Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 260
Filter
Add more filters

Publication year range
1.
Annu Rev Immunol ; 40: 295-321, 2022 04 26.
Article in English | MEDLINE | ID: mdl-35471841

ABSTRACT

Macrophages are first responders for the immune system. In this role, they have both effector functions for neutralizing pathogens and sentinel functions for alerting other immune cells of diverse pathologic threats, thereby initiating and coordinating a multipronged immune response. Macrophages are distributed throughout the body-they circulate in the blood, line the mucosal membranes, reside within organs, and survey the connective tissue. Several reviews have summarized their diverse roles in different physiological scenarios and in the initiation or amplification of different pathologies. In this review, we propose that both the effector and the sentinel functions of healthy macrophages rely on three hallmark properties: response specificity, context dependence, and stimulus memory. When these hallmark properties are diminished, the macrophage's biological functions are impaired, which in turn results in increased risk for immune dysregulation, manifested by immune deficiency or autoimmunity. We review the evidence and the molecular mechanisms supporting these functional hallmarks.


Subject(s)
Immunity, Innate , Macrophages , Animals , Humans
2.
Genes Dev ; 37(9-10): 351-353, 2023 05 01.
Article in English | MEDLINE | ID: mdl-37253615

ABSTRACT

The core promoter determines not only where gene transcription initiates but also the transcriptional activity in both basal and enhancer-induced conditions. Multiple short sequence elements within the core promoter have been identified in different species, but how they function together and to what extent they are truly species-specific has remained unclear. In this issue of Genes & Development, Vo ngoc and colleagues (pp. 377-382) report undertaking massively parallel measurements of synthetic core promoters to generate a large data set of their activities that informs a statistical learning model to identify the sequence differences of human and Drosophila core promoters. This machine learning model was then applied to design gene core promoters that are particularly specific for the human transcriptional machinery.


Subject(s)
Artificial Intelligence , Drosophila Proteins , Animals , Humans , Promoter Regions, Genetic/genetics , Drosophila/genetics , Drosophila/metabolism , Drosophila Proteins/metabolism , Transcription, Genetic
3.
Immunity ; 54(9): 1915-1932, 2021 09 14.
Article in English | MEDLINE | ID: mdl-34525335

ABSTRACT

Immune sentinel cells initiate immune responses to pathogens and tissue injury and are capable of producing highly stimulus-specific responses. Insight into the mechanisms underlying such specificity has come from the identification of regulatory factors and biochemical pathways, as well as the definition of signaling circuits that enable combinatorial and temporal coding of information. Here, we review the multi-layered molecular mechanisms that underlie stimulus-specific gene expression in macrophages. We categorize components of inflammatory and anti-pathogenic signaling pathways into five layers of regulatory control and discuss unifying mechanisms determining signaling characteristics at each layer. In this context, we review mechanisms that enable combinatorial and temporal encoding of information, identify recurring regulatory motifs and principles, and present strategies for integrating experimental and computational approaches toward the understanding of signaling specificity in innate immunity.


Subject(s)
Immunity, Innate/immunology , Macrophages/immunology , Animals , Humans
4.
Immunity ; 54(5): 916-930.e7, 2021 05 11.
Article in English | MEDLINE | ID: mdl-33979588

ABSTRACT

Macrophages initiate inflammatory responses via the transcription factor NFκB. The temporal pattern of NFκB activity determines which genes are expressed and thus, the type of response that ensues. Here, we examined how information about the stimulus is encoded in the dynamics of NFκB activity. We generated an mVenus-RelA reporter mouse line to enable high-throughput live-cell analysis of primary macrophages responding to host- and pathogen-derived stimuli. An information-theoretic workflow identified six dynamical features-termed signaling codons-that convey stimulus information to the nucleus. In particular, oscillatory trajectories were a hallmark of responses to cytokine but not pathogen-derived stimuli. Single-cell imaging and RNA sequencing of macrophages from a mouse model of Sjögren's syndrome revealed inappropriate responses to stimuli, suggestive of confusion of two NFκB signaling codons. Thus, the dynamics of NFκB signaling classify immune threats through six signaling codons, and signal confusion based on defective codon deployment may underlie the etiology of some inflammatory diseases.


