175 results on '"Kate Schroder"'
Search Results
2. The prevalence of hypoxemia among pediatric and adult patients presenting to healthcare facilities in low- and middle-income countries: protocol for a systematic review and meta-analysis
- Author
-
Felix Lam, Rami Subhi, Jason Houdek, Kate Schroder, Audrey Battu, and Hamish Graham
- Subjects
Systematic review ,Meta-analysis ,Hypoxemia ,Pulse oximetry ,Medicine - Abstract
Abstract Background Hypoxemia is a severe condition associated with high rates of mortality, particularly in low- and middle-income countries (LMICs) with poor access to oxygen therapy. Despite its clinical significance, there have been few studies to describe the burden of hypoxemia. Thus, the primary objective of this study is to systematically describe the prevalence of hypoxemia among pediatric and adult patients in low- and middle-income countries. Methods/design Standard systematic review methods will be used. Bibliographic databases (MEDLINE, EMBASE, CINAHL) will be searched from 1998 onwards. The search strategy aims to identify studies that have measured peripheral blood oxygen saturation (SpO2) in children and adults presenting to health facilities in LMICs. Studies will be included if oxygen saturation measurements by pulse oximetry were measured. No studies will be excluded based on study design though patients recruited from intensive care units and post-operative care will be excluded. The primary outcome is the prevalence of hypoxemia on presentation to the healthcare facility. We define hypoxemia on the basis of SpO2 measurements, and use a threshold of SpO2 less than 90% at sea level though allow for a lower threshold for studies conducted at higher altitude and where justified. Standardized tools will be used to extract data on number of patients with SpO2 measurements, number of patients with hypoxemia, patient population characteristics, and study characteristics. Quality of the included studies will be assessed using the “Checklist for Prevalence Studies” developed by the Joanna Briggs Institute. If there are enough studies to do so, we will conduct meta-analysis using a random effects model to estimate prevalence of hypoxemia and conduct subgroup analyses by age and disease groups. Discussion Hypoxemia is a critical condition and understanding the burden of hypoxemia may support decision-making in LMICs to deploy pulse oximeters and oxygen treatments more efficiently to address diseases and patient populations with the highest burden. Previous studies on hypoxemia prevalence have focused too narrowly on a few diseases or specific patient populations (e.g., pneumonia in children under five) whereas any effort to improve access to oxygen requires understanding of the potential demand for oxygen for all diseases and population groups. Governments, UN agencies, donors, and NGOs are investing strongly to improve oxygen systems in LMICs. Effective oxygen system planning requires estimation of oxygen need, informed by robust data on hypoxemia prevalence and admission patterns at all the levels of the health system. This study aims to fill that gap by providing comprehensive estimates of hypoxemia prevalence. Systematic review registration PROSPERO CRD42019136622 .
- Published
- 2020
- Full Text
- View/download PDF
3. The complex interplay between endoplasmic reticulum stress and the NLRP3 inflammasome: a potential therapeutic target for inflammatory disorders
- Author
-
Wai Chin Chong, Madhur D Shastri, Gregory M Peterson, Rahul P Patel, Prabuddha S Pathinayake, Kamal Dua, Nicole G Hansbro, Alan C Hsu, Peter A Wark, Shakti Dhar Shukla, Matt D Johansen, Kate Schroder, and Philip M Hansbro
- Subjects
endoplasmic reticulum stress ,inflammasome ,inflammatory disorder ,NLRP3 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Inflammation is the result of a complex network of cellular and molecular interactions and mechanisms that facilitate immune protection against intrinsic and extrinsic stimuli, particularly pathogens, to maintain homeostasis and promote tissue healing. However, dysregulation in the immune system elicits excess/abnormal inflammation resulting in unintended tissue damage and causes major inflammatory diseases including asthma, chronic obstructive pulmonary disease, atherosclerosis, inflammatory bowel diseases, sarcoidosis and rheumatoid arthritis. It is now widely accepted that both endoplasmic reticulum (ER) stress and inflammasomes play critical roles in activating inflammatory signalling cascades. Notably, evidence is mounting for the involvement of ER stress in exacerbating inflammasome‐induced inflammatory cascades, which may provide a new axis for therapeutic targeting in a range of inflammatory disorders. Here, we comprehensively review the roles, mechanisms and interactions of both ER stress and inflammasomes, as well as their interconnected relationships in inflammatory signalling cascades. We also discuss novel therapeutic strategies that are being developed to treat ER stress‐ and inflammasome‐related inflammatory disorders.
- Published
- 2021
- Full Text
- View/download PDF
4. Endothelial cells are not productively infected by SARS‐CoV‐2
- Author
-
Lilian Schimmel, Keng Yih Chew, Claudia J Stocks, Teodor E Yordanov, Patricia Essebier, Arutha Kulasinghe, James Monkman, Anna Flavia Ribeiro dosSantos Miggiolaro, Caroline Cooper, Lucia deNoronha, Kate Schroder, Anne Karine Lagendijk, Larisa I Labzin, Kirsty R Short, and Emma J Gordon
- Subjects
blood vessels ,COVID‐19 ,endothelial cells ,inflammation ,SARS‐CoV‐2 ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract Objectives Thrombotic and microvascular complications are frequently seen in deceased COVID‐19 patients. However, whether this is caused by direct viral infection of the endothelium or inflammation‐induced endothelial activation remains highly contentious. Methods Here, we use patient autopsy samples, primary human endothelial cells and an in vitro model of the pulmonary epithelial–endothelial cell barrier. Results We show that primary human endothelial cells express very low levels of the SARS‐CoV‐2 receptor ACE2 and the protease TMPRSS2, which blocks their capacity for productive viral infection, and limits their capacity to produce infectious virus. Accordingly, endothelial cells can only be infected when they overexpress ACE2, or are exposed to very high concentrations of SARS‐CoV‐2. We also show that SARS‐CoV‐2 does not infect endothelial cells in 3D vessels under flow conditions. We further demonstrate that in a co‐culture model endothelial cells are not infected with SARS‐CoV‐2. Endothelial cells do however sense and respond to infection in the adjacent epithelial cells, increasing ICAM‐1 expression and releasing pro‐inflammatory cytokines. Conclusions Taken together, these data suggest that in vivo, endothelial cells are unlikely to be infected with SARS‐CoV‐2 and that infection may only occur if the adjacent pulmonary epithelium is denuded (basolateral infection) or a high viral load is present in the blood (apical infection). In such a scenario, whilst SARS‐CoV‐2 infection of the endothelium can occur, it does not contribute to viral amplification. However, endothelial cells may still play a key role in SARS‐CoV‐2 pathogenesis by sensing adjacent infection and mounting a pro‐inflammatory response to SARS‐CoV‐2.
- Published
- 2021
- Full Text
- View/download PDF
5. The rOX‐stars of inflammation: links between the inflammasome and mitochondrial meltdown
- Author
-
Caroline L Holley and Kate Schroder
- Subjects
infection ,inflammasome ,mitochondria ,NLRP3 ,ROS ,Immunologic diseases. Allergy ,RC581-607 - Abstract
Abstract The nod‐like receptor protein 3 (NLRP3) inflammasome drives inflammation in response to mitochondrial dysfunction. As metabolic powerhouses with prokaryotic ancestry, mitochondria are a cache for danger‐associated molecular patterns and pathogen‐associated molecular pattern‐like molecules that elicit potent innate immune responses. Persistent mitochondrial damage caused by infection, or genetic or environmental factors, can lead to inappropriate or sustained inflammasome signalling. Here, we review the features of mitochondria that drive inflammatory signalling, with a particular focus on mitochondrial activation of the NLRP3 inflammasome. Given that mitochondrial network dynamics, metabolic activity and redox state are all intricately linked to each other and to NLRP3 inflammasome activity, we highlight the importance of a holistic approach to investigations of NLRP3 activation by dysfunctional mitochondria.
- Published
- 2020
- Full Text
- View/download PDF
6. Interleukin-1β Maturation Triggers Its Relocation to the Plasma Membrane for Gasdermin-D-Dependent and -Independent Secretion
- Author
-
Mercedes Monteleone, Amanda C. Stanley, Kaiwen W. Chen, Darren L. Brown, Jelena S. Bezbradica, Jessica B. von Pein, Caroline L. Holley, Dave Boucher, Melanie R. Shakespear, Ronan Kapetanovic, Verena Rolfes, Matthew J. Sweet, Jennifer L. Stow, and Kate Schroder
- Subjects
Biology (General) ,QH301-705.5 - Abstract
Summary: IL-1β requires processing by caspase-1 to generate the active, pro-inflammatory cytokine. Acute IL-1β secretion from inflammasome-activated macrophages requires caspase-1-dependent GSDMD cleavage, which also induces pyroptosis. Mechanisms of IL-1β secretion by pyroptotic and non-pyroptotic cells, and the precise functions of caspase-1 and GSDMD therein, are unresolved. Here, we show that, while efficient early secretion of endogenous IL-1β from primary non-pyroptotic myeloid cells in vitro requires GSDMD, later IL-1β release in vitro and in vivo proceeds independently of GSDMD. IL-1β maturation is sufficient for slow, caspase-1/GSDMD-independent secretion of ectopic IL-1β from resting, non-pyroptotic macrophages, but the speed of IL-1β release is boosted by inflammasome activation, via caspase-1 and GSDMD. IL-1β cleavage induces IL-1β enrichment at PIP2-enriched plasma membrane ruffles, and this is a prerequisite for IL-1β secretion and is mediated by a polybasic motif within the cytokine. We thus reveal a mechanism in which maturation-induced IL-1β trafficking facilitates its unconventional secretion. : Interleukin-1β is a potent pro-inflammatory cytokine whose dysregulated production drives a myriad of human diseases. Monteleone et al. uncover the trafficking mechanisms driving the unconventional secretion of mature interleukin-1β in non-pyroptotic and pyroptotic myeloid cells and reveal functions for caspase-1 and GSDMD therein. Keywords: interleukin-1, unconventional protein secretion, inflammasome, caspase-1, macrophage, neutrophil, gasdermin, pyroptosis, trafficking, phosphoinositides
- Published
- 2018
- Full Text
- View/download PDF
7. XIAP Loss Triggers RIPK3- and Caspase-8-Driven IL-1β Activation and Cell Death as a Consequence of TLR-MyD88-Induced cIAP1-TRAF2 Degradation
- Author
-
Kate E. Lawlor, Rebecca Feltham, Monica Yabal, Stephanie A. Conos, Kaiwen W. Chen, Stephanie Ziehe, Carina Graß, Yifan Zhan, Tan A. Nguyen, Cathrine Hall, Angelina J. Vince, Simon M. Chatfield, Damian B. D’Silva, Kenneth C. Pang, Kate Schroder, John Silke, David L. Vaux, Philipp J. Jost, and James E. Vince
- Subjects
Toll-like receptor ,NLRP3 ,RIPK3 ,caspase-8 ,TNFR2 ,XIAP ,cIAP1 ,necroptosis ,interferon ,autoinflammatory disease ,Biology (General) ,QH301-705.5 - Abstract
X-linked Inhibitor of Apoptosis (XIAP) deficiency predisposes people to pathogen-associated hyperinflammation. Upon XIAP loss, Toll-like receptor (TLR) ligation triggers RIPK3-caspase-8-mediated IL-1β activation and death in myeloid cells. How XIAP suppresses these events remains unclear. Here, we show that TLR-MyD88 causes the proteasomal degradation of the related IAP, cIAP1, and its adaptor, TRAF2, by inducing TNF and TNF Receptor 2 (TNFR2) signaling. Genetically, we define that myeloid-specific cIAP1 loss promotes TLR-induced RIPK3-caspase-8 and IL-1β activity in the absence of XIAP. Importantly, deletion of TNFR2 in XIAP-deficient cells limited TLR-MyD88-induced cIAP1-TRAF2 degradation, cell death, and IL-1β activation. In contrast to TLR-MyD88, TLR-TRIF-induced interferon (IFN)β inhibited cIAP1 loss and consequent cell death. These data reveal how, upon XIAP deficiency, a TLR-TNF-TNFR2 axis drives cIAP1-TRAF2 degradation to allow TLR or TNFR1 activation of RIPK3-caspase-8 and IL-1β. This mechanism may explain why XIAP-deficient patients can exhibit symptoms reminiscent of patients with activating inflammasome mutations.
