Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 406
Filter
Add more filters











Publication year range
1.
Cell Rep ; 43(9): 114706, 2024 Sep 04.
Article in English | MEDLINE | ID: mdl-39235945

ABSTRACT

To gain insight into how an adjuvant impacts vaccination responses, we use systems immunology to study human H5N1 influenza vaccination with or without the adjuvant AS03, longitudinally assessing 14 time points including multiple time points within the first day after prime and boost. We develop an unsupervised computational framework to discover high-dimensional response patterns, which uncover adjuvant- and immunogenicity-associated early response dynamics, including some that differ post prime versus boost. With or without adjuvant, some vaccine-induced transcriptional patterns persist to at least 100 days after initial vaccination. Single-cell profiling of surface proteins, transcriptomes, and chromatin accessibility implicates transcription factors in the erythroblast-transformation-specific (ETS) family as shaping these long-lasting signatures, primarily in classical monocytes but also in CD8+ naive-like T cells. These cell-type-specific signatures are elevated at baseline in high-antibody responders in an independent vaccination cohort, suggesting that antigen-agnostic baseline immune states can be modulated by vaccine antigens alone to enhance future responses.

2.
Cell Rep Methods ; : 100856, 2024 Aug 30.
Article in English | MEDLINE | ID: mdl-39243752

ABSTRACT

The ongoing co-circulation of multiple severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) strains necessitates advanced methods such as high-throughput multiplex pseudovirus systems for evaluating immune responses to different variants, crucial for developing updated vaccines and neutralizing antibodies (nAbs). We have developed a quadri-fluorescence (qFluo) pseudovirus platform by four fluorescent reporters with different spectra, allowing simultaneous measurement of the nAbs against four variants in a single test. qFluo shows high concordance with the classical single-reporter assay when testing monoclonal antibodies and human plasma. Utilizing qFluo, we assessed the immunogenicities of the spike of BA.5, BQ.1.1, XBB.1.5, and CH.1.1 in hamsters. An analysis of cross-neutralization against 51 variants demonstrated superior protective immunity from XBB.1.5, especially against prevalent strains such as "FLip" and JN.1, compared to BA.5. Our finding partially fills the knowledge gap concerning the immunogenic efficacy of the XBB.1.5 vaccine against current dominant variants, being instrumental in vaccine-strain decisions and insight into the evolutionary path of SARS-CoV-2.

3.
Cell Rep ; 43(9): 114720, 2024 Sep 07.
Article in English | MEDLINE | ID: mdl-39244752

ABSTRACT

Macrophages are major host cells for the protozoan Leishmania parasite. Depending on their activation state, they either contribute to the detection and elimination of Leishmania spp. or promote parasite resilience. Here, we report that the activation of the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2) in macrophages plays a pivotal role in the progression of Leishmania infantum infection by controlling inflammation and redox balance of macrophages. We also highlight the involvement of the NOX2/reactive oxygen species (ROS) axis in early Nrf2 activation and, subsequently, prostaglandin E2 (PGE2)/EP2r signaling in the sustenance of Nrf2 activation upon infection. Moreover, we establish a ferroptosis-like process within macrophages as a cell death program of L. infantum and the protective effect of Nrf2 in macrophages against L. infantum death. Altogether, these results identify Nrf2 as a critical factor for the susceptibility of L. infantum infection, highlighting Nrf2 as a promising pharmacological target for the development of therapeutic approaches for the treatment of visceral leishmaniasis.

4.
Cell Rep ; 43(8): 114583, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39110597

ABSTRACT

Vast shotgun metagenomics data remain an underutilized resource for novel enzymes. Artificial intelligence (AI) has increasingly been applied to protein mining, but its conventional performance evaluation is interpolative in nature, and these trained models often struggle to extrapolate effectively when challenged with unknown data. In this study, we present a framework (DeepMineLys [deep mining of phage lysins from human microbiome]) based on the convolutional neural network (CNN) to identify phage lysins from three human microbiome datasets. When validated with an independent dataset, our method achieved an F1-score of 84.00%, surpassing existing methods by 20.84%. We expressed 16 lysin candidates from the top 100 sequences in E. coli, confirming 11 as active. The best one displayed an activity 6.2-fold that of lysozyme derived from hen egg white, establishing it as the most potent lysin from the human microbiome. Our study also underscores several important issues when applying AI to biology questions. This framework should be applicable for mining other proteins.


