Your browser doesn't support javascript.
loading
: 20 | 50 | 100
1 - 7 de 7
1.
Nat Metab ; 4(5): 524-533, 2022 05.
Article En | MEDLINE | ID: mdl-35655024

Since its discovery in inflammatory macrophages, itaconate has attracted much attention due to its antimicrobial and immunomodulatory activity1-3. However, instead of investigating itaconate itself, most studies used derivatized forms of itaconate and thus the role of non-derivatized itaconate needs to be scrutinized. Mesaconate, a metabolite structurally very close to itaconate, has never been implicated in mammalian cells. Here we show that mesaconate is synthesized in inflammatory macrophages from itaconate. We find that both, non-derivatized itaconate and mesaconate dampen the glycolytic activity to a similar extent, whereas only itaconate is able to repress tricarboxylic acid cycle activity and cellular respiration. In contrast to itaconate, mesaconate does not inhibit succinate dehydrogenase. Despite their distinct impact on metabolism, both metabolites exert similar immunomodulatory effects in pro-inflammatory macrophages, specifically a reduction of interleukin (IL)-6 and IL-12 secretion and an increase of CXCL10 production in a manner that is independent of NRF2 and ATF3. We show that a treatment with neither mesaconate nor itaconate impairs IL-1ß secretion and inflammasome activation. In summary, our results identify mesaconate as an immunomodulatory metabolite in macrophages, which interferes to a lesser extent with cellular metabolism than itaconate.


Macrophages , Succinates , Animals , Inflammasomes , Macrophages/drug effects , Macrophages/metabolism , Mice , RAW 264.7 Cells , Succinates/metabolism , Succinates/pharmacology
2.
iScience ; 24(11): 103300, 2021 Nov 19.
Article En | MEDLINE | ID: mdl-34746710

Pathogenic viruses induce metabolic changes in host cells to secure the availability of biomolecules and energy to propagate. Influenza A virus (IAV) and severe acute respiratory syndrome corona virus 2 (SARS-CoV-2) both infect the human airway epithelium and are important human pathogens. The metabolic changes induced by these viruses in a physiologically relevant human model and how this affects innate immune responses to limit viral propagation are not well known. Using an ex vivo model of pseudostratified primary human airway epithelium, we here demonstrate that infection with both IAV and SARS-CoV-2 resulted in distinct metabolic changes including increases in lactate dehydrogenase A (LDHA) expression and LDHA-mediated lactate formation. Interestingly, LDHA regulated both basal and induced mitochondrial anti-viral signaling protein (MAVS)-dependent type I interferon (IFN) responses to promote IAV, but not SARS-CoV-2, replication. Our data demonstrate that LDHA and lactate promote IAV but not SARS-CoV-2 replication by inhibiting MAVS-dependent induction of type I IFN in primary human airway epithelium.

3.
Front Immunol ; 12: 635738, 2021.
Article En | MEDLINE | ID: mdl-33679790

Detection of pathogen-derived DNA or RNA species by cellular nucleic acid sensors prompts release of anti-microbial interferons and cytokines. In contrast to their protective anti-microbial functions, inappropriate or excessive activation of nucleic acid sensors can cause inflammatory diseases. Nucleic acid sensing is therefore tightly controlled by regulatory factors acting through both transcriptional and post-transcriptional mechanisms. Recently, it has become clearer that metabolic pathways-previously thought to be unconnected with immune responses-can influence nucleic acid sensing. This regulation can be observed when immune system cells undergo metabolic reprogramming in response to stimulation with pathogen-associated molecular patterns such as lipopolysaccharide from gram negative bacteria. Metabolic reprogramming leads to accumulation and secretion of metabolites, which have been mostly viewed as end-products of processes providing cellular energy and building blocks. However, metabolites have now been identified as important regulators of nucleic acid sensing. This mini-review aims to outline current knowledge on regulation of central nucleic acid sensing pathways by metabolites during metabolic reprogramming.


Cellular Reprogramming , DNA/metabolism , Energy Metabolism , Pathogen-Associated Molecular Pattern Molecules/metabolism , RNA/metabolism , Receptors, Pattern Recognition/metabolism , Animals , DNA/immunology , Humans , Ligands , Pathogen-Associated Molecular Pattern Molecules/immunology , RNA/immunology , Receptors, Pattern Recognition/immunology , Signal Transduction , Toll-Like Receptors/metabolism
4.
Antiviral Res ; 185: 104988, 2021 01.
Article En | MEDLINE | ID: mdl-33248195

