 10.1016/j.gene.2023.147184] [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: 08/19/2022] [Revised: 11/27/2022] [Accepted: 01/04/2023] [Indexed: 01/09/2023]
Abstract
Amphibians such as the wood frogs,Rana sylvatica, are a primary example of a freeze-tolerant vertebrate that undergoes whole body freezing. Multiple adaptations including sequestering 65-70% of total body water as extracellular/extra organ ice and producing massive amounts of glucose as a cryoprotectant support this. Interestingly, the high glucose levels induced in response to freezing can amplify oxidative stress's effects (reactive oxygen species, ROS) and induce inflammation and mitochondrial dysfunction. Since both freezing and dehydration stress (independent of freezing) can render wood frogs hyperglycemic, this study focussed on these two stresses to elucidate the role of a scaffold protein thioredoxin interacting protein (TXNIP), which localizes in multiple compartments inside the cell under hyperglycemic conditions and mediate diverse stress responses. The results from this study suggest a stress-specific response of TXNIP in inducing the cell-damaging pathway of inflammasome activation via its cytoplasmic localization during freezing. Interestingly, mitochondrial localization of TXNIP did not leads to increase in its binding to thioredoxin 2 (TRX-2) and activating the dysfunction of this organelle by releasing a mitochondrial protein cytochrome c (Cyt c) in cytoplasm under both freezing and dehydration stresses. Post-translational modifications of TXNIP hinted on changes in the regulating proteins involved in the inflammasome and mitochondrial dysfunction pathways, whereas sequential differences (cytosine residues) of amphibian TXNIP (compared to mammalian) assessed via 3D-modeling attributed to its weak binding to TRX-2. Overall, this study summarizes differential role of proteins activated under freeze and dehydration induced hyperglycemic response in freeze tolerant wood frogs.
Collapse
|
26
|
Pereira RB, Rahali FZ, Nehme R, Falleh H, Jemaa MB, Sellami IH, Ksouri R, Bouhallab S, Ceciliani F, Abdennebi-Najar L, Pereira DM. Anti-inflammatory activity of essential oils from Tunisian aromatic and medicinal plants and their major constituents in THP-1 macrophages. Food Res Int 2023; 167:112678. [PMID: 37087210 DOI: 10.1016/j.foodres.2023.112678] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2022] [Revised: 01/26/2023] [Accepted: 03/09/2023] [Indexed: 03/17/2023]
Abstract
In this study, the capacity of eight essential oils (EOs), sage (Salvia officinalis), coriander (Coriandrum sativum), rosemary (Rosmarinus officinalis), black cumin (Nigella sativa), prickly juniper (Juniperus oxycedrus), geranium (Pelargonium graveolens), oregano (Origanum vulgare) and wormwood (Artemisia herba-alba), on the inhibition of NF-κB activation was screened at concentrations up to 0.25 µL/mL using THP-1 human macrophages bearing a NF-κB reporter. This screening selected coriander, geranium, and wormwood EOs as the most active, which later evidenced the ability to decrease over 50 % IL-6, IL-1β, TNF-α and COX-2 mRNA expression in LPS-stimulated THP-1 macrophages. The chemical composition of selected EOs was performed by gas chromatography-mass spectrometry (GC-MS). The two major constituents (>50 % of each EO) were tested at the same concentrations presented in each EO. It was demonstrated that the major compound or the binary mixtures of the two major compounds could explain the anti-inflammatory effects reported for the crude EOs. Additionally, the selected EOs also inhibit>50 % caspase-1 activity. However, this effect could not be attributed to the major components (except for β-citronellol/geranium oil, 40 %/65 % caspase-1 inhibition), suggesting, in addition to potential synergistic effects, the presence of minor compounds with caspase-1 inhibitory activity. These results demonstrated the potential use of the EOs obtained from Tunisian flora as valuable sources of anti-inflammatory agents providing beneficial health effects by reducing the levels of inflammatory mediators involved in the genesis of several diseases.
Collapse
|
27
|
Xu R, Zhao H, Qi J, Yao G, He Y, Lu Y, Zhu Q, Wang Y, Ding Y, Zhu Z, Li X, Vankelecom H, Sun Y. Local glucose elevation activates pyroptosis via NLRP3 inflammasome in ovarian granulosa cells of overweight patients. FASEB J 2023; 37:e22807. [PMID: 36826432 DOI: 10.1096/fj.202201796rr] [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: 11/04/2022] [Revised: 01/23/2023] [Accepted: 01/25/2023] [Indexed: 02/25/2023]
Abstract
Overweight, with an increasing prevalence worldwide, significantly impairs the clinical outcomes following in vitro fertilization (IVF). Hyperglycemia, hyperlipidemia, and metabolic disorders are always accompanied by the majority of overweight patients. The association between granulosa cell function and metabolic alterations in follicular fluid including lipids, proteins, and growth factors has been extensively documented. However, the effects of higher glucose level on ovarian granulosa cells (GCs), remain largely unknown. In this study, we identified that overweight women had elevated follicular glucose level which profoundly activated NLRP3 inflammasome and pyroptosis. An in vitro correlation between follicular high glucose, NLRP3 inflammasome and pyroptosis was also established. More importantly, in granulosa cells of overweight patients, the activation of the NLRP3 inflammasome and pyroptosis induced by high glucose was involved in the dysregulation of estradiol synthesis. Our study may provide new options to interpretate and improve IVF outcomes in overweight women.