Subject(s)
Bacteriophages , Microbiota , Humans , Bacteriophages/genetics , Bacteriophages/metabolism , Data Mining , Viral Proteins/metabolism , Neural Networks, Computer , Animals , Muramidase/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism
5.
Cell Rep ; 43(8): 114585, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39110590

ABSTRACT

Previous studies have demonstrated that gut microbiota dysbiosis promotes the development of mastitis. The interaction of the vagus nerve and gut microbiota endows host homeostasis and regulates disease development, but whether the vagus nerve participates in the pathogenesis of mastitis is unclear. Here, vagotomized mice exhibit disruption of the blood-milk barrier and mammary gland inflammation. Notably, mastitis and barrier damage caused by vagotomy are dependent on the gut microbiota, as evidenced by antibiotic treatment and fecal microbiota transplantation. Vagotomy significantly alters the gut microbial composition and tryptophan metabolism and reduces the 5-hydroxyindole acetic acid (5-HIAA) level. Supplementation with 5-HIAA alleviates vagotomy-induced mastitis, which is associated with the activation of the aryl hydrocarbon receptor (AhR) and subsequent inhibition of the NF-κB pathway. Collectively, our findings indicate the important role of the vagus-mediated gut-mammary axis in the pathogenesis of mastitis and imply a potential strategy for the treatment of mastitis by targeting the vagus-gut microbiota interaction.


Subject(s)
Gastrointestinal Microbiome , Mastitis , Tryptophan , Vagotomy , Animals , Tryptophan/metabolism , Female , Mice , Mastitis/metabolism , Mastitis/microbiology , Receptors, Aryl Hydrocarbon/metabolism , Vagus Nerve/metabolism , NF-kappa B/metabolism , Dysbiosis/microbiology , Dysbiosis/metabolism , Mice, Inbred C57BL , Fecal Microbiota Transplantation , Mammary Glands, Animal/microbiology , Mammary Glands, Animal/metabolism , Mammary Glands, Animal/pathology
6.
Cell Rep ; 43(8): 114608, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39120972

ABSTRACT

Ubiquitination is essential for the proteasomal turnover of IRF3, the central factor mediating the antiviral innate immune response. However, the spatiotemporal regulation of IRF3 ubiquitination for the precise activation and timely resolution of innate immunity remains unclear. Here, we identified BRCA1-associated protein-1 (BAP1) and ubiquitin-protein ligase E3C (UBE3C) as the key deubiquitinase and ubiquitinase for temporal control of IRF3 stability during viral infection. In the early stage, BAP1 dominates and removes K48-linked ubiquitination of IRF3 in the nucleus, preventing its proteasomal degradation and facilitating efficient interferon (IFN)-ß production. In the late stage, E3 ligase UBE3C, induced by IFN-ß, specifically mediates IRF3 ubiquitination and promotes its proteasomal degradation. Overall, the sequential interactions with BAP1 and UBE3C govern IRF3 stability during innate response, ensuring effective viral clearance and inflammation resolution. Our findings provide insights into the temporal control of innate signaling and suggest potential interventions in viral infection.


Subject(s)
Immunity, Innate , Interferon Regulatory Factor-3 , Tumor Suppressor Proteins , Ubiquitin Thiolesterase , Ubiquitin-Protein Ligases , Ubiquitination , Interferon Regulatory Factor-3/metabolism , Ubiquitin-Protein Ligases/metabolism , Humans , Ubiquitin Thiolesterase/metabolism , Ubiquitin Thiolesterase/genetics , Tumor Suppressor Proteins/metabolism , Animals , HEK293 Cells , Mice , Interferon-beta/metabolism , Proteolysis , Mice, Inbred C57BL , Signal Transduction , Proteasome Endopeptidase Complex/metabolism
7.
Cell Rep ; 43(8): 114601, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39126650