Subject(s)
Codon/genetics , Macrophages/physiology , NF-kappa B/genetics , Signal Transduction/genetics , Animals , Cells, Cultured , Cytokines/genetics , Disease Models, Animal , Gene Expression Regulation/genetics , Inflammation/genetics , Mice , Mice, Inbred C57BL , Sjogren's Syndrome/genetics , Transcription Factor RelA/genetics
5.
Immunity ; 50(3): 616-628.e6, 2019 03 19.
Article in English | MEDLINE | ID: mdl-30850343

ABSTRACT

Humoral immunity depends on efficient activation of B cells and their subsequent differentiation into antibody-secreting cells (ASCs). The transcription factor NFκB cRel is critical for B cell proliferation, but incorporating its known regulatory interactions into a mathematical model of the ASC differentiation circuit prevented ASC generation in simulations. Indeed, experimental ectopic cRel expression blocked ASC differentiation by inhibiting the transcription factor Blimp1, and in wild-type (WT) cells cRel was dynamically repressed during ASC differentiation by Blimp1 binding the Rel locus. Including this bi-stable circuit of mutual cRel-Blimp1 antagonism into a multi-scale model revealed that dynamic repression of cRel controls the switch from B cell proliferation to ASC generation phases and hence the respective cell population dynamics. Our studies provide a mechanistic explanation of how dysregulation of this bi-stable circuit might result in pathologic B cell population phenotypes and thus offer new avenues for diagnostic stratification and treatment.


Subject(s)
B-Lymphocytes/immunology , Cell Differentiation/immunology , Cell Proliferation/physiology , NF-kappa B/immunology , Animals , Antibody-Producing Cells/immunology , Cell Line , Female , Gene Expression Regulation/immunology , HEK293 Cells , Humans , Immunity, Humoral/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred C57BL
6.
Cell ; 155(2): 448-61, 2013 Oct 10.
Article in English | MEDLINE | ID: mdl-24120141

ABSTRACT

Highly networked signaling hubs are often associated with disease, but targeting them pharmacologically has largely been unsuccessful in the clinic because of their functional pleiotropy. Motivated by the hypothesis that a dynamic signaling code confers functional specificity, we investigated whether dynamic features may be targeted pharmacologically to achieve therapeutic specificity. With a virtual screen, we identified combinations of signaling hub topologies and dynamic signal profiles that are amenable to selective inhibition. Mathematical analysis revealed principles that may guide stimulus-specific inhibition of signaling hubs, even in the absence of detailed mathematical models. Using the NFκB signaling module as a test bed, we identified perturbations that selectively affect the response to cytokines or pathogen components. Together, our results demonstrate that the dynamics of signaling may serve as a pharmacological target, and we reveal principles that delineate the opportunities and constraints of developing stimulus-specific therapeutic agents aimed at pleiotropic signaling hubs.


Subject(s)
Molecular Targeted Therapy , Signal Transduction/drug effects , Animals , Computer Simulation , Drug Discovery , Drug Evaluation, Preclinical , Humans , NF-kappa B/metabolism
7.
Cell ; 149(3): 630-41, 2012 Apr 27.
Article in English | MEDLINE | ID: mdl-22541433

ABSTRACT

In female mouse embryos, somatic cells undergo a random form of X chromosome inactivation (XCI), whereas extraembryonic trophoblast cells in the placenta undergo imprinted XCI, silencing exclusively the paternal X chromosome. Initiation of imprinted XCI requires a functional maternal allele of the X-linked gene Rnf12, which encodes the ubiquitin ligase Rnf12/RLIM. We find that knockout (KO) of Rnf12 in female mammary glands inhibits alveolar differentiation and milk production upon pregnancy, with alveolar cells that lack RLIM undergoing apoptosis as they begin to differentiate. Genetic analyses demonstrate that these functions are mediated primarily by the paternal Rnf12 allele due to nonrandom maternal XCI in mammary epithelial cells. These results identify paternal Rnf12/RLIM as a critical survival factor for milk-producing alveolar cells and, together with population models, reveal implications of transgenerational epigenetic inheritance.


