Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 54
Filtrar
Más filtros













Base de datos
Intervalo de año de publicación
1.
Med ; 5(2): 132-147.e7, 2024 Feb 09.
Artículo en Inglés | MEDLINE | ID: mdl-38272035

RESUMEN

BACKGROUND: Transforming growth factor ß (TGF-ß) is implicated as a key mediator of pathological fibrosis, but its pleiotropic activity in a range of homeostatic functions presents challenges to its safe and effective therapeutic targeting. There are three isoforms of TGF-ß, TGF-ß1, TGF-ß2, and TGF-ß3, which bind to a common receptor complex composed of TGF-ßR1 and TGF-ßR2 to induce similar intracellular signals in vitro. We have recently shown that the cellular expression patterns and activation thresholds of TGF-ß2 and TGF-ß3 are distinct from those of TGF-ß1 and that selective short-term TGF-ß2 and TGF-ß3 inhibition can attenuate fibrosis in vivo without promoting excessive inflammation. Isoform-selective inhibition of TGF-ß may therefore provide a therapeutic opportunity for patients with chronic fibrotic disorders. METHODS: Transcriptomic profiling of skin biopsies from patients with systemic sclerosis (SSc) from multiple clinical trials was performed to evaluate the role of TGF-ß3 in this disease. Antibody humanization, biochemical characterization, crystallization, and pre-clinical experiments were performed to further characterize an anti-TGF-ß3 antibody. FINDINGS: In the skin of patients with SSc, TGF-ß3 expression is uniquely correlated with biomarkers of TGF-ß signaling and disease severity. Crystallographic studies establish a structural basis for selective TGF-ß3 inhibition with a potent and selective monoclonal antibody that attenuates fibrosis effectively in vivo at clinically translatable exposures. Toxicology studies suggest that, as opposed to pan-TGF-ß inhibitors, this anti-TGF-ß3 antibody has a favorable safety profile for chronic administration. CONCLUSION: We establish a rationale for targeting TGF-ß3 in SSc with a favorable therapeutic index. FUNDING: This study was funded by Genentech, Inc.


Asunto(s)
Esclerodermia Sistémica , Factor de Crecimiento Transformador beta3 , Humanos , Factor de Crecimiento Transformador beta3/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Fibrosis , Esclerodermia Sistémica/tratamiento farmacológico , Isoformas de Proteínas/metabolismo
2.
iScience ; 26(11): 108133, 2023 Nov 17.
Artículo en Inglés | MEDLINE | ID: mdl-37867940

RESUMEN

Systemic sclerosis (SSc) interstitial lung disease (ILD) is among the leading causes of SSc-related morbidity and mortality. Tocilizumab (TCZ, anti-IL6RA) has demonstrated a reduced rate of pulmonary function decline in two phase 2/3 trials (faSScinate and focuSSced) in SSc-ILD patients. We performed transcriptome analysis of skin biopsy samples collected in the studies to decipher gene networks that were potentially associated with clinical responses to TCZ treatment. One module correlated with disease progression showed pharmacodynamic changes with TCZ treatment, and was characterized by plasma cell (PC) genes. PC signature gene expression levels were also significantly increased in both fibrotic SSc and IPF lungs compared to controls. scRNAseq analyses confirmed that PC signature genes were co-expressed in CD38 and CD138 expressing PC subsets in SSc lungs. These data provide insights into the potential role of PC in disease progression and mechanisms of action of TCZ in fibrotic interstitial lung diseases.

3.
Clin Immunol ; 254: 109695, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37479123

RESUMEN

Drug development for systemic sclerosis (SSc) benefits from understanding the relationship between disease and circulating biomarkers to enable activities such as patient stratification and evaluation of therapeutic response. We measured biomarkers in serum from SSc patients from a phase 3 trial of tocilizumab (focuSSced) and compared baseline levels with healthy controls (HCs). Several baseline biomarkers appeared elevated in SSc patients compared to HCs, suggesting activation of epithelial damage, inflammation, fibrosis, and extracellular matrix (ECM) remodeling. Baseline correlations among both periostin/COMP and ECM biomarker subsets implicated their participation in fibroblast activation. Tocilizumab treatment modulated serum biomarkers of macrophage activation, inflammation, and ECM turnover, including collagen formation and degradation neoepitopes. Baseline CRP, periostin, and SP-D showed prognostic trends for worsening lung function, and IL-6, COMP, periostin, and Pro-C3 showed prognostic trends for worsening skin thickness. These prognostic results warrant confirmation in additional patient cohorts to verify their utility.