- Published
- 2017
- Full Text
- View/download PDF
8. Multiple inflammasomes may regulate the interleukin-1-driven inflammation in protracted bacterial bronchitis
- Author
-
Alice C-H. Chen, Hai B. Tran, Yang Xi, Stephanie T. Yerkovich, Katherine J. Baines, Susan J. Pizzutto, Melanie Carroll, Avril A.B. Robertson, Matthew A. Cooper, Kate Schroder, Jodie L. Simpson, Peter G. Gibson, Greg Hodge, Ian B. Masters, Helen M. Buntain, Helen L. Petsky, Samantha J. Prime, Anne B. Chang, Sandra Hodge, and John W. Upham
- Subjects
Medicine - Abstract
Protracted bacterial bronchitis (PBB) in young children is characterised by prolonged wet cough, prominent airway interleukin (IL)-1β expression and infection, often with nontypeable Haemophilus influenzae (NTHi). The mechanisms responsible for IL-1-driven inflammation in PBB are poorly understood. We hypothesised that the inflammation in PBB involves the NLRP3 and/or AIM2 inflammasome/IL-1β axis. Lung macrophages obtained from bronchoalveolar lavage (BAL), peripheral blood mononuclear cells (PBMCs), blood monocytes and monocyte-derived macrophages from patients with PBB and age-matched healthy controls were cultured in control medium or exposed to live NTHi. In healthy adult PBMCs, CD14+ monocytes contributed to 95% of total IL-1β-producing cells upon NTHi stimulation. Stimulation of PBB PBMCs with NTHi significantly increased IL-1β expression (p
- Published
- 2018
- Full Text
- View/download PDF
9. The Neutrophil NLRC4 Inflammasome Selectively Promotes IL-1β Maturation without Pyroptosis during Acute Salmonella Challenge
- Author
-
Kaiwen W. Chen, Christina J. Groß, Flor Vásquez Sotomayor, Katryn J. Stacey, Jurg Tschopp, Matthew J. Sweet, and Kate Schroder
- Subjects
Biology (General) ,QH301-705.5 - Abstract
The macrophage NLRC4 inflammasome drives potent innate immune responses against Salmonella by eliciting caspase-1-dependent proinflammatory cytokine production (e.g., interleukin-1β [IL-1β]) and pyroptotic cell death. However, the potential contribution of other cell types to inflammasome-mediated host defense against Salmonella was unclear. Here, we demonstrate that neutrophils, typically viewed as cellular targets of IL-1β, themselves activate the NLRC4 inflammasome during acute Salmonella infection and are a major cell compartment for IL-1β production during acute peritoneal challenge in vivo. Importantly, unlike macrophages, neutrophils do not undergo pyroptosis upon NLRC4 inflammasome activation. The resistance of neutrophils to pyroptotic death is unique among inflammasome-signaling cells so far described and allows neutrophils to sustain IL-1β production at a site of infection without compromising the crucial inflammasome-independent antimicrobial effector functions that would be lost if neutrophils rapidly lysed upon caspase-1 activation. Inflammasome pathway modification in neutrophils thus maximizes host proinflammatory and antimicrobial responses during pathogen challenge.
- Published
- 2014
- Full Text
- View/download PDF
10. IL-1 Contributes to the Anti-Cancer Efficacy of Ingenol Mebutate.
- Author
-
Thuy T Le, Kresten Skak, Kate Schroder, Wayne A Schroder, Glen M Boyle, Carly J Pierce, and Andreas Suhrbier
- Subjects
Medicine ,Science - Abstract
Ingenol mebutate is approved for the topical treatment of actinic keratoses and may ultimately also find utility in treating skin cancers. Here we show that relapse rates of subcutaneous B16 melanoma tumours treated topically with ingenol mebutate were not significantly different in C57BL/6 and Rag1-/- mice, suggesting B and T cells do not play a major role in the anti-cancer efficacy of ingenol mebutate. Relapse rates were, however, significantly increased in MyD88-/- mice and in C57BL/6 mice treated with the anti-IL-1 agent, anakinra. Ingenol mebutate treatment induces a pronounced infiltration of neutrophils, which have been shown to have anti-cancer activity in mice. Herein we provide evidence that IL-1 promotes neutrophil recruitment to the tumour, decreases apoptosis of infiltrating neutrophils and increases neutrophil tumour killing activity. These studies suggest IL-1, via its action on neutrophils, promotes the anti-cancer efficacy of ingenol mebutate, with ingenol mebutate treatment causing both IL-1β induction and IL-1α released from keratinocytes.
- Published
- 2016
- Full Text
- View/download PDF
11. Development of a DNA barcode tagging method for monitoring dynamic changes in gene expression by using an ultra high-throughput sequencer
- Author
-
Norihiro Maeda, Hiromi Nishiyori, Mari Nakamura, Chika Kawazu, Mitsuyoshi Murata, Hiromi Sano, Kengo Hayashida, Shiro Fukuda, Michihira Tagami, Akira Hasegawa, Kayoko Murakami, Kate Schroder, Katharine Irvine, David A. Hume, Yoshihide Hayashizaki, Piero Carninci, and Harukazu Suzuki
- Subjects
Biology (General) ,QH301-705.5 - Abstract
CAGE (cap analysis of gene expression) is a method for identifying transcription start sites by sequencing the first 20 or 21 nucleotides from the 5′ end of capped transcripts, allowing genome-wide promoter analyses to be performed. The potential of the CAGE as a form of expression profiling was limited previously by sequencing technology and the labor-intensive protocol. Here we describe an improved CAGE method for use with a next generation sequencer. This modified method allows the identification of the RNA source of each CAGE tag within a pooled library by introducing DNA tags (barcodes). The method not only drastically improves the sequencing capacity, but also contributes to savings in both time and budget. Additionally, this pooled CAGE tag method enables the dynamic changes in promoter usage and gene expression to be monitored.
- Published
- 2008
- Full Text
- View/download PDF
12. Macrophage activation and differentiation signals regulate schlafen-4 gene expression: evidence for Schlafen-4 as a modulator of myelopoiesis.
- Author
-
Wendy J van Zuylen, Valerie Garceau, Adi Idris, Kate Schroder, Katharine M Irvine, Jane E Lattin, Dmitry A Ovchinnikov, Andrew C Perkins, Andrew D Cook, John A Hamilton, Paul J Hertzog, Katryn J Stacey, Stuart Kellie, David A Hume, and Matthew J Sweet
- Subjects
Medicine ,Science - Abstract
BackgroundThe ten mouse and six human members of the Schlafen (Slfn) gene family all contain an AAA domain. Little is known of their function, but previous studies suggest roles in immune cell development. In this report, we assessed Slfn regulation and function in macrophages, which are key cellular regulators of innate immunity.Methodology/principal findingsMultiple members of the Slfn family were up-regulated in mouse bone marrow-derived macrophages (BMM) by the Toll-like Receptor (TLR)4 agonist lipopolysaccharide (LPS), the TLR3 agonist Poly(I∶C), and in disease-affected joints in the collagen-induced model of rheumatoid arthritis. Of these, the most inducible was Slfn4. TLR agonists that signal exclusively through the MyD88 adaptor protein had more modest effects on Slfn4 mRNA levels, thus implicating MyD88-independent signalling and autocrine interferon (IFN)-β in inducible expression. This was supported by the substantial reduction in basal and LPS-induced Slfn4 mRNA expression in IFNAR-1⁻/⁻ BMM. LPS causes growth arrest in macrophages, and other Slfn family genes have been implicated in growth control. Slfn4 mRNA levels were repressed during macrophage colony-stimulating factor (CSF-1)-mediated differentiation of bone marrow progenitors into BMM. To determine the role of Slfn4 in vivo, we over-expressed the gene specifically in macrophages in mice using a csf1r promoter-driven binary expression system. Transgenic over-expression of Slfn4 in myeloid cells did not alter macrophage colony formation or proliferation in vitro. Monocyte numbers, as well as inflammatory macrophages recruited to the peritoneal cavity, were reduced in transgenic mice that specifically over-expressed Slfn4, while macrophage numbers and hematopoietic activity were increased in the livers and spleens.ConclusionsSlfn4 mRNA levels were up-regulated during macrophage activation but down-regulated during differentiation. Constitutive Slfn4 expression in the myeloid lineage in vivo perturbs myelopoiesis. We hypothesise that the down-regulation of Slfn4 gene expression during macrophage differentiation is a necessary step in development of this lineage.
- Published
- 2011
- Full Text
- View/download PDF
13. Stoking inflammasome fires in the COVID-19 neighborhood
- Author
-
Larisa I. Labzin and Kate Schroder
- Subjects
Virology ,Parasitology ,Microbiology - Published
- 2023
- Full Text
- View/download PDF
14. NLRP3 and pyroptosis blockers for treating inflammatory diseases
- Author
-
Pablo Pelegrin, Rebecca Coll, and Kate Schroder
- Subjects
Inflammation ,drug-like candidates ,Pharmacology ,integumentary system ,diabetes ,Inflammasomes ,gasdermin D ,NASH ,Toxicology ,stroke ,small-molecule inhibitors ,NLRP3 ,inflammasome ,NLR Family, Pyrin Domain-Containing 3 Protein ,intestinal inflammation ,Pyroptosis ,Humans ,neurodegenerative diseases - Abstract
The nucleotide-binding oligomerization domain, leucine-rich repeat and pyrin domain-containing protein 3 (NLRP3) inflammasome has emerged as a key mediator of pathological inflammation in many diseases and is an exciting drug target. Here, we review the molecular basis of NLRP3 inhibition by drug-like small molecules under development as novel therapeutics. We also summarize recent strategies to block pyroptosis as a novel approach to suppress chronic inflammation. Major recent developments in this area include the elucidation of mechanisms of action (MoAs) by which small molecules block NLRP3 inflammasome assembly and gasdermin D (GSDMD)-induced pyroptosis. We also discuss the status of clinical trials using agents that block specific components of the NLRP3 pathway, including their potential clinical applications for the treatment of many diseases.