Subject(s)
Cell Survival , Mammary Glands, Animal/cytology , Ubiquitin-Protein Ligases/metabolism , Animals , Epigenesis, Genetic , Female , Genomic Imprinting , Male , Mammary Glands, Animal/physiology , Mice , Pregnancy , Ubiquitin-Protein Ligases/genetics , X Chromosome Inactivation
8.
Proc Natl Acad Sci U S A ; 121(30): e2309686121, 2024 Jul 23.
Article in English | MEDLINE | ID: mdl-39024115

ABSTRACT

Antibody responses require the proliferative expansion of B cells controlled by affinity-dependent signals. Yet, proliferative bursts are heterogeneous, varying between 0 and 8 divisions in response to the same stimulus. NFκB cRel is activated in response to immune stimulation in B cells and is genetically required for proliferation. Here, we asked whether proliferative heterogeneity is controlled by natural variations in cRel abundance. We developed a fluorescent reporter mTFP1-cRel for the direct observation of cRel in live proliferating B cells. We found that cRel is heterogeneously distributed among naïve B cells, which are enriched for high expressors in a heavy-tailed distribution. We found that high cRel expressors show faster activation of the proliferative program, but do not sustain it well, with population expansion decaying earlier. With a mathematical model of the molecular network, we showed that cRel heterogeneity arises from balancing positive feedback by autoregulation and negative feedback by its inhibitor IκBε, confirmed by mouse knockouts. Using live-cell fluorescence microscopy, we showed that increased cRel primes B cells for early proliferation via higher basal expression of the cell cycle driver cMyc. However, peak cMyc induction amplitude is constrained by incoherent feedforward regulation, decoding the fold change of cRel activity to terminate the proliferative burst. This results in a complex nonlinear, nonmonotonic relationship between cRel expression and the extent of proliferation. These findings emphasize the importance of direct observational studies to complement gene knockout results and to learn about quantitative relationships between biological processes and their key regulators in the context of natural variations.


Subject(s)
B-Lymphocytes , Cell Proliferation , NF-kappa B , Animals , B-Lymphocytes/immunology , B-Lymphocytes/metabolism , Mice , NF-kappa B/metabolism , Mice, Knockout , Mice, Inbred C57BL , Proto-Oncogene Proteins c-rel/metabolism , Proto-Oncogene Proteins c-rel/genetics
9.
Mol Syst Biol ; 20(8): 898-932, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38872050

ABSTRACT

Macrophages sense pathogens and orchestrate specific immune responses. Stimulus specificity is thought to be achieved through combinatorial and dynamical coding by signaling pathways. While NFκB dynamics are known to encode stimulus information, dynamical coding in other signaling pathways and their combinatorial coordination remain unclear. Here, we established live-cell microscopy to investigate how NFκB and p38 dynamics interface in stimulated macrophages. Information theory and machine learning revealed that p38 dynamics distinguish cytokine TNF from pathogen-associated molecular patterns and high doses from low, but contributed little to information-rich NFκB dynamics when both pathways are considered. This suggests that immune response genes benefit from decoding immune signaling dynamics or combinatorics, but not both. We found that the heterogeneity of the two pathways is surprisingly uncorrelated. Mathematical modeling revealed potential sources of uncorrelated heterogeneity in the branched pathway network topology and predicted it to drive gene expression variability. Indeed, genes dependent on both p38 and NFκB showed high scRNAseq variability and bimodality. These results identify combinatorial signaling as a mechanism to restrict NFκB-AND-p38-responsive inflammatory cytokine expression to few cells.


