Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 51
Filter
1.
Cell ; 187(14): 3506-3530, 2024 Jul 11.
Article in English | MEDLINE | ID: mdl-38996486

ABSTRACT

Fibrotic interstitial lung diseases (fILDs) have poor survival rates and lack effective therapies. Despite evidence for immune mechanisms in lung fibrosis, immunotherapies have been unsuccessful for major types of fILD. Here, we review immunological mechanisms in lung fibrosis that have the potential to impact clinical practice. We first examine innate immunity, which is broadly involved across fILD subtypes. We illustrate how innate immunity in fILD involves a complex interplay of multiple cell subpopulations and molecular pathways. We then review the growing evidence for adaptive immunity in lung fibrosis to provoke a re-examination of its role in clinical fILD. We close with future directions to address key knowledge gaps in fILD pathobiology: (1) longitudinal studies emphasizing early-stage clinical disease, (2) immune mechanisms of acute exacerbations, and (3) next-generation immunophenotyping integrating spatial, genetic, and single-cell approaches. Advances in these areas are essential for the future of precision medicine and immunotherapy in fILD.


Subject(s)
Immunity, Innate , Lung Diseases, Interstitial , Humans , Lung Diseases, Interstitial/immunology , Lung Diseases, Interstitial/pathology , Animals , Adaptive Immunity , Immunotherapy , Pulmonary Fibrosis/immunology , Pulmonary Fibrosis/pathology , Lung/pathology , Lung/immunology
2.
Cell ; 176(4): 897-912.e20, 2019 02 07.
Article in English | MEDLINE | ID: mdl-30686579

ABSTRACT

A complete chart of cis-regulatory elements and their dynamic activity is necessary to understand the transcriptional basis of differentiation and function of an organ system. We generated matched epigenome and transcriptome measurements in 86 primary cell types that span the mouse immune system and its differentiation cascades. This breadth of data enable variance components analysis that suggests that genes fall into two distinct classes, controlled by either enhancer- or promoter-driven logic, and multiple regression that connects genes to the enhancers that regulate them. Relating transcription factor (TF) expression to the genome-wide accessibility of their binding motifs classifies them as predominantly openers or closers of local chromatin accessibility, pinpointing specific cis-regulatory elements where binding of given TFs is likely functionally relevant, validated by chromatin immunoprecipitation sequencing (ChIP-seq). Overall, this cis-regulatory atlas provides a trove of information on transcriptional regulation through immune differentiation and a foundational scaffold to define key regulatory events throughout the immunological genome.


Subject(s)
Immune System/immunology , Immune System/metabolism , Regulatory Elements, Transcriptional/genetics , Animals , Binding Sites/genetics , Chromatin , Chromatin Immunoprecipitation/methods , Enhancer Elements, Genetic/genetics , Epigenomics/methods , Gene Expression Regulation/genetics , Mice , Mice, Inbred C57BL , Promoter Regions, Genetic/genetics , Protein Binding/genetics , Transcription Factors/metabolism , Transcriptome/genetics
3.
Nat Immunol ; 22(7): 914-927, 2021 07.
Article in English | MEDLINE | ID: mdl-34099919

ABSTRACT

To better define the control of immune system regulation, we generated an atlas of microRNA (miRNA) expression from 63 mouse immune cell populations and connected these signatures with assay for transposase-accessible chromatin using sequencing (ATAC-seq), chromatin immunoprecipitation followed by sequencing (ChIP-seq) and nascent RNA profiles to establish a map of miRNA promoter and enhancer usage in immune cells. miRNA complexity was relatively low, with >90% of the miRNA compartment of each population comprising <75 miRNAs; however, each cell type had a unique miRNA signature. Integration of miRNA expression with chromatin accessibility revealed putative regulatory elements for differentially expressed miRNAs, including miR-21a, miR-146a and miR-223. The integrated maps suggest that many miRNAs utilize multiple promoters to reach high abundance and identified dominant and divergent miRNA regulatory elements between lineages and during development that may be used by clustered miRNAs, such as miR-99a/let-7c/miR-125b, to achieve distinct expression. These studies, with web-accessible data, help delineate the cis-regulatory elements controlling miRNA signatures of the immune system.