- Published
- 2022
- Full Text
- View/download PDF
15. Response to Comment on 'Inflammasome inhibition prevents α-synuclein pathology and dopaminergic neurodegeneration in mice'
- Author
-
Eduardo A. Albornoz, Richard Gordon, Vinod Kumar, Avril A. B. Robertson, Kate Schroder, and Trent M. Woodruff
- Subjects
General Medicine - Abstract
We have replicated our original finding of elevated cleaved caspase-1 in mouse brains and neuroprotection by an NLRP3 inflammasome inhibitor in two mouse models of Parkinson’s disease.
- Published
- 2023
- Full Text
- View/download PDF
16. Macrophage ACE2 is necessary for SARS-CoV-2 replication and subsequent cytokine responses that restrict continued virion release
- Author
-
Larisa I. Labzin, Keng Yih Chew, Kathrin Eschke, Xiaohui Wang, Tyron Esposito, Claudia J. Stocks, James Rae, Ralph Patrick, Helen Mostafavi, Brittany Hill, Teodor E. Yordanov, Caroline L. Holley, Stefan Emming, Svenja Fritzlar, Francesca L. Mordant, Daniel P. Steinfort, Kanta Subbarao, Christian M. Nefzger, Anne K. Lagendijk, Emma J. Gordon, Robert G. Parton, Kirsty R. Short, Sarah L. Londrigan, and Kate Schroder
- Subjects
Cell Biology ,Molecular Biology ,Biochemistry - Abstract
Macrophages are key cellular contributors to the pathogenesis of COVID-19, the disease caused by the virus SARS-CoV-2. The SARS-CoV-2 entry receptor ACE2 is present only on a subset of macrophages at sites of SARS-CoV-2 infection in humans. Here, we investigated whether SARS-CoV-2 can enter macrophages, replicate, and release new viral progeny; whether macrophages need to sense a replicating virus to drive cytokine release; and, if so, whether ACE2 is involved in these mechanisms. We found that SARS-CoV-2 could enter, but did not replicate within, ACE2-deficient human primary macrophages and did not induce proinflammatory cytokine expression. By contrast, ACE2 overexpression in human THP-1–derived macrophages permitted SARS-CoV-2 entry, processing and replication, and virion release. ACE2-overexpressing THP-1 macrophages sensed active viral replication and triggered proinflammatory, antiviral programs mediated by the kinase TBK-1 that limited prolonged viral replication and release. These findings help elucidate the role of ACE2 and its absence in macrophage responses to SARS-CoV-2 infection.
- Published
- 2023
- Full Text
- View/download PDF
17. Inflammasomes and the IL-1 Family in Bone Homeostasis and Disease
- Author
-
Hsu-Wen Tseng, Selwin Gabriel Samuel, Kate Schroder, Jean-Pierre Lévesque, and Kylie A Alexander
- Subjects
Inflammasomes ,Endocrinology, Diabetes and Metabolism ,Interleukin-1beta ,NLR Family, Pyrin Domain-Containing 3 Protein ,Animals ,Homeostasis ,Humans ,Signal Transduction - Abstract
Purpose of ReviewInflammasomes are multimeric protein structures with crucial roles in host responses against infections and injuries. The importance of inflammasome activation goes beyond host defense as a dysregulated inflammasome and subsequent secretion of IL-1 family members is believed to be involved in the pathogenesis of various diseases, some of which also produce skeletal manifestations. The purpose of this review is to summarize recent developments in the understanding of inflammasome regulation and IL-1 family members in bone physiology and pathology and current therapeutics will be discussed.Recent FindingsSmall animal models have been vital to help understand how the inflammasome regulates bone dynamics. Animal models with gain or loss of function in various inflammasome components or IL-1 family signaling have illustrated how these systems can impact numerous bone pathologies and have been utilized to test new inflammasome therapeutics.SummaryIt is increasingly clear that a tightly regulated inflammasome is required not only for host defense but for skeletal homeostasis, as a dysregulated inflammasome is linked to diseases of pathological bone accrual and loss. Given the complexities of inflammasome activation and redundancies in IL-1 activation and secretion, targeting these pathways is at times challenging. Ongoing research into inflammasome-mediated mechanisms will allow the development of new therapeutics for inflammasome/IL-1 diseases.
- Published
- 2022
- Full Text
- View/download PDF
18. Inflammatory Caspases: Toward a Unified Model for Caspase Activation by Inflammasomes
- Author
-
Connie Ross, Amy H. Chan, Jessica B. von Pein, Madhavi P. Maddugoda, Dave Boucher, and Kate Schroder
- Subjects
Cell Death ,Inflammasomes ,Caspases ,Caspase 1 ,Immunology ,Pyroptosis ,Animals ,Humans ,Immunology and Allergy - Abstract
Inflammasomes are inflammatory signaling complexes that provide molecular platforms to activate the protease function of inflammatory caspases. Caspases-1, -4, -5, and -11 are inflammatory caspases activated by inflammasomes to drive lytic cell death and inflammatory mediator production, thereby activating host-protective and pathological immune responses. Here, we comprehensively review the mechanisms that govern the activity of inflammatory caspases. We discuss inflammatory caspase activation and deactivation mechanisms, alongside the physiological importance of caspase activity kinetics. We also examine mechanisms of caspase substrate selection and how inflammasome and cell identities influence caspase activity and resultant inflammatory and pyroptotic cellular programs. Understanding how inflammatory caspases are regulated may offer new strategies for treating infection and inflammasome-driven disease.
- Published
- 2022
- Full Text
- View/download PDF
19. Aim2 suppresses cigarette smoke‐induced neutrophil recruitment, neutrophil caspase‐1 activation and anti‐Ly6G‐mediated neutrophil depletion
- Author
-
Chantal Donovan, Richard Y Kim, Izabela Galvao, Andrew G Jarnicki, Alexandra C Brown, Bernadette Jones‐Freeman, Henry M Gomez, Ridhima Wadhwa, Elinor Hortle, Ranjith Jayaraman, Haroon Khan, Sophie Pickles, Priyanka Sahu, Vrushali Chimankar, Xiaofan Tu, Md Khadem Ali, Jemma R Mayall, Duc H Nguyen, Kurtis F Budden, Vinod Kumar, Kate Schroder, Avril AB Robertson, Matthew A Cooper, Peter AB Wark, Brian G Oliver, Jay C Horvat, and Philip M Hansbro
- Subjects
DNA-Binding Proteins ,Mice, Inbred C57BL ,Mice ,Neutrophil Infiltration ,Neutrophils ,Caspase 1 ,Immunology ,Animals ,0601 Biochemistry and Cell Biology, 1107 Immunology ,Immunology and Allergy ,Cell Biology ,Cigarette Smoking - Abstract
Increased inflammasome responses are strongly implicated in inflammatory diseases; however, their specific roles are incompletely understood. Therefore, we sought to examine the roles of nucleotide-binding oligomerization domain-like receptor (NLR) family, pyrin domain-containing 3 (NLRP3) and absent in melanoma-2 (AIM2) inflammasomes in cigarette smoke-induced inflammation in a model of experimental chronic obstructive pulmonary disease (COPD). We targeted NLRP3 with the inhibitor MCC950 given prophylactically or therapeutically and examined Aim2
- Published
- 2022
- Full Text
- View/download PDF
20. Apical extrusion prevents apoptosis from activating an acute inflammatory program in epithelia
- Author
-
Kinga Duszyc, Jessica B. von Pein, Divya Ramnath, Denni Currin-Ross, Suzie Verma, Fayth Lim, Matthew J. Sweet, Kate Schroder, and Alpha S. Yap
- Abstract
Apoptosis is traditionally considered to be an immunologically silent form of cell death. Multiple mechanisms exist to ensure that apoptosis does not stimulate the immune system to cause inflammation or autoimmunity. Against this expectation, we now report epithelia are programmed to provoke, rather than suppress, inflammation in response to apoptosis. We found that an acute inflammatory response led by neutrophils occurs in zebrafish and cell culture when apoptotic epithelial cells cannot be expelled from the monolayer by apical extrusion. This reflects an intrinsic circuit where ATP released from apoptotic cells stimulates epithelial cells in the immediate vicinity to produce IL-8. As the epithelial barrier is compromised when apical extrusion fails, this juxta-apoptotic proinflammatory pathway may represent an early-response mechanism at sites of potential microbial ingress. Conversely, apical extrusion prevents inappropriate epithelial inflammation by physically eliminating apoptotic cells before they can activate this proinflammatory circuit.
- Published
- 2023
- Full Text
- View/download PDF
21. HDAC7 is an immunometabolic switch triaging danger signals for engagement of antimicrobial versus inflammatory responses in macrophages
- Author
-
Kaustav Das Gupta, Divya Ramnath, Jessica B. von Pein, James E. B. Curson, Yizhuo Wang, Rishika Abrol, Asha Kakkanat, Shayli Varasteh Moradi, Kimberley S. Gunther, Ambika M. V. Murthy, Claudia J. Stocks, Ronan Kapetanovic, Robert C. Reid, Abishek Iyer, Zoe C. Ilka, William M. Nauseef, Manuel Plan, Lin Luo, Jennifer L. Stow, Kate Schroder, Denuja Karunakaran, Kirill Alexandrov, Melanie R. Shakespear, Mark A. Schembri, David P. Fairlie, and Matthew J. Sweet
- Subjects
Multidisciplinary - Abstract
The immune system must be able to respond to a myriad of different threats, each requiring a distinct type of response. Here, we demonstrate that the cytoplasmic lysine deacetylase HDAC7 in macrophages is a metabolic switch that triages danger signals to enable the most appropriate immune response. Lipopolysaccharide (LPS) and soluble signals indicating distal or far-away danger trigger HDAC7-dependent glycolysis and proinflammatory IL-1β production. In contrast, HDAC7 initiates the pentose phosphate pathway (PPP) for NADPH and reactive oxygen species (ROS) production in response to the more proximal threat of nearby bacteria, as exemplified by studies on uropathogenic Escherichia coli (UPEC). HDAC7-mediated PPP engagement via 6-phosphogluconate dehydrogenase (6PGD) generates NADPH for antimicrobial ROS production, as well as D-ribulose-5-phosphate (RL5P) that both synergizes with ROS for UPEC killing and suppresses selective inflammatory responses. This dual functionality of the HDAC7-6PGD-RL5P axis prioritizes responses to proximal threats. Our findings thus reveal that the PPP metabolite RL5P has both antimicrobial and immunomodulatory activities and that engagement of enzymes in catabolic versus anabolic metabolic pathways triages responses to different types of danger for generation of inflammatory versus antimicrobial responses, respectively.
- Published
- 2023
- Full Text
- View/download PDF
22. Caspase-4 dimerisation and D289 auto-processing elicits an interleukin-1β converting enzyme
- Author
-
Amy H. Chan, Kassandra Vezyrgiannis, Jessica B. Von Pein, Xiaohui Wang, Larisa I Labzin, Dave Boucher, and Kate Schroder
- Abstract
The non-canonical inflammasome is a signalling complex critical for cell defence against cytosolic Gram-negative bacteria. A key step in the human non-canonical inflammasome pathway involves unleashing the proteolytic activity of caspase-4 within this complex. Caspase-4 induces inflammatory responses by cleaving gasdermin-D (GSDMD) to initiate pyroptosis, although the molecular mechanisms that activate caspase-4 and govern its capacity to cleave substrates are poorly defined. Caspase-11, the murine counterpart of caspase-4, acquires protease activity within the non-canonical inflammasome by forming a dimer that self-cleaves at D285 to directly cleave GSDMD. These cleavage events trigger signalling via the NLRP3-ASC-caspase-1 axis, leading to downstream cleavage of the pro-interleukin-1β (pro-IL-1β) cytokine precursor. Here, we show that caspase-4 first dimerises then self-cleaves at two sites – D270 and D289 – in the interdomain linker to acquire full proteolytic activity, cleave GSDMD and induce cell death. Surprisingly, caspase-4 dimerisation and self-cleavage at D289 generates a caspase-4 p34/p9 protease species that directly cleaves pro-IL-1β, resulting in its maturation and secretion independently of the NLRP3 inflammasome in primary human myeloid and epithelial cells. Our study thus elucidates the key molecular events that underpin signalling by the caspase-4 inflammasome, and identifies IL-1β as a natural substrate of caspase-4.