ABSTRACT

Fungal pathogens such as Candida albicans pose a significant threat to human health with limited treatment options available. One strategy to expand the therapeutic target space is to identify genes important for pathogen growth in host-relevant environments. Here, we leverage a pooled functional genomic screening strategy to identify genes important for fitness of C. albicans in diverse conditions. We identify an essential gene with no known Saccharomyces cerevisiae homolog, C1_09670C, and demonstrate that it encodes subunit 3 of replication factor A (Rfa3). Furthermore, we apply computational analyses to identify functionally coherent gene clusters and predict gene function. Through this approach, we predict the cell-cycle-associated function of C3_06880W, a previously uncharacterized gene required for fitness specifically at elevated temperatures, and follow-up assays confirm that C3_06880W encodes Iml3, a component of the C. albicans kinetochore with roles in virulence in vivo. Overall, this work reveals insights into the vulnerabilities of C. albicans.


Subject(s)
Candida albicans , Fungal Proteins , Candida albicans/genetics , Candida albicans/pathogenicity , Fungal Proteins/genetics , Fungal Proteins/metabolism , Genetic Fitness , Genomics/methods , Virulence/genetics , Genome, Fungal , Humans
8.
Cell Rep ; 43(8): 114635, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39154338

ABSTRACT

Early childhood caries (ECC) is influenced by microbial and host factors, including social, behavioral, and oral health. In this cross-sectional study, we analyze interkingdom dynamics in the dental plaque microbiome and its association with host variables. We use 16S rRNA and ITS1 amplicon sequencing on samples collected from preschool children and analyze questionnaire data to examine the social determinants of oral health. The results indicate a significant enrichment of Streptococcus mutans and Candida dubliniensis in ECC samples, in contrast to Neisseria oralis in caries-free children. Our interkingdom correlation analysis reveals that Candida dubliniensis is strongly correlated with both Neisseria bacilliformis and Prevotella veroralis in ECC. Additionally, ECC shows significant associations with host variables, including oral health status, age, place of residence, and mode of childbirth. This study provides empirical evidence associating the oral microbiome with socioeconomic and behavioral factors in relation to ECC, offering insights for developing targeted prevention strategies.


Subject(s)
Dental Caries , Dental Plaque , Microbiota , Socioeconomic Factors , Humans , Dental Caries/microbiology , Dental Plaque/microbiology , Child, Preschool , Female , Male , Cross-Sectional Studies , RNA, Ribosomal, 16S/genetics
9.
Cell Rep ; 43(8): 114641, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39154339

ABSTRACT

Caspase-8-dependent pyroptosis has been shown to mediate host protection from Yersinia infection. For this mode of cell death, the kinase activity of receptor-interacting protein kinase 1 (RIPK1) is required, but the autophosphorylation sites required to drive caspase-8 activation have not been determined. Here, we show that non-canonical autophosphorylation of RIPK1 at threonine 169 (T169) is necessary for caspase-8-mediated pyroptosis. Mice with alanine in the T169 position are highly susceptible to Yersinia dissemination. Mechanistically, the delayed formation of a complex containing RIPK1, ZBP1, Fas-associated protein with death domain (FADD), and caspase-8 abrogates caspase-8 maturation in T169A mice and leads to the eventual activation of RIPK3-dependent necroptosis in vivo; however, this is insufficient to protect the host, suggesting that timely pyroptosis during early response is specifically required to control infection. These results position RIPK1 T169 phosphorylation as a driver of pyroptotic cell death critical for host defense.


Subject(s)
Pyroptosis , Receptor-Interacting Protein Serine-Threonine Kinases , Yersinia Infections , Animals , Receptor-Interacting Protein Serine-Threonine Kinases/metabolism , Phosphorylation , Yersinia Infections/metabolism , Yersinia Infections/microbiology , Mice , Caspase 8/metabolism , Mice, Inbred C57BL , Yersinia/metabolism , Humans
10.
Cell Rep ; 43(8): 114631, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39154342