Asunto(s)
Esclerodermia Sistémica , Humanos , Esclerodermia Sistémica/tratamiento farmacológico , Fibrosis , Biomarcadores , Matriz Extracelular , Inflamación
4.
Cell Metab ; 34(9): 1377-1393.e8, 2022 09 06.
Artículo en Inglés | MEDLINE | ID: mdl-35987202

RESUMEN

Fibrosis is the major risk factor associated with morbidity and mortality in patients with non-alcoholic steatohepatitis (NASH)-driven chronic liver disease. Although numerous efforts have been made to identify the mediators of the initiation of liver fibrosis, the molecular underpinnings of fibrosis progression remain poorly understood, and therapies to arrest liver fibrosis progression are elusive. Here, we identify a pathway involving WNT1-inducible signaling pathway protein 1 (WISP1) and myocardin-related transcription factor (MRTF) as a central mechanism driving liver fibrosis progression through the integrin-dependent transcriptional reprogramming of myofibroblast cytoskeleton and motility. In mice, WISP1 deficiency protects against fibrosis progression, but not fibrosis onset. Moreover, the therapeutic administration of a novel antibody blocking WISP1 halted the progression of existing liver fibrosis in NASH models. These findings implicate the WISP1-MRTF axis as a crucial determinant of liver fibrosis progression and support targeting this pathway by antibody-based therapy for the treatment of NASH fibrosis.


Asunto(s)
Enfermedad del Hígado Graso no Alcohólico , Factores de Transcripción , Animales , Hígado/metabolismo , Cirrosis Hepática/metabolismo , Ratones , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/metabolismo , Proteínas Nucleares , Transducción de Señal , Transactivadores , Factores de Transcripción/metabolismo
5.
Sci Transl Med ; 14(641): eabl8146, 2022 04 20.
Artículo en Inglés | MEDLINE | ID: mdl-35442706

RESUMEN

Asthma and inflammatory airway diseases restrict airflow in the lung, compromising gas exchange and lung function. Inhaled corticosteroids (ICSs) can reduce inflammation, control symptoms, and improve lung function; however, a growing number of patients with severe asthma do not benefit from ICS. Using bronchial airway epithelial brushings from patients with severe asthma or primary human cells, we delineated a corticosteroid-driven fibroblast growth factor (FGF)-dependent inflammatory axis, with FGF-responsive fibroblasts promoting downstream granulocyte colony-stimulating factor (G-CSF) production, hyaluronan secretion, and neutrophilic inflammation. Allergen challenge studies in mice demonstrate that the ICS, fluticasone propionate, inhibited type 2-driven eosinophilia but induced a concomitant increase in FGFs, G-CSF, hyaluronan, and neutrophil infiltration. We developed a model of steroid-induced neutrophilic inflammation mediated, in part, by induction of an FGF-dependent epithelial-mesenchymal axis, which may explain why some individuals do not benefit from ICS. In further proof-of-concept experiments, we found that combination therapy with pan-FGF receptor inhibitors and corticosteroids prevented both eosinophilic and steroid-induced neutrophilic inflammation. Together, these results establish FGFs as therapeutic targets for severe asthma patients who do not benefit from ICS.


Asunto(s)
Asma , Factores de Crecimiento de Fibroblastos , Corticoesteroides/farmacología , Corticoesteroides/uso terapéutico , Animales , Fluticasona/farmacología , Fluticasona/uso terapéutico , Factor Estimulante de Colonias de Granulocitos/uso terapéutico , Humanos , Ácido Hialurónico , Inflamación/tratamiento farmacológico , Ratones
6.
Sci Transl Med ; 13(605)2021 08 04.
Artículo en Inglés | MEDLINE | ID: mdl-34349032

RESUMEN

Transforming growth factor-ß (TGFß) is a key driver of fibrogenesis. Three TGFß isoforms (TGFß1, TGFß2, and TGFß3) in mammals have distinct functions in embryonic development; however, the postnatal pathological roles and activation mechanisms of TGFß2 and TGFß3 have not been well characterized. Here, we show that the latent forms of TGFß2 and TGFß3 can be activated by integrin-independent mechanisms and have lower activation thresholds compared to TGFß1. Unlike TGFB1, TGFB2 and TGFB3 expression is increased in human lung and liver fibrotic tissues compared to healthy control tissues. Thus, TGFß2 and TGFß3 may play a pathological role in fibrosis. Inducible conditional knockout mice and anti-TGFß isoform-selective antibodies demonstrated that TGFß2 and TGFß3 are independently involved in mouse fibrosis models in vivo, and selective TGFß2 and TGFß3 inhibition does not lead to the increased inflammation observed with pan-TGFß isoform inhibition. A cocrystal structure of a TGFß2-anti-TGFß2/3 antibody complex reveals an allosteric isoform-selective inhibitory mechanism. Therefore, inhibiting TGFß2 and/or TGFß3 while sparing TGFß1 may alleviate fibrosis without toxicity concerns associated with pan-TGFß blockade.