- Published
- 2023
- Full Text
- View/download PDF
23. Interleukin‐1 Is Overexpressed in Injured Muscles Following Spinal Cord Injury and Promotes Neurogenic Heterotopic Ossification
- Author
-
Irina Kulina, Kylie A. Alexander, Beulah Jose, Allison R. Pettit, Jean-Pierre Levesque, Kate Schroder, Cedryck Vaquette, Gethin P. Thomas, François Genêt, Jules Gueguen, Whitney Fleming, Dorothée Girard, Marjorie Salga, Susan M. Millard, Lawrie Wheeler, Hsu-Wen Tseng, and Sébastien Banzet
- Subjects
Pathology ,medicine.medical_specialty ,Endocrinology, Diabetes and Metabolism ,Interleukin-1beta ,Inflammation ,Pathogenesis ,Mice ,medicine ,Animals ,Humans ,Orthopedics and Sports Medicine ,Spinal cord injury ,Spinal Cord Injuries ,CD68 ,business.industry ,Muscles ,Ossification, Heterotopic ,Interleukin ,Spinal cord ,medicine.disease ,medicine.anatomical_structure ,Spinal Cord ,Tumor necrosis factor alpha ,medicine.symptom ,Interleukin 1 receptor, type I ,business ,Interleukin-1 - Abstract
Neurogenic heterotopic ossifications (NHOs) form in periarticular muscles following severe spinal cord (SCI) and traumatic brain injuries. The pathogenesis of NHO is poorly understood with no effective preventive treatment. The only curative treatment remains surgical resection of pathological NHOs. In a mouse model of SCI-induced NHO that involves a transection of the spinal cord combined with a muscle injury, a differential gene expression analysis revealed that genes involved in inflammation such as interleukin-1β (IL-1β) were overexpressed in muscles developing NHO. Using mice knocked-out for the gene encoding IL-1 receptor (IL1R1) and neutralizing antibodies for IL-1α and IL-1β, we show that IL-1 signaling contributes to NHO development following SCI in mice. Interestingly, other proteins involved in inflammation that were also overexpressed in muscles developing NHO, such as colony-stimulating factor-1, tumor necrosis factor or C-C chemokine ligand-2 did not promote NHO development. Finally using NHO biopsies from SCI and TBI patients, we show that IL-1β is expressed by CD68+ macrophages. IL-1α and IL-1β produced by activated human monocytes promote calcium mineralization and RUNX2 expression in fibro-adipogenic progenitors isolated from muscles surrounding NHOs. Altogether these data suggest that interleukin-1 promotes NHO development in both humans and mice. This article is protected by copyright. All rights reserved.
- Published
- 2021
- Full Text
- View/download PDF
24. Parkinson's disease: connecting mitochondria to inflammasomes
- Author
-
Grace M.E.P. Lawrence, Caroline L. Holley, and Kate Schroder
- Subjects
Mammals ,Inflammasomes ,Dopaminergic Neurons ,Immunology ,NLR Family, Pyrin Domain-Containing 3 Protein ,Immunology and Allergy ,Animals ,Humans ,Parkinson Disease ,Microglia ,Mitochondria - Abstract
Activated microglia foster a neurotoxic, inflammatory environment in the mammalian central nervous system (CNS) that drives the pathology of neurodegenerative diseases including Parkinson's disease (PD). Moreover, mitochondrial fission promotes microglial inflammatory responses in vitro. Given that the NLRP3 inflammasome and mitochondria are central regulators of both inflammation and PD, we explore potential functions for the NLRP3 inflammasome and mitochondrial dynamics in PD. Specifically, we propose that inducible microglial mitochondrial fission can promote NLRP3-dependent neuroinflammation in hereditary and idiopathic PD. Further in-depth exploration of this topic can prompt valuable discoveries of the underlying molecular mechanisms of PD neuroinflammation, identify novel candidate anti-inflammatory therapeutics for PD, and ideally provide better outcomes for PD patients.
- Published
- 2022
25. Come on mtDNA, light my fire
- Author
-
Grace M.E.P. Lawrence, Caroline L. Holley, and Kate Schroder
- Subjects
Infectious Diseases ,Inflammasomes ,Immunology ,NLR Family, Pyrin Domain-Containing 3 Protein ,Immunology and Allergy ,DNA, Mitochondrial ,Mitochondria ,Signal Transduction - Abstract
Oxidized mitochondrial DNA (ox-mtDNA) activates NLRP3 inflammasome signaling through an ill-defined mechanism. In this issue of Immunity, Xian et al. reveal FEN1 endonuclease cleaves ox-mtDNA into fragments that escape mitochondria, igniting NLRP3 and cGAS-STING signaling and inflammation.
- Published
- 2022
26. The NLRP3 inflammasome triggers sterile neuroinflammation and Alzheimer’s disease
- Author
-
Madhavi P. Maddugoda, Kate Schroder, Sabrina Sofia Burgener, Jürgen Götz, and Mark T Milner
- Subjects
0301 basic medicine ,Inflammasomes ,Immunology ,Inflammation ,Pathogenesis ,03 medical and health sciences ,0302 clinical medicine ,Alzheimer Disease ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Humans ,Immunology and Allergy ,Serum amyloid A ,Cognitive decline ,Neuroinflammation ,integumentary system ,business.industry ,Inflammasome ,medicine.disease ,030104 developmental biology ,Tumor necrosis factor alpha ,medicine.symptom ,business ,Dysbiosis ,Neuroscience ,030215 immunology ,medicine.drug - Abstract
To maintain homeostasis, an organism must detect and resolve sterile tissue damage. The NLRP3 inflammasome coordinates such processes to clear tissue damage and induce repair. Dysregulated NLRP3 inflammasome activity, however, drives many conditions including Alzheimer's disease (AD). Recent reports posit that β-amyloid and tau aggregates trigger destructive NLRP3 inflammasome signalling in the brain, leading to AD pathophysiology and cognitive decline. Other endogenous molecules (e.g. TNF, ATP, serum amyloid A), as well as dysbiosis, can induce peripheral or central inflammation and thereby promote microglial NLRP3 inflammasome signalling and resultant AD. The NLRP3 inflammasome is thus emerging as a critical driver of sterile neuroinflammation and the resultant pathogenesis and progression of AD.
- Published
- 2021
- Full Text
- View/download PDF
27. Update of the FANTOM web resource: from mammalian transcriptional landscape to its dynamic regulation.
- Author
-
Hideya Kawaji, Jessica Severin, Marina Lizio, Alistair R. R. Forrest, Erik van Nimwegen, Michael Rehli, Kate Schroder, Katharine M. Irvine, Harukazu Suzuki, Piero Carninci, Yoshihide Hayashizaki, and Carsten O. Daub
- Published
- 2011
- Full Text
- View/download PDF
28. Neutrophil-Derived S100A8/A9 Amplify Granulopoiesis After Myocardial Infarction
- Author
-
Ashish Dhyani, Hind Lal, Gerard Pernes, Chander Raman, Annas Al-Sharea, Ahmed Abdel-Latif, Rahul Annabathula, James E. Hudson, Gopalkrishna Sreejit, Beatriz Y Hanaoka, Sunil K. Noothi, Kate Schroder, Prabhakara R Nagareddy, Gregory A. Quaife-Ryan, Enzo R. Porrello, Maria B. Grant, Dragana Dragoljevic, Susan S. Smyth, Baskaran Athmanathan, and Andrew J. Murphy
- Subjects
0303 health sciences ,Myocardial ischemia ,business.industry ,Inflammation ,Inflammasome ,030204 cardiovascular system & hematology ,medicine.disease ,Granulopoiesis ,Calgranulin A ,03 medical and health sciences ,0302 clinical medicine ,Physiology (medical) ,Immunology ,medicine ,Myelopoiesis ,Myocardial infarction ,medicine.symptom ,Cardiology and Cardiovascular Medicine ,S100a8 a9 ,business ,030304 developmental biology ,medicine.drug - Abstract
Background:Myocardial infarction (MI) triggers myelopoiesis, resulting in heightened production of neutrophils. However, the mechanisms that sustain their production and recruitment to the injured heart are unclear.Methods:Using a mouse model of the permanent ligation of the left anterior descending artery and flow cytometry, we first characterized the temporal and spatial effects of MI on different myeloid cell types. We next performed global transcriptome analysis of different cardiac cell types within the infarct to identify the drivers of the acute inflammatory response and the underlying signaling pathways. Using a combination of genetic and pharmacological strategies, we identified the sequelae of events that led to MI-induced myelopoiesis. Cardiac function was assessed by echocardiography. The association of early indexes of neutrophilia with major adverse cardiovascular events was studied in a cohort of patients with acute MI.Results:Induction of MI results in rapid recruitment of neutrophils to the infarct, where they release specific alarmins, S100A8 and S100A9. These alarmins bind to the Toll-like receptor 4 and prime the nod-like receptor family pyrin domain-containing 3 inflammasome in naïve neutrophils and promote interleukin-1β secretion. The released interleukin-1β interacts with its receptor (interleukin 1 receptor type 1) on hematopoietic stem and progenitor cells in the bone marrow and stimulates granulopoiesis in a cell-autonomous manner. Genetic or pharmacological strategies aimed at disruption of S100A8/A9 and their downstream signaling cascade suppress MI-induced granulopoiesis and improve cardiac function. Furthermore, in patients with acute coronary syndrome, higher neutrophil count on admission and after revascularization correlates positively with major adverse cardiovascular disease outcomes.Conclusions:Our study provides novel evidence for the primary role of neutrophil-derived alarmins (S100A8/A9) in dictating the nature of the ensuing inflammatory response after myocardial injury. Therapeutic strategies aimed at disruption of S100A8/A9 signaling or their downstream mediators (eg, nod-like receptor family pyrin domain-containing 3 inflammasome, interleukin-1β) in neutrophils suppress granulopoiesis and may improve cardiac function in patients with acute coronary syndrome.