ABSTRACT

Enzymatic modification of DNA nucleobases can coordinate gene expression, nuclease protection, or mutagenesis. We recently discovered a clade of phage-specific cytosine methyltransferase (MT) and 5-methylpyrimidine dioxygenase (5mYOX) enzymes that produce 5-hydroxymethylcytosine (5hmC) as a precursor for enzymatic hypermodifications on viral genomes. Here, we identify phage MT- and 5mYOX-associated glycosyltransferases (GTs) that catalyze linkage of diverse sugars to 5hmC nucleobase substrates. Metavirome mining revealed thousands of biosynthetic gene clusters containing enzymes with predicted roles in cytosine sugar hypermodification. We developed a platform for high-throughput screening of GT-containing pathways, relying on the Escherichia coli metabolome as a substrate pool. We successfully reconstituted several pathways and isolated diverse sugar modifications appended to cytosine, including mono-, di-, or tri-saccharides comprised of hexoses, N-acetylhexosamines, or heptose. These findings expand our knowledge of hypermodifications on nucleic acids and the origins of corresponding sugar-installing enzymes.


Subject(s)
Glycosyltransferases , Polysaccharides , Polysaccharides/metabolism , Glycosyltransferases/metabolism , Glycosyltransferases/genetics , Escherichia coli/genetics , Escherichia coli/metabolism , 5-Methylcytosine/metabolism , 5-Methylcytosine/analogs & derivatives , DNA/metabolism
11.
Cell Rep ; 43(8): 114624, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39154341

ABSTRACT

Chlamydia trachomatis, a leading cause of bacterial sexually transmitted infections, creates a specialized intracellular replicative niche by translocation and insertion of a diverse array of effectors (Incs [inclusion membrane proteins]) into the inclusion membrane. Here, we characterize IncE, a multifunctional Inc that encodes two non-overlapping short linear motifs (SLiMs) within its short cytosolic C terminus. The proximal SLiM, by mimicking just a small portion of an R-N-ethylmaleimide-sensitive factor adaptor protein receptor (SNARE) motif, binds and recruits syntaxin (STX)7- and STX12-containing vesicles to the inclusion. The distal SLiM mimics the sorting nexin (SNX)5 and SNX6 cargo binding site to recruit SNX6-containing vesicles to the inclusion. By simultaneously binding two distinct vesicle classes, IncE brings these vesicles in close apposition with each other at the inclusion to facilitate C. trachomatis intracellular development. Our work suggests that Incs may have evolved SLiMs to enable rapid evolution in a limited protein space to disrupt host cell processes.


Subject(s)
Bacterial Proteins , Chlamydia trachomatis , Chlamydia trachomatis/metabolism , Humans , Bacterial Proteins/metabolism , HeLa Cells , Amino Acid Motifs , Protein Transport , Sorting Nexins/metabolism , Sorting Nexins/genetics , Qa-SNARE Proteins/metabolism , Protein Binding
12.
Cell Rep ; 43(8): 114571, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39093698

ABSTRACT

Rice stripe virus (RSV) establishes infection in the ovaries of its vector insect, Laodelphax striatellus. We demonstrate that RSV infection delays ovarian maturation by inhibiting membrane localization of the vitellogenin receptor (VgR), thereby reducing the vitellogenin (Vg) accumulation essential for egg development. We identify the host protein L. striatellus Rab1 protein (LsRab1), which directly interacts with RSV nucleocapsid protein (NP) within nurse cells. LsRab1 is required for VgR surface localization and ovarian Vg accumulation. RSV inhibits LsRab1 function through two mechanisms: NP binding LsRab1 prevents GTP binding, and NP binding LsRab1-GTP complexes stimulates GTP hydrolysis, forming an inactive LsRab1 form. Through this dual inhibition, RSV infection prevents LsRab1 from facilitating VgR trafficking to the cell membrane, leading to inefficient Vg uptake. The Vg-VgR pathway is present in most oviparous animals, and the mechanisms detailed here provide insights into the vertical transmission of other insect-transmitted viruses of medical and agricultural importance.