Subject(s)
Inflammation , Macrophages , NF-kappa B , Signal Transduction , p38 Mitogen-Activated Protein Kinases , p38 Mitogen-Activated Protein Kinases/metabolism , NF-kappa B/metabolism , Macrophages/metabolism , Macrophages/immunology , Inflammation/metabolism , Inflammation/genetics , Inflammation/immunology , Animals , Mice , Cytokines/metabolism , Machine Learning , Gene Expression Regulation , Humans , Tumor Necrosis Factor-alpha/metabolism
10.
EMBO Rep ; 24(7): e55986, 2023 Jul 05.
Article in English | MEDLINE | ID: mdl-37212045

ABSTRACT

Tumor necrosis factor (TNF) is a key inflammatory cytokine that warns recipient cells of a nearby infection or tissue damage. Acute exposure to TNF activates characteristic oscillatory dynamics of the transcription factor NFκB and induces a characteristic gene expression program; these are distinct from the responses of cells directly exposed to pathogen-associated molecular patterns (PAMPs). Here, we report that tonic TNF exposure is critical for safeguarding TNF's specific functions. In the absence of tonic TNF conditioning, acute exposure to TNF causes (i) NFκB signaling dynamics that are less oscillatory and more like PAMP-responsive NFκB dynamics, (ii) immune gene expression that is more similar to the Pam3CSK4 response program, and (iii) broader epigenomic reprogramming that is characteristic of PAMP-responsive changes. We show that the absence of tonic TNF signaling effects subtle changes to TNF receptor availability and dynamics such that enhanced pathway activity results in non-oscillatory NFκB. Our results reveal tonic TNF as a key tissue determinant of the specific cellular responses to acute paracrine TNF exposure, and their distinction from responses to direct exposure to PAMPs.


Subject(s)
Pathogen-Associated Molecular Pattern Molecules , Tumor Necrosis Factor-alpha , Pathogen-Associated Molecular Pattern Molecules/metabolism , Tumor Necrosis Factor-alpha/pharmacology , Tumor Necrosis Factor-alpha/metabolism , Signal Transduction , NF-kappa B/metabolism , Macrophages/metabolism
11.
Proc Natl Acad Sci U S A ; 119(4)2022 01 25.
Article in English | MEDLINE | ID: mdl-35058362

ABSTRACT

Immune cells infiltrate the peripheral nervous system (PNS) after injury and with autoimmunity, but their net effect is divergent. After injury, immune cells are reparative, while in inflammatory neuropathies (e.g., Guillain Barré Syndrome and chronic inflammatory demyelinating polyneuropathy), immune cells are proinflammatory and promote autoimmune demyelination. An understanding of immune cell phenotypes that distinguish these conditions may, therefore, reveal new therapeutic targets for switching immune cells from an inflammatory role to a reparative state. In an autoimmune regulator (Aire)-deficient mouse model of inflammatory neuropathy, we used single-cell RNA sequencing of sciatic nerves to discover a transcriptionally heterogeneous cellular landscape, including multiple myeloid, innate lymphoid, and lymphoid cell types. Analysis of cell-cell ligand-receptor interactions uncovered a macrophage-mediated tumor necrosis factor-α (TNF-α) signaling axis that is induced by interferon-γ and required for initiation of autoimmune demyelination. Developmental trajectory visualization suggested that TNF-α signaling is associated with metabolic reprogramming of macrophages and polarization of macrophages from a reparative state in injury to a pathogenic, inflammatory state in autoimmunity. Autocrine TNF-α signaling induced macrophage expression of multiple genes (Clec4e, Marcksl1, Cxcl1, and Cxcl10) important in immune cell activation and recruitment. Genetic and antibody-based blockade of TNF-α/TNF-α signaling ameliorated clinical neuropathy, peripheral nerve infiltration, and demyelination, which provides preclinical evidence that the TNF-α axis may be effectively targeted to resolve inflammatory neuropathies.