Collapse
Affiliation(s)
- Rui Xu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hanting Zhao
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Jia Qi
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Guangxin Yao
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yaqiong He
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yao Lu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Qinling Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Yuan Wang
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Ying Ding
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Zhenyi Zhu
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Xinyu Li
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| | - Hugo Vankelecom
- Department of Development and Regeneration, Cluster Stem Cell Biology and Embryology, Research Unit of Stem Cell Research, University of Leuven (KU Leuven), Leuven, Belgium
| | - Yun Sun
- Center for Reproductive Medicine, Ren Ji Hospital, Shanghai Jiao Tong University School of Medicine, Shanghai, China.,Shanghai Key Laboratory for Assisted Reproduction and Reproductive Genetics, Shanghai, China
| |
Collapse
|
28
|
Nandi D, Forster J, Ramesh A, Nguyen A, Bharadwaj H, Kulkarni A. Nanoreporter for Real-Time Monitoring of Inflammasome Activity and Targeted Therapy. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2023; 10:e2204900. [PMID: 36603165 PMCID: PMC9951342 DOI: 10.1002/advs.202204900] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 11/18/2022] [Indexed: 06/17/2023]
Abstract
Inflammasome activation is associated with a myriad of inflammatory diseases. However, existing methods provides a limited understanding of spatiotemporal kinetics of inflammasome activation, with restricted scope for early detection of associated treatment efficacy. This limitation offers an opportunity for the development of biocompatible in-vivo inflammasome monitoring tools with translational prospects. To achieve this, they report developing a pair of lipid-based nanoparticle systems, a reporter nanoparticle consisting of a caspase-1 activatable probe alone, and a theranostic nanoparticle combining the probe with an inflammasome-inhibiting drug. This biocompatible platform enhances the probe's residence time in circulation by preventing its opsonization and allowing its sustained release over time. Their results demonstrate the specificity of reporter nanoparticles towards caspase-1 activity and provides early-on monitoring of inflammasome activation both in-vitro as well as in-vivo. Additionally, the delivery of disulfiram, an inflammasome-inhibiting drug, along with reporter probe using theranostic nanoparticles enables real-time tracking of treatment efficacy in the gouty-arthritis inflammatory model. In summary, they report an unparalleled pair of the inflammasome-associated reporter and theranostic platforms suited not only for diagnostic applications but can also detect inflammasome-targeted treatment efficiency in real-time. These findings establish two novel, sensitive nanotools for non-invasive evaluation of inflammasome-targeted immunotherapy.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
| | - James Forster
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anujan Ramesh
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Anh Nguyen
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
| | - Hariharan Bharadwaj
- Department of PathologyUMass ChanMedical School‐BaystateSpringfieldMA01107USA
| | - Ashish Kulkarni
- Department of Chemical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Department of Veterinary and Animal SciencesUniversity of MassachusettsAmherstMA01003USA
- Department of Biomedical EngineeringUniversity of MassachusettsAmherstMA01003USA
- Center for Bioactive DeliveryInstitute for Applied Life SciencesUniversity of MassachusettsAmherstMA01003USA
| |
Collapse
|
29
|
Ren X, Tao M, Liu X, Zhang L, Li M, Hai Z. Caspase-1-responsive fluorescence biosensors for monitoring endogenous inflammasome activation. Biosens Bioelectron 2023; 219:114812. [PMID: 36272346 DOI: 10.1016/j.bios.2022.114812] [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: 06/14/2022] [Revised: 09/27/2022] [Accepted: 10/11/2022] [Indexed: 11/07/2022]
Abstract
The activation of inflammasome leads to secretion of inflammatory factors and cell pyroptosis that are critical in the pathogenesis of various chronic and acute inflammatory diseases. Recruitment and activation of caspase-1 is a marker of inflammasome activation. However, there is still lack of real-time and efficient methods to detect the activation of inflammasome, especially in vivo. Herein, we developed two activatable caspase-1-responsive fluorescence biosensors, WEHD-HCy and YVAD-HCy, to specifically monitor the activation of inflammasome in vivo. Our in vitro study demonstrated that WEHD-HCy and YVAD-HCy can sensitively and specifically respond to caspase-1 activation. Moreover, these biosensors can efficiency and specifically activated in the common inflammatory disease model, including inflammatory bowel disease, Salmonella infection, and acute arthritis. In particular, WEHD-HCy is more advantageous than YVAD-HCy to specifically image of caspase-1 activity both in vitro and in vivo. These caspase-1-responsive fluorescence biosensors provide an efficient, rapid, and in situ tool for monitoring inflammasome activation, and have the potential to be suitable for clinical diagnosis of various inflammatory diseases associated with inflammasome activation.
Collapse
Affiliation(s)
- Xingxing Ren
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China
| | - Menglin Tao
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Xiaoming Liu
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China
| | - Lele Zhang
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China
| | - Mingsong Li
- Department of Gastroenterology, Third Affiliated Hospital of Guangzhou Medical University, Guangzhou, 510145, China.
| | - Zijuan Hai
- Key Laboratory of Structure and Functional Regulation of Hybrid Materials, Ministry of Education, Institutes of Physical Science and Information Technology, Anhui University, Hefei, Anhui, 230601, China.
| |
Collapse
|
30
|
Paerewijck O, Lamkanfi M. The human inflammasomes. Mol Aspects Med 2022; 88:101100. [PMID: 35696786 DOI: 10.1016/j.mam.2022.101100] [Citation(s) in RCA: 42] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2022] [Revised: 05/25/2022] [Accepted: 06/01/2022] [Indexed: 12/14/2022]
Abstract
Two decades of inflammasome research has led to a vast body of knowledge on the complex regulatory mechanisms and pathological roles of canonical and non-canonical inflammasome activation in a plethora of research models of primarily rodent origin. More recently, the field has made notable progress in characterizing human-specific inflammasomes and their regulation mechanisms, including an expansion of inflammasome biology to adaptive immune cells. These exciting developments in basic research have been accompanied by potentially transformative results from large clinical trials and translational efforts to develop inflammasome-targeted small molecule inhibitors for therapeutic use. Here, we will discuss recent findings in the field with a specific emphasis on activation mechanisms of human inflammasomes and their potential role in auto-inflammatory, metabolic and neoplastic diseases.
Collapse
Affiliation(s)
- Oonagh Paerewijck
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, B-9000, Belgium
| | - Mohamed Lamkanfi
- Laboratory of Medical Immunology, Department of Internal Medicine and Paediatrics, Ghent University, Ghent, B-9000, Belgium.
| |
Collapse
|
31
|
Buscetta M, Cristaldi M, Cimino M, La Mensa A, Dino P, Bucchieri F, Rappa F, Amato S, Aronica TS, Pace E, Bertani A, Cipollina C. Cigarette smoke promotes inflammasome-independent activation of caspase-1 and -4 leading to gasdermin D cleavage in human macrophages. FASEB J 2022; 36:e22525. [PMID: 36004615 DOI: 10.1096/fj.202200837r] [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: 06/06/2022] [Revised: 08/02/2022] [Accepted: 08/17/2022] [Indexed: 11/11/2022]
Abstract
Mechanisms and consequences of gasdermin D (GSDMD) activation in cigarette smoke (CS)-associated inflammation and lung disease are unknown. GSDMD is a downstream effector of caspase-1, -8, and -4. Upon cleavage, GSDMD generates pores into cell membranes. Different degrees of GSDMD activation are associated with a range of physiological outputs ranging from cell hyperactivation to pyroptosis. We have previously reported that in human monocyte-derived macrophages CS extract (CSE) inhibits the NLRP3 inflammasome and shifts the response to lipopolysaccharide (LPS) towards the TLR4-TRIF axis leading to activation of caspase-8, which, in turn, activates caspase-1. In the present work, we investigated whether other ASC-dependent inflammasomes could be involved in caspase activation by CSE and whether caspase activation led to GSDMD cleavage and other downstream effects. Presented results demonstrate that CSE promoted ASC-independent activation of caspase-1 leading to GSDMD cleavage and increased cell permeability, in the absence of cell death. GSDMD cleavage was strongly enhanced upon stimulation with LPS+CSE, suggesting a synergistic effect between the two stimuli. Noteworthy, CSE promoted LPS internalization leading to caspase-4 activation, thus contributing to increased GSDMD cleavage. Caspase-dependent GSDMD cleavage was associated with mitochondrial superoxide generation. Increased cleaved GSDMD was found in lung macrophages of smokers compared to ex-smokers and non-smoking controls. Our findings revealed that ASC-independent activation of caspase-1, -4, and -8 and GSDMD cleavage upon exposure to CS may contribute to macrophage dysfunction and feed the chronic inflammation observed in the smokers' lung.