- Published
- 2020
- Full Text
- View/download PDF
29. Macrophages only sense infectious SARS-CoV-2 when they express sufficient ACE2 to permit viral entry, where rapid cytokine responses then limit viral replication
- Author
-
Larisa I Labzin, Keng Yih Chew, Kathrin Eschke, Xiaohui Wang, Tyron Esposito, Claudia J Stocks, James Rae, Ralph Patrick, Helen Mostafavi, Brittany Hill, Teodor E. Yordanov, Caroline L Holley, Stefan Emming, Svenja Fritzlar, Francesca L. Mordant, Daniel P. Steinfort, Kanta Subbarao, Christian M. Nefzger, Anne K Lagendijk, Emma Gordon, Robert Parton, Kirsty R. Short, Sarah L. Londrigan, and Kate Schroder
- Abstract
Macrophages are key cellular contributors to COVID-19 pathogenesis. Whether SARS-CoV-2 can enter macrophages, replicate and release new viral progeny remains controversial. Similarly, whether macrophages need to sense replicating virus to drive cytokine release is also unclear. Macrophages are heterogeneous cells poised to respond to their local microenvironment, and accordingly, the SARS-CoV-2 entry receptor ACE2 is only present on a subset of macrophages at sites of human infection. Here, we use in vitro approaches to investigate how SARS-CoV-2 interacts with ACE2-negative and ACE2-positive human macrophages and determine how these macrophage populations sense and respond to SARS-CoV-2. We show that SARS-CoV-2 does not replicate within ACE2-negative human macrophages and does not induce pro-inflammatory cytokine expression. By contrast, ACE2 expression in human macrophages permits SARS-CoV-2 entry, replication, and virion release. ACE2-expressing macrophages sense replicating virus to trigger pro-inflammatory and anti-viral programs that limit virus release. These combined findings resolve several controversies regarding macrophage-SARS-CoV-2 interactions and identify a signaling circuit by which macrophages sense SARS-CoV-2 cell entry and respond by restricting viral replication.One sentence summaryLack of macrophage ACE2 expression precludes SARS-CoV-2 entry and sensing, while ACE2-expressing macrophages sense intramacrophage SARS-CoV-2 replication to induce rapid anti-viral responses that limit new virion release.
- Published
- 2022
- Full Text
- View/download PDF
30. Caging NLRP3 tames inflammasome activity
- Author
-
Rebecca Coll and Kate Schroder
- Subjects
Cytosol ,Inflammasomes ,Cryoelectron Microscopy ,NLR Family, Pyrin Domain-Containing 3 Protein ,General Biochemistry, Genetics and Molecular Biology - Abstract
How the danger sensor NLRP3 is activated is intensively debated. Using cryo-electron microscopy (EM) approaches, Andreeva and colleagues made the remarkable discovery that inactive NLRP3 forms a double ring of 12-16 monomers that shield its pyrin domains from the cytosol. We discuss this surprising new mechanism of inflammasome regulation.
- Published
- 2021
- Full Text
- View/download PDF
31. Author response for 'Interleukin‐1 is overexpressed in injured muscles following spinal cord injury and promotes neurogenic heterotopic ossification'
- Author
-
François Genêt, Beulah Jose, Kate Schroder, Whitney Fleming, Hsu-Wen Tseng, Sébastien Banzet, Gethin P. Thomas, Kylie A. Alexander, Jean-Pierre Levesque, Irina Kulina, Cedryck Vaquette, Dorothée Girard, Lawrie Wheeler, Jules Gueguen, Allison R. Pettit, Susan M. Millard, and Marjorie Salga
- Subjects
Pathology ,medicine.medical_specialty ,business.industry ,Neurogenic heterotopic ossification ,Medicine ,Interleukin ,business ,medicine.disease ,Spinal cord injury - Published
- 2021
- Full Text
- View/download PDF
32. Streptolysins are the primary inflammasome activators in macrophages during Streptococcus pyogenes infection
- Author
-
Timothy C. Barnett, Stephan Brouwer, Mercedes Monteleone, Victor Nizet, Mark J. Walker, Amanda J. Cork, Kate Schroder, and Johanna Richter
- Subjects
Inflammasomes ,Streptococcus pyogenes ,Immunology ,Interleukin-1beta ,medicine.disease_cause ,Pyrin domain ,Proinflammatory cytokine ,03 medical and health sciences ,Mice ,Streptococcal Infections ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Immunology and Allergy ,Animals ,Secretion ,030304 developmental biology ,0303 health sciences ,030306 microbiology ,Chemistry ,Macrophages ,Inflammasome ,Cell Biology ,3. Good health ,Cell biology ,Streptolysins ,Streptolysin ,Lipoteichoic acid ,Inflammasome complex ,medicine.drug - Abstract
Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes an array of infectious diseases in humans. Accumulating clinical evidence suggests that proinflammatory interleukin (IL)-1β signaling plays an important role in GAS disease progression. The host regulates the production and secretion of IL-1β via the cytosolic inflammasome pathway. Activation of the NLR family pyrin domain-containing 3 (NLRP3) inflammasome complex requires two signals: a priming signal that stimulates increased transcription of genes encoding the components of the inflammasome pathway, and an activating signal that induces assembly of the inflammasome complex. Here we show that GAS-derived lipoteichoic acid can provide a priming signal for NLRP3 inflammasome activation. As only few GAS-derived proteins have been associated with inflammasome-dependent IL-1β signaling, we investigated novel candidates that might play a role in activating the inflammasome pathway by infecting mouse bone marrow-derived macrophages and human THP-1 macrophage-like cells with a panel of isogenic GAS mutant strains. We found that the cytolysins streptolysin O (SLO) and streptolysin S are the main drivers of IL-1β release in proliferating logarithmic phase GAS. Using a mutant form of recombinant SLO, we confirmed that bacterial pore formation on host cell membranes is a key mechanism required for inflammasome activation. Our results suggest that streptolysins are major determinants of GAS-induced inflammation and present an attractive target for therapeutic intervention.
- Published
- 2021
33. The microglial NLRP3 inflammasome is activated by amyotrophic lateral sclerosis proteins
- Author
-
Vandana Deora, Trent M. Woodruff, Richard D. Gordon, Eduardo A. Albornoz, Cyril J. Jagaraj, John D. Lee, Avril A. B. Robertson, Justin J. Yerbury, Julie D. Atkin, Kate Schroder, Mark E. Cooper, and Luke McAlary
- Subjects
0301 basic medicine ,Inflammasomes ,Mice, Transgenic ,Biology ,Protein aggregation ,03 medical and health sciences ,Cellular and Molecular Neuroscience ,Superoxide Dismutase-1 ,0302 clinical medicine ,Downregulation and upregulation ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Humans ,Secretion ,Neuroinflammation ,integumentary system ,Microglia ,Amyotrophic Lateral Sclerosis ,Neurodegeneration ,Inflammasome ,medicine.disease ,Cell biology ,Disease Models, Animal ,030104 developmental biology ,medicine.anatomical_structure ,Neurology ,Disease Progression ,Inflammasome complex ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Microglial NLRP3 inflammasome activation is emerging as a key contributor to neuroinflammation during neurodegeneration. Pathogenic protein aggregates such as β-amyloid and α-synuclein trigger microglial NLRP3 activation, leading to caspase-1 activation and IL-1β secretion. Both caspase-1 and IL-1β contribute to disease progression in the mouse SOD1G93A model of amyotrophic lateral sclerosis (ALS), suggesting a role for microglial NLRP3. Prior studies, however, suggested SOD1G93A mice microglia do not express NLRP3, and SOD1G93A protein generated IL-1β in microglia independent to NLRP3. Here, we demonstrate using Nlrp3-GFP gene knock-in mice that microglia express NLRP3 in SOD1G93A mice. We show that both aggregated and soluble SOD1G93A activates inflammasome in primary mouse microglia leading caspase-1 and IL-1β cleavage, ASC speck formation, and the secretion of IL-1β in a dose- and time-dependent manner. Importantly, SOD1G93A was unable to induce IL-1β secretion from microglia deficient for Nlrp3, or pretreated with the specific NLRP3 inhibitor MCC950, confirming NLRP3 as the key inflammasome complex mediating SOD1-induced microglial IL-1β secretion. Microglial NLRP3 upregulation was also observed in the TDP-43Q331K ALS mouse model, and TDP-43 wild-type and mutant proteins could also activate microglial inflammasomes in a NLRP3-dependent manner. Mechanistically, we identified the generation of reactive oxygen species and ATP as key events required for SOD1G93A -mediated NLRP3 activation. Taken together, our data demonstrate that ALS microglia express NLRP3, and that pathological ALS proteins activate the microglial NLRP3 inflammasome. NLRP3 inhibition may therefore be a potential therapeutic approach to arrest microglial neuroinflammation and ALS disease progression.
- Published
- 2019
- Full Text
- View/download PDF
34. Inhibitors of class I histone deacetylases attenuate thioacetamide‐induced liver fibrosis in mice by suppressing hepatic type 2 inflammation
- Author
-
Matthew J. Sweet, Abishek Iyer, David P. Fairlie, Elizabeth E. Powell, Zhixuan Loh, Robert Reid, Rebecca L. Fitzsimmons, Divya Ramnath, Jennifer L. Stow, Kate Schroder, Katharine M. Irvine, Andrew D. Clouston, and Praveer Gupta
- Subjects
Liver Cirrhosis ,0301 basic medicine ,Cell ,Inflammation ,Thioacetamide ,Ligands ,Histone Deacetylases ,Mice ,03 medical and health sciences ,chemistry.chemical_compound ,0302 clinical medicine ,Immune system ,Fibrosis ,medicine ,Animals ,Pharmacology ,business.industry ,Anti-Inflammatory Agents, Non-Steroidal ,Innate lymphoid cell ,medicine.disease ,Research Papers ,Histone Deacetylase Inhibitors ,Mice, Inbred C57BL ,030104 developmental biology ,medicine.anatomical_structure ,chemistry ,Hepatocyte ,Hepatic stellate cell ,Cancer research ,Female ,medicine.symptom ,business ,030217 neurology & neurosurgery - Abstract
BACKGROUND AND PURPOSE: Chronic liver diseases feature excessive collagen and matrix protein deposition or crosslinking that characterises fibrosis, leads to scar tissue, and disrupts liver functions. There is no effective treatment. This study investigated whether treatment with selective histone deacetylase (HDAC) inhibitors might specifically reduce type 2 inflammation in the injured liver, thereby attenuating fibrogenesis in mice. EXPERIMENTAL APPROACH: Thioacetamide (TAA) was used to induce hepatic inflammation, fibrosis, and liver damage in female C57BL/6 mice, similar to the clinical features of chronic human liver disease. We used eight inhibitors of different human HDAC enzymes to probe histological (IHC and TUNEL), biochemical and immunological changes (flow cytometry, qPCR, Legendplex, and elisa) in pathology, fibrosis, hepatic immune cell flux, and inflammatory cytokine expression. KEY RESULTS: Inhibitors of class I, but not class II, HDAC enzymes potently suppressed chronic hepatic inflammation and fibrosis in mice, attenuating accumulation and activation of IL‐33‐dependent, but not IL‐25‐dependent, group 2 innate lymphoid cells (ILC2) and inhibiting type 2 inflammation that drives hepatic stellate cells to secrete excessive collagen and matrix proteins. CONCLUSIONS AND IMPLICATIONS: The results show that potent and selective inhibitors of class I only HDAC enzymes profoundly inhibit hepatocyte death and type 2 inflammation to prevent TAA‐induced liver fibrosis in mice. The specific HDAC enzymes identified here may be key promoters of inflammation in chronic liver fibrosis.
