Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 111
Filter
Add more filters

Publication year range
1.
Cell ; 165(3): 551-65, 2016 Apr 21.
Article in English | MEDLINE | ID: mdl-27040498

ABSTRACT

Systemic lupus erythematosus (SLE) is an autoimmune disease characterized by loss of tolerance to nucleic acids and highly diverse clinical manifestations. To assess its molecular heterogeneity, we longitudinally profiled the blood transcriptome of 158 pediatric patients. Using mixed models accounting for repeated measurements, demographics, treatment, disease activity (DA), and nephritis class, we confirmed a prevalent IFN signature and identified a plasmablast signature as the most robust biomarker of DA. We detected gradual enrichment of neutrophil transcripts during progression to active nephritis and distinct signatures in response to treatment in different nephritis subclasses. Importantly, personalized immunomonitoring uncovered individual correlates of disease activity that enabled patient stratification into seven groups, supported by patient genotypes. Our study uncovers the molecular heterogeneity of SLE and provides an explanation for the failure of clinical trials. This approach may improve trial design and implementation of tailored therapies in genetically and clinically complex autoimmune diseases. PAPERCLIP.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Adolescent , Child , Female , Humans , Longitudinal Studies , Lupus Erythematosus, Systemic/immunology , Lupus Erythematosus, Systemic/pathology , Lupus Erythematosus, Systemic/therapy , Lupus Nephritis/genetics , Lupus Nephritis/immunology , Neutrophils/immunology , Polymorphism, Single Nucleotide , Precision Medicine , Transcriptome
2.
Nat Immunol ; 17(5): 495-504, 2016 May.
Article in English | MEDLINE | ID: mdl-27019227

ABSTRACT

Aberrant nucleic acids generated during viral replication are the main trigger for antiviral immunity, and mutations that disrupt nucleic acid metabolism can lead to autoinflammatory disorders. Here we investigated the etiology of X-linked reticulate pigmentary disorder (XLPDR), a primary immunodeficiency with autoinflammatory features. We discovered that XLPDR is caused by an intronic mutation that disrupts the expression of POLA1, which encodes the catalytic subunit of DNA polymerase-α. Unexpectedly, POLA1 deficiency resulted in increased production of type I interferons. This enzyme is necessary for the synthesis of RNA:DNA primers during DNA replication and, strikingly, we found that POLA1 is also required for the synthesis of cytosolic RNA:DNA, which directly modulates interferon activation. Together this work identifies POLA1 as a critical regulator of the type I interferon response.


Subject(s)
DNA Polymerase I/metabolism , DNA/biosynthesis , Interferon Type I/metabolism , RNA/biosynthesis , Base Sequence , Cells, Cultured , Cytosol/metabolism , DNA/genetics , DNA Polymerase I/genetics , Family Health , Female , Fibroblasts/cytology , Fibroblasts/metabolism , Gene Expression Profiling , Genetic Diseases, X-Linked/genetics , Genetic Diseases, X-Linked/metabolism , HEK293 Cells , HeLa Cells , Humans , Immunoblotting , Male , Microscopy, Confocal , Mutation , Oligonucleotide Array Sequence Analysis , Pedigree , Pigmentation Disorders/genetics , Pigmentation Disorders/metabolism , RNA/genetics , Reverse Transcriptase Polymerase Chain Reaction
4.
Nat Immunol ; 14(1): 61-71, 2013 Jan.
Article in English | MEDLINE | ID: mdl-23160154

ABSTRACT

The sensing of viral nucleic acids by the innate immune system triggers the production of type I interferons, which activates interferon-stimulated genes (ISGs) and directs a multifaceted antiviral response. ISGs can also be activated through interferon-independent pathways, although the precise mechanisms remain elusive. Here we found that the cytosolic exonuclease Trex1 regulated the activation of a subset of ISGs independently of interferon. Both Trex1(-/-) mouse cells and Trex1-mutant human cells had high expression of genes encoding antiviral molecules ('antiviral genes') and were refractory to viral infection. The interferon-independent activation of antiviral genes in Trex1(-/-) cells required the adaptor STING, the kinase TBK1 and the transcription factors IRF3 and IRF7. We also found that Trex1-deficient cells had an expanded lysosomal compartment, altered subcellular localization of the transcription factor TFEB and diminished activity of the regulator mTORC1. Together our data identify Trex1 as a regulator of lysosomal biogenesis and interferon-independent activation of antiviral genes and show that dysregulation of lysosomes can elicit innate immune responses.