Subject(s)
Receptors, Cell Surface , Tenuivirus , rab1 GTP-Binding Proteins , Animals , Female , rab1 GTP-Binding Proteins/metabolism , Tenuivirus/physiology , Tenuivirus/metabolism , Receptors, Cell Surface/metabolism , Egg Proteins/metabolism , Insect Proteins/metabolism , Insect Proteins/genetics , Vitellogenins/metabolism , Nucleocapsid Proteins/metabolism , Hemiptera/virology , Hemiptera/metabolism , Ovary/virology , Ovary/metabolism , Protein Binding , Protein Transport , Cell Membrane/metabolism , Cell Membrane/virology , Plant Diseases/virology
13.
Cell Rep ; 43(8): 114566, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39088320

ABSTRACT

Novel insecticides were recently introduced to counter pyrethroid resistance threats in African malaria vectors. To prolong their effectiveness, potential cross-resistance from promiscuous pyrethroid metabolic resistance mechanisms must be elucidated. Here, we demonstrate that the duplicated P450s CYP6P9a/-b, proficient pyrethroid metabolizers, reduce neonicotinoid efficacy in Anopheles funestus while enhancing the potency of chlorfenapyr. Transgenic expression of CYP6P9a/-b in Drosophila confirmed that flies expressing both genes were significantly more resistant to neonicotinoids than controls, whereas the contrasting pattern was observed for chlorfenapyr. This result was also confirmed by RNAi knockdown experiments. In vitro expression of recombinant CYP6P9a and metabolism assays established that it significantly depletes both clothianidin and chlorfenapyr, with metabolism of chlorfenapyr producing the insecticidally active intermediate metabolite tralopyril. This study highlights the risk of cross-resistance between pyrethroid and neonicotinoid and reveals that chlorfenapyr-based control interventions such as Interceptor G2 could remain efficient against some P450-based resistant mosquitoes.


Subject(s)
Anopheles , Cytochrome P-450 Enzyme System , Guanidines , Insecticide Resistance , Insecticides , Malaria , Neonicotinoids , Pyrethrins , Thiazoles , Animals , Thiazoles/pharmacology , Guanidines/pharmacology , Insecticide Resistance/genetics , Anopheles/drug effects , Anopheles/genetics , Pyrethrins/pharmacology , Pyrethrins/metabolism , Neonicotinoids/pharmacology , Insecticides/pharmacology , Cytochrome P-450 Enzyme System/metabolism , Cytochrome P-450 Enzyme System/genetics , Substrate Specificity , Mosquito Vectors/drug effects , Mosquito Vectors/genetics , Insect Proteins/metabolism , Insect Proteins/genetics
14.
Cell Rep ; 43(8): 114572, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39116202

ABSTRACT

Antibiotics cause collateral damage to resident microbes that is associated with various health risks. To date, studies have largely focused on the impacts of antibiotics on large intestinal and fecal microbiota. Here, we employ a gastrointestinal (GI) tract-wide integrated multiomic approach to show that amoxicillin (AMX) treatment reduces bacterial abundance, bile salt hydrolase activity, and unconjugated bile acids in the small intestine (SI). Losses of fatty acids (FAs) and increases in acylcarnitines in the large intestine (LI) correspond with spatially distinct expansions of Proteobacteria. Parasutterella excrementihominis engage in FA biosynthesis in the SI, while multiple Klebsiella species employ FA oxidation during expansion in the LI. We subsequently demonstrate that restoration of unconjugated bile acids can mitigate losses of commensals in the LI while also inhibiting the expansion of Proteobacteria during AMX treatment. These results suggest that the depletion of bile acids and lipids may contribute to AMX-induced dysbiosis in the lower GI tract.


Subject(s)
Amoxicillin , Bile Acids and Salts , Bile Acids and Salts/metabolism , Animals , Amoxicillin/pharmacology , Mice , Gastrointestinal Microbiome/drug effects , Mice, Inbred C57BL , Anti-Bacterial Agents/pharmacology , Proteobacteria/metabolism , Proteobacteria/drug effects , Fatty Acids/metabolism , Male , Microbiota/drug effects
15.
Cell Rep ; 43(8): 114602, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39137112