Subject(s)
Gene Expression Profiling , Immune System/metabolism , MicroRNAs/genetics , Promoter Regions, Genetic , Transcriptome , Animals , Cells, Cultured , Chromatin Immunoprecipitation , Computational Biology , Gene Expression Regulation, Developmental , Immune System/cytology , Immune System/immunology , Male , Mice , Mice, Inbred C57BL , Mice, Transgenic , MicroRNAs/metabolism , RNA-Seq
4.
Am J Respir Crit Care Med ; 208(11): 1177-1195, 2023 12 01.
Article in English | MEDLINE | ID: mdl-37756440

ABSTRACT

Rationale: Despite the importance of inflammation in chronic obstructive pulmonary disease (COPD), the immune cell landscape in the lung tissue of patients with mild-moderate disease has not been well characterized at the single-cell and molecular level. Objectives: To define the immune cell landscape in lung tissue from patients with mild-moderate COPD at single-cell resolution. Methods: We performed single-cell transcriptomic, proteomic, and T-cell receptor repertoire analyses on lung tissue from patients with mild-moderate COPD (n = 5, Global Initiative for Chronic Obstructive Lung Disease I or II), emphysema without airflow obstruction (n = 5), end-stage COPD (n = 2), control (n = 6), or donors (n = 4). We validated in an independent patient cohort (N = 929) and integrated with the Hhip+/- murine model of COPD. Measurements and Main Results: Mild-moderate COPD lungs have increased abundance of two CD8+ T cell subpopulations: cytotoxic KLRG1+TIGIT+CX3CR1+ TEMRA (T effector memory CD45RA+) cells, and DNAM-1+CCR5+ T resident memory (TRM) cells. These CD8+ T cells interact with myeloid and alveolar type II cells via IFNG and have hyperexpanded T-cell receptor clonotypes. In an independent cohort, the CD8+KLRG1+ TEMRA cells are increased in mild-moderate COPD lung compared with control or end-stage COPD lung. Human CD8+KLRG1+ TEMRA cells are similar to CD8+ T cells driving inflammation in an aging-related murine model of COPD. Conclusions: CD8+ TEMRA cells are increased in mild-moderate COPD lung and may contribute to inflammation that precedes severe disease. Further study of these CD8+ T cells may have therapeutic implications for preventing severe COPD.


Subject(s)
CD8-Positive T-Lymphocytes , Pulmonary Disease, Chronic Obstructive , Humans , Animals , Mice , Disease Models, Animal , Proteomics , Lung/metabolism , Inflammation , Receptors, Antigen, T-Cell
5.
Circulation ; 146(18): 1344-1356, 2022 11.
Article in English | MEDLINE | ID: mdl-36036760