Subject(s)
Antigens, Viral/immunology , Exodeoxyribonucleases/metabolism , Lysosomes/physiology , Phosphoproteins/metabolism , RNA Virus Infections/immunology , RNA Viruses/immunology , Animals , Exodeoxyribonucleases/genetics , HeLa Cells , Humans , Immunity, Active/genetics , Interferons/immunology , Mice , Mice, Knockout , Mutation/genetics , Organelle Biogenesis , Phosphoproteins/genetics , RNA, Small Interfering/genetics
5.
Proc Natl Acad Sci U S A ; 118(7)2021 02 16.
Article in English | MEDLINE | ID: mdl-33568531

ABSTRACT

Recent studies have identified thousands of long noncoding RNAs (lncRNAs) in mammalian genomes that regulate gene expression in different biological processes. Although lncRNAs have been identified in a variety of immune cells and implicated in immune response, the biological function and mechanism of the majority remain unexplored, especially in sepsis. Here, we identify a role for a lncRNA-gastric adenocarcinoma predictive long intergenic noncoding RNA (GAPLINC)-previously characterized for its role in cancer, now in the context of innate immunity, macrophages, and LPS-induced endotoxic shock. Transcriptome analysis of macrophages from humans and mice reveals that GAPLINC is a conserved lncRNA that is highly expressed following macrophage differentiation. Upon inflammatory activation, GAPLINC is rapidly down-regulated. Macrophages depleted of GAPLINC display enhanced expression of inflammatory genes at baseline, while overexpression of GAPLINC suppresses this response. Consistent with GAPLINC-depleted cells, Gaplinc knockout mice display enhanced basal levels of inflammatory genes and show resistance to LPS-induced endotoxic shock. Mechanistically, survival is linked to increased levels of nuclear NF-κB in Gaplinc knockout mice that drives basal expression of target genes typically only activated following inflammatory stimulation. We show that this activation of immune response genes prior to LPS challenge leads to decreased blood clot formation, which protects Gaplinc knockout mice from multiorgan failure and death. Together, our results identify a previously unknown function for GAPLINC as a negative regulator of inflammation and uncover a key role for this lncRNA in modulating endotoxic shock.


Subject(s)
Immunity, Innate , Shock, Septic/immunology , Animals , Cells, Cultured , Female , Humans , Lipopolysaccharides/toxicity , Male , Mice , Mice, Inbred C57BL , NF-kappa B/metabolism , RNA, Long Noncoding/genetics , RNA, Long Noncoding/metabolism , Shock, Septic/etiology , Shock, Septic/genetics , THP-1 Cells , Transcriptome
6.
Ann Rheum Dis ; 81(12): 1712-1721, 2022 12.
Article in English | MEDLINE | ID: mdl-35977808

ABSTRACT

OBJECTIVES: Families that contain multiple siblings affected with childhood onset of systemic lupus erythematosus (SLE) likely have strong genetic predispositions. We performed whole exome sequencing (WES) to identify familial rare risk variants and to assess their effects in lupus. METHODS: Sanger sequencing validated the two ultra-rare, predicted pathogenic risk variants discovered by WES and identified additional variants in 562 additional patients with SLE. Effects of a splice site variant and a frameshift variant were assessed using a Minigene assay and CRISPR/Cas9-mediated knock-in (KI) mice, respectively. RESULTS: The two familial ultra-rare, predicted loss-of-function (LOF) SAT1 variants exhibited X-linked recessive Mendelian inheritance in two unrelated African-American families. Each LOF variant was transmitted from the heterozygous unaffected mother to her two sons with childhood-onset SLE. The p.Asp40Tyr variant affected a splice donor site causing deleterious transcripts. The young hemizygous male and homozygous female Sat1 p.Glu92Leufs*6 KI mice spontaneously developed splenomegaly, enlarged glomeruli with leucocyte infiltration, proteinuria and elevated expression of type I interferon-inducible genes. SAT1 is highly expressed in neutrophils and encodes spermidine/spermine-N1-acetyltransferase 1 (SSAT1), a rate-limiting enzyme in polyamine catabolism. Young male KI mice exhibited neutrophil defects and decreased proportions of Foxp3 +CD4+ T-cell subsets. Circulating neutrophil counts and proportions of Foxp3 +CD4+ T cells correlated with decreased plasma levels of spermine in treatment-naive, incipient SLE patients. CONCLUSIONS: We identified two novel SAT1 LOF variants, showed the ability of the frameshift variant to confer murine lupus, highlighted the pathogenic role of dysregulated polyamine catabolism and identified SAT1 LOF variants as new monogenic causes for SLE.