ABSTRACT

Klebsiella aerogenes is an understudied and clinically important pathogen. We therefore investigate its population structure by genome analysis aligned with metadata. We sequence 130 non-duplicated K. aerogenes clinical isolates and identify two inter-patient transmission events. We then retrieve all publicly available K. aerogenes genomes (n = 1,026, accessed by January 1, 2023) and analyze them with our 130 genomes. We develop a core-genome multi-locus sequence-typing scheme. We find that K. aerogenes is a species complex comprising four phylogroups undergoing evolutionary divergence, likely forming three species. We delineate remarkable clonal diversity and identify three worldwide-distributed carbapenemase-encoding clonal clusters, representing high-risk lineages. We uncover that K. aerogenes has an open genome equipped by a large arsenal of antimicrobial resistance genes. We identify two genetic regions specific for K. aerogenes, encoding a type VI secretion system and flagella/chemotaxis for motility, respectively, both contributing to the virulence. These results provide much-needed insights into the population structure and pan-genomes of K. aerogenes.


Subject(s)
Enterobacter aerogenes , Genome, Bacterial , Virulence/genetics , Humans , Enterobacter aerogenes/genetics , Enterobacter aerogenes/drug effects , Enterobacter aerogenes/pathogenicity , Drug Resistance, Bacterial/genetics , Phylogeny , Genomics/methods , Virulence Factors/genetics , Anti-Bacterial Agents/pharmacology , Bacterial Proteins/genetics , Bacterial Proteins/metabolism , Klebsiella Infections/microbiology , Klebsiella Infections/epidemiology
16.
Cell Rep ; 43(9): 114694, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39196777

ABSTRACT

Subgenomic flavivirus RNAs (sfRNAs) are structured RNAs encoded by flaviviruses that promote viral infection by inhibiting cellular RNA decay machinery. Herein, we analyze sfRNA production and localization using single-molecule RNA fluorescence in situ hybridization (smRNA-FISH) throughout West Nile virus, Zika virus, or dengue virus serotype 2 infection. We observe that sfRNAs are generated during the RNA replication phase of viral infection in the cytosol and accumulate in processing bodies (P-bodies), which contain RNA decay machinery such as XRN1 and Dcp1b. However, upon activation of the host antiviral endoribonuclease, ribonuclease L (RNase L), sfRNAs re-localize to ribonucleoprotein complexes known as RNase L-induced bodies (RLBs). RLB-mediated sequestration of sfRNAs reduces sfRNA association with RNA decay machinery in P-bodies, which coincides with increased viral RNA decay. These findings establish a functional role for RLBs in enhancing the cell-mediated decay of viral RNA by sequestering functional viral RNA decay products.

17.
Cell Rep Methods ; 4(8): 100832, 2024 Aug 19.
Article in English | MEDLINE | ID: mdl-39111313

ABSTRACT

Existing models of the human skin have aided our understanding of skin health and disease. However, they currently lack a microbial component, despite microbes' demonstrated connections to various skin diseases. Here, we present a robust, standardized model of the skin microbial community (SkinCom) to support in vitro and in vivo investigations. Our methods lead to the formation of an accurate, reproducible, and diverse community of aerobic and anaerobic bacteria. Subsequent testing of SkinCom on the dorsal skin of mice allowed for DNA and RNA recovery from both the applied SkinCom and the dorsal skin, highlighting its practicality for in vivo studies and -omics analyses. Furthermore, 66% of the responses to common cosmetic chemicals in vitro were in agreement with a human trial. Therefore, SkinCom represents a valuable, standardized tool for investigating microbe-metabolite interactions and facilitates the experimental design of in vivo studies targeting host-microbe relationships.


Subject(s)
Bacteria , Host Microbial Interactions , Microbiota , Models, Biological , Skin , Skin/microbiology , Microbiota/drug effects , Humans , Animals , Mice , Bacteria/drug effects , Cosmetics/pharmacology , Host Microbial Interactions/drug effects
18.
Cell Rep ; 43(8): 114493, 2024 Aug 27.
Article in English | MEDLINE | ID: mdl-39028622

ABSTRACT

Severe malnutrition is associated with infections, namely lower respiratory tract infections (LRTIs), diarrhea, and sepsis, and underlies the high risk of morbidity and mortality in children under 5 years of age. Dysregulations in neutrophil responses in the acute phase of infection are speculated to underlie these severe adverse outcomes; however, very little is known about their biology in this context. Here, in a lipopolysaccharide-challenged low-protein diet (LPD) mouse model, as a model of malnutrition, we show that protein deficiency disrupts neutrophil mitochondrial dynamics and ATP generation to obstruct the neutrophil differentiation cascade. This promotes the accumulation of atypical immature neutrophils that are incapable of optimal antimicrobial response and, in turn, exacerbate systemic pathogen spread and the permeability of the alveolocapillary membrane with the resultant lung damage. Thus, this perturbed response may contribute to higher mortality risk in malnutrition. We also offer a nutritional therapeutic strategy, nicotinamide, to boost neutrophil-mediated immunity in LPD-fed mice.