ABSTRACT

BACKGROUND: The efficacy and safety of prophylactic full-dose anticoagulation and antiplatelet therapy in critically ill COVID-19 patients remain uncertain. METHODS: COVID-PACT (Prevention of Arteriovenous Thrombotic Events in Critically-ill COVID-19 Patients Trial) was a multicenter, 2×2 factorial, open-label, randomized-controlled trial with blinded end point adjudication in intensive care unit-level patients with COVID-19. Patients were randomly assigned to a strategy of full-dose anticoagulation or standard-dose prophylactic anticoagulation. Absent an indication for antiplatelet therapy, patients were additionally randomly assigned to either clopidogrel or no antiplatelet therapy. The primary efficacy outcome was the hierarchical composite of death attributable to venous or arterial thrombosis, pulmonary embolism, clinically evident deep venous thrombosis, type 1 myocardial infarction, ischemic stroke, systemic embolic event or acute limb ischemia, or clinically silent deep venous thrombosis, through hospital discharge or 28 days. The primary efficacy analyses included an unmatched win ratio and time-to-first event analysis while patients were on treatment. The primary safety outcome was fatal or life-threatening bleeding. The secondary safety outcome was moderate to severe bleeding. Recruitment was stopped early in March 2022 (≈50% planned recruitment) because of waning intensive care unit-level COVID-19 rates. RESULTS: At 34 centers in the United States, 390 patients were randomly assigned between anticoagulation strategies and 292 between antiplatelet strategies (382 and 290 in the on-treatment analyses). At randomization, 99% of patients required advanced respiratory therapy, including 15% requiring invasive mechanical ventilation; 40% required invasive ventilation during hospitalization. Comparing anticoagulation strategies, a greater proportion of wins occurred with full-dose anticoagulation (12.3%) versus standard-dose prophylactic anticoagulation (6.4%; win ratio, 1.95 [95% CI, 1.08-3.55]; P=0.028). Results were consistent in time-to-event analysis for the primary efficacy end point (full-dose versus standard-dose incidence 19/191 [9.9%] versus 29/191 [15.2%]; hazard ratio, 0.56 [95% CI, 0.32-0.99]; P=0.046). The primary safety end point occurred in 4 (2.1%) on full dose and in 1 (0.5%) on standard dose (P=0.19); the secondary safety end point occurred in 15 (7.9%) versus 1 (0.5%; P=0.002). There was no difference in all-cause mortality (hazard ratio, 0.91 [95% CI, 0.56-1.48]; P=0.70). There were no differences in the primary efficacy or safety end points with clopidogrel versus no antiplatelet therapy. CONCLUSIONS: In critically ill patients with COVID-19, full-dose anticoagulation, but not clopidogrel, reduced thrombotic complications with an increase in bleeding, driven primarily by transfusions in hemodynamically stable patients, and no apparent excess in mortality. REGISTRATION: URL: https://www. CLINICALTRIALS: gov; Unique identifier: NCT04409834.


Subject(s)
COVID-19 , Thrombosis , Venous Thrombosis , Humans , Critical Illness , Thrombosis/drug therapy , Clopidogrel/therapeutic use , Hemorrhage/chemically induced , Anticoagulants/adverse effects , Venous Thrombosis/drug therapy , Venous Thrombosis/epidemiology , Venous Thrombosis/prevention & control , Platelet Aggregation Inhibitors/adverse effects , Treatment Outcome
6.
Nat Immunol ; 12(12): 1202-11, 2011 Oct 30.
Article in English | MEDLINE | ID: mdl-22037601

ABSTRACT

Invariant natural killer T cells (iNKT cells) have a prominent role during infection and other inflammatory processes, and these cells can be activated through their T cell antigen receptors by microbial lipid antigens. However, increasing evidence shows that they are also activated in situations in which foreign lipid antigens would not be present, which suggests a role for lipid self antigen. We found that an abundant endogenous lipid, ß-D-glucopyranosylceramide (ß-GlcCer), was a potent iNKT cell self antigen in mouse and human and that its activity depended on the composition of the N-acyl chain. Furthermore, ß-GlcCer accumulated during infection and in response to Toll-like receptor agonists, contributing to iNKT cell activation. Thus, we propose that recognition of ß-GlcCer by the invariant T cell antigen receptor translates innate danger signals into iNKT cell activation.


Subject(s)
Autoantigens/immunology , Bacterial Infections/immunology , Glycosphingolipids/immunology , Natural Killer T-Cells/immunology , Animals , Autoimmunity/immunology , Cell Line , Glycosphingolipids/metabolism , Humans , Lymphocyte Activation/immunology , Lymphoid Tissue/immunology , Lymphoid Tissue/metabolism , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Knockout , Receptors, Antigen, T-Cell/immunology , Receptors, Antigen, T-Cell/metabolism
7.
JAMA ; 329(14): 1183-1196, 2023 04 11.
Article in English | MEDLINE | ID: mdl-37039790