Subject(s)
Genetic Diseases, X-Linked , Lupus Erythematosus, Systemic , Animals , Child , Female , Humans , Male , Mice , Genetic Predisposition to Disease , Homozygote , Lupus Erythematosus, Systemic/genetics , Spermine/blood , Genetic Diseases, X-Linked/genetics , Acetyltransferases/genetics
7.
PLoS Genet ; 15(4): e1008092, 2019 04.
Article in English | MEDLINE | ID: mdl-31022184

ABSTRACT

Human leukocyte antigen (HLA) is a key genetic factor conferring risk of systemic lupus erythematosus (SLE), but precise independent localization of HLA effects is extremely challenging. As a result, the contribution of specific HLA alleles and amino-acid residues to the overall risk of SLE and to risk of specific autoantibodies are far from completely understood. Here, we dissected (a) overall SLE association signals across HLA, (b) HLA-peptide interaction, and (c) residue-autoantibody association. Classical alleles, SNPs, and amino-acid residues of eight HLA genes were imputed across 4,915 SLE cases and 13,513 controls from Eastern Asia. We performed association followed by conditional analysis across HLA, assessing both overall SLE risk and risk of autoantibody production. DR15 alleles HLA-DRB1*15:01 (P = 1.4x10-27, odds ratio (OR) = 1.57) and HLA-DQB1*06:02 (P = 7.4x10-23, OR = 1.55) formed the most significant haplotype (OR = 2.33). Conditioned protein-residue signals were stronger than allele signals and mapped predominantly to HLA-DRB1 residue 13 (P = 2.2x10-75) and its proxy position 11 (P = 1.1x10-67), followed by HLA-DRB1-37 (P = 4.5x10-24). After conditioning on HLA-DRB1, novel associations at HLA-A-70 (P = 1.4x10-8), HLA-DPB1-35 (P = 9.0x10-16), HLA-DQB1-37 (P = 2.7x10-14), and HLA-B-9 (P = 6.5x10-15) emerged. Together, these seven residues increased the proportion of explained heritability due to HLA to 2.6%. Risk residues for both overall disease and hallmark autoantibodies (i.e., nRNP: DRB1-11, P = 2.0x10-14; DRB1-13, P = 2.9x10-13; DRB1-30, P = 3.9x10-14) localized to the peptide-binding groove of HLA-DRB1. Enrichment for specific amino-acid characteristics in the peptide-binding groove correlated with overall SLE risk and with autoantibody presence. Risk residues were in primarily negatively charged side-chains, in contrast with rheumatoid arthritis. We identified novel SLE signals in HLA Class I loci (HLA-A, HLA-B), and localized primary Class II signals to five residues in HLA-DRB1, HLA-DPB1, and HLA-DQB1. These findings provide insights about the mechanisms by which the risk residues interact with each other to produce autoantibodies and are involved in SLE pathophysiology.


Subject(s)
Amino Acid Sequence , Autoantibodies/immunology , Disease Susceptibility , Histocompatibility Antigens Class II/chemistry , Histocompatibility Antigens Class II/immunology , Histocompatibility Antigens Class I/chemistry , Histocompatibility Antigens Class I/immunology , Lupus Erythematosus, Systemic/etiology , Alleles , Amino Acid Substitution , Asian People , Female , Genetic Predisposition to Disease , Genetic Variation , Histocompatibility Antigens Class I/genetics , Histocompatibility Antigens Class II/genetics , Humans , Male , Odds Ratio , Polymorphism, Single Nucleotide
8.
J Clin Immunol ; 41(5): 1031-1047, 2021 07.
Article in English | MEDLINE | ID: mdl-33656624

ABSTRACT

PURPOSE: The human antibody repertoire forms in response to infections, the microbiome, vaccinations, and environmental exposures. The specificity of such antibody responses was compared among a cohort of toddlers to identify differences between seropositive versus seronegative responses. METHODS: An assessment of the serum IgM and IgG antibody reactivities in 197 toddlers of 1- and 2-years of age was performed with a microfluidic array containing 110 distinct antigens. Longitudinal profiling was done from years 1 to 2. Seropositivity to RNA and DNA viruses; bacteria; live attenuated, inactive, and subunit vaccines; and autoantigens was compared. A stratification was developed based on quantitative variations in the IgG responses. Clinical presentations and previously known genetic risk alleles for various immune system conditions were investigated in relation to IgG responses. RESULTS: IgG reactivities stratified toddlers into low, moderate, and high responder groups. The high group (17%) had elevated IgG responses to multiple RNA and DNA viruses (e.g., respiratory syncytial virus, Epstein-Barr virus, adenovirus, Coxsackievirus) and this correlated with increased responses to live attenuated viral vaccines and certain autoantigens. This high group was more likely to be associated with gestational diabetes and an older age. Genetic analyses identified polymorphisms in the IL2RB, TNFSF4, and INS genes in two high responder individuals that were associated with their elevated cytokine levels and clinical history of eczema and asthma. CONCLUSION: Serum IgG profiling of toddlers reveals correlations between the magnitude of the antibody responses towards viruses, live attenuated vaccines, and certain autoantigens. A low responder group had much weaker responses overall, including against vaccines. The serum antibody screen also identifies individuals with IgG responses to less common infections (West Nile virus, parvovirus, tuberculosis). The characterization of the antibody responses in combination with the identification of genetic risk alleles provides an opportunity to identify children with increased risk of clinical disease.