Collapse
Affiliation(s)
| | | | | | - Agnese La Mensa
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Paola Dino
- Fondazione RiMED, Palermo, Italy.,Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Fabio Bucchieri
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Francesca Rappa
- Department of Biomedicine, Neurosciences and Advanced Diagnostics (BiND), University of Palermo, Palermo, Italy
| | - Santina Amato
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Tommaso Silvano Aronica
- Azienda di Rilievo Nazionale ed Alta Specializzazione Ospedali (A.R.N.A.S) "Civico Di Cristina Benfratelli", Palermo, Italy
| | - Elisabetta Pace
- Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| | | | - Chiara Cipollina
- Fondazione RiMED, Palermo, Italy.,Istituto di Farmacologia Traslazionale (IFT)-CNR, Palermo, Italy
| |
Collapse
|
32
|
The role of NLRP3 inflammasome in psychotropic drug-induced hepatotoxicity. Cell Death Dis 2022; 8:313. [PMID: 35810159 PMCID: PMC9271040 DOI: 10.1038/s41420-022-01109-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2022] [Revised: 06/22/2022] [Accepted: 06/28/2022] [Indexed: 11/29/2022]
Abstract
Increased medical application of psychotropic drugs raised attention concerning their toxicological effects. In fact, more than 160 psychotropic drugs including antidepressants and antipsychotics, have been shown to cause liver side effects, but the underlying mechanisms are still poorly understood. Here, we discovered that fluoxetine, a common antidepressant, was specifically sensed by NLRP3 inflammasome, whose subsequent activation resulted in the maturation of caspase-1 and IL-1β, as well as gasdermin D (GSDMD) cleavage, which could be completely abrogated by a selective NLRP3 inhibitor MCC950 or Nlrp3 knockout (Nlrp3−/−). Mechanistically, mitochondrial damage and the subsequent mitochondrial reactive oxygen species (mtROS) accumulation were crucial upstream signaling events in fluoxetine-triggered NLRP3 inflammasome activation. In fluoxetine hepatotoxicity models, mice showed the alterations of aminotransferase levels, hepatic inflammation and hepatocyte death in an NLRP3-dependent manner, and MCC950 pretreatment could reverse these side effects of fluoxetine. Notably, we also found that multiple antidepressants, such as amitriptyline, paroxetine, and imipramine, and antipsychotics, such as asenapine, could specifically trigger the NLRP3 inflammasome activation. Collectively, our findings implicate multiple psychotropic drugs may act as danger signals sensed by the NLRP3 inflammasome and result in hepatic injury.
Collapse
|
33
|
Abstract
Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection induces inflammatory response, cytokine storm, venous thromboembolism, coagulopathy, and multiple organ damage. Resting endothelial cells prevent coagulation, control blood flow, and inhibit inflammation. However, it remains unknown how SARS-CoV-2 induces strong molecular signals in distant cells for immunopathogenesis. In this study, we examined the consequence of human endothelial cells, microvascular endothelial cells (HMEC-1), and liver endothelial cells (TMNK-1) to exosomes isolated from plasma of mild or severe COVID-19 patients. We observed a significant induction of NLRP3, caspase-1, and interleukin-1β (IL-1β) mRNA expression in endothelial cells following exposure to exosomes from severe COVID-19 patients compared with that from patients with mild disease or healthy donors. Activation of caspase-1 was noted in the endothelial cell culture medium following exposure to the COVID-19 exosomes. Furthermore, COVID-19 exosomes significantly induced mature IL-1β secretion in both HMEC-1 and TMNK-1 endothelial cell culture medium. Thus, our results demonstrated for the first time that exosomes from COVID-19 plasma trigger NLRP3 inflammasome in endothelial cells of distant organs resulting in IL-1β secretion and inflammatory response. IMPORTANCE Severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) infection is a global health problem. Although the vaccine controls infection, understanding the molecular mechanism of pathogenesis will help in developing future therapies. Furthermore, several investigators predicted the involvement of endothelial cell-related inflammation in SARS-CoV-2 infection and using extracellular vesicles as a cargo to carry a drug or vaccine for combating SARS-CoV-2 infection. However, the mechanism by which endothelial cells are inflamed remains unknown. Our present study highlights that exosomes from severe COVID-19 patients can enhance inflammasome activity in distant endothelial cells for augmentation of immunopathogenesis and opens an avenue for developing therapies.
Collapse
|
34
|
Rademacher F, Bartels J, Gläser R, Rodewald M, Schubert S, Drücke D, Rohde H, Harder J. Staphylococcus epidermidis-derived protease Esp mediates proteolytic activation of pro-IL-1beta in human keratinocytes. J Invest Dermatol 2022; 142:2756-2765.e8. [PMID: 35490742 DOI: 10.1016/j.jid.2022.04.010] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 04/03/2022] [Accepted: 04/07/2022] [Indexed: 11/16/2022]
Abstract
The gram-positive bacterium Staphylococcus epidermidis (SE) is an abundant skin commensal. It plays an important role in cutaneous defense by activation of IL-1 signaling. In keratinocytes, SE induces the release of mature IL-1beta. IL-1beta serves as an important cytokine of host defense. It contains an N-terminal prodomain that has to be cleaved off to generate active mature IL-1beta. Typically, processing and release of IL-1beta are associated with inflammasome assembly and activation of the protease caspase-1. Here we report that bacterial challenge of keratinocytes with SE induced the release of mature IL-1beta in a caspase-1-independent manner. Instead, the SE -derived serine protease Esp was identified as a pro-IL-1beta processing factor leading to a proteolytic maturation of active IL-1beta. Esp production and secretion by various SE strains correlated with their capacity to induce release of mature IL-1beta in human primary keratinocytes. Reconstitution of Esp-lacking SE strains with Esp enhanced their capacity to induce IL-1beta release in keratinocytes and skin. Intracellular abundance of pro-IL-1beta and cytotoxic effects of SE suggest release of pro-IL-1beta during injury followed by extracellular Esp-mediated processing to mature IL-1beta. These findings provide further insights into how a skin commensal interacts with keratinocytes to activate cutaneous host innate defense.