Subject(s)
Neuroinflammatory Diseases/etiology , Neuroinflammatory Diseases/metabolism , Peripheral Nervous System Diseases/etiology , Peripheral Nervous System Diseases/metabolism , Polyendocrinopathies, Autoimmune/complications , Tumor Necrosis Factor-alpha/metabolism , Adoptive Transfer , Animals , Antibodies, Monoclonal/pharmacology , Autocrine Communication , Biomarkers , Cytokines/metabolism , Disease Models, Animal , Disease Susceptibility , Gene Expression Profiling , Inflammation Mediators/metabolism , Macrophages/immunology , Macrophages/metabolism , Mice , Mice, Transgenic , Neuroinflammatory Diseases/drug therapy , Neuroinflammatory Diseases/pathology , Paracrine Communication , Peripheral Nervous System Diseases/drug therapy , Peripheral Nervous System Diseases/pathology , Polyendocrinopathies, Autoimmune/genetics , Receptors, Tumor Necrosis Factor/deficiency , Sciatic Nerve/immunology , Sciatic Nerve/metabolism , Sciatic Nerve/pathology , Signal Transduction , Tumor Necrosis Factor-alpha/antagonists & inhibitors
12.
Mol Syst Biol ; 19(5): e11294, 2023 05 09.
Article in English | MEDLINE | ID: mdl-36929731

ABSTRACT

Type I interferons (IFN) induce powerful antiviral and innate immune responses via the transcription factor, IFN-stimulated gene factor (ISGF3). However, in some pathological contexts, type I IFNs are responsible for exacerbating inflammation. Here, we show that a high dose of IFN-ß also activates an inflammatory gene expression program in contrast to IFN-λ3, a type III IFN, which elicits only the common antiviral gene program. We show that the inflammatory gene program depends on a second, potentiated phase in ISGF3 activation. Iterating between mathematical modeling and experimental analysis, we show that the ISGF3 activation network may engage a positive feedback loop with its subunits IRF9 and STAT2. This network motif mediates stimulus-specific ISGF3 dynamics that are dependent on ligand, dose, and duration of exposure, and when engaged activates the inflammatory gene expression program. Our results reveal a previously underappreciated dynamical control of the JAK-STAT/IRF signaling network that may produce distinct biological responses and suggest that studies of type I IFN dysregulation, and in turn therapeutic remedies, may focus on feedback regulators within it.


Subject(s)
Gene Expression Regulation , Transcription Factors , Feedback , Antiviral Agents , Signal Transduction
13.
Nat Immunol ; 13(12): 1162-70, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23086447

ABSTRACT

The NF-κB protein RelB controls dendritic cell (DC) maturation and may be targeted therapeutically to manipulate T cell responses in disease. Here we report that RelB promoted DC activation not as the expected RelB-p52 effector of the noncanonical NF-κB pathway, but as a RelB-p50 dimer regulated by canonical IκBs, IκBα and IκBɛ. IκB control of RelB minimized spontaneous maturation but enabled rapid pathogen-responsive maturation. Computational modeling of the NF-κB signaling module identified control points of this unexpected cell type-specific regulation. Fibroblasts that we engineered accordingly showed DC-like RelB control. Canonical pathway control of RelB regulated pathogen-responsive gene expression programs. This work illustrates the potential utility of systems analyses in guiding the development of combination therapeutics for modulating DC-dependent T cell responses.


Subject(s)
Dendritic Cells/immunology , Dendritic Cells/metabolism , Lymphocyte Activation , NF-kappa B/metabolism , Transcription Factor RelB/metabolism , Animals , Cell Differentiation/genetics , Cell Line , Fibroblasts/metabolism , Gene Expression Regulation , I-kappa B Kinase/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , NF-kappa B/genetics , Protein Multimerization , Signal Transduction , Toll-Like Receptor 2/metabolism , Toll-Like Receptor 4/metabolism , Toll-Like Receptor 9/metabolism , Transcription Factor RelB/genetics
14.
Chemistry ; 30(45): e202400580, 2024 Aug 12.
Article in English | MEDLINE | ID: mdl-38838081