Subject(s)
Antibodies, Viral/blood , Autoantigens/immunology , Bacteria/immunology , DNA Viruses/immunology , Immunoglobulin G/blood , RNA Viruses/immunology , Vaccines/immunology , Child, Preschool , Cytokines/blood , Female , Genotype , Humans , Immunoglobulin M/blood , Infant , Male , Microfluidic Analytical Techniques
9.
Oncologist ; 25(8): e1242-e1245, 2020 08.
Article in English | MEDLINE | ID: mdl-32400023

ABSTRACT

Immune-related adverse events induced by immune checkpoint inhibitor (ICI) therapy may affect diverse organ systems, including skeletal and cardiac muscle. ICI-associated myositis may result in substantial morbidity and occasional mortality. We present a case of a patient with advanced non-small cell lung cancer who developed grade 4 myositis with concurrent myocarditis early after initiation of anti-programmed death ligand 1 therapy (durvalumab). Autoantibody analysis revealed marked increases in anti-3-hydroxy-3-methylglutaryl-coenzyme A reductase antibody levels that preceded clinical toxicity, and further increased during toxicity. Notably, the patient had a history of intolerable statin myopathy, which had resolved clinically after statin discontinuation and prior to ICI initiation. This case demonstrates a potential association between statin exposure, autoantibodies, and ICI-associated myositis.


Subject(s)
Carcinoma, Non-Small-Cell Lung , Hydroxymethylglutaryl-CoA Reductase Inhibitors , Lung Neoplasms , Myositis , Humans , Hydroxymethylglutaryl-CoA Reductase Inhibitors/adverse effects , Immune Checkpoint Inhibitors , Myositis/chemically induced , Myositis/drug therapy
10.
Oncologist ; 25(5): e753-e757, 2020 05.
Article in English | MEDLINE | ID: mdl-32167195

ABSTRACT

Immune checkpoint inhibitor (ICI)-induced immune-related adverse events (irAEs) may affect almost any organ system and occur at any point during therapy. Autoantibody analysis may provide insight into the mechanism, nature, and timing of these events. We report a case of ICI-induced late-onset Raynaud's-like phenomenon in a patient receiving combination immunotherapy. A 53-year-old woman with advanced non-small lung cancer received combination anti-cytotoxic T-lymphocyte antigen 4 and anti-programmed death 1 ICI therapy. She developed early (hypophysitis at 4 months) and late (Raynaud's at >20 months) irAEs. Longitudinal assessment of 124 autoantibodies was correlated with toxicity. Although autoantibody levels were generally stable for the first 18 months of therapy, shortly before the development of Raynaud's, a marked increase in multiple autoantibodies was observed. This case highlights the potential for delayed autoimmune toxicities and provides potential biologic insights into the dynamic nature of these events. KEY POINTS: A patient treated with dual anti-PD1 and anti-CTLA4 therapy developed Raynaud's-like signs and symptoms more than 18 months after starting therapy. In this case, autoantibody changes became apparent shortly before onset of clinical toxicity. This case highlights the potential for late-onset immune-related adverse events checkpoint inhibitors, requiring continuous clinical vigilance. The optimal duration of checkpoint inhibitor therapy in patients with profound and prolonged responses remains unclear.


Subject(s)
Immunotherapy , Lung Neoplasms , Autoantibodies , Female , Humans , Immunologic Factors , Immunotherapy/adverse effects , Middle Aged
11.
Am J Respir Crit Care Med ; 200(3): 370-380, 2019 08 01.
Article in English | MEDLINE | ID: mdl-30694692

ABSTRACT

Rationale: There is poor understanding about protective immunity and the pathogenesis of cavitation in patients with tuberculosis.Objectives: To map pathophysiological pathways at anatomically distinct positions within the human tuberculosis cavity.Methods: Biopsies were obtained from eight predetermined locations within lung cavities of patients with multidrug-resistant tuberculosis undergoing therapeutic surgical resection (n = 14) and healthy lung tissue from control subjects without tuberculosis (n = 10). RNA sequencing, immunohistochemistry, and bacterial load determination were performed at each cavity position. Differentially expressed genes were normalized to control subjects without tuberculosis, and ontologically mapped to identify a spatially compartmentalized pathophysiological map of the cavity. In silico perturbation using a novel distance-dependent dynamical sink model was used to investigate interactions between immune networks and bacterial burden, and to integrate these identified pathways.Measurements and Main Results: The median (range) lung cavity volume on positron emission tomography/computed tomography scans was 50 cm3 (15-389 cm3). RNA sequence reads (31% splice variants) mapped to 19,049 annotated human genes. Multiple proinflammatory pathways were upregulated in the cavity wall, whereas a downregulation "sink" in the central caseum-fluid interface characterized 53% of pathways including neuroendocrine signaling, calcium signaling, triggering receptor expressed on myeloid cells-1, reactive oxygen and nitrogen species production, retinoic acid-mediated apoptosis, and RIG-I-like receptor signaling. The mathematical model demonstrated that neuroendocrine, protein kinase C-θ, and triggering receptor expressed on myeloid cells-1 pathways, and macrophage and neutrophil numbers, had the highest correlation with bacterial burden (r > 0.6), whereas T-helper effector systems did not.Conclusions: These data provide novel insights into host immunity to Mycobacterium tuberculosis-related cavitation. The pathways defined may serve as useful targets for the design of host-directed therapies, and transmission prevention interventions.