- Published
- 2019
- Full Text
- View/download PDF
35. MCC950 directly targets the NLRP3 ATP-hydrolysis motif for inflammasome inhibition
- Author
-
Avril A. B. Robertson, James A. Fraser, Michael P. Jennings, Jessica L. Chitty, Kate Schroder, James R. Hill, Rebecca C. Coll, Dave Boucher, Christopher J. Day, Alina Zamoshnikova, and Nicholas L. Massey
- Subjects
0303 health sciences ,integumentary system ,Chemistry ,030302 biochemistry & molecular biology ,Inflammasome ,Cell Biology ,Cell biology ,03 medical and health sciences ,ATP hydrolysis ,NACHT domain ,Molecular targets ,medicine ,Molecular Biology ,030304 developmental biology ,medicine.drug - Abstract
Inhibition of the NLRP3 inflammasome is a promising strategy for the development of new treatments for inflammatory diseases. MCC950 is a potent and specific small-molecule inhibitor of the NLRP3 pathway, but its molecular target is not defined. Here, we show that MCC950 directly interacts with the Walker B motif within the NLRP3 NACHT domain, thereby blocking ATP hydrolysis and inhibiting NLRP3 activation and inflammasome formation.
- Published
- 2019
- Full Text
- View/download PDF
36. Inflammasome activation and <scp>IL</scp> ‐1β signalling in group A Streptococcus disease
- Author
-
Mark J. Walker, Kate Schroder, Johanna Richter, and Stephan Brouwer
- Subjects
Inflammasomes ,Streptococcus pyogenes ,Interleukin-1beta ,Immunology ,Inflammasome ,Inflammation ,Disease ,Biology ,medicine.disease_cause ,Microbiology ,Group A ,Signalling ,Streptococcal Infections ,Virology ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Humans ,Secretion ,medicine.symptom ,Pathogen ,Signal Transduction ,medicine.drug - Abstract
Group A Streptococcus (GAS) is a Gram-positive bacterial pathogen that causes significant morbidity and mortality worldwide. Recent clinical evidence suggests that the inflammatory marker interleukin-1β (IL-1β) plays an important role in GAS disease progression, and presents a potential target for therapeutic intervention. Interaction with GAS activates the host inflammasome pathway to stimulate production and secretion of IL-1β, but GAS can also stimulate IL-1β production in an inflammasome-independent manner. This review highlights progress that has been made in understanding the importance of host cell inflammasomes and IL-1 signalling in GAS disease, and explores challenges and unsolved problems in this host-pathogen interaction. TAKE AWAY: Inflammasome signalling during GAS infection is an emerging field of research. GAS modulates the NLRP3 inflammasome pathway through multiple mechanisms. SpeB contributes to IL-1β production independently of the inflammasome pathway. IL-1β signalling can be host-protective, but also drive severe GAS disease.
- Published
- 2021
- Full Text
- View/download PDF
37. Vincristine-induced peripheral neuropathy is driven by canonical NLRP3 activation and IL-1β release
- Author
-
Alexander Mueller, Alexandra V. Smith, Lena Batoon, Mercedes Monteleone, Kate Schroder, Cheyenne J. Sandrock, Jennifer R. Deuis, Grace Pamo Lawrence, Brandon J. Wainwright, Elissa Tolson, Amanda Mayor, Evelyn Israel Nadar, Irina Vetter, Christelle Adolphe, Jean-Pierre Levesque, Bryan Tay, Hana Starobova, and Allison R. Pettit
- Subjects
0301 basic medicine ,Chemotherapy ,Anakinra ,Vincristine ,business.industry ,medicine.medical_treatment ,Immunology ,Inflammasome ,medicine.disease ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Allodynia ,Peripheral neuropathy ,Tumor progression ,Knockout mouse ,medicine ,Cancer research ,Immunology and Allergy ,medicine.symptom ,business ,030217 neurology & neurosurgery ,medicine.drug - Abstract
Vincristine is an important component of many regimens used for pediatric and adult malignancies, but it causes a dose-limiting sensorimotor neuropathy for which there is no effective treatment. This study aimed to delineate the neuro-inflammatory mechanisms contributing to the development of mechanical allodynia and gait disturbances in a murine model of vincristine-induced neuropathy, as well as to identify novel treatment approaches. Here, we show that vincristine-induced peripheral neuropathy is driven by activation of the NLRP3 inflammasome and subsequent release of interleukin-1β from macrophages, with mechanical allodynia and gait disturbances significantly reduced in knockout mice lacking NLRP3 signaling pathway components, or after treatment with the NLRP3 inhibitor MCC950. Moreover, treatment with the IL-1 receptor antagonist anakinra prevented the development of vincristine-induced neuropathy without adversely affecting chemotherapy efficacy or tumor progression in patient-derived medulloblastoma xenograph models. These results detail the neuro-inflammatory mechanisms leading to vincristine-induced peripheral neuropathy and suggest that repurposing anakinra may be an effective co-treatment strategy to prevent vincristine-induced peripheral neuropathy.
- Published
- 2021
- Full Text
- View/download PDF
38. Endothelial cells elicit a pro-inflammatory response to SARS-CoV-2 without productive viral infection
- Author
-
Keng Yih Chew, Emma Gordon, Claudia J. Stocks, Essebier T, Kate Schroder, Lilian Schimmel, Teodor E. Yordanov, James Monkman, Anne K. Lagendijk, Arutha Kulasinghe, dos Santos Miggiolaro Afr, Kirsty R. Short, de Noronha L, and Larisa I. Labzin
- Subjects
Endothelium ,business.industry ,Cell ,Inflammation ,Endothelial activation ,Pathogenesis ,medicine.anatomical_structure ,In vivo ,Immunology ,medicine ,medicine.symptom ,business ,Receptor ,Viral load - Abstract
ObjectivesThrombotic and microvascular complications are frequently seen in deceased COVID-19 patients. However, whether this is caused by direct viral infection of the endothelium or inflammation-induced endothelial activation remains highly contentious.MethodsHere, we use patient autopsy samples, primary human endothelial cells and an in vitro model of the pulmonary epithelial-endothelial cell barrier to show that primary human endothelial cells express very low levels the SARS-CoV-2 receptor ACE2 and the protease TMPRSS2.ResultsAccordingly, endothelial cells can only be infected when SARS-CoV-2 is present at very high concentrations. However, this is not a productive infection (i.e. no infectious virus is produced) and viral entry induces an inflammatory response. We also show that SARS-CoV-2 does not infect endothelial cells in 3D vessels under flow conditions. We further demonstrate that in a co-culture model endothelial cells are not infected with SARS-CoV-2. They do however sense and respond to infection in the adjacent epithelial cells, increasing ICAM-1 expression and releasing pro-inflammatory cytokines.ConclusionsTaken together, these data suggest that in vivo, endothelial cells are unlikely to be infected with SARS-CoV-2 and that infection is only likely to occur if the adjacent pulmonary epithelium is denuded (basolateral infection) or a high viral load is present in the blood (apical infection). In such a scenario, whilst SARS-CoV-2 infection of the endothelium can occur, it does not contribute to viral amplification. However, endothelial cells are still likely to play a key role in SARS-CoV-2 pathogenesis by sensing adjacent infection and mounting a pro-inflammatory response to SARS-CoV-2.
- Published
- 2021
- Full Text
- View/download PDF
39. Endothelial cells are not productively infected by SARS‐CoV‐2
- Author
-
Arutha Kulasinghe, Anna Flavia Ribeiro dos Santos Miggiolaro, Keng Yih Chew, Lilian Schimmel, Patricia Essebier, Claudia J. Stocks, Kate Schroder, Emma Gordon, James Monkman, Kirsty R. Short, Teodor E. Yordanov, Larisa I. Labzin, Caroline Cooper, Lucia de Noronha, Anne K. Lagendijk, Graduate School, ACS - Atherosclerosis & ischemic syndromes, and AII - Infectious diseases
- Subjects
Endothelium ,viruses ,Immunology ,Cell ,Inflammation ,030204 cardiovascular system & hematology ,Biology ,SARS‐CoV‐2 ,Pathogenesis ,Endothelial activation ,blood vessels ,03 medical and health sciences ,0302 clinical medicine ,In vivo ,COVID‐19 ,medicine ,Immunology and Allergy ,Receptor ,General Nursing ,030304 developmental biology ,0303 health sciences ,SARS-CoV-2 ,fungi ,COVID-19 ,Original Articles ,RC581-607 ,endothelial cells ,3. Good health ,medicine.anatomical_structure ,inflammation ,Original Article ,medicine.symptom ,Immunologic diseases. Allergy ,Viral load - Abstract
Objectives Thrombotic and microvascular complications are frequently seen in deceased COVID‐19 patients. However, whether this is caused by direct viral infection of the endothelium or inflammation‐induced endothelial activation remains highly contentious. Methods Here, we use patient autopsy samples, primary human endothelial cells and an in vitro model of the pulmonary epithelial–endothelial cell barrier. Results We show that primary human endothelial cells express very low levels of the SARS‐CoV‐2 receptor ACE2 and the protease TMPRSS2, which blocks their capacity for productive viral infection, and limits their capacity to produce infectious virus. Accordingly, endothelial cells can only be infected when they overexpress ACE2, or are exposed to very high concentrations of SARS‐CoV‐2. We also show that SARS‐CoV‐2 does not infect endothelial cells in 3D vessels under flow conditions. We further demonstrate that in a co‐culture model endothelial cells are not infected with SARS‐CoV‐2. Endothelial cells do however sense and respond to infection in the adjacent epithelial cells, increasing ICAM‐1 expression and releasing pro‐inflammatory cytokines. Conclusions Taken together, these data suggest that in vivo, endothelial cells are unlikely to be infected with SARS‐CoV‐2 and that infection may only occur if the adjacent pulmonary epithelium is denuded (basolateral infection) or a high viral load is present in the blood (apical infection). In such a scenario, whilst SARS‐CoV‐2 infection of the endothelium can occur, it does not contribute to viral amplification. However, endothelial cells may still play a key role in SARS‐CoV‐2 pathogenesis by sensing adjacent infection and mounting a pro‐inflammatory response to SARS‐CoV‐2., We show that endothelial cells are not productively infected with SARS‐CoV‐2 but mount a pro‐inflammatory response to the virus.