ABSTRACT

IMPORTANCE: Overactivation of the renin-angiotensin system (RAS) may contribute to poor clinical outcomes in patients with COVID-19. Objective: To determine whether angiotensin-converting enzyme (ACE) inhibitor or angiotensin receptor blocker (ARB) initiation improves outcomes in patients hospitalized for COVID-19. DESIGN, SETTING, AND PARTICIPANTS: In an ongoing, adaptive platform randomized clinical trial, 721 critically ill and 58 non-critically ill hospitalized adults were randomized to receive an RAS inhibitor or control between March 16, 2021, and February 25, 2022, at 69 sites in 7 countries (final follow-up on June 1, 2022). INTERVENTIONS: Patients were randomized to receive open-label initiation of an ACE inhibitor (n = 257), ARB (n = 248), ARB in combination with DMX-200 (a chemokine receptor-2 inhibitor; n = 10), or no RAS inhibitor (control; n = 264) for up to 10 days. MAIN OUTCOMES AND MEASURES: The primary outcome was organ support-free days, a composite of hospital survival and days alive without cardiovascular or respiratory organ support through 21 days. The primary analysis was a bayesian cumulative logistic model. Odds ratios (ORs) greater than 1 represent improved outcomes. RESULTS: On February 25, 2022, enrollment was discontinued due to safety concerns. Among 679 critically ill patients with available primary outcome data, the median age was 56 years and 239 participants (35.2%) were women. Median (IQR) organ support-free days among critically ill patients was 10 (-1 to 16) in the ACE inhibitor group (n = 231), 8 (-1 to 17) in the ARB group (n = 217), and 12 (0 to 17) in the control group (n = 231) (median adjusted odds ratios of 0.77 [95% bayesian credible interval, 0.58-1.06] for improvement for ACE inhibitor and 0.76 [95% credible interval, 0.56-1.05] for ARB compared with control). The posterior probabilities that ACE inhibitors and ARBs worsened organ support-free days compared with control were 94.9% and 95.4%, respectively. Hospital survival occurred in 166 of 231 critically ill participants (71.9%) in the ACE inhibitor group, 152 of 217 (70.0%) in the ARB group, and 182 of 231 (78.8%) in the control group (posterior probabilities that ACE inhibitor and ARB worsened hospital survival compared with control were 95.3% and 98.1%, respectively). CONCLUSIONS AND RELEVANCE: In this trial, among critically ill adults with COVID-19, initiation of an ACE inhibitor or ARB did not improve, and likely worsened, clinical outcomes. TRIAL REGISTRATION: ClinicalTrials.gov Identifier: NCT02735707.


Subject(s)
Angiotensin Receptor Antagonists , Angiotensin-Converting Enzyme Inhibitors , COVID-19 Drug Treatment , COVID-19 , Renin-Angiotensin System , Female , Humans , Male , Middle Aged , Angiotensin Receptor Antagonists/pharmacology , Angiotensin Receptor Antagonists/therapeutic use , Angiotensin-Converting Enzyme Inhibitors/pharmacology , Angiotensin-Converting Enzyme Inhibitors/therapeutic use , Bayes Theorem , COVID-19/therapy , Renin-Angiotensin System/drug effects , Hospitalization , COVID-19 Drug Treatment/methods , Critical Illness , Receptors, Chemokine/antagonists & inhibitors
8.
Thorax ; 77(10): 1041-1044, 2022 10.
Article in English | MEDLINE | ID: mdl-35907639

ABSTRACT

Although interstitial lung disease (ILD) causes significant morbidity and mortality in rheumatoid arthritis (RA), it is difficult to predict the development or progression of ILD, emphasising the need for improved discovery through minimally invasive diagnostic tests. Aptamer-based proteomic profiling was used to assess 1321 proteins from 159 patients with rheumatoid arthritis with interstitial lung disease (RA-ILD), RA without ILD, idiopathic pulmonary fibrosis and healthy controls. Differential expression and gene set enrichment analyses revealed molecular signatures that are strongly associated with the presence and severity of RA-ILD and provided insight into unexplored pathways of disease. These warrant further study as non-invasive diagnostic tools and future therapeutic targets.