ABSTRACT

We report the syntheses of tin(II) salts of the types [L1SnX]SnX3 [L1=2,6-{(i-PrO)2(O)P}2C5H3N: 1, X=Cl; 2, X=Br], [L2SnCl]SnCl3 [L2=2-{(i-PrO)Ph(O)P}-6-{(i-PrO)2(O)P}C5H3N: 3], [L3SnX]SnX3 [L3=2,6-{MeO(O)C}2C5H3N: 4, X=Cl; 5, X=Br], [L4SnX]SnX3 [L4=2,6-{Et2N(O)C}2C5H3N: 6, X=Cl; 7, X=Br]. These compounds were obtained by addition of SnX2 to the corresponding ligand inducing autoionization of the respective tin(II) halide. The thermal stability of 1, 3, and 4 was elucidated, giving, under ester cleavage and cyclisation, the tin(II) derivatives 8-12. The reaction of [L1SnCl]SnCl3 (1) with W(CO)4(thf)2 afforded the tungsten tetracarbonyl complex [{L1SnCl}{SnCl3}W(CO)4] (13), representing the first example in which a tin(II) stannate anion and a tin(II) stannylium cation simultaneously coordinate to a transition metal centre. The compounds were characterized by single crystal X-ray diffraction analyses and in part by elemental analyses, IR and NMR spectroscopy, electrospray ionization mass spectrometry. DFT calculations accompany the experimental work.

15.
Cell ; 138(1): 114-28, 2009 Jul 10.
Article in English | MEDLINE | ID: mdl-19596239

ABSTRACT

We describe a broad mechanistic framework for the transcriptional induction of mammalian primary response genes by Toll-like receptors and other stimuli. One major class of primary response genes is characterized by CpG-island promoters, which facilitate promiscuous induction from constitutively active chromatin without a requirement for SWI/SNF nucleosome remodeling complexes. The low nucleosome occupancy at promoters in this class can be attributed to the assembly of CpG islands into unstable nucleosomes, which may lead to SWI/SNF independence. Another major class consists of non-CpG-island promoters that assemble into stable nucleosomes, resulting in SWI/SNF dependence and a requirement for transcription factors that promote selective nucleosome remodeling. Some stimuli, including serum and tumor necrosis factor-alpha, exhibit a strong bias toward activation of SWI/SNF-independent CpG-island genes. In contrast, interferon-beta is strongly biased toward SWI/SNF-dependent non-CpG-island genes. By activating a diverse set of transcription factors, Toll-like receptors induce both classes and others for an optimal response to microbial pathogens.


Subject(s)
Chromatin Assembly and Disassembly , CpG Islands , Transcriptional Activation , Animals , CD4-Positive T-Lymphocytes/metabolism , Chromosomal Proteins, Non-Histone/metabolism , Humans , Interferon Regulatory Factor-3/metabolism , Lipopolysaccharides/immunology , Mice , Nucleosomes/metabolism , Promoter Regions, Genetic , Transcription Factors/metabolism , Transcription, Genetic
16.
Proc Natl Acad Sci U S A ; 118(10)2021 03 09.
Article in English | MEDLINE | ID: mdl-33649198