- Published
- 2021
40. Data-driven normalization strategies for high-throughput quantitative RT-PCR.
- Author
-
Jessica Cara Mar, Yasumasa Kimura, Kate Schroder, Katharine M. Irvine, Yoshihide Hayashizaki, Harukazu Suzuki, David A. Hume, and John Quackenbush
- Published
- 2009
- Full Text
- View/download PDF
41. Placental inflammasome signaling: Protection for mother and baby
- Author
-
Kate Schroder and Sabrina Sofia Burgener
- Subjects
0301 basic medicine ,Inflammasomes ,Placenta ,Innate Immunity and Inflammation ,Immunology ,Mothers ,Article ,Infectious Disease and Host Defense ,03 medical and health sciences ,Fetus ,0302 clinical medicine ,Pregnancy ,Second trimester ,Humans ,Immunology and Allergy ,Medicine ,reproductive and urinary physiology ,business.industry ,Inflammasome ,medicine.disease ,Trophoblasts ,030104 developmental biology ,medicine.anatomical_structure ,embryonic structures ,Female ,Signal transduction ,business ,030215 immunology ,medicine.drug - Abstract
Immune profiling of secreted factors from human placentas across gestation identifies inflammasome hyperactivation in placental trophoblasts resulting in constitutive IL-1β and IL-18 secretion. Trophoblast-secreted IL-1β primes circulating monocytes for inflammasome activation and is important for control of placental Listeria monocytogenes infection., The human placenta is a dynamic organ that modulates physiological adaptations to pregnancy. To define the immunological signature of the human placenta, we performed unbiased profiling of secreted immune factors from human chorionic villi isolated from placentas at mid and late stages of pregnancy. We show that placental trophoblasts constitutively secrete the inflammasome-associated cytokines IL-1β and IL-18, which is blocked by NLRP3 inflammasome inhibitors and occurs without detectable gasdermin D cleavage. We further show that placenta-derived IL-1β primes monocytes for inflammasome induction to protect against Listeria monocytogenes infection. Last, we show that the human placenta responds to L. monocytogenes infection through additional inflammasome activation and that inhibition of this pathway sensitizes villi to infection. Our results thus identify the inflammasome as an important mechanism by which the human placenta regulates systemic and local immunity during pregnancy to defend against L. monocytogenes infection., Graphical Abstract
- Published
- 2020
- Full Text
- View/download PDF
42. The complex interplay between endoplasmic reticulum stress and the NLRP3 inflammasome: a potential therapeutic target for inflammatory disorders
- Author
-
Alan Hsu, Matt D. Johansen, Kamal Dua, Prabuddha S. Pathinayake, Kate Schroder, Philip M. Hansbro, Madhur D. Shastri, Peter A. B. Wark, Nicole G. Hansbro, Gregory M. Peterson, Wai Chin Chong, Shakti D. Shukla, and Rahul P. Patel
- Subjects
lcsh:Immunologic diseases. Allergy ,0301 basic medicine ,Immunology ,Inflammation ,Review ,Therapeutic targeting ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,NLRP3 ,inflammasome ,medicine ,1107 Immunology, 1115 Pharmacology and Pharmaceutical Sciences ,inflammatory disorder ,Immunology and Allergy ,General Nursing ,business.industry ,Endoplasmic reticulum ,Inflammasome ,medicine.disease ,030104 developmental biology ,030220 oncology & carcinogenesis ,Rheumatoid arthritis ,Unfolded protein response ,endoplasmic reticulum stress ,medicine.symptom ,lcsh:RC581-607 ,business ,Homeostasis ,medicine.drug - Abstract
Inflammation is the result of a complex network of cellular and molecular interactions and mechanisms that facilitate immune protection against intrinsic and extrinsic stimuli, particularly pathogens, to maintain homeostasis and promote tissue healing. However, dysregulation in the immune system elicits excess/abnormal inflammation resulting in unintended tissue damage and causes major inflammatory diseases including asthma, chronic obstructive pulmonary disease, atherosclerosis, inflammatory bowel diseases, sarcoidosis and rheumatoid arthritis. It is now widely accepted that both endoplasmic reticulum (ER) stress and inflammasomes play critical roles in activating inflammatory signalling cascades. Notably, evidence is mounting for the involvement of ER stress in exacerbating inflammasome‐induced inflammatory cascades, which may provide a new axis for therapeutic targeting in a range of inflammatory disorders. Here, we comprehensively review the roles, mechanisms and interactions of both ER stress and inflammasomes, as well as their interconnected relationships in inflammatory signalling cascades. We also discuss novel therapeutic strategies that are being developed to treat ER stress‐ and inflammasome‐related inflammatory disorders., In this Review, we discuss the state‐of‐the‐art understanding of the pathways and factors involved in ER stress and inflammasome activation. We describe how these pathways induce inflammatory responses and are involved in chronic inflammatory diseases. We discuss new links between ER stress inflammasome activity and inflammation, and potential new therapeutic approaches to suppress ER stress and inflammasome‐induced inflammation.
- Published
- 2020
43. Neutrophil Extracellular Traps in Host Defense
- Author
-
Kate Schroder and Sabrina Sofia Burgener
- Subjects
Vasculitis ,Host (biology) ,Neutrophils ,NADPH Oxidases ,Neutrophil extracellular traps ,Biology ,Extracellular Traps ,General Biochemistry, Genetics and Molecular Biology ,Immunity, Innate ,Cell biology ,Autoimmune Diseases ,Histones ,medicine.anatomical_structure ,Anti-Infective Agents ,Immune System ,medicine ,Extracellular ,Animals ,Humans ,Bone marrow ,Intracellular ,Perspectives ,Peptide Hydrolases ,Signal Transduction - Abstract
Neutrophils are produced in the bone marrow and then patrol blood vessels from which they can be rapidly recruited to a site of infection. Neutrophils bind, engulf, and efficiently kill invading microbes via a suite of defense mechanisms. Diverse extracellular and intracellular microbes induce neutrophils to extrude neutrophil extracellular traps (NETs) through the process of NETosis. Here, we review the signaling mechanisms and cell biology underpinning the key NETosis pathways during infection and the antimicrobial functions of NETs in host defense.
- Published
- 2020
44. Lipopolysaccharide promotes Drp1-dependent mitochondrial fission and associated inflammatory responses in macrophages
- Author
-
Ronan Kapetanovic, Takashi Okada, Matthew J. Sweet, Kate Schroder, Jost de Bruin, Grace Mep Lawrence, Justin C. St. John, David P. Fairlie, Syeda Farhana Afroz, Divya Ramnath, Antje Blumenthal, and James E. B. Curson
- Subjects
0301 basic medicine ,Dynamins ,Lipopolysaccharides ,Toll‐like receptor ,Immunology ,macrophage ,Mitochondrion ,Mitochondrial Dynamics ,Outstanding Observation ,Mitochondrial Proteins ,03 medical and health sciences ,DNM1L ,Mice ,0302 clinical medicine ,Immunology and Allergy ,Animals ,Humans ,Cells, Cultured ,Toll-like receptor ,Chemistry ,mitochondrial fission ,Macrophages ,lipopolysaccharide ,Cell Biology ,Fibroblasts ,Cell biology ,Toll-Like Receptor 4 ,030104 developmental biology ,Mitochondrial biogenesis ,mitochondrial fusion ,Myeloid Differentiation Factor 88 ,TLR4 ,Phosphorylation ,Mitochondrial fission ,Dynamin‐related protein 1 ,030215 immunology - Abstract
Mitochondria have a multitude of functions, including energy generation and cell signaling. Recent evidence suggests that mitochondrial dynamics (i.e. the balance between mitochondrial fission and fusion) also regulate immune functions. Here, we reveal that lipopolysaccharide (LPS) stimulation increases mitochondrial numbers in mouse bone marrow‐derived macrophages (BMMs) and human monocyte‐derived macrophages. In BMMs, this response requires Toll‐like receptor 4 (Tlr4) and the TLR adaptor protein myeloid differentiation primary response 88 (MyD88) but is independent of mitochondrial biogenesis. Consistent with this phenomenon being a consequence of mitochondrial fission, the dynamin‐related protein 1 (Drp1) GTPase that promotes mitochondrial fission is enriched on mitochondria in LPS‐activated macrophages and is required for the LPS‐mediated increase in mitochondrial numbers in both BMMs and mouse embryonic fibroblasts. Pharmacological agents that skew toward mitochondrial fusion also abrogated this response. LPS triggered acute Drp1 phosphorylation at serine 635 (S635), followed by sustained Drp1 dephosphorylation at serine 656 (S656), in BMMs. LPS‐induced S656 dephosphorylation was abrogated in MyD88‐deficient BMMs, suggesting that this post‐translational modification is particularly important for Tlr4‐inducible fission. Pharmacological or genetic targeting of Tlr4‐inducible fission had selective effects on inflammatory mediator production, with LPS‐inducible mitochondrial fission promoting the expression and/or secretion of a subset of inflammatory mediators in BMMs and mouse embryonic fibroblasts. Thus, triggering of Tlr4 results in MyD88‐dependent activation of Drp1, leading to inducible mitochondrial fission and subsequent inflammatory responses in macrophages., Here, we demonstrate that lipopolysaccharide (LPS) acts via a Tlr4–MyD88–Drp1 axis to initiate mitochondrial fission and selective inflammatory responses in macrophages. We also show that LPS promotes sequential dynamin‐related protein 1 (Drp1) serine phosphorylation (S635) and dephosphorylation (S656), with the latter modification requiring the adaptor molecule myeloid differentiation primary response 88 (MyD88). We did not observe LPS‐inducible mitochondrial DNA replication in the same cells, suggesting that LPS‐inducible mitochondrial fission can occur independently of mitochondrial DNA replication.
- Published
- 2020
45. Rapid lamellipodial responses by neighbor cells drive epithelial sealing in response to pyroptotic cell death
- Author
-
Alexis Bonfim-Melo, Ivar Noordstra, Shafali Gupta, Amy H. Chan, Mathew J.K. Jones, Kate Schroder, and Alpha S. Yap
- Subjects
Cell Death ,Inflammasomes ,Pyroptosis ,Humans ,Apoptosis ,Epithelial Cells ,Pseudopodia ,Models, Biological ,Epithelium ,General Biochemistry, Genetics and Molecular Biology - Abstract
Cell injury poses a substantial challenge for epithelia homeostasis. Several cellular processes preserve epithelial barriers in response to apoptosis, but less is known about other forms of cell death, such as pyroptosis. Here we use an inducible caspase-1 system to analyze how colon epithelial monolayers respond to pyroptosis. We confirm that sporadic pyroptotic cells are physically eliminated from confluent monolayers by apical extrusion. This is accompanied by a transient defect in barrier function at the site of the pyroptotic cells. By visualizing cell shape changes and traction patterns in combination with cytoskeletal inhibitors, we show that rapid lamellipodial responses in the neighbor cells are responsible for correcting the leakage and resealing the barrier. Cell contractility is not required for this resealing response, in contrast to the response to apoptosis. Therefore, pyroptosis elicits a distinct homeostatic response from the epithelium that is driven by the stimulation of lamellipodia in neighbor cells.
- Published
- 2022
- Full Text
- View/download PDF
46. The Salmonella pathogenicity island-2 subverts human NLRP3 and NLRC4 inflammasome responses
- Author
-
Damien Bierschenk, Kate Schroder, Seth L. Masters, Fiona Moghaddas, Paul J. Baker, Dave Boucher, and Mercedes Monteleone
- Subjects
Salmonella typhimurium ,0301 basic medicine ,Salmonella ,Genomic Islands ,Inflammasomes ,Interleukin-1beta ,Immunology ,Caspase-11 ,Biology ,medicine.disease_cause ,Microbiology ,Type three secretion system ,03 medical and health sciences ,0302 clinical medicine ,Immune system ,NLRC4 ,NLR Family, Pyrin Domain-Containing 3 Protein ,Pyroptosis ,medicine ,Animals ,Humans ,Immunology and Allergy ,Myeloid Cells ,Innate immune system ,Cell Death ,Macrophages ,Calcium-Binding Proteins ,Inflammasome ,Cell Biology ,CARD Signaling Adaptor Proteins ,Mice, Inbred C57BL ,030104 developmental biology ,030220 oncology & carcinogenesis ,medicine.drug - Abstract
Inflammasomes are signaling hubs that activate inflammatory caspases to drive cytokine maturation and cell lysis. Inflammasome activation by Salmonella Typhimurium infection or Salmonella-derived molecules is extensively studied in murine myeloid cells. Salmonella-induced inflammasome signaling in human innate immune cells, is however, poorly characterized. Here, we show that Salmonella mutation to inactivate the Salmonella pathogenicity island-2 type III secretion system (SPI2 T3SS) potentiates S. Typhimurium-induced inflammasome responses from primary human macrophages, resulting in strong IL-1β production and macrophage death. Inactivation of the SPI1 T3SS diminished human macrophage responses to WT and ΔSPI2 Salmonella. Salmonella ΔSPI2 elicited a mixed inflammasome response from human myeloid cells, in which NLR family CARD-domain containing protein 4 (NLRC4) and NLR family PYRIN-domain containing protein 3 (NLRP3) perform somewhat redundant functions in generating IL-1β and inducing pyroptosis. Our data suggest that Salmonella employs the SPI2 T3SS to subvert SPI1-induced NLRP3 and NLRC4 inflammasome responses in human primary macrophages, in a species-specific immune evasion mechanism. The Gram-negative bacterium Salmonella Typhimurium suppresses human macrophage inflammasome responses via the Salmonella Pathogenicity Island 2.