Subject(s)
Mycobacterium tuberculosis/isolation & purification , Sequence Analysis, RNA , Tuberculosis, Multidrug-Resistant/microbiology , Tuberculosis, Pulmonary/microbiology , Adolescent , Adult , Aged , Case-Control Studies , Female , Humans , Male , Middle Aged , RNA, Viral/isolation & purification , Young Adult
12.
Br J Cancer ; 120(1): 63-68, 2019 01.
Article in English | MEDLINE | ID: mdl-30377338

ABSTRACT

BACKGROUND: Up to 40% of cancer patients on immune checkpoint inhibitors develop clinically significant immune-related adverse events (irAEs). The role of host immune status and function in predisposing patients to the development of irAEs remains unknown. METHODS: Sera from 65 patients receiving immune checkpoint inhibitors and 13 healthy controls were evaluated for 40 cytokines at pre-treatment, after 2-3 weeks and after 6 weeks and analysed for correlation with the development of irAEs. RESULTS: Of the 65 cancer patients enrolled, 55% were women; the mean age was 65 years and 98% received anti-PD1/PDL1 therapy. irAEs occurred in 35% of cases. Among healthy controls, cytokine levels were stable over time and lower than those in cancer patients at baseline. Significant increases in CXCL9, CXCL10, CXCL11 and CXCL13 occurred 2 weeks post treatment, and in CXCL9, CXCL10, CXCL11, CXCL13, IL-10 and CCL26 at 6 weeks post treatment. Patients who developed irAEs had lower levels of CXCL9, CXCL10, CXCL11 and CXCL19 at baseline and exhibited greater increases in CXCL9 and CXCL10 levels at post treatment compared to patients without irAEs. CONCLUSIONS: Patients who developed irAEs have lower baseline levels and greater post-treatment increases in multiple cytokine levels, suggesting that underlying immune dysregulation may be associated with heightened risk for irAEs.


Subject(s)
Drug-Related Side Effects and Adverse Reactions/immunology , Immunotherapy/adverse effects , Neoplasms/therapy , Aged , Cytokines/genetics , Cytokines/immunology , Drug-Related Side Effects and Adverse Reactions/pathology , Female , Humans , Male , Middle Aged , Neoplasms/immunology , Neoplasms/pathology , Risk Factors
13.
Immunity ; 32(2): 253-65, 2010 Feb 26.
Article in English | MEDLINE | ID: mdl-20153220

ABSTRACT

CD4(+) T cells deficient in signaling lymphocyte activation molecule (SLAM)-associated protein (SAP) exhibit a selective impairment in adhesion to antigen-presenting B cells but not dendritic cells (DCs), resulting in defective germinal center formation. However, the nature of this selective adhesion defect remained unclear. We found that whereas T cell:DC interactions were primarily integrin dependent, T cell:B cell interactions had both an early integrin-dependent phase and a sustained phase that also required SAP. We further found that the SLAM family member CD84 was required for prolonged T cell:B cell contact, optimal T follicular helper function, and germinal center formation in vivo. Moreover, both CD84 and another SLAM member, Ly108, mediated T cell adhesion and participated in stable T cell:B cell interactions in vitro. Our results reveal insight into the dynamic regulation of T cell:B cell interactions and identify SLAM family members as critical components of sustained T cell:B cell adhesion required for productive humoral immunity.


Subject(s)
B-Lymphocytes/metabolism , Cell Adhesion/immunology , Germinal Center/metabolism , Intracellular Signaling Peptides and Proteins/metabolism , T-Lymphocytes/metabolism , Animals , Antigens, CD/genetics , Antigens, CD/immunology , Antigens, CD/metabolism , Antigens, Ly/immunology , Antigens, Ly/metabolism , B-Lymphocytes/immunology , B-Lymphocytes/pathology , Cell Adhesion/genetics , Cell Differentiation/genetics , Cell Differentiation/immunology , Cell Proliferation , Cells, Cultured , Germinal Center/pathology , Interferon-gamma/metabolism , Interleukins/metabolism , Intracellular Signaling Peptides and Proteins/genetics , Intracellular Signaling Peptides and Proteins/immunology , Lymphocyte Activation/genetics , Mice , Mice, Inbred C57BL , Mice, Knockout , Receptors, Cell Surface/genetics , Receptors, Cell Surface/immunology , Receptors, Cell Surface/metabolism , Signaling Lymphocytic Activation Molecule Associated Protein , Signaling Lymphocytic Activation Molecule Family , Signaling Lymphocytic Activation Molecule Family Member 1 , T-Lymphocytes/immunology , T-Lymphocytes/pathology
14.
Am J Respir Crit Care Med ; 198(9): 1208-1219, 2018 11 01.
Article in English | MEDLINE | ID: mdl-29877726