ABSTRACT

Persistent methicillin-resistant Staphylococcus aureus (MRSA) bacteremia is life threatening and occurs in up to 30% of MRSA bacteremia cases despite appropriate antimicrobial therapy. Isolates of MRSA that cause antibiotic-persistent methicillin-resistant S. aureus bacteremia (APMB) typically have in vitro antibiotic susceptibilities equivalent to those causing antibiotic-resolving methicillin-resistant S. aureus bacteremia (ARMB). Thus, persistence reflects host-pathogen interactions occurring uniquely in context of antibiotic therapy in vivo. However, host factors and mechanisms involved in APMB remain unclear. We compared DNA methylomes in circulating immune cells from patients experiencing APMB vs. ARMB. Overall, methylation signatures diverged in the distinct patient cohorts. Differentially methylated sites intensified proximate to transcription factor binding sites, primarily in enhancer regions. In APMB patients, significant hypomethylation was observed in binding sites for CCAAT enhancer binding protein-ß (C/EBPß) and signal transducer/activator of transcription 1 (STAT1). In contrast, hypomethylation in ARMB patients localized to glucocorticoid receptor and histone acetyltransferase p300 binding sites. These distinct methylation signatures were enriched in neutrophils and achieved a mean area under the curve of 0.85 when used to predict APMB using a classification model. These findings validated by targeted bisulfite sequencing (TBS-seq) differentiate epigenotypes in patients experiencing APMB vs. ARMB and suggest a risk stratification strategy for antibiotic persistence in patients treated for MRSA bacteremia.


Subject(s)
Bacteremia/metabolism , DNA Methylation , Methicillin-Resistant Staphylococcus aureus/metabolism , Response Elements , Staphylococcal Infections/metabolism , Anti-Bacterial Agents/administration & dosage , Bacteremia/drug therapy , CCAAT-Enhancer-Binding Protein-beta/metabolism , Female , Humans , Male , Middle Aged , STAT1 Transcription Factor/metabolism , Staphylococcal Infections/drug therapy , p300-CBP Transcription Factors/metabolism
17.
J Allergy Clin Immunol ; 152(5): 1261-1272, 2023 11.
Article in English | MEDLINE | ID: mdl-37460023

ABSTRACT

BACKGROUND: Autoimmune diseases are leading causes of ill health and morbidity and have diverse etiology. Two signaling pathways are key drivers of autoimmune pathology, interferon and nuclear factor-κB (NF-κB)/RelA, defining the 2 broad labels of interferonopathies and relopathies. Prior work has established that genetic loss of function of the NF-κB subunit RelB leads to autoimmune and inflammatory pathology in mice and humans. OBJECTIVE: We sought to characterize RelB-deficient autoimmunity by unbiased profiling of the responses of immune sentinel cells to stimulus and to determine the functional role of dysregulated gene programs in the RelB-deficient pathology. METHODS: Transcriptomic profiling was performed on fibroblasts and dendritic cells derived from patients with RelB deficiency and knockout mice, and transcriptomic responses and pathology were assessed in mice deficient in both RelB and the type I interferon receptor. RESULTS: We found that loss of RelB in patient-derived fibroblasts and mouse myeloid cells results in elevated induction of hundreds of interferon-stimulated genes. Removing hyperexpression of the interferon-stimulated gene program did not ameliorate the autoimmune pathology of RelB knockout mice. Instead, we found that RelB suppresses a different set of inflammatory response genes in a manner that is independent of interferon signaling but associated with NF-κB binding motifs. CONCLUSION: Although transcriptomic profiling would describe RelB-deficient autoimmune disease as an interferonopathy, the genetic evidence indicates that the pathology in mice is interferon-independent.


Subject(s)
Autoimmune Diseases , NF-kappa B , Animals , Humans , Mice , Autoimmune Diseases/genetics , Interferons/genetics , Mice, Knockout , NF-kappa B/metabolism , Signal Transduction , Transcription Factor RelB/genetics
18.
J Infect Dis ; 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38001039