Subject(s)
Arthritis, Rheumatoid , Idiopathic Pulmonary Fibrosis , Lung Diseases, Interstitial , Humans , Proteomics , Lung Diseases, Interstitial/complications , Idiopathic Pulmonary Fibrosis/genetics , Idiopathic Pulmonary Fibrosis/complications , Arthritis, Rheumatoid/genetics , Arthritis, Rheumatoid/complications
9.
Am J Respir Cell Mol Biol ; 61(6): 737-746, 2019 12.
Article in English | MEDLINE | ID: mdl-31461627

ABSTRACT

The impact of lipotoxicity on the development of lung fibrosis is unclear. Saturated fatty acids, such as palmitic acid (PA), activate endoplasmic reticulum (ER) stress, a cellular stress response associated with the development of idiopathic pulmonary fibrosis (IPF). We tested the hypothesis that PA increases susceptibility to lung epithelial cell death and experimental fibrosis by modulating ER stress. Total liquid chromatography and mass spectrometry were used to measure fatty acid content in IPF lungs. Wild-type mice were fed a high-fat diet (HFD) rich in PA or a standard diet and subjected to bleomycin-induced lung injury. Lung fibrosis was determined by hydroxyproline content. Mouse lung epithelial cells were treated with PA. ER stress and cell death were assessed by Western blotting, TUNEL staining, and cell viability assays. IPF lungs had a higher level of PA compared with controls. Bleomycin-exposed mice fed an HFD had significantly increased pulmonary fibrosis associated with increased cell death and ER stress compared with those fed a standard diet. PA increased apoptosis and activation of the unfolded protein response in lung epithelial cells. This was attenuated by genetic deletion and chemical inhibition of CD36, a fatty acid transporter. In conclusion, consumption of an HFD rich in saturated fat increases susceptibility to lung fibrosis and ER stress, and PA mediates lung epithelial cell death and ER stress via CD36. These findings demonstrate that lipotoxicity may have a significant impact on the development of lung injury and fibrosis by enhancing pro-death ER stress pathways.


Subject(s)
Diet, High-Fat/adverse effects , Endoplasmic Reticulum Stress/drug effects , Palmitic Acid/toxicity , Pulmonary Fibrosis/chemically induced , Animals , Apoptosis/drug effects , CD36 Antigens/deficiency , CD36 Antigens/physiology , Epithelial Cells/drug effects , Lung/metabolism , Lung/pathology , Male , Mice , Mice, Inbred C57BL , Palmitic Acid/administration & dosage , Palmitic Acid/pharmacokinetics , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology
11.
Semin Immunol ; 27(1): 26-32, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25841627

ABSTRACT

Invariant natural killer T (iNKT) cells are innate T cells that express a semi-invariant T cell receptor (TCR) and recognize lipid antigens presented by CD1d molecules. As part of innate immunity, iNKT cells rapidly produce large amounts of cytokines after activation and regulate the function of innate and adaptive immune cells in antimicrobial immunity, tumor rejection and inflammatory diseases. Global transcriptional profiling has advanced our understanding of all aspects of iNKT cell biology. In this review, we discuss transcriptional analyses of iNKT cell development, functional subsets of iNKT cells, and global comparisons of iNKT cells to other innate and adaptive immune cells. Global transcriptional analysis revealed that iNKT cells have a transcriptional profile distinct from NK cells and MHC-restricted T cells, both during thymic development and in the periphery. The transcription factors EGR2 and PLZF (and microRNA like miR-150) are key regulators of the iNKT cell transcriptome during development. PLZF is one of several factors that control the homing and maintenance of organ-specific iNKT cell populations. As in MHC-restricted T cells, specific transcription factors are characteristic of functional subsets of iNKT cells, such as the transcription factor T-bet in the NKT1 subset. Exciting future directions for global transcriptional analyses include iNKT cells in disease models, diverse NKT cells and human studies.


Subject(s)
Natural Killer T-Cells/metabolism , Transcription, Genetic , Animals , Early Growth Response Protein 2/metabolism , Humans , Inflammation/metabolism , Kruppel-Like Transcription Factors/metabolism , MicroRNAs/metabolism , Transcriptome
12.
Resuscitation ; 198: 110142, 2024 May.
Article in English | MEDLINE | ID: mdl-38342294