Asunto(s)
Factor de Crecimiento Transformador beta2 , Factor de Crecimiento Transformador beta3 , Animales , Modelos Animales de Enfermedad , Femenino , Fibrosis , Humanos , Ratones , Isoformas de Proteínas/metabolismo , Factor de Crecimiento Transformador beta2/metabolismo , Factor de Crecimiento Transformador beta3/metabolismo
7.
Cell Rep Med ; 1(8): 100140, 2020 11 17.
Artículo en Inglés | MEDLINE | ID: mdl-33294861

RESUMEN

Progressive lung fibrosis is a major cause of mortality in systemic sclerosis (SSc) patients, but the underlying mechanisms remain unclear. We demonstrate that immune complexes (ICs) activate human monocytes to promote lung fibroblast migration partly via osteopontin (OPN) secretion, which is amplified by autocrine monocyte colony stimulating factor (MCSF) and interleukin-6 (IL-6) activity. Bulk and single-cell RNA sequencing demonstrate that elevated OPN expression in SSc lung tissue is enriched in macrophages, partially overlapping with CCL18 expression. Serum OPN is elevated in SSc patients with interstitial lung disease (ILD) and prognosticates future lung function deterioration in SSc cohorts. Serum OPN levels decrease following tocilizumab (monoclonal anti-IL-6 receptor) treatment, confirming the connection between IL-6 and OPN in SSc patients. Collectively, these data suggest a plausible link between autoantibodies and lung fibrosis progression, where circulating OPN serves as a systemic proxy for IC-driven profibrotic macrophage activity, highlighting its potential as a promising biomarker in SSc ILD.


Asunto(s)
Células Mieloides/metabolismo , Osteopontina/metabolismo , Esclerodermia Sistémica/metabolismo , Autoanticuerpos/metabolismo , Biomarcadores/metabolismo , Línea Celular , Quimiocinas CC/metabolismo , Progresión de la Enfermedad , Fibrosis/metabolismo , Humanos , Interleucina-6/metabolismo , Pulmón/metabolismo , Enfermedades Pulmonares Intersticiales/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo
8.
Mucosal Immunol ; 12(5): 1174-1186, 2019 09.
Artículo en Inglés | MEDLINE | ID: mdl-31308480

RESUMEN

There continues to be a major need for more effective inflammatory bowel disease (IBD) therapies. IL-13Rα2 is a decoy receptor that binds the cytokine IL-13 with high affinity and diminishes its STAT6-mediated effector functions. Previously, we found that IL-13Rα2 was necessary for IBD in mice deficient in the anti-inflammatory cytokine IL-10. Here, we tested for the first time a therapeutic antibody specifically targeting IL-13Rα2. We also used the antibody and Il13ra2-/- mice to dissect the role of IL-13Rα2 in IBD pathogenesis and recovery. Il13ra2-/- mice were modestly protected from induction of dextran sodium sulfate (DSS)-induced colitis. Following a 7-day recovery period, Il13ra2-/- mice or wild-type mice administered the IL-13Rα2-neutralizing antibody had significantly improved colon health compared to control mice. Neutralizing IL-13Rα2 to increase IL-13 bioavailability promoted resolution of IBD even if neutralization occurred only during recovery. To link our observations in mice to a large human cohort, we conducted a phenome-wide association study of a more active variant of IL-13 (R130Q) that has reduced affinity for IL-13Rα2. Human subjects carrying R130Q reported a lower risk for Crohn's disease. Our findings endorse moving anti-IL-13Rα2 into preclinical drug development with the goal of accelerating recovery and maintaining remission in Crohn's disease patients.


Asunto(s)
Antiinflamatorios/farmacología , Anticuerpos Monoclonales/farmacología , Enfermedades Inflamatorias del Intestino/metabolismo , Subunidad alfa2 del Receptor de Interleucina-13/antagonistas & inhibidores , Subunidad alfa2 del Receptor de Interleucina-13/metabolismo , Animales , Enfermedad de Crohn/etiología , Enfermedad de Crohn/metabolismo , Enfermedad de Crohn/patología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Susceptibilidad a Enfermedades , Eosinófilos/inmunología , Eosinófilos/metabolismo , Mutación con Ganancia de Función , Variación Genética , Humanos , Inmunidad , Enfermedades Inflamatorias del Intestino/tratamiento farmacológico , Enfermedades Inflamatorias del Intestino/etiología , Enfermedades Inflamatorias del Intestino/patología , Subunidad alfa2 del Receptor de Interleucina-13/genética , Ratones , Oportunidad Relativa
9.
JCI Insight ; 52019 06 18.
Artículo en Inglés | MEDLINE | ID: mdl-31211697