ABSTRACT

RATIONALE: Acquired resistance is an important driver of multidrug-resistant tuberculosis (TB), even with good treatment adherence. However, exactly what initiates the resistance and how it arises remain poorly understood. OBJECTIVES: To identify the relationship between drug concentrations and drug susceptibility readouts (minimum inhibitory concentrations [MICs]) in the TB cavity. METHODS: We recruited patients with medically incurable TB who were undergoing therapeutic lung resection while on treatment with a cocktail of second-line anti-TB drugs. On the day of surgery, antibiotic concentrations were measured in the blood and at seven prespecified biopsy sites within each cavity. Mycobacterium tuberculosis was grown from each biopsy site, MICs of each drug identified, and whole-genome sequencing performed. Spearman correlation coefficients between drug concentration and MIC were calculated. MEASUREMENTS AND MAIN RESULTS: Fourteen patients treated for a median of 13 months (range, 5-31 mo) were recruited. MICs and drug resistance-associated single-nucleotide variants differed between the different geospatial locations within each cavity, and with pretreatment and serial sputum isolates, consistent with ongoing acquisition of resistance. However, pretreatment sputum MIC had an accuracy of only 49.48% in predicting cavitary MICs. There were large concentration-distance gradients for each antibiotic. The location-specific concentrations inversely correlated with MICs (P < 0.05) and therefore acquired resistance. Moreover, pharmacokinetic/pharmacodynamic exposures known to amplify drug-resistant subpopulations were encountered in all positions. CONCLUSIONS: These data inform interventional strategies relevant to drug delivery, dosing, and diagnostics to prevent the development of acquired resistance. The role of high intracavitary penetration as a biomarker of antibiotic efficacy, when assessing new regimens, requires clarification.


Subject(s)
Antitubercular Agents/pharmacology , Lung/drug effects , Lung/pathology , Tuberculosis, Multidrug-Resistant/drug therapy , Tuberculosis, Multidrug-Resistant/pathology , Adolescent , Adult , Antitubercular Agents/therapeutic use , Biopsy , Dose-Response Relationship, Drug , Drug Therapy, Combination , Female , Humans , Male , Microbial Sensitivity Tests , Middle Aged , Prospective Studies , Young Adult
15.
Nucleic Acids Res ; 45(8): 4590-4605, 2017 05 05.
Article in English | MEDLINE | ID: mdl-28334891

ABSTRACT

RAD51, a multifunctional protein, plays a central role in DNA replication and homologous recombination repair, and is known to be involved in cancer development. We identified a novel role for RAD51 in innate immune response signaling. Defects in RAD51 lead to the accumulation of self-DNA in the cytoplasm, triggering a STING-mediated innate immune response after replication stress and DNA damage. In the absence of RAD51, the unprotected newly replicated genome is degraded by the exonuclease activity of MRE11, and the fragmented nascent DNA accumulates in the cytosol, initiating an innate immune response. Our data suggest that in addition to playing roles in homologous recombination-mediated DNA double-strand break repair and replication fork processing, RAD51 is also implicated in the suppression of innate immunity. Thus, our study reveals a previously uncharacterized role of RAD51 in initiating immune signaling, placing it at the hub of new interconnections between DNA replication, DNA repair, and immunity.


Subject(s)
DNA Replication , DNA-Binding Proteins/genetics , DNA/genetics , Membrane Proteins/genetics , Rad51 Recombinase/genetics , Recombinational DNA Repair/genetics , Cell Line, Tumor , DNA/immunology , DNA Breaks, Double-Stranded/drug effects , DNA-Binding Proteins/immunology , Epithelial Cells/cytology , Epithelial Cells/drug effects , Epithelial Cells/immunology , Fibroblasts/cytology , Fibroblasts/drug effects , Fibroblasts/immunology , Genes, Reporter , Humans , Hydroxamic Acids/pharmacology , Immunity, Innate , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , MRE11 Homologue Protein , Membrane Proteins/immunology , Pyrimidinones/pharmacology , Rad51 Recombinase/deficiency , Rad51 Recombinase/immunology , Recombinational DNA Repair/immunology , Signal Transduction/genetics , Signal Transduction/immunology , Thiones/pharmacology , Vorinostat , Red Fluorescent Protein
16.
J Immunol ; 196(4): 1507-16, 2016 Feb 15.
Article in English | MEDLINE | ID: mdl-26773143