Collapse
Affiliation(s)
| | | | - Regine Gläser
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Meno Rodewald
- Department of Dermatology, Kiel University, Kiel, Germany
| | - Sabine Schubert
- Institute of Infection Medicine, Kiel University, Kiel, Germany
| | - Daniel Drücke
- Department of Reconstructive Surgery, University Hospital Schleswig-Holstein, Campus Kiel, Kiel, Germany
| | - Holger Rohde
- Institute for Medical Microbiology, Virology and Hygiene, University Medical Center Hamburg-Eppendorf (UKE), Hamburg, Germany
| | - Jürgen Harder
- Department of Dermatology, Kiel University, Kiel, Germany.
| |
Collapse
|
35
|
Tang H, Ye Y, Li L, Zhou Y, Hou L, Ren S, Xu Y. A20 alleviated caspase-1-mediated pyroptosis and inflammation stimulated by Porphyromonas gingivalis lipopolysaccharide and nicotine through autophagy enhancement. Hum Cell 2022; 35:803-816. [PMID: 35212946 DOI: 10.1007/s13577-022-00678-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Accepted: 01/19/2022] [Indexed: 01/16/2023]
Abstract
Periodontitis is the leading cause of tooth loss, and patients with smoking habits are at an increased risk of developing periodontitis. A20 (the tumor necrosis factor alpha-induced protein 3, TNFAIP3) is one of the key regulators of inflammation and cell death in numerous tissues. Emerging researches indicated A20 as a fundamental molecule in the periodontal tissue. This study was to evaluate the role of A20 against cell death and inflammation in periodontitis and to elucidate the underlying mechanisms. In our study, western blot, autophagy detection, and transmission electron microscopy showed that lipopolysaccharide from Porphyromonas gingivalis (Pg.LPS) and nicotine (NI) could enhance the activation of autophagy. Pg.LPS and NI induce the pyroptosis of human periodontal ligament cells (hPDLCs), as evidenced by the decrease of membrane integrity and the increase of NLRP3, GSDMD, GSDMD-N, caspase-1 activity, and the pro-inflammatory cytokines of IL-1β, IL-6, TNF-α. Further researches were focused on that A20, an ubiquitin-editing enzyme, was linked to hPDLCs pyroptosis. Overexpression or silencing A20 could diminish or aggravate pyroptosis in hPDLCs by the modulation of autophagy. The above results demonstrated that A20 dictated the cross-talk between pyroptosis and autophagy. Overexpression of A20 enhanced autophagy to reduce pyroptosis, and thus alleviating inflammation, suggesting that A20 may be a potent target in the treatment of periodontitis.
Collapse
Affiliation(s)
- Hui Tang
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yu Ye
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Lu Li
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yi Zhou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Liguang Hou
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Shuangshuang Ren
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China
| | - Yan Xu
- Jiangsu Key Laboratory of Oral Diseases, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China. .,Department of Periodontics, Affiliated Hospital of Stomatology, Nanjing Medical University, 1 # Shanghai Road, Nanjing, Jiangsu, 210029, People's Republic of China.
| |
Collapse
|
36
|
Pyroptosis-Mediated Periodontal Disease. Int J Mol Sci 2021; 23:ijms23010372. [PMID: 35008798 PMCID: PMC8745163 DOI: 10.3390/ijms23010372] [Citation(s) in RCA: 36] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/23/2021] [Accepted: 12/27/2021] [Indexed: 12/17/2022] Open
Abstract
Pyroptosis is a caspase-dependent process relevant to the understanding of beneficial host responses and medical conditions for which inflammation is central to the pathophysiology of the disease. Pyroptosis has been recently suggested as one of the pathways of exacerbated inflammation of periodontal tissues. Hence, this focused review aims to discuss pyroptosis as a pathological mechanism in the cause of periodontitis. The included articles presented similarities regarding methods, type of cells applied, and cell stimulation, as the outcomes also point to the same direction considering the cellular events. The collected data indicate that virulence factors present in the diseased periodontal tissues initiate the inflammasome route of tissue destruction with caspase activation, cleavage of gasdermin D, and secretion of interleukins IL-1β and IL-18. Consequently, removing periopathogens’ virulence factors that trigger pyroptosis is a potential strategy to combat periodontal disease and regain tissue homeostasis.
Collapse
|
37
|
Procházková M, Killinger M, Prokeš L, Klepárník K. Miniaturized bioluminescence technology for single-cell quantification of caspase-3/7. J Pharm Biomed Anal 2021; 209:114512. [PMID: 34891005 DOI: 10.1016/j.jpba.2021.114512] [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: 08/24/2021] [Revised: 11/26/2021] [Accepted: 11/27/2021] [Indexed: 11/25/2022]
Abstract
Correct determination of the instantaneous level and changes of relevant proteins inside individual cells is essential for correct interpretation and understanding of physiological, diagnostic, and therapeutic events. Thus, single-cell analyses are important for quantification of natural cellular heterogeneity, which cannot be evaluated from averaged data of a cell population measurements. Here, we developed an original highly sensitive and selective instrumentation and methodology based on homogeneous single-step bioluminescence assay to quantify caspases and evaluate their heterogeneity in individual cells. Individual suspended cells are selected under microscope and reliably transferred into the 7 µl detection vials by a micromanipulator. The sensitivity of the method is given by implementation of photomultiplying tube with a cooled photocathode working in the photon counting mode. By optimization of our device and methodology, the limits of detection and quantitation were decreased down to 2.1 and 7.0 fg of recombinant caspase-3, respectively. These masses are lower than average amounts of caspase-3/7 in individual apoptotic and even non-apoptotic cells. As a proof of concept, the content of caspase-3/7 in single treated and untreated HeLa cells was determined to be 154 and 25 fg, respectively. Based on these results, we aim to use the technology for investigations of non-apoptotic functions of caspases.
Collapse
Affiliation(s)
- Markéta Procházková
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, Brno 602 00, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kotlářská 267/2, Brno 611 37, Czech Republic.
| | - Michael Killinger
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, Brno 602 00, Czech Republic; Department of Chemistry, Faculty of Science, Masaryk University, Kotlářská 267/2, Brno 611 37, Czech Republic.
| | - Lubomír Prokeš
- Department of Physics, Chemistry and Vocational Education, Faculty of Education, Masaryk University, Poříčí 7, Brno 603 00, Czech Republic.
| | - Karel Klepárník
- Department of Bioanalytical Instrumentation, Institute of Analytical Chemistry, v.v.i., Czech Academy of Sciences, Veveří 97, Brno 602 00, Czech Republic.
| |
Collapse
|
38
|
Ulrich C, Kneser L, Fiedler R, Beckert J, Wildgrube S, Seibert E, Fick S, Schäfer C, Markau S, Trojanowicz B, Girndt M. Pyroptosis: A Common Feature of Immune Cells of Haemodialysis Patients. Toxins (Basel) 2021; 13:toxins13120839. [PMID: 34941677 PMCID: PMC8704801 DOI: 10.3390/toxins13120839] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Revised: 11/22/2021] [Accepted: 11/23/2021] [Indexed: 12/02/2022] Open
Abstract
NLRP-3 inflammasome activation can result in interleukin-1β (IL-1β) release and inflammatory cell death (pyroptosis). Caspase-1 is able to trigger both processes. However, other caspases, caspase-4, -5 and -8, are believed to initiate pyroptosis without affecting IL-1 secretion. In this study, we evaluated two cardiovascular risk groups, haemodialysis patients (HD) and patients with intact kidney function but high blood pressure (BP), to analyse the mechanisms driving pyroptosis. Twenty HD were age-, gender- and diabetes-matched to BP. We found a common pyroptotic pattern in both patient groups, at which pyroptosis rates but not IL-1 β levels were significantly higher in monocytes (HD vs. BP: p < 0.05), granulocytes (p < 0.01) and lymphocytes (p < 0.01) of HD patients. As uremic toxins are drivers of inflammation and regulated cell death, we applied a monocyte- and macrophage-like THP-1 model system to demonstrate that the protein-bound uremic toxin indoxyl sulfate (IS) is an inducer of pyroptotic cell death, particularly engaging caspase-4/caspase-5 and to a lesser extent caspase-8 and caspase-1. These data suggest that the uremic toxin IS can mediate pyroptosis in HD patients and the inflammatory caspase-4 and/or caspase-5 contribute to pyroptosis rates to a higher extent in comparison to caspase-1.