RESUMEN

The lung is a relatively quiescent organ during homeostasis, but has a remarkable capacity for repair after injury. Alveolar epithelial type I cells (AEC1s) line airspaces and mediate gas exchange. After injury, they are regenerated by differentiation from their progenitors - alveolar epithelial type II cells (AEC2s) - which also secrete surfactant to maintain surface tension and alveolar patency. While recent studies showed that the maintenance of AEC2 stemness is Wnt dependent, the molecular mechanisms underlying AEC2-AEC1 differentiation in adult lung repair are still incompletely understood. Here we show that WWTR1 (TAZ) plays a crucial role in AEC differentiation. Using an in vitro organoid culture system, we found that tankyrase inhibition can efficiently block AEC2-AEC1 differentiation, and this effect was due to the inhibition of TAZ. In a bleomycin induced lung injury model, conditional deletion of TAZ in AEC2s dramatically reduced AEC1 regeneration during recovery, leading to exacerbated alveolar lesions and fibrosis. In patients with idiopathic pulmonary fibrosis (IPF), decreased blood levels of RAGE, a biomarker of AEC1 health, were associated with more rapid disease progression. Our findings implicate TAZ as a critical factor involved in AEC2 to AEC1 differentiation, and hence the maintenance of alveolar integrity after injury.


Asunto(s)
Células Epiteliales Alveolares/efectos de los fármacos , Células Epiteliales Alveolares/metabolismo , Diferenciación Celular/efectos de los fármacos , Diferenciación Celular/fisiología , Lesión Pulmonar/metabolismo , Transactivadores/metabolismo , Transactivadores/farmacología , Proteínas Adaptadoras Transductoras de Señales , Animales , Bleomicina/efectos adversos , Modelos Animales de Enfermedad , Humanos , Fibrosis Pulmonar Idiopática/patología , Pulmón/patología , Lesión Pulmonar/inducido químicamente , Lesión Pulmonar/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Organogénesis/efectos de los fármacos , Organogénesis/fisiología , Organoides/metabolismo , Regeneración/fisiología , Células Madre/metabolismo , Transactivadores/genética , Transcriptoma , beta Catenina/genética , beta Catenina/metabolismo
10.
Lancet Respir Med ; 6(8): 615-626, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30072107

RESUMEN

BACKGROUND: Heterogeneity in the progression of idiopathic pulmonary fibrosis (IPF) might reflect diversity in underlying pathobiology, and represents a major challenge in the prediction of clinical progression and treatment benefit. Previous studies have found peripheral blood concentrations of several protein biomarkers to be prognostic for overall survival duration in patients with IPF, but these findings have generally not been directly compared and replicated between cohorts. We aimed to use the pivotal trials for pirfenidone to evaluate prognostic and predictive properties of biomarkers across multiple endpoints, and whether they are modulated by pirfenidone treatment. METHODS: We did post-hoc analyses of test and replication cohorts from CAPACITY 004 (NCT00287716), CAPACITY 006 (NCT00287729), and ASCEND (NCT01366209) trials for the plasma proteins CCL13, CCL17, CCL18, CXCL13, CXCL14, COMP, interleukin 13, MMP3, MMP7, osteopontin, periostin, and YKL40. Eligible participants had IPF and received pirfenidone 2403 mg/day or placebo in CAPACITY (test cohort) or ASCEND (replication cohort), were aged 40-80 years, and without missing biomarker data at baseline. To identify biomarkers that were consistently prognostic for clinical outcome measures, the primary analysis was the association between biomarker concentrations at baseline and absolute change in percentage of predicted forced vital capacity (FVC%pred) at 12 months (CAPACITY week 48, ASCEND week 52) in the placebo group. Biomarkers within the test cohort that met predefined success criteria of a prognostic p value less than 0·10 from multivariate analysis were further assessed in the replication cohort. Furthermore, the predictive effect size (ie, biomarkers that were predictive for benefit from pirfenidone) was calculated as the difference in FVC%pred treatment effect (pirfenidone in relation to placebo) between high versus low biomarker subgroups at week 48 (test cohort) or week 52 (replication cohort). FINDINGS: Several baseline biomarkers (CCL13, CCL18, COMP, CXCL13, CXCL14, periostin, and YKL40) were prognostic for progression outcomes in the placebo groups of the test cohort. However, only CCL18 was consistently prognostic for absolute change in percentage of FVC%pred in both the test (p=0·032) and replication (p=0·004) cohorts. Pirfenidone treatment benefit was consistent regardless of baseline biomarker concentration. INTERPRETATION: Blood CCL18 concentrations were the most consistent predictor of disease progression across IPF cohorts with potential to inform new target discovery and clinical trial design. Future validation of these findings in prospective studies is warranted. FUNDING: Genentech Inc.