Subject(s)
Mitochondria , Neutrophils , Animals , Neutrophils/metabolism , Neutrophils/immunology , Mitochondria/metabolism , Mice , Mice, Inbred C57BL , Lipopolysaccharides/pharmacology , Adenosine Triphosphate/metabolism , Cell Differentiation , Protein Deficiency/metabolism , Male
19.
Cell Rep ; 43(7): 114435, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985673

ABSTRACT

Cell membranes mediate interactions between life and its environment, with lipids determining their properties. Understanding how cells adjust their lipidomes to tune membrane properties is crucial yet poorly defined due to the complexity of most organisms. We used quantitative shotgun lipidomics to study temperature adaptation in the simple organism Mycoplasma mycoides and the minimal cell JCVI-syn3B. We show that lipid abundances follow a universal logarithmic distribution across eukaryotes and bacteria, with comparable degrees of lipid remodeling for adaptation regardless of lipidomic or organismal complexity. Lipid features analysis demonstrates head-group-specific acyl chain remodeling as characteristic of lipidome adaptation; its deficiency in Syn3B is associated with impaired homeoviscous adaptation. Temporal analysis reveals a two-stage cold adaptation process: swift cholesterol and cardiolipin shifts followed by gradual acyl chain modifications. This work provides an in-depth analysis of lipidome adaptation in minimal cells, laying a foundation to probe the design principles of living membranes.


Subject(s)
Adaptation, Physiological , Lipidomics , Mycoplasma mycoides , Temperature , Mycoplasma mycoides/metabolism , Lipids/chemistry , Lipid Metabolism , Cholesterol/metabolism
20.
Cell Rep ; 43(7): 114478, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-38985668

ABSTRACT

Lyssavirus is a kind of neurotropic pathogen that needs to evade peripheral host immunity to enter the central nervous system to accomplish infection. NLRP3 inflammasome activation is essential for the host to defend against pathogen invasion. This study demonstrates that the matrix protein (M) of lyssavirus can inhibit both the priming step and the activation step of NLRP3 inflammasome activation. Specifically, M of lyssavirus can compete with NEK7 for binding to NLRP3, which restricts downstream apoptosis-associated speck-like protein containing a CARD (ASC) oligomerization. The serine amino acid at the 158th site of M among lyssavirus is critical for restricting ASC oligomerization. Moreover, recombinant lab-attenuated lyssavirus rabies (rabies lyssavirus [RABV]) with G158S mutation at M decreases interleukin-1ß (IL-1ß) production in bone-marrow-derived dendritic cells (BMDCs) to facilitate lyssavirus invasion into the brain thereby elevating pathogenicity in mice. Taken together, this study reveals a common mechanism by which lyssavirus inhibits NLRP3 inflammasome activation to evade host defenses.


Subject(s)
Dendritic Cells , Inflammasomes , Interleukin-1beta , Mice, Inbred C57BL , NLR Family, Pyrin Domain-Containing 3 Protein , Viral Matrix Proteins , NLR Family, Pyrin Domain-Containing 3 Protein/metabolism , Animals , Inflammasomes/metabolism , Inflammasomes/immunology , Mice , Viral Matrix Proteins/metabolism , Humans , Interleukin-1beta/metabolism , Dendritic Cells/metabolism , Dendritic Cells/immunology , Dendritic Cells/virology , Lyssavirus/metabolism , Lyssavirus/immunology , NIMA-Related Kinases/metabolism , Protein Binding , CARD Signaling Adaptor Proteins/metabolism , HEK293 Cells
SELECTION OF CITATIONS
SEARCH DETAIL