Collapse
Affiliation(s)
- Christof Ulrich
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
- Correspondence: ; Tel.: +49-345-557-3386
| | - Leonie Kneser
- Agaplesion Ev. Klinikum Schaumburg, 57392 Oberkirchen, Germany;
| | - Roman Fiedler
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| | - Julia Beckert
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| | | | - Eric Seibert
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
- Nephrologisches Zentrum Villingen-Schwenningen, 78054 Villingen-Schwenningen, Germany
| | - Sylvia Fick
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| | - Christoph Schäfer
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| | - Silke Markau
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| | - Bogusz Trojanowicz
- Department of Visceral, Vascular and Endocrine Surgery, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany;
| | - Matthias Girndt
- Department of Internal Medicine II, Martin Luther University Halle-Wittenberg, 06120 Halle, Germany; (R.F.); (J.B.); (E.S.); (S.F.); (C.S.); (S.M.); (M.G.)
| |
Collapse
|
39
|
Wittmann N, Behrendt AK, Mishra N, Bossaller L, Meyer-Bahlburg A. Instructions for Flow Cytometric Detection of ASC Specks as a Readout of Inflammasome Activation in Human Blood. Cells 2021; 10:2880. [PMID: 34831104 PMCID: PMC8616555 DOI: 10.3390/cells10112880] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2021] [Revised: 10/14/2021] [Accepted: 10/21/2021] [Indexed: 02/06/2023] Open
Abstract
Inflammasome activation is linked to the aggregation of the adaptor protein ASC into a multiprotein complex, known as the ASC speck. Redistribution of cytosolic ASC to this complex has been widely used as a readout for inflammasome activation and precedes the downstream proteolytic release of the proinflammatory cytokines, IL-1β and IL-18. Although inflammasomes are important for many diseases such as periodic fever syndromes, COVID-19, gout, sepsis, atherosclerosis and Alzheimer's disease, only a little knowledge exists on the precise and cell type specific occurrence of inflammasome activation in patient samples ex vivo. In this report, we provide detailed information about the optimal conditions to reliably identify inflammasome activated monocytes by ASC speck formation using a modified flow cytometric method introduced by Sester et al. in 2015. Since no protocol for optimal sample processing exists, we tested human blood samples for various conditions including anticoagulant, time and temperature, the effect of one freeze-thaw cycle for PBMC storage, and the fast generation of a positive control. We believe that this flow cytometric protocol will help researchers to perform high quality translational research in multicenter studies, and therefore provide a basis for investigating the role of the inflammasome in the pathogenesis of various diseases.
Collapse
Affiliation(s)
- Nico Wittmann
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Ann-Kathrin Behrendt
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| | - Neha Mishra
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Lukas Bossaller
- Section of Rheumatology, Department of Medicine A, University Medicine, University of Greifswald, 17489 Greifswald, Germany;
| | - Almut Meyer-Bahlburg
- Pediatric Rheumatology, Department Pediatric and Adolescent Medicine, University Medicine, University of Greifswald, 17489 Greifswald, Germany; (N.W.); (A.-K.B.)
| |
Collapse
|
40
|
Nandi D, Farid NSS, Karuppiah HAR, Kulkarni A. Imaging Approaches to Monitor Inflammasome Activation. J Mol Biol 2021; 434:167251. [PMID: 34537231 DOI: 10.1016/j.jmb.2021.167251] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Revised: 09/09/2021] [Accepted: 09/10/2021] [Indexed: 10/20/2022]
Abstract
Inflammasomes are a critical component of innate immune response which plays an important role in the pathogenesis of various chronic and acute inflammatory disease conditions. An inflammasome complex consists of a multimeric protein assembly triggered by any form of pathogenic or sterile insult, resulting in caspase-1 activation. This active enzyme is further known to activate downstream pro-inflammatory cytokines along with a pore-forming protein, eventually leading to a lytic cell death called pyroptosis. Understanding the spatiotemporal kinetics of essential inflammasome components provides a better interpretation of the complex signaling underlying inflammation during several disease pathologies. This can be attained via in-vitro and in-vivo imaging platforms, which not only provide a basic understanding of molecular signaling but are also crucial to develop and screen targeted therapeutics. To date, numerous studies have reported platforms to image different signaling components participating in inflammasome activation. Here, we review several elements of inflammasome signaling, a common molecular mechanism combining these elements and their respective imaging tools. We anticipate that future needs will include developing new inflammasome imaging systems that can be utilized as clinical tools for diagnostics and monitoring treatment responses.
Collapse
Affiliation(s)
- Dipika Nandi
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA. https://twitter.com/dipikanandi24
| | - Noorul Shaheen Sheikh Farid
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/Shaheen30n
| | - Hayat Anu Ranjani Karuppiah
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA. https://twitter.com/AnuHayat
| | - Ashish Kulkarni
- Department of Chemical Engineering, University of Massachusetts, Amherst, MA, USA; Department of Veterinary and Animal Sciences, University of Massachusetts, Amherst, MA, USA; Department of Biomedical Engineering, University of Massachusetts, Amherst, MA, USA; Center for Bioactive Delivery, Institute for Applied Life Sciences, University of Massachusetts, Amherst, MA 01003, USA.
| |
Collapse
|
41
|
Lowes DJ, Miao J, Al-Waqfi RA, Avad KA, Hevener KE, Peters BM. Identification of Dual-Target Compounds with Antifungal and Anti-NLRP3 Inflammasome Activity. ACS Infect Dis 2021; 7:2522-2535. [PMID: 34260210 PMCID: PMC11344480 DOI: 10.1021/acsinfecdis.1c00270] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Invasive and superficial infections caused by the Candida species result in significant global morbidity and mortality. As the pathogenicity of these organisms is intimately intertwined with host immune response, therapies to target both the fungus and host inflammation may be warranted. Structural similarities exist between established inhibitors of the NLRP3 inflammasome and those of fungal acetohydroxyacid synthase (AHAS). Therefore, we leveraged this information to conduct an in silico molecular docking screen to find novel polypharmacologic inhibitors of these targets that resulted in the identification of 12 candidate molecules. Of these, compound 10 significantly attenuated activation of the NLPR3 inflammasome by LPS + ATP, while also demonstrating growth inhibitory activity against C. albicans that was alleviated in the presence of exogenous branched chain amino acids, consistent with targeting of fungal AHAS. SAR studies delineated an essential molecular scaffold required for dual activity. Ultimately, 10 and its analog 10a resulted in IC50 (IL-1β release) and MIC50 (fungal growth) values with low μM potency against several Candida species. Collectively, this work demonstrates promising potential of dual-target approaches for improved management of fungal infections.