ABSTRACT

AIM: We sought to investigate the relationship between mechanical cardiopulmonary resuscitation (CPR) during in-hospital cardiac arrest and survival to hospital discharge. METHODS: Utilizing the prospectively collected American Heart Association's Get With The Guidelines database, we performed an observational study. Data from 153 institutions across the United States were reviewed with a total of 351,125 patients suffering cardiac arrest between 2011 and 2019 were screened. After excluding patients with cardiac arrests lasting less than 5 minutes, and patients who had incomplete data, a total of 111,143 patients were included. Our primary exposure was mechanical vs. manual CPR, and the primary outcome was survival to hospital discharge. Multivariate logistic regression models and propensity weighted analyses were used. RESULTS: 11.8% of patients who received mechanical CPR survived to hospital discharge versus 16.9% in the manual CPR group. Patients who received mechanical CPR had a lower probability of survival to discharge compared to patients who received manual CPR (OR 0.66 95% CI 0.58-0.75; p < 0.001). This association persisted with multi-variable adjustment (OR 0.57 95% CI 0.46-0.70, p < 0.0001) and propensity weighted analysis (OR 0.68 95% CI 0.44-0 0.92, p < 0.0001). Mechanical CPR was associated with decrease likelihood of return of spontaneous circulation after multivariate adjustment (OR 0.68, 95% CI 0.60-0.76; p < 0.001). CONCLUSIONS: Mechanical CPR was associated with a decreased likelihood of survival to hospital discharge and ROSC compared to manual CPR. This finding should be interpreted within the context of important limitations of this study and randomized trials are needed to better investigate this relationship.


Subject(s)
Cardiopulmonary Resuscitation , Heart Arrest , Humans , Cardiopulmonary Resuscitation/methods , Cardiopulmonary Resuscitation/statistics & numerical data , Male , Female , Heart Arrest/therapy , Heart Arrest/mortality , Aged , Middle Aged , United States/epidemiology , Patient Discharge/statistics & numerical data , Cohort Studies , Propensity Score
13.
STAR Protoc ; 5(1): 102874, 2024 Mar 15.
Article in English | MEDLINE | ID: mdl-38310512

ABSTRACT

Immunophenotyping of out-of-hospital cardiac arrest (OHCA) patients is of increasing interest but has challenges. Here, we describe steps for the design of the clinical cohort, planning patient enrollment and sample collection, and ethical review of the study protocol. We detail procedures for blood sample collection and cryopreservation of peripheral blood mononuclear cells (PBMCs). We detail steps to modulate immune checkpoints in OHCA PBMC ex vivo. This protocol also has relevance for immunophenotyping other types of critical illness. For complete details on the use and execution of this protocol, please refer to Tamura et al. (2023).1.


Subject(s)
Leukocytes, Mononuclear , Out-of-Hospital Cardiac Arrest , Humans , Immunophenotyping , Out-of-Hospital Cardiac Arrest/diagnosis , Cryopreservation
14.
bioRxiv ; 2024 Apr 28.
Article in English | MEDLINE | ID: mdl-38712233

ABSTRACT

Myofibroblast differentiation, essential for driving extracellular matrix synthesis in pulmonary fibrosis, requires increased glycolysis. While glycolytic cells must export lactate, the contributions of lactate transporters to myofibroblast differentiation are unknown. In this study, we investigated how MCT1 and MCT4, key lactate transporters, influence myofibroblast differentiation and experimental pulmonary fibrosis. Our findings reveal that inhibiting MCT1 or MCT4 reduces TGFß-stimulated pulmonary myofibroblast differentiation in vitro and decreases bleomycin-induced pulmonary fibrosis in vivo. Through comprehensive metabolic analyses, including bioenergetics, stable isotope tracing, metabolomics, and imaging mass spectrometry in both cells and mice, we demonstrate that inhibiting lactate transport enhances oxidative phosphorylation, reduces reactive oxygen species production, and diminishes glucose metabolite incorporation into fibrotic lung regions. Furthermore, we introduce VB253, a novel MCT4 inhibitor, which ameliorates pulmonary fibrosis in both young and aged mice, with comparable efficacy to established antifibrotic therapies. These results underscore the necessity of lactate transport for myofibroblast differentiation, identify MCT1 and MCT4 as promising pharmacologic targets in pulmonary fibrosis, and support further evaluation of lactate transport inhibitors for patients for whom limited therapeutic options currently exist.