- Published
- 2018
- Full Text
- View/download PDF
47. Interleukin-1β Maturation Triggers Its Relocation to the Plasma Membrane for Gasdermin-D-Dependent and -Independent Secretion
- Author
-
Ronan Kapetanovic, Darren L. Brown, Matthew J. Sweet, Dave Boucher, Caroline L. Holley, Jelena S. Bezbradica, Jennifer L. Stow, Kate Schroder, Kaiwen W. Chen, Verena Rolfes, Amanda C. Stanley, Mercedes Monteleone, Jessica B. von Pein, and Melanie R. Shakespear
- Subjects
0301 basic medicine ,medicine.medical_treatment ,Interleukin-1beta ,Caspase 1 ,Endogeny ,Transfection ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,medicine ,Humans ,Secretion ,lcsh:QH301-705.5 ,Unconventional protein secretion ,Chemistry ,Cell Membrane ,Intracellular Signaling Peptides and Proteins ,Pyroptosis ,Interleukin ,Inflammasome ,Phosphate-Binding Proteins ,Cell biology ,caspase-1 ,gasdermin ,inflammasome ,interleukin-1 ,macrophage ,neutrophil ,phosphoinositides ,pyroptosis ,trafficking ,unconventional protein secretion ,030104 developmental biology ,Cytokine ,lcsh:Biology (General) ,medicine.drug - Abstract
Summary: IL-1β requires processing by caspase-1 to generate the active, pro-inflammatory cytokine. Acute IL-1β secretion from inflammasome-activated macrophages requires caspase-1-dependent GSDMD cleavage, which also induces pyroptosis. Mechanisms of IL-1β secretion by pyroptotic and non-pyroptotic cells, and the precise functions of caspase-1 and GSDMD therein, are unresolved. Here, we show that, while efficient early secretion of endogenous IL-1β from primary non-pyroptotic myeloid cells in vitro requires GSDMD, later IL-1β release in vitro and in vivo proceeds independently of GSDMD. IL-1β maturation is sufficient for slow, caspase-1/GSDMD-independent secretion of ectopic IL-1β from resting, non-pyroptotic macrophages, but the speed of IL-1β release is boosted by inflammasome activation, via caspase-1 and GSDMD. IL-1β cleavage induces IL-1β enrichment at PIP2-enriched plasma membrane ruffles, and this is a prerequisite for IL-1β secretion and is mediated by a polybasic motif within the cytokine. We thus reveal a mechanism in which maturation-induced IL-1β trafficking facilitates its unconventional secretion. : Interleukin-1β is a potent pro-inflammatory cytokine whose dysregulated production drives a myriad of human diseases. Monteleone et al. uncover the trafficking mechanisms driving the unconventional secretion of mature interleukin-1β in non-pyroptotic and pyroptotic myeloid cells and reveal functions for caspase-1 and GSDMD therein. Keywords: interleukin-1, unconventional protein secretion, inflammasome, caspase-1, macrophage, neutrophil, gasdermin, pyroptosis, trafficking, phosphoinositides
- Published
- 2018
- Full Text
- View/download PDF
48. Cutting Edge: Blockade of Inhibitor of Apoptosis Proteins Sensitizes Neutrophils to TNF- but Not Lipopolysaccharide-Mediated Cell Death and IL-1β Secretion
- Author
-
Kaiwen W. Chen, Jelena S. Bezbradica, Dave Boucher, Jessica B. von Pein, Kate E. Lawlor, Ben A. Croker, James E Vince, Motti Gerlic, and Kate Schroder
- Subjects
Lipopolysaccharides ,0301 basic medicine ,Programmed cell death ,Inflammasomes ,Neutrophils ,Necroptosis ,Interleukin-1beta ,Immunology ,Ripoptosome ,Apoptosis ,Inhibitor of apoptosis ,Inhibitor of Apoptosis Proteins ,Mice ,Necrosis ,03 medical and health sciences ,0302 clinical medicine ,NLR Family, Pyrin Domain-Containing 3 Protein ,medicine ,Animals ,Immunology and Allergy ,Inflammation ,Cell Death ,Chemistry ,Macrophages ,Inflammasome ,Cell biology ,Mice, Inbred C57BL ,Toll-Like Receptor 4 ,030104 developmental biology ,Receptors, Tumor Necrosis Factor, Type I ,Caspases ,Tumor Necrosis Factors ,Tumor necrosis factor alpha ,Signal transduction ,Ripoptosome assembly ,Signal Transduction ,030215 immunology ,medicine.drug - Abstract
The mammalian inhibitor of apoptosis proteins (IAPs) are key regulators of cell death and inflammation. A major function of IAPs is to block the formation of a cell death–inducing complex, termed the ripoptosome, which can trigger caspase-8–dependent apoptosis or caspase-independent necroptosis. Recent studies report that upon TLR4 or TNF receptor 1 (TNFR1) signaling in macrophages, the ripoptosome can also induce NLRP3 inflammasome formation and IL-1β maturation. Whether neutrophils have the capacity to assemble a ripoptosome to induce cell death and inflammasome activation during TLR4 and TNFR1 signaling is unclear. In this study, we demonstrate that murine neutrophils can signal via TNFR1-driven ripoptosome assembly to induce both cell death and IL-1β maturation. However, unlike macrophages, neutrophils suppress TLR4-dependent cell death and NLRP3 inflammasome activation during IAP inhibition via deficiencies in the CD14/TRIF arm of TLR4 signaling.
- Published
- 2018
- Full Text
- View/download PDF
49. IFN Regulatory Factor 3 Balances Th1 and T Follicular Helper Immunity during Nonlethal Blood-Stage Plasmodium Infection
- Author
-
Christian R. Engwerda, Arya Sheela Nair, Rebecca Austin, Shannon E. Best, Daniel Fernandez-Ruiz, Lianne I. M. Lansink, Ashraful Haque, Gayle M. Davey, Megan S. F. Soon, Chelsea L. Edwards, William R. Heath, Kate Schroder, Lily Fogg, Geoffrey R. Hill, Steven W. Lane, Jane A. C. Wilson, Ismail Sebina, Urijah N. Liligeto, Andreas Suhrbier, and Kylie R. James
- Subjects
0301 basic medicine ,Innate immune system ,Cellular differentiation ,Immunology ,Priming (immunology) ,Germinal center ,biochemical phenomena, metabolism, and nutrition ,Biology ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,medicine.anatomical_structure ,Immune system ,Immunity ,medicine ,Immunology and Allergy ,Memory B cell ,B cell ,030215 immunology - Abstract
Differentiation of CD4+ Th cells is critical for immunity to malaria. Several innate immune signaling pathways have been implicated in the detection of blood-stage Plasmodium parasites, yet their influence over Th cell immunity remains unclear. In this study, we used Plasmodium-reactive TCR transgenic CD4+ T cells, termed PbTII cells, during nonlethal P. chabaudi chabaudi AS and P. yoelii 17XNL infection in mice, to examine Th cell development in vivo. We found no role for caspase1/11, stimulator of IFN genes, or mitochondrial antiviral-signaling protein, and only modest roles for MyD88 and TRIF-dependent signaling in controlling PbTII cell expansion. In contrast, IFN regulatory factor 3 (IRF3) was important for supporting PbTII expansion, promoting Th1 over T follicular helper (Tfh) differentiation, and controlling parasites during the first week of infection. IRF3 was not required for early priming by conventional dendritic cells, but was essential for promoting CXCL9 and MHC class II expression by inflammatory monocytes that supported PbTII responses in the spleen. Thereafter, IRF3-deficiency boosted Tfh responses, germinal center B cell and memory B cell development, parasite-specific Ab production, and resolution of infection. We also noted a B cell–intrinsic role for IRF3 in regulating humoral immune responses. Thus, we revealed roles for IRF3 in balancing Th1- and Tfh-dependent immunity during nonlethal infection with blood-stage Plasmodium parasites.
- Published
- 2018
- Full Text
- View/download PDF
50. SCIMP is a spatiotemporal transmembrane scaffold for Erk1/2 to direct pro-inflammatory signaling in TLR-activated macrophages
- Author
-
Matthew J. Sweet, Richard Lucas, Lin Luo, James E. B. Curson, Kaustav Das Gupta, Yvette W. H. Koh, Nicholas D. Condon, Neeraj Tuladhar, Kate Schroder, Jennifer L. Stow, Liping Liu, Sabrina Sofia Burgener, and Evan Ingley
- Subjects
MAP Kinase Signaling System ,medicine.medical_treatment ,Mice, Transgenic ,Inflammation ,General Biochemistry, Genetics and Molecular Biology ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Animals ,Macrophage ,Phosphorylation ,030304 developmental biology ,0303 health sciences ,Toll-like receptor ,Mitogen-Activated Protein Kinase 3 ,Innate immune system ,Chemistry ,Kinase ,Macrophages ,Toll-Like Receptors ,Transmembrane protein ,Cell biology ,Toll-Like Receptor 4 ,Cytokine ,030220 oncology & carcinogenesis ,TLR4 ,Cytokines ,medicine.symptom ,Signal Transduction - Abstract
Immune cells are armed with Toll-like receptors (TLRs) for sensing and responding to pathogens and other danger cues. The role of extracellular-signal-regulated kinases 1/2 (Erk1/2) in TLR signaling remains enigmatic, with both pro- and anti-inflammatory functions described. We reveal here that the immune-specific transmembrane adaptor SCIMP is a direct scaffold for Erk1/2 in TLR pathways, with high-resolution, live-cell imaging revealing that SCIMP guides the spatial and temporal recruitment of Erk2 to membrane ruffles and macropinosomes for pro-inflammatory TLR4 signaling. SCIMP-deficient mice display defects in Erk1/2 recruitment to TLR4, c-Fos activation, and pro-inflammatory cytokine production, with these effects being phenocopied by Erk1/2 signaling inhibition. Our findings thus delineate a selective role for SCIMP as a key scaffold for the membrane recruitment of Erk1/2 kinase to initiate TLR-mediated pro-inflammatory responses in macrophages.
- Published
- 2021
- Full Text
- View/download PDF
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.