ABSTRACT

BACKGROUND: Clinical outcomes in bacterial bloodstream infections (BSI) are influenced by multiple factors, including bacterial species, host immunity, and antibiotic therapy. However, the mechanisms by which such factors influence outcomes and their potential biomarkers are poorly understood. We aimed to identify bacterial- and antibiotic-specific host transcriptional signatures in patients with bacterial BSI. METHODS: RNA-Seq was performed on blood from patients with BSI due to prototypic Gram-negative vs. Gram-positive pathogens: Escherichia coli (n = 30) or Klebsiella pneumoniae (n = 28) vs. methicillin-susceptible Staphylococcus aureus [MSSA] (n = 24) or methicillin-resistant S. aureus (MRSA) (n = 58). Patients were matched by age, gender, and race. RESULTS: No significant host transcriptome differences were detected in patients with E. coli versus K. pneumoniae BSI, so these were considered together as Gram-negative BSI. Relative to S. aureus BSI, patients with Gram-negative BSI had increased activation of the classical complement system. However, the most significant signal was a reduction in host transcriptional signatures involving mitochondrial energy transduction and oxidative burst in MRSA vs. MSSA. This attenuated host transcriptional signature remained after controlling for antibiotic therapy. CONCLUSIONS: Given importance of immune cellular energetics and reactive oxygen species in eliminating hematogenous or intracellular MRSA, these findings may offer insights into its persistence relative to other bacterial BSI.

19.
Biophys J ; 122(5): 817-834, 2023 03 07.
Article in English | MEDLINE | ID: mdl-36710493

ABSTRACT

Necroptosis is a form of regulated cell death associated with degenerative disorders, autoimmune and inflammatory diseases, and cancer. To better understand the biochemical mechanisms regulating necroptosis, we constructed a detailed computational model of tumor necrosis factor-induced necroptosis based on known molecular interactions from the literature. Intracellular protein levels, used as model inputs, were quantified using label-free mass spectrometry, and the model was calibrated using Bayesian parameter inference to experimental protein time course data from a well-established necroptosis-executing cell line. The calibrated model reproduced the dynamics of phosphorylated mixed lineage kinase domain-like protein, an established necroptosis reporter. A subsequent dynamical systems analysis identified four distinct modes of necroptosis signal execution, distinguished by rate constant values and the roles of the RIP1 deubiquitinating enzymes A20 and CYLD. In one case, A20 and CYLD both contribute to RIP1 deubiquitination, in another RIP1 deubiquitination is driven exclusively by CYLD, and in two modes either A20 or CYLD acts as the driver with the other enzyme, counterintuitively, inhibiting necroptosis. We also performed sensitivity analyses of initial protein concentrations and rate constants to identify potential targets for modulating necroptosis sensitivity within each mode. We conclude by associating numerous contrasting and, in some cases, counterintuitive experimental results reported in the literature with one or more of the model-predicted modes of necroptosis execution. In all, we demonstrate that a consensus pathway model of tumor necrosis factor-induced necroptosis can provide insights into unresolved controversies regarding the molecular mechanisms driving necroptosis execution in numerous cell types under different experimental conditions.


Subject(s)
Cues , Necroptosis , Humans , Necrosis/metabolism , Necrosis/pathology , Bayes Theorem , Tumor Necrosis Factor-alpha/pharmacology , Apoptosis
20.
Semin Cell Dev Biol ; 115: 27-36, 2021 07.
Article in English | MEDLINE | ID: mdl-33386235

ABSTRACT

The control over iron availability is crucial under homeostatic conditions and even more in the case of an infection. This results from diverse properties of iron: first, iron is an important trace element for the host as well as for the pathogen for various cellular and metabolic processes, second, free iron catalyzes Fenton reaction and is therefore producing reactive oxygen species as a part of the host defense machinery, third, iron exhibits important effects on immune cell function and differentiation and fourth almost every immune activation in turn impacts on iron metabolism and spatio-temporal iron distribution. The central importance of iron in the host and microbe interplay and thus for the course of infections led to diverse strategies to restrict iron for invading pathogens. In this review, we focus on how iron restriction to the pathogen is a powerful innate immune defense mechanism of the host called "nutritional immunity". Important proteins in the iron-host-pathogen interplay will be discussed as well as the influence of iron on the efficacy of innate and adaptive immunity. Recently described processes like ferritinophagy and ferroptosis are further covered in respect to their impact on inflammation and infection control and how they impact on our understanding of the interaction of host and pathogen.


Subject(s)
Host-Pathogen Interactions/immunology , Immunity, Innate/immunology , Iron/metabolism , Humans
SELECTION OF CITATIONS
SEARCH DETAIL