15.
bioRxiv ; 2024 May 23.
Article in English | MEDLINE | ID: mdl-38826450

ABSTRACT

Fibrosis drives end-organ damage in many diseases. However, clinical trials targeting individual upstream activators of fibroblasts, such as TGFß, have largely failed. Here, we target the leukemia inhibitory factor receptor (LIFR) as a "master amplifier" of multiple upstream activators of lung fibroblasts. In idiopathic pulmonary fibrosis (IPF), the most common fibrotic lung disease, we found that lung myofibroblasts had high LIF expression. Further, TGFß1, one of the key drivers of fibrosis, upregulated LIF expression in IPF fibroblasts. In vitro anti-LIFR antibody blocking on human IPF lung fibroblasts reduced induction of profibrotic genes downstream of TGFß1, IL-4 and IL-13. Further, siRNA silencing of LIFR in IPF precision cut lung slices reduced expression of fibrotic proteins. Together, we find that LIFR drives an autocrine positive feedback loop that amplifies and sustains pathogenic activation of IPF fibroblasts downstream of multiple external stimuli, implicating LIFR as a therapeutic target in fibrosis. Significance Statement: Fibroblasts have a central role in the pathogenesis of fibrotic diseases. However, due to in part to multiple profibrotic stimuli, targeting a single activator of fibroblasts, like TGFß, has not yielded successful clinical treatments. We hypothesized that a more effective therapeutic strategy is identifying a downstream "master amplifier" of a range of upstream profibrotic stimuli. This study identifies the leukemia inhibitory factor receptor (LIFR) on fibrotic lung fibroblasts amplifies multiple profibrotic stimuli, such as IL-13 and TGFß. Blocking LIFR reduced fibrosis in ex vivo lung tissue from patients with idiopathic pulmonary fibrosis (IPF). LIFR, acting as a master amplifier downstream of fibroblast activation, offers an alternative therapeutic strategy for fibrotic diseases.

16.
Front Immunol ; 14: 1269335, 2023.
Article in English | MEDLINE | ID: mdl-37942334

ABSTRACT

Introduction: Severe respiratory illness is the most prominent manifestation of patients infected with SARS-CoV-2, and yet the molecular mechanisms underlying severe lung disease in COVID-19 affected patients still require elucidation. Human leukocyte antigen class I (HLA-I) expression is crucial for antigen presentation and the host's response to SARS-CoV-2. Methods: To gain insights into the immune response and molecular pathways involved in severe lung disease, we performed immunopeptidomic and proteomic analyses of lung tissues recovered at four COVID-19 autopsy and six non-COVID-19 transplants. Results: We found signals of tissue injury and regeneration in lung fibroblast and alveolar type I/II cells, resulting in the production of highly immunogenic self-antigens within the lungs of COVID-19 patients. We also identified immune activation of the M2c macrophage as the primary source of HLA-I presentation and immunogenicity in this context. Additionally, we identified 28 lung signatures that can serve as early plasma markers for predicting infection and severe COVID-19 disease. These protein signatures were predominantly expressed in macrophages and epithelial cells and were associated with complement and coagulation cascades. Discussion: Our findings emphasize the significant role of macrophage-mediated immunity in the development of severe lung disease in COVID-19 patients.


Subject(s)
COVID-19 , Humans , COVID-19/pathology , SARS-CoV-2 , Proteomics , Lung , Biopsy
17.
Med ; 4(7): 432-456.e6, 2023 Jul 14.
Article in English | MEDLINE | ID: mdl-37257452