Pandemic spread of emerging human pathogenic viruses, such as the current SARS-CoV-2, poses both an immediate and future challenge to human health and society. Currently, effective treatment of infection with SARS-CoV-2 is limited and broad spectrum antiviral therapies to meet other emerging pandemics are absent leaving the World population largely unprotected. Here, we have identified distinct members of the family of polyether ionophore antibiotics with potent ability to inhibit SARS-CoV-2 replication and cytopathogenicity in cells. Several compounds from this class displayed more than 100-fold selectivity between viral-induced cytopathogenicity and inhibition of cell viability, however the compound X-206 displayed >500-fold selectivity and was furthermore able to inhibit viral replication even at sub-nM levels. The antiviral mechanism of the polyether ionophores is currently not understood in detail. We demonstrate, e.g. through unbiased bioactivity profiling, that their effects on the host cells differ from those of cationic amphiphiles such as hydroxychloroquine. Collectively, our data suggest that polyether ionophore antibiotics should be subject to further investigations as potential broad-spectrum antiviral agents.


Anti-Bacterial Agents/pharmacology , Antiviral Agents/pharmacology , COVID-19 Drug Treatment , Ethers, Cyclic/pharmacology , Ionophores/pharmacology , SARS-CoV-2/drug effects , Animals , Chlorocebus aethiops , Humans , Vero Cells , Virus Replication/drug effects
5.
Nat Commun ; 11(1): 4938, 2020 10 02.
Article En | MEDLINE | ID: mdl-33009401

Antiviral strategies to inhibit Severe Acute Respiratory Syndrome Coronavirus 2 (SARS-CoV2) and the pathogenic consequences of COVID-19 are urgently required. Here, we demonstrate that the NRF2 antioxidant gene expression pathway is suppressed in biopsies obtained from COVID-19 patients. Further, we uncover that NRF2 agonists 4-octyl-itaconate (4-OI) and the clinically approved dimethyl fumarate (DMF) induce a cellular antiviral program that potently inhibits replication of SARS-CoV2 across cell lines. The inhibitory effect of 4-OI and DMF extends to the replication of several other pathogenic viruses including Herpes Simplex Virus-1 and-2, Vaccinia virus, and Zika virus through a type I interferon (IFN)-independent mechanism. In addition, 4-OI and DMF limit host inflammatory responses to SARS-CoV2 infection associated with airway COVID-19 pathology. In conclusion, NRF2 agonists 4-OI and DMF induce a distinct IFN-independent antiviral program that is broadly effective in limiting virus replication and in suppressing the pro-inflammatory responses of human pathogenic viruses, including SARS-CoV2.


Anti-Inflammatory Agents/pharmacology , Antiviral Agents/pharmacology , Betacoronavirus/drug effects , Coronavirus Infections/drug therapy , Dimethyl Fumarate/agonists , NF-E2-Related Factor 2/metabolism , Pneumonia, Viral/drug therapy , Succinates/agonists , Adult , Antioxidants/pharmacology , Betacoronavirus/metabolism , COVID-19 , Coronavirus Infections/virology , Dimethyl Fumarate/pharmacology , Female , Gene Expression , Gene Knockdown Techniques , Humans , Interferon Type I , Lung/pathology , Male , NF-E2-Related Factor 2/genetics , Pandemics , Pneumonia, Viral/virology , SARS-CoV-2 , Signal Transduction/drug effects , Succinates/pharmacology , Virus Replication/drug effects
7.
EMBO Rep ; 21(12): e51252, 2020 12 03.
Article En | MEDLINE | ID: mdl-33112036

Respiratory infections, like the current COVID-19 pandemic, target epithelial cells in the respiratory tract. Alveolar macrophages (AMs) are tissue-resident macrophages located within the lung. They play a key role in the early phases of an immune response to respiratory viruses. AMs are likely the first immune cells to encounter SARS-CoV-2 during an infection, and their reaction to the virus will have a profound impact on the outcome of the infection. Interferons (IFNs) are antiviral cytokines and among the first cytokines produced upon viral infection. In this study, AMs from non-infectious donors are challenged with SARS-CoV-2. We demonstrate that challenged AMs are incapable of sensing SARS-CoV-2 and of producing an IFN response in contrast to other respiratory viruses, like influenza A virus and Sendai virus, which trigger a robust IFN response. The absence of IFN production in AMs upon challenge with SARS-CoV-2 could explain the initial asymptotic phase observed during COVID-19 and argues against AMs being the sources of pro-inflammatory cytokines later during infection.


COVID-19/immunology , Macrophages, Alveolar/immunology , Macrophages, Alveolar/virology , SARS-CoV-2/immunology , Antiviral Agents/immunology , COVID-19/virology , Cells, Cultured , Cytokines/immunology , Epithelial Cells/immunology , Epithelial Cells/virology , Humans , Immune Evasion , Interferon Type I/immunology , Lung/immunology , Lung/virology , Pandemics
...