Asunto(s)
Antiinflamatorios no Esteroideos/administración & dosificación , Ensayos Clínicos como Asunto/normas , Fibrosis Pulmonar Idiopática/tratamiento farmacológico , Piridonas/administración & dosificación , Anciano , Biomarcadores/sangre , Progresión de la Enfermedad , Femenino , Estudios de Seguimiento , Humanos , Fibrosis Pulmonar Idiopática/genética , Masculino , Modelos de Riesgos Proporcionales
11.
Lancet Respir Med ; 6(8): 603-614, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-29891356

RESUMEN

BACKGROUND: Idiopathic pulmonary fibrosis (IPF) risk has a strong genetic component. Studies have implicated variations at several loci, including TERT, surfactant genes, and a single nucleotide polymorphism at chr11p15 (rs35705950) in the intergenic region between TOLLIP and MUC5B. Patients with IPF who have risk alleles at rs35705950 have longer survival from the time of IPF diagnosis than do patients homozygous for the non-risk allele, whereas patients with shorter telomeres have shorter survival times. We aimed to assess whether rare protein-altering variants in genes regulating telomere length are enriched in patients with IPF homozygous for the non-risk alleles at rs35705950. METHODS: Between Nov 1, 2014, and Nov 1, 2016, we assessed blood samples from patients aged 40 years or older and of European ancestry with sporadic IPF from three international phase 3 clinical trials (INSPIRE, CAPACITY, ASCEND), one phase 2 study (RIFF), and US-based observational studies (Vanderbilt Clinical Interstitial Lung Disease Registry and the UCSF Interstitial Lung Disease Clinic registry cohorts) at the Broad Institute (Cambridge, MA, USA) and Human Longevity (San Diego, CA, USA). We also assessed blood samples from non-IPF controls in several clinical trials. We did whole-genome sequencing to assess telomere length and identify rare protein-altering variants, stratified by rs35705950 genotype. We also assessed rare functional variation in TERT exons and compared telomere length and disease progression across genotypes. FINDINGS: We assessed samples from 1510 patients with IPF and 1874 non-IPF controls. 30 (3%) of 1046 patients with an rs35705950 risk allele had a rare protein-altering variant in TERT compared with 34 (7%) of 464 non-risk allele carriers (odds ratio 0·40 [95% CI 0·24-0·66], p=0·00039). Subsequent analyses identified enrichment of rare protein-altering variants in PARN and RTEL1, and rare variation in TERC in patients with IPF compared with controls. We expanded our study population to provide a more accurate estimation of rare variant frequency in these four loci, and to calculate telomere length. The proportion of patients with at least one rare variant in TERT, PARN, TERC, or RTEL1 was higher in patients with IPF than in controls (149 [9%] of 1739 patients vs 205 [2%] of 8645 controls, p=2·44 × 10-8). Patients with IPF who had a variant in any of the four identified telomerase component genes had telomeres that were 3·69-16·10% shorter than patients without a variant in any of the four genes and had an earlier mean age of disease onset than patients without one or more variants (65·1 years [SD 7·8] vs 67·1 years [7·9], p=0·004). In the placebo arms of clinical trials, shorter telomeres were significantly associated with faster disease progression (1·7% predicted forced vital capacity per kb per year, p=0·002). Pirfenidone had treatment benefit regardless of telomere length (p=4·24 × 10-8 for telomere length lower than the median, p=0·0044 for telomere length greater than the median). INTERPRETATION: Rare protein-altering variants in TERT, PARN, TERC, and RTEL1 are enriched in patients with IPF compared with controls, and, in the case of TERT, particularly in individuals without a risk allele at the rs35705950 locus. This suggests that multiple genetic factors contribute to sporadic IPF, which might implicate distinct mechanisms of pathogenesis and disease progression. FUNDING: Genentech, National Institutes of Health, Francis Family Foundation, Pulmonary Fibrosis Foundation, Nina Ireland Program for Lung Health, US Department of Veterans Affairs.


Asunto(s)
Fibrosis Pulmonar Idiopática/sangre , Mucina 5B/sangre , Homeostasis del Telómero/genética , Anciano , Estudios de Casos y Controles , Ensayos Clínicos como Asunto , Femenino , Humanos , Fibrosis Pulmonar Idiopática/genética , Masculino , Persona de Mediana Edad , Secuenciación Completa del Genoma
12.
J Immunol ; 199(3): 847-853, 2017 08 01.
Artículo en Inglés | MEDLINE | ID: mdl-28652398

RESUMEN

Many chronic liver disorders are characterized by dysregulated immune responses and hepatocyte death. We used an in vivo model to study the immune response to necrotic liver injury and found that necrotic liver cells induced eosinophil recruitment. Necrotic liver induced eosinophil IL-1ß and IL-18 secretion, degranulation, and cell death. Caspase-1 inhibitors blocked all of these responses. Caspase-1-mediated cell death with accompanying cytokine release is the hallmark of a novel form of cell death termed pyroptosis. To confirm this response in a disease model, we isolated eosinophils from the livers of Schistosoma mansoni-infected mice. S. mansoni eggs lodge in the hepatic sinusoids of infected mice, resulting in hepatocyte death, inflammation, and progressive liver fibrosis. This response is typified by massive eosinophilia, and we were able to confirm pyroptosis in the infiltrating eosinophils. This demonstrated that pyroptosis is a cellular pathway used by eosinophils in response to large-scale hepatic cell death.