ABSTRACT

BACKGROUND: Most patients hospitalized after cardiac arrest (CA) die because of neurological injury. The systemic inflammatory response after CA is associated with neurological injury and mortality but remains poorly defined. METHODS: We determine the innate immune network induced by clinical CA at single-cell resolution. FINDINGS: Immune cell states diverge as early as 6 h post-CA between patients with good or poor neurological outcomes 30 days after CA. Nectin-2+ monocyte and Tim-3+ natural killer (NK) cell subpopulations are associated with poor outcomes, and interactome analysis highlights their crosstalk via cytokines and immune checkpoints. Ex vivo studies of peripheral blood cells from CA patients demonstrate that immune checkpoints are a compensatory mechanism against inflammation after CA. Interferon γ (IFNγ)/interleukin-10 (IL-10) induced Nectin-2 on monocytes; in a negative feedback loop, Nectin-2 suppresses IFNγ production by NK cells. CONCLUSIONS: The initial hours after CA may represent a window for therapeutic intervention in the resolution of inflammation via immune checkpoints. FUNDING: This work was supported by funding from the American Heart Association, Brigham and Women's Hospital Department of Medicine, the Evergreen Innovation Fund, and the National Institutes of Health.


Subject(s)
Cytokines , Transcriptome , United States , Humans , Female , Cytokines/pharmacology , Nectins/genetics , Killer Cells, Natural , Inflammation
18.
Nat Med ; 11(11): 1180-7, 2005 Nov.
Article in English | MEDLINE | ID: mdl-16208318

ABSTRACT

Host defense against viruses probably depends on targeted death of infected host cells and then clearance of cellular corpses by macrophages. For this process to be effective, the macrophage must presumably avoid its own virus-induced death. Here we identify one such mechanism. We show that mice lacking the chemokine Ccl5 are immune compromised to the point of delayed viral clearance, excessive airway inflammation and respiratory death after mouse parainfluenza or human influenza virus infection. Virus-inducible levels of Ccl5 are required to prevent apoptosis of virus-infected mouse macrophages in vivo and mouse and human macrophages ex vivo. The protective effect of Ccl5 requires activation of the Ccr5 chemokine receptor and consequent bilateral activation of G(alphai)-PI3K-AKT and G(alphai)-MEK-ERK signaling pathways. The antiapoptotic action of chemokine signaling may therefore allow scavengers to finally stop the host cell-to-cell infectious process.


Subject(s)
Apoptosis , Chemokines, CC/metabolism , Macrophages, Alveolar/metabolism , Receptors, CCR5/metabolism , Signal Transduction , Animals , Antibodies, Monoclonal/metabolism , Blotting, Western , Cell Survival , Cells, Cultured , Chemokine CCL5 , Chemokines, CC/genetics , Fluorescein-5-isothiocyanate , Fluorescent Antibody Technique, Indirect , Fluorescent Dyes , Immunohistochemistry , Macrophages, Alveolar/virology , Mice , Mice, Knockout , Microscopy, Fluorescence , Respirovirus Infections , Sendai virus , Virus Replication
19.
Elife ; 112022 01 28.
Article in English | MEDLINE | ID: mdl-35089131

ABSTRACT

A healthcare center widely sharing its internal guidelines on how to treat COVID-19 patients "just wasn't done." As the pandemic raged at a Boston hospital, the next generation of clinical leaders pushed for change.


Subject(s)
Access to Information , COVID-19/therapy , Laboratories, Hospital/standards , Leadership , Access to Information/ethics , Access to Information/psychology , Boston
20.
Cell Rep Med ; 3(3): 100533, 2022 03 15.
Article in English | MEDLINE | ID: mdl-35474746

ABSTRACT

The rapid pace of the COVID-19 pandemic precluded traditional approaches to evaluating clinical research and guidelines. We highlight notable successes and pitfalls of clinicians' new approaches to managing evidence amidst an unprecedented crisis. In "Era 1" (early 2020), clinicians relied on anecdote and social media, which democratized conversations on guidelines, but also led clinicians astray. "Era 2" (approximately late 2020 to early 2021) saw preprints that accelerated new interventions but suffered from a surfeit of poor-quality data. In the current era, clinicians consolidate the evidentiary gains of Era 2 with living, online clinical guidelines, but the public suffers from misinformation. The COVID-19 pandemic is a laboratory on how clinicians adapt to an absence of clinical guidance amidst an informational and healthcare crisis. Challenges remain as we integrate new approaches to innovations made in the traditional guideline process to confront both the long tail of COVID-19 and future pandemics.


Subject(s)
COVID-19 , Social Media , Communication , Humans , Pandemics
SELECTION OF CITATIONS
SEARCH DETAIL