Collapse
Affiliation(s)
- David J Lowes
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Jian Miao
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Rand A Al-Waqfi
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kristiana A Avad
- Graduate Program in Pharmaceutical Sciences, College of Graduate Health Sciences, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Doctor of Pharmacy Program, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Kirk E Hevener
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| | - Brian M Peters
- Department of Clinical Pharmacy and Translational Science, College of Pharmacy, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
- Department of Microbiology, Immunology, and Biochemistry, College of Medicine, University of Tennessee Health Science Center, Memphis, Tennessee 38163, United States
| |
Collapse
|
42
|
Yang G, Lu C, Mei Z, Sun X, Han J, Qian J, Liang Y, Pan Z, Kong D, Xu S, Liu Z, Gao Y, Qi G, Shou Y, Chen S, Cao Z, Zhao Y, Lin C, Zhao Y, Geng Y, Ma W, Yan X. Association of Cancer Stem Cell Radio-Resistance Under Ultra-High Dose Rate FLASH Irradiation With Lysosome-Mediated Autophagy. Front Cell Dev Biol 2021; 9:672693. [PMID: 33996830 PMCID: PMC8116574 DOI: 10.3389/fcell.2021.672693] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2021] [Accepted: 04/06/2021] [Indexed: 12/03/2022] Open
Abstract
Cancer stem cell (CSC) is thought to be the major cause of radio-resistance and relapse post radiotherapy (RT). Recently ultra-high dose rate “FLASH-RT” evokes great interest for its decreasing normal tissue damages while maintaining tumor responses compared with conventional dose rate RT. However, the killing effect and mechanism of FLASH irradiation (FLASH-IR) on CSC and normal cancer cell are still unclear. Presently the radiation induced death profile of CSC and normal cancer cell were studied. Cells were irradiated with FLASH-IR (∼109 Gy/s) at the dose of 6–9 Gy via laser-accelerated nanosecond particles. Then the ratio of apoptosis, pyroptosis and necrosis were determined. The results showed that FLASH-IR can induce apoptosis, pyroptosis and necrosis in both CSC and normal cancer cell with different ratios. And CSC was more resistant to radiation than normal cancer cell under FLASH-IR. Further experiments tracing lysosome and autophagy showed that CSCs had higher levels of lysosome and autophagy. Taken together, our results suggested that the radio-resistance of CSC may associate with the increase of lysosome-mediated autophagy, and the decrease of apoptosis, necrosis and pyroptosis. To our limited knowledge, this is the first report shedding light on the killing effects and death pathways of CSC and normal cancer cell under FLASH-IR. By clarifying the death pathways of CSC and normal cancer cell under FLASH-IR, it may help us improve the understanding of the radio-resistance of CSC and thus help to optimize the future clinical FLASH treatment plan.
Collapse
Affiliation(s)
- Gen Yang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Chunyang Lu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Zhusong Mei
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Xiaoyi Sun
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Jintao Han
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Jing Qian
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Yulan Liang
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Zhuo Pan
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Defeng Kong
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Shirui Xu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Zhipeng Liu
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Ying Gao
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Guijun Qi
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Yinren Shou
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Shiyou Chen
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Zhengxuan Cao
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Ye Zhao
- Teaching and Research Section of Nuclear Medicine, School of Basic Medical Sciences, Anhui Medical University, Hefei, China
| | - Chen Lin
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Yanying Zhao
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Yixing Geng
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Wenjun Ma
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China
| | - Xueqing Yan
- State Key Laboratory of Nuclear Physics and Technology, School of Physics, CAPT, Peking University, Beijing, China.,Collaborative Innovation Center of Extreme Optics, Shanxi University, Taiyuan, China
| |
Collapse
|
43
|
Ding H, Li Y, Li X, Liu X, Chen S, Liu M, Zeng H. Treatment with 7% and 10% CO 2 enhanced expression of IL-1β, TNF-α, and IL-6 in hypoxic cultures of human whole blood. J Int Med Res 2021; 48:300060520912105. [PMID: 32264730 PMCID: PMC7144675 DOI: 10.1177/0300060520912105] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Objective This study investigated whether hypercapnia influenced the inflammatory response of hypoxic blood. Methods Human whole blood was cultured with 0.2% oxygen (O2) and treated with 5%, 7%, or 10% carbon dioxide (CO2). Interleukin (IL)-1β, tumor necrosis factor (TNF)-α, and IL-6 were evaluated in whole blood cultures. Reactive oxygen species (ROS) production and expression levels of caspase-1 and IL-1β were evaluated in THP-1 monocytic cells. Results IL-1β, TNF-α, and IL-6 levels were higher in the hypoxia + 7% CO2 group than in the hypoxia + 5% CO2 group. The hypoxia + 10% CO2 group had the highest IL-1β, TNF-α, and IL-6 levels, compared with the hypoxia + 7% CO2 and hypoxia + 5% CO2 groups. Expression levels of IL-1β, TNF-α, and IL-6 were significantly negatively correlated with pH levels in the cell culture medium. Treatment with 7% and 10% CO2 increased the production of ROS and the expression of caspase-1 and IL-1β in hypoxia-activated THP-1 cells. Conclusions High levels of CO2 treatment increased expression levels of IL-1β, TNF-α, and IL-6 in hypoxic whole blood cultures. High levels of CO2-induced ROS overproduction and NLRP3 inflammasome activation in monocytes may comprise a target to mitigate the inflammatory response of hypoxic blood.
Collapse
Affiliation(s)
- Hongguang Ding
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Ya Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China.,School of Medicine, South China University of Technology, Guangzhou, China
| | - Xusheng Li
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Xinqiang Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Shenglong Chen
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Mengting Liu
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| | - Hongke Zeng
- Department of Emergency and Critical Care Medicine, Guangdong Provincial People's Hospital, Guangdong Academy of Medical Sciences, Guangzhou, China
| |
Collapse
|
44
|
Xu G, Fu S, Zhan X, Wang Z, Zhang P, Shi W, Qin N, Chen Y, Wang C, Niu M, Guo Y, Wang J, Bai Z, Xiao X. Echinatin effectively protects against NLRP3 inflammasome-driven diseases by targeting HSP90. JCI Insight 2021; 6:134601. [PMID: 33350984 PMCID: PMC7934863 DOI: 10.1172/jci.insight.134601] [Citation(s) in RCA: 87] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2019] [Accepted: 12/02/2020] [Indexed: 12/14/2022] Open
Abstract
Aberrant activation of NLRP3 inflammasome has been implicated in a variety of human inflammatory diseases, but currently, no pharmacological NLRP3 inhibitor has been approved. In this study, we showed that echinatin, the ingredient of the traditional herbal medicine licorice, effectively suppresses the activation of NLRP3 inflammasome in vitro and in vivo. Further investigation revealed that echinatin exerts its inhibitory effect on NLRP3 inflammasome by binding to heat-shock protein 90 (HSP90), inhibiting its ATPase activity and disrupting the association between the cochaperone SGT1 and HSP90-NLRP3. Importantly, in vivo experiments demonstrated that administration of echinatin obviously inhibits NLRP3 inflammasome activation and ameliorates LPS-induced septic shock and dextran sodium sulfate-induced (DSS-induced) colitis in mice. Moreover, echinatin exerted favorable pharmacological effects on liver inflammation and fibrosis in a mouse model of nonalcoholic steatohepatitis (NASH). Collectively, our study identifies echinatin as a potentially novel inhibitor of NLRP3 inflammasome, and its use may be developed as a therapeutic approach for the treatment of NLRP3-driven diseases.