Asunto(s)
Caspasa 1/metabolismo , Eosinófilos/fisiología , Hepatocitos/patología , Hígado/inmunología , Piroptosis , Esquistosomiasis mansoni/fisiopatología , Animales , Inhibidores de Caspasas/farmacología , Muerte Celular , Movimiento Celular , Modelos Animales de Enfermedad , Eosinofilia , Eosinófilos/inmunología , Eosinófilos/metabolismo , Interleucina-18/inmunología , Interleucina-18/metabolismo , Interleucina-1beta/inmunología , Interleucina-1beta/metabolismo , Hígado/parasitología , Hígado/patología , Cirrosis Hepática/fisiopatología , Ratones , Necrosis , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología
13.
Sci Transl Med ; 9(396)2017 06 28.
Artículo en Inglés | MEDLINE | ID: mdl-28659437

RESUMEN

Nonalcoholic fatty liver disease (NAFLD) is now the most common progressive liver disease in developed countries and is the second leading indication for liver transplantation due to the extensive fibrosis it causes. NAFLD progression is thought to be tied to chronic low-level type 1 inflammation originating in the adipose tissue during obesity; however, the specific immunological mechanisms regulating the progression of NAFLD-associated fibrosis in the liver are unclear. To investigate the immunopathogenesis of NAFLD more completely, we investigated adipose dysfunction, nonalcoholic steatohepatitis (NASH), and fibrosis in mice that develop polarized type 1 or type 2 immune responses. Unexpectedly, obese interleukin-10 (IL-10)/IL-4-deficient mice (type 1-polarized) were highly resistant to NASH. This protection was associated with an increased hepatic interferon-γ (IFN-γ) signature. Conversely, IFN-γ-deficient mice progressed rapidly to NASH with evidence of fibrosis dependent on transforming growth factor-ß (TGF-ß) and IL-13 signaling. Unlike increasing type 1 inflammation and the marked loss of eosinophils seen in expanding adipose tissue, progression of NASH was associated with increasing eosinophilic type 2 liver inflammation in mice and human patient biopsies. Finally, simultaneous inhibition of TGF-ß and IL-13 signaling attenuated the fibrotic machinery more completely than TGF-ß alone in NAFLD-associated fibrosis. Thus, although type 2 immunity maintains healthy metabolic signaling in adipose tissues, it exacerbates the progression of NAFLD collaboratively with TGF-ß in the liver.


Asunto(s)
Progresión de la Enfermedad , Inmunidad , Enfermedades Metabólicas/inmunología , Enfermedades Metabólicas/prevención & control , Enfermedad del Hígado Graso no Alcohólico/inmunología , Enfermedad del Hígado Graso no Alcohólico/prevención & control , Factor de Crecimiento Transformador beta/metabolismo , Tejido Adiposo/patología , Animales , Dieta Alta en Grasa , Eosinófilos/patología , Humanos , Inflamación/patología , Interferón gamma/deficiencia , Interferón gamma/metabolismo , Cirrosis Hepática/patología , Masculino , Ratones Endogámicos C57BL , Enfermedad del Hígado Graso no Alcohólico/patología , Obesidad/patología
14.
Sci Rep ; 6: 39714, 2016 12 22.
Artículo en Inglés | MEDLINE | ID: mdl-28004808

RESUMEN

Pulmonary fibrosis is a potentially lethal late adverse event of thoracic irradiation. Prior research indicates that unrestrained TGF-ß1 and/or type 2 cytokine-driven immune responses promote fibrosis following radiation injury, but the full spectrum of factors governing this pathology remains unclear. Interleukin 13 (IL-13) is a key factor in fibrotic disease associated with helminth infection, but it is unclear whether it plays a similar role in radiation-induced lung fibrosis. Using a mouse model, we tested the hypothesis that IL-13 drives the progression of radiation-induced pulmonary fibrosis. Irradiated lungs from wild-type c57BL/6NcR mice accumulated alternatively-activated macrophages, displayed elevated levels of IL-13, and extensive fibrosis, whereas IL-13 deficient mice were resistant to these changes. Furthermore, plasma from irradiated wild-type mice showed a transient increase in the IL-13 saturated fraction of the circulating decoy receptor IL-13Rα2. Finally, we determined that therapeutic neutralization of IL-13, during the period of IL-13Rα2 saturation was sufficient to protect mice from lung fibrosis. Taken together, our results demonstrate that IL-13 is a major regulator of radiation-induced lung injury and demonstrates that strategies focusing on IL-13 may be useful in screening for timely delivery of anti-IL-13 therapeutics.