ABSTRACT

C57BL/6 mice bearing the Sle2(z) lupus-susceptibility congenic interval on chromosome 4 display high titers of polyclonal autoantibodies with generalized B cell hyperactivity, hallmarks of systemic lupus erythematosus. In B6.Sle2(z)HEL(Ig).sHEL BCR-transgenic mice, Sle2(z) did not breach central tolerance, but it led to heightened expression of endogenous Ig H and L chains in splenic B cells, upregulation of RAG, and serological polyreactivity, suggestive of excessive receptor revision. Fatty acid amide hydrolase (FAAH), a gene in the minimal subcongenic interval generated through recombinant mapping, was found to be upregulated in Sle2(z) B cells by microarray analysis, Western blot, and functional assays. Pharmacological inhibition of FAAH reversed the increase in receptor revision, RAG expression, and polyreactive autoantibodies in lupus-prone mice. These studies indicate that increased peripheral BCR revision, or selective peripheral expansion of BCR-revised B cells, may lead to systemic autoimmunity and that FAAH is a lupus-susceptibility gene that might regulate this process.


Subject(s)
Amidohydrolases/metabolism , B-Lymphocyte Subsets/immunology , Lupus Erythematosus, Systemic/immunology , Receptors, Antigen, B-Cell/immunology , Amidohydrolases/genetics , Animals , Autoantibodies/immunology , B-Lymphocytes/immunology , B-Lymphocytes/physiology , Genetic Predisposition to Disease , Immune Tolerance/genetics , Lupus Erythematosus, Systemic/genetics , Mice, Inbred C57BL , Mice, Transgenic , Microarray Analysis , Receptors, Antigen, B-Cell/metabolism , Spleen/cytology , Spleen/immunology
17.
Proc Natl Acad Sci U S A ; 112(45): 13994-9, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26508631

ABSTRACT

Recognition of pathogen-associated molecular patterns by Toll-like receptors (TLRs) on dendritic cells (DCs) leads to DC maturation, a process involving up-regulation of MHC and costimulatory molecules and secretion of proinflammatory cytokines. All TLRs except TLR3 achieve these outcomes by using the signaling adaptor myeloid differentiation factor 88. TLR4 and TLR3 can both use the Toll-IL-1 receptor domain-containing adaptor inducing IFN-ß (TRIF)-dependent signaling pathway leading to IFN regulatory factor 3 (IRF3) activation and induction of IFN-ß and -α4. The TRIF signaling pathway, downstream of both of these TLRs, also leads to DC maturation, and it has been proposed that the type I IFNs act in cis to induce DC maturation and subsequent effects on adaptive immunity. The present study was designed to understand the molecular mechanisms of TRIF-mediated DC maturation. We have discovered that TLR4-TRIF-induced DC maturation was independent of both IRF3 and type I IFNs. In contrast, TLR3-mediated DC maturation was completely dependent on type I IFN feedback. We found that differential activation of mitogen-activated protein kinases by the TLR4- and TLR3-TRIF axes determined the type I IFN dependency for DC maturation. In addition, we found that the adjuvanticity of LPS to induce T-cell activation is completely independent of type I IFNs. The important distinction between the TRIF-mediated signaling pathways of TLR4 and TLR3 discovered here could have a major impact in the design of future adjuvants that target this pathway.


Subject(s)
Adaptor Proteins, Vesicular Transport/metabolism , Dendritic Cells/immunology , Gene Expression Regulation/immunology , Signal Transduction/physiology , Toll-Like Receptor 3/metabolism , Toll-Like Receptor 4/metabolism , Animals , Base Sequence , Blotting, Western , CD4-Positive T-Lymphocytes/metabolism , Cell Proliferation/physiology , Dendritic Cells/cytology , Flow Cytometry , Lipopolysaccharides , Mice , Mice, Knockout , Molecular Sequence Data , Sequence Analysis, RNA
18.
Proc Natl Acad Sci U S A ; 112(45): E6195-204, 2015 Nov 10.
Article in English | MEDLINE | ID: mdl-26512111

ABSTRACT

Glomerulonephritis is a common and debilitating feature of systemic lupus erythematosus (SLE). The precise immune mechanisms that drive the progression from benign autoimmunity to glomerulonephritis are largely unknown. Previous investigations have shown that a moderate increase of the innate Toll-like receptor 7 (TLR7) is sufficient for the development of nephritis. In these systems normalization of B-cell TLR7 expression or temporal depletion of plasmacytoid dendritic cells (pDCs) slow progression; however, the critical cell that is responsible for driving full immunopathology remains unidentified. In this investigation we have shown that conventional DC expression of TLR7 is essential for severe autoimmunity in the Sle1Tg7 model of SLE. We show that a novel expanding CD11b(+) conventional DC subpopulation dominates the infiltrating renal inflammatory milieu, localizing to the glomeruli. Moreover, exposure of human myeloid DCs to IFN-α or Flu increases TLR7 expression, suggesting they may have a role in self-RNA recognition pathways in clinical disease. To our knowledge, this study is the first to highlight the importance of conventional DC-TLR7 expression for kidney pathogenesis in a murine model of SLE.