Collapse
Affiliation(s)
- Guang Xu
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,Integrative Medical Centre, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Shubin Fu
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China.,Jiujiang Institute for Food and Drug Control, Jiujiang, China
| | - Xiaoyan Zhan
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,Integrative Medical Centre, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Zhilei Wang
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,School of Pharmacy, Chengdu University of Traditional Chinese Medicine, Chengdu, China
| | - Ping Zhang
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,Integrative Medical Centre, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Wei Shi
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Nan Qin
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China.,School of Pharmacy, Jiangxi University of Traditional Chinese Medicine, Nanchang, China
| | - Yuanyuan Chen
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Chunyu Wang
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Ming Niu
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Yuming Guo
- Integrative Medical Centre, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Jiabo Wang
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Zhaofang Bai
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| | - Xiaohe Xiao
- Military Institute of Chinese Materia, the Fifth Medical Centre, General Hospital of PLA, Beijing, China
| |
Collapse
|
45
|
Makoni NJ, Nichols MR. The intricate biophysical puzzle of caspase-1 activation. Arch Biochem Biophys 2021; 699:108753. [PMID: 33453207 DOI: 10.1016/j.abb.2021.108753] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2020] [Revised: 12/22/2020] [Accepted: 01/05/2021] [Indexed: 12/13/2022]
Abstract
This review takes a closer look at the structural components of the molecules involved in the processes leading to caspase-1 activation. Interleukins 1β and 18 (IL-1β, IL-18) are well-known proinflammatory cytokines that are produced following cleavage of their respective precursor proteins by the cysteine protease caspase-1. Active caspase-1 is the final step of the NLRP3 inflammasome, a three-protein intracellular complex involved in inflammation and induction of pyroptosis (a proinflammatory cell-death process). NLRP3 activators facilitate assembly of the inflammasome complex and subsequent activation of caspase-1 by autoproteolysis. However, the definitive structural components of active caspase-1 are still unclear and new data add to the complexity of this process. This review outlines the historical and recent findings that provide supporting evidence for the structural aspects of caspase-1 autoproteolysis and activation.
Collapse
Affiliation(s)
- Nyasha J Makoni
- Department of Chemistry & Biochemistry, University of Missouri-St. Louis, St. Louis, MO, USA
| | - Michael R Nichols
- Department of Chemistry & Biochemistry, University of Missouri-St. Louis, St. Louis, MO, USA.
| |
Collapse
|
46
|
Healy LD, Fernández JA, Mosnier LO, Griffin JH. Activated protein C and PAR1-derived and PAR3-derived peptides are anti-inflammatory by suppressing macrophage NLRP3 inflammasomes. J Thromb Haemost 2021; 19:269-280. [PMID: 33049092 PMCID: PMC7790994 DOI: 10.1111/jth.15133] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Revised: 09/15/2020] [Accepted: 10/05/2020] [Indexed: 12/13/2022]
Abstract
Essentials Activated protein C (APC) is a serine protease with anticoagulant and cytoprotective effects. We tested whether APC or non-canonical PAR-derived peptides suppress inflammasome activity. APC or PAR1- and PAR3-derived peptides restrict inflammasome-dependent caspase-1 activity. Combined PAR1-derived and PAR3-derived peptides synergistically suppress caspase-1 activity. ABSTRACT: Background Activated protein C (APC) has been shown to restrict murine inflammasome activity. However, whether APC can exert anti-inflammatory activity in part through suppression of inflammasome activation in human systems is unknown. Objectives Studies were made to determine whether either APC or protease activated receptor (PAR)-derived peptides can reduce NLRP3 inflammasome activity in differentiated human THP-1 macrophage-like cells or in primary human monocytes stimulated to activate the inflammasome. Methods Human THP-1 cells or primary human monocytes were differentiated, treated with APC or PAR-derived peptides, and then stimulated with lipopolysaccharide and ATP to induce caspase-1 activity, a product of inflammasome activation. Results Activated protein C or noncanonical PAR1-derived or PAR3-derived peptides significantly reduced caspase-1 activity, detection of fluorescent NLRP3, and IL-1β release from THP-1 cells. At low concentrations where no effect was observed for each individual peptide, combinations of the PAR1-derived peptide and the PAR3-derived peptide resulted in a significant synergistic decrease in caspase-1 and IL-1β release. Caspase-1 activity was also reduced in primary human monocytes. Studies using blocking antibodies and small molecule PAR1 inhibitors suggest that EPCR, PAR1, and PAR3 each play roles in the observed anti-inflammatory effects. Several shortened versions of the PAR1- and PAR3-derived peptide reduced caspase-1 activity and exhibited synergistic anti-inflammatory effects. Conclusions The results indicate that both APC and certain PAR1- and PAR3-derived peptides, which are biased agonists for PAR1 or PAR3, can reduce inflammasome activity in stimulated human monocytes as measured by caspase-1 activity and IL-1β release and that PAR-derived biased peptide agonist combinations are synergistically anti-inflammatory.
Collapse
Affiliation(s)
- Laura D Healy
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - José A Fernández
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - Laurent O Mosnier
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| | - John H Griffin
- Department of Molecular Medicine, The Scripps Research Institute, La Jolla, CA, USA
| |
Collapse
|
47
|
Abstract
Tumor necrosis factor superfamily (TNFSF) ligands and receptors have distinctive structural characters that link them to cell growth, cell survival, or cell death. Some of these can activate both inflammatory and apoptotic pathways, depending on target cell types and other extrinsic stimuli. Many of the TNF receptor superfamily molecules are expressed in cells of the immune system, which may be central to autoimmune and inflammatory diseases as well as cancer. However, the function of TNFSF members is not just restricted to immune cells. Members of TNFSF have been linked to an array of pathophysiologies, including cancer, neurologic, cardiovascular, pulmonary, autoimmune, and metabolic diseases. TNF-α of TNFSF is a pro-inflammatory cytokine produced by macrophages/monocytes, widely implicated in the pathogenesis of inflammatory disorders. In view of these facts, TNF-α has been recommended as an important target for discovering drugs for autoimmune and inflammatory diseases and cancer. Various cell-based assays to understand the role of TNF-α in inflammation and to estimate the concentrations of TNF-α levels in body fluids such as plasma, synovium, etc., are being followed by researchers. In this chapter, methods of cell viability assay, ELISA assay, RT-PCR, and western blot analysis for estimating LPS-induced TNF-α protein expressions are described in detail.