Asunto(s)
Interleucina-13/farmacología , Lesión Pulmonar , Fibrosis Pulmonar , Traumatismos Experimentales por Radiación , Animales , Lesión Pulmonar/inmunología , Lesión Pulmonar/patología , Lesión Pulmonar/prevención & control , Ratones , Ratones Noqueados , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Fibrosis Pulmonar/prevención & control , Traumatismos Experimentales por Radiación/inmunología , Traumatismos Experimentales por Radiación/patología , Traumatismos Experimentales por Radiación/prevención & control
15.
Immunity ; 45(1): 145-58, 2016 07 19.
Artículo en Inglés | MEDLINE | ID: mdl-27421703

RESUMEN

Fibroproliferative diseases are driven by dysregulated tissue repair responses and are a major cause of morbidity and mortality because they affect nearly every organ system. Type 2 cytokine responses are critically involved in tissue repair; however, the mechanisms that regulate beneficial regeneration versus pathological fibrosis are not well understood. Here, we have shown that the type 2 effector cytokine interleukin-13 simultaneously, yet independently, directed hepatic fibrosis and the compensatory proliferation of hepatocytes and biliary cells in progressive models of liver disease induced by interleukin-13 overexpression or after infection with Schistosoma mansoni. Using transgenic mice with interleukin-13 signaling genetically disrupted in hepatocytes, cholangiocytes, or resident tissue fibroblasts, we have revealed direct and distinct roles for interleukin-13 in fibrosis, steatosis, cholestasis, and ductular reaction. Together, these studies show that these mechanisms are simultaneously controlled but distinctly regulated by interleukin-13 signaling. Thus, it may be possible to promote interleukin-13-dependent hepatobiliary expansion without generating pathological fibrosis. VIDEO ABSTRACT.


Asunto(s)
Hígado Graso/inmunología , Fibroblastos/inmunología , Interleucina-13/metabolismo , Cirrosis Hepática Biliar/inmunología , Hígado/patología , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/inmunología , Animales , Ácidos y Sales Biliares/biosíntesis , Proliferación Celular , Células Cultivadas , Fibrosis , Humanos , Interleucina-13/genética , Interleucina-13/inmunología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Transducción de Señal , Células Th2/inmunología
16.
J Exp Med ; 213(8): 1409-18, 2016 07 25.
Artículo en Inglés | MEDLINE | ID: mdl-27432938

RESUMEN

Innate lymphoid cells (ILC) play an important role in many immune processes, including control of infections, inflammation, and tissue repair. To date, little is known about the metabolism of ILC and whether these cells can metabolically adapt in response to environmental signals. Here we show that type 2 innate lymphoid cells (ILC2), important mediators of barrier immunity, predominantly depend on fatty acid (FA) metabolism during helminth infection. Further, in situations where an essential nutrient, such as vitamin A, is limited, ILC2 sustain their function and selectively maintain interleukin 13 (IL-13) production via increased acquisition and utilization of FA. Together, these results reveal that ILC2 preferentially use FAs to maintain their function in the context of helminth infection or malnutrition and propose that enhanced FA usage and FA-dependent IL-13 production by ILC2 could represent a host adaptation to maintain barrier immunity under dietary restriction.


Asunto(s)
Ácidos Grasos/inmunología , Helmintiasis/inmunología , Inmunidad Innata , Linfocitos/inmunología , Desnutrición/inmunología , Animales , Helmintiasis/genética , Helmintiasis/parasitología , Interleucina-13/inmunología , Desnutrición/genética , Desnutrición/parasitología , Ratones , Ratones Noqueados
17.
Sci Transl Med ; 8(337): 337ra65, 2016 05 04.
Artículo en Inglés | MEDLINE | ID: mdl-27147589

RESUMEN

Thymic stromal lymphopoietin (TSLP), interleukin-25 (IL-25), and IL-33 are important initiators of type 2-associated mucosal inflammation and immunity. However, their role in the maintenance of progressive type 2 inflammation and fibrosis is much less clear. Using chronic models of helminth infection and allergic lung inflammation, we show that collective disruption of TSLP, IL-25, and IL-33 signaling suppresses chronic and progressive type 2 cytokine-driven inflammation and fibrosis. In a schistosome lung granuloma model or during chronic Schistosoma mansoni infection in the liver, individual ablation of TSLP, IL-25, or IL-33/ST2 had no impact on the development of IL-4/IL-13-dependent inflammation or fibrosis. However, significant reductions in granuloma-associated eosinophils, hepatic fibrosis, and IL-13-producing type 2 innate lymphoid cells (ILC2s) were observed when signaling of all three mediators was simultaneously disrupted. Combined blockade through monoclonal antibody (mAb) treatment also reduced IL-5 and IL-13 expression during primary and secondary granuloma formation in the lungs. In a model of chronic house dust mite-induced allergic lung inflammation, combined mAb treatment did not decrease established inflammation or fibrosis. TSLP/IL-33 double-knockout mice treated with anti-IL-25 mAb during priming, however, displayed decreased inflammation, mucus production, and lung remodeling in the chronic phase. Together, these studies reveal partially redundant roles for TSLP, IL-25, and IL-33 in the maintenance of type 2 pathology and suggest that in some settings, early combined targeting of these mediators is necessary to ameliorate progressive type 2-driven disease.


Asunto(s)
Citocinas/metabolismo , Fibrosis/inmunología , Inflamación/inmunología , Inflamación/terapia , Interleucina-17/metabolismo , Interleucina-33/metabolismo , Neoplasias Pulmonares/inmunología , Animales , Anticuerpos Monoclonales/uso terapéutico , Citocinas/antagonistas & inhibidores , Citocinas/genética , Femenino , Fibrosis/tratamiento farmacológico , Fibrosis/terapia , Granuloma/tratamiento farmacológico , Granuloma/inmunología , Granuloma/parasitología , Granuloma/terapia , Inflamación/tratamiento farmacológico , Interleucina-13/antagonistas & inhibidores , Interleucina-13/genética , Interleucina-13/metabolismo , Interleucina-17/antagonistas & inhibidores , Interleucina-17/genética , Interleucina-33/antagonistas & inhibidores , Interleucina-33/genética , Interleucina-4/antagonistas & inhibidores , Interleucina-4/genética , Interleucina-4/metabolismo , Neoplasias Pulmonares/tratamiento farmacológico , Neoplasias Pulmonares/parasitología , Neoplasias Pulmonares/terapia , Masculino , Ratones , Ratones Endogámicos BALB C , Ratones Endogámicos C57BL , Ratones Noqueados , Schistosoma mansoni/inmunología , Schistosoma mansoni/patogenicidad , Linfopoyetina del Estroma Tímico
18.
Nat Immunol ; 17(5): 538-44, 2016 May.
Artículo en Inglés | MEDLINE | ID: mdl-27043413

RESUMEN

Acidic mammalian chitinase (AMCase) is known to be induced by allergens and helminths, yet its role in immunity is unclear. Using AMCase-deficient mice, we show that AMCase deficiency reduced the number of group 2 innate lymphoid cells during allergen challenge but was not required for establishment of type 2 inflammation in the lung in response to allergens or helminths. In contrast, AMCase-deficient mice showed a profound defect in type 2 immunity following infection with the chitin-containing gastrointestinal nematodes Nippostrongylus brasiliensis and Heligmosomoides polygyrus bakeri. The impaired immunity was associated with reduced mucus production and decreased intestinal expression of the signature type 2 response genes Il13, Chil3, Retnlb, and Clca1. CD103(+) dendritic cells, which regulate T cell homing, were also reduced in mesenteric lymph nodes of infected AMCase-deficient mice. Thus, AMCase functions as a critical initiator of protective type 2 responses to intestinal nematodes but is largely dispensable for allergic responses in the lung.


Asunto(s)
Quitinasas/inmunología , Tracto Gastrointestinal/inmunología , Inmunidad/inmunología , Infecciones por Strongylida/inmunología , Animales , Quitinasas/genética , Quitinasas/metabolismo , Canales de Cloruro/genética , Canales de Cloruro/inmunología , Canales de Cloruro/metabolismo , Citometría de Flujo , Tracto Gastrointestinal/metabolismo , Tracto Gastrointestinal/parasitología , Expresión Génica/inmunología , Hormonas Ectópicas/genética , Hormonas Ectópicas/inmunología , Hormonas Ectópicas/metabolismo , Interacciones Huésped-Parásitos/inmunología , Hipersensibilidad/genética , Hipersensibilidad/inmunología , Hipersensibilidad/metabolismo , Inmunidad/genética , Péptidos y Proteínas de Señalización Intercelular , Interleucina-13/genética , Interleucina-13/inmunología , Interleucina-13/metabolismo , Lectinas/genética , Lectinas/inmunología , Lectinas/metabolismo , Pulmón/inmunología , Pulmón/metabolismo , Pulmón/patología , Ratones Endogámicos C57BL , Ratones Noqueados , Microscopía Fluorescente , Nematospiroides dubius/inmunología , Nematospiroides dubius/fisiología , Nippostrongylus/inmunología , Nippostrongylus/fisiología , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Infecciones por Strongylida/metabolismo , Infecciones por Strongylida/parasitología , beta-N-Acetilhexosaminidasas/genética , beta-N-Acetilhexosaminidasas/inmunología , beta-N-Acetilhexosaminidasas/metabolismo
19.
J Pathol ; 239(3): 344-54, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27125685

RESUMEN

Persistent or dysregulated IL-13 responses are key drivers of fibrosis in multiple organ systems, and this identifies this cytokine as an important therapeutic target. Nevertheless, the mechanisms by which IL-13 blockade leads to the amelioration of fibrosis remain unclear. Because IFN-γ exhibits potent anti-fibrotic activity, and IL-4Rα signalling antagonizes IFN-γ effector function, compensatory increases in IFN-γ activity following IL-13/IL-4Rα blockade might contribute to the reduction in