Subject(s)
Dendritic Cells/metabolism , Lupus Nephritis/physiopathology , Toll-Like Receptor 7/metabolism , Up-Regulation , Analysis of Variance , Animals , Base Sequence , CD11b Antigen/metabolism , DNA Primers/genetics , Flow Cytometry , Gene Expression Profiling/methods , Humans , Image Processing, Computer-Assisted , Kidney Glomerulus/cytology , Kidney Glomerulus/pathology , Lupus Nephritis/metabolism , Mice , Microscopy, Confocal , Molecular Sequence Data , Real-Time Polymerase Chain Reaction , Sequence Analysis, RNA , Statistics, Nonparametric
19.
Article in English | MEDLINE | ID: mdl-28584143

ABSTRACT

Linezolid has an excellent sterilizing effect in tuberculosis patients but high adverse event rates. The dose that would maximize efficacy and minimize toxicity is unknown. We performed linezolid dose-effect and dose-scheduling studies in the hollow fiber system model of tuberculosis (HFS-TB) for sterilizing effect. HFS-TB units were treated with several doses to mimic human-like linezolid intrapulmonary pharmacokinetics and repetitively sampled for drug concentration, total bacterial burden, linezolid-resistant subpopulations, and RNA sequencing over 2 months. Linezolid-resistant isolates underwent whole-genome sequencing. The expression of genes encoding efflux pumps in the first 1 to 2 weeks revealed the same exposure-response patterns as the linezolid-resistant subpopulation. Linezolid-resistant isolates from the 2nd month of therapy revealed mutations in several efflux pump/transporter genes and a LuxR-family transcriptional regulator. Linezolid sterilizing effect was linked to the ratio of unbound 0- to 24-h area under the concentration-time curve (AUC0-24) to MIC. Optimal microbial kill was achieved at an AUC0-24/MIC ratio of 119. The optimal sterilizing effect dose for clinical use was identified using Monte Carlo simulations. Clinical doses of 300 and 600 mg/day (or double the dose every other day) achieved this target in 87% and >99% of 10,000 patients, respectively. The susceptibility breakpoint identified was 2 mg/liter. The simulations identified that a 300-mg/day dose did not achieve AUC0-24s associated with linezolid toxicity, while 600 mg/day achieved those AUC0-24s in <20% of subjects. The linezolid dose of 300 mg/day performed well and should be compared to 600 mg/day or 1,200 mg every other day in clinical trials.


Subject(s)
Antitubercular Agents/therapeutic use , Linezolid/therapeutic use , Mycobacterium tuberculosis/drug effects , Protein Synthesis Inhibitors/therapeutic use , Tuberculosis, Pulmonary/drug therapy , Antitubercular Agents/adverse effects , Dose-Response Relationship, Drug , Drug Resistance, Bacterial/genetics , Genome, Bacterial/genetics , Humans , Linezolid/adverse effects , Membrane Transport Proteins/genetics , Microbial Sensitivity Tests , Repressor Proteins/genetics , Trans-Activators/genetics
20.
Am J Hum Genet ; 94(4): 586-98, 2014 Apr 03.
Article in English | MEDLINE | ID: mdl-24702955

ABSTRACT

Efforts to identify lupus-associated causal variants in the FAM167A/BLK locus on 8p21 are hampered by highly associated noncausal variants. In this report, we used a trans-population mapping and sequencing strategy to identify a common variant (rs922483) in the proximal BLK promoter and a tri-allelic variant (rs1382568) in the upstream alternative BLK promoter as putative causal variants for association with systemic lupus erythematosus. The risk allele (T) at rs922483 reduced proximal promoter activity and modulated alternative promoter usage. Allelic differences at rs1382568 resulted in altered promoter activity in B progenitor cell lines. Thus, our results demonstrated that both lupus-associated functional variants contribute to the autoimmune disease association by modulating transcription of BLK in B cells and thus potentially altering immune responses.


Subject(s)
Lupus Erythematosus, Systemic/genetics , Promoter Regions, Genetic , Transcription, Genetic , src-Family Kinases/genetics , Alleles , Chromosomes, Human, Pair 8 , Electrophoretic Mobility Shift Assay , Female , Genetic Predisposition to Disease , Haplotypes , Humans , Male , Polymorphism, Single Nucleotide
SELECTION OF CITATIONS
SEARCH DETAIL