Collapse
|
48
|
Zhang H, Zahid A, Ismail H, Tang Y, Jin T, Tao J. An overview of disease models for NLRP3 inflammasome over-activation. Expert Opin Drug Discov 2020; 16:429-446. [PMID: 33131335 DOI: 10.1080/17460441.2021.1844179] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Introduction: Inflammatory reactions, including those mediated by the NLRP3 inflammasome, maintain the body's homeostasis by removing pathogens, repairing damaged tissues, and adapting to stressed environments. However, uncontrolled activation of the NLRP3 inflammasome tends to cause various diseases using different mechanisms. Recently, many inhibitors of the NLRP3 inflammasome have been reported and many are being developed. In order to assess their efficacy, specificity, and mechanism of action, the screening process of inhibitors requires various types of cell and animal models of NLRP3-associated diseases.Areas covered: In the following review, the authors give an overview of the cell and animal models that have been used during the research and development of various inhibitors of the NLRP3 inflammasome.Expert opinion: There are many NLRP3 inflammasome inhibitors, but most of the inhibitors have poor specificity and often influence other inflammatory pathways. The potential risk for cross-reaction is high; therefore, the development of highly specific inhibitors is essential. The selection of appropriate cell and animal models, and combined use of different models for the evaluation of these inhibitors can help to clarify the target specificity and therapeutic effects, which is beneficial for the development and application of drugs targeting the NLRP3 inflammasome.
Collapse
Affiliation(s)
- Hongliang Zhang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Ayesha Zahid
- Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Hazrat Ismail
- MOE Key Laboratory for Cellular Dynamics & Anhui Key Laboratory for Chemical Biology, CAS Center for Excellence in Molecular Cell Science. Hefei National Science Center for Physical Sciences at Microscale. University of Science and Technology of China, Hefei, China
| | - Yujie Tang
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| | - Tengchuan Jin
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Division of Molecular Medicine, Hefei National Laboratory for Physical Sciences at Microscale, CAS Key Laboratory of Innate Immunity and Chronic Disease, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,CAS Center for Excellence in Molecular Cell Science, Shanghai, China
| | - Jinhui Tao
- Department of Rheumatology and Immunology, the First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China
| |
Collapse
|
49
|
Shi C, Killoran MP, Hall MP, Otto P, Wood MG, Strauss E, Encell LP, Machleidt T, Wood KV, Kirkland TA. 5,5-Dialkylluciferins are thermal stable substrates for bioluminescence-based detection systems. PLoS One 2020; 15:e0243747. [PMID: 33315907 PMCID: PMC7735563 DOI: 10.1371/journal.pone.0243747] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2020] [Accepted: 11/26/2020] [Indexed: 11/18/2022] Open
Abstract
Firefly luciferase-based ATP detection assays are frequently used as a sensitive, cost-efficient method for monitoring hygiene in many industrial settings. Solutions of detection reagent, containing a mixture of a substrate and luciferase enzyme that produces photons in the presence of ATP, are relatively unstable and maintain only a limited shelf life even under refrigerated conditions. It is therefore common for the individual performing a hygiene test to manually prepare fresh reagent at the time of monitoring. To simplify sample processing, a liquid detection reagent with improved thermal stability is needed. The engineered firefly luciferase, Ultra-Glo™, fulfills one aspect of this need and has been valuable for hygiene monitoring because of its high resistance to chemical and thermal inactivation. However, solutions containing both Ultra-Glo™ luciferase and its substrate luciferin gradually lose the ability to effectively detect ATP over time. We demonstrate here that dehydroluciferin, a prevalent oxidative breakdown product of luciferin, is a potent inhibitor of Ultra-Glo™ luciferase and that its formation in the detection reagent is responsible for the decreased ability to detect ATP. We subsequently found that dialkylation at the 5-position of luciferin (e.g., 5,5-dimethylluciferin) prevents degradation to dehydroluciferin and improves substrate thermostability in solution. However, since 5,5-dialkylluciferins are poorly utilized by Ultra-Glo™ luciferase as substrates, we used structural optimization of the luciferin dialkyl modification and protein engineering of Ultra-Glo™ to develop a luciferase/luciferin pair that shows improved total reagent stability in solution at ambient temperature. The results of our studies outline a novel luciferase/luciferin system that could serve as foundations for the next generation of bioluminescence ATP detection assays with desirable reagent stability.
Collapse
Affiliation(s)
- Ce Shi
- Promega Biosciences, Inc., San Luis Obispo, California, United States of America
| | | | - Mary P. Hall
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Paul Otto
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Monika G. Wood
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Ethan Strauss
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Lance P. Encell
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Thomas Machleidt
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Keith V. Wood
- Promega Corporation, Madison, Wisconsin, United States of America
| | - Thomas A. Kirkland
- Promega Biosciences, Inc., San Luis Obispo, California, United States of America
| |
Collapse
|
50
|
Terahara K, Iwabuchi R, Iwaki R, Takahashi Y, Tsunetsugu-Yokota Y. Substantial induction of non-apoptotic CD4 T-cell death during the early phase of HIV-1 infection in a humanized mouse model. Microbes Infect 2020; 23:104767. [PMID: 33049386 DOI: 10.1016/j.micinf.2020.10.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Revised: 10/05/2020] [Accepted: 10/06/2020] [Indexed: 02/07/2023]
Abstract
Several mechanisms underline induction of CD4 T-cell death by human immunodeficiency virus (HIV) infection. For a long time, apoptosis was considered central to cell death involved in the depletion of CD4 T cells during HIV infection. However, which types of cell death are induced during the early phase of HIV infection in vivo remains unclear. In this study, CD4 T-cell death induced in early HIV infection was characterized using humanized mice challenged with CCR5-tropic (R5) or CXCR4-tropic (X4) HIV-1. Results showed that CD4 T-cell death was induced in the spleen 3 days post-challenge with both R5 and X4 HIV-1. Although cell death without caspase-1 and caspase-3/7 activation was preferentially observed, caspase-1+ pyroptosis was also significantly induced within the memory subpopulation by R5 or X4 HIV-1 and the naïve subpopulation by X4 HIV-1. In contrast, caspase-3/7+ apoptosis was not enhanced by either R5 or X4 HIV-1. Furthermore, phosphorylated mixed lineage kinase domain-like protein+ necroptosis was induced by only X4 HIV-1. These findings indicate that various types of non-apoptotic CD4 T-cell death, such as pyroptosis and necroptosis, are induced during the early phase of HIV infection in vivo.
Collapse
Affiliation(s)
- Kazutaka Terahara
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan.
| | - Ryutaro Iwabuchi
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan; Department of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsucho, Shinjuku-ku, Tokyo, 162-8480, Japan
| | - Rieko Iwaki
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yoshimasa Takahashi
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan
| | - Yasuko Tsunetsugu-Yokota
- Department of Immunology, National Institute of Infectious Diseases, 1-23-1 Toyama, Shinjuku-ku, Tokyo, 162-8640, Japan; Department of Medical Technology, School of Human Sciences, Tokyo University of Technology, 5-23-22 Nishikamata, Ota-ku, Tokyo, 144-8535, Japan
| |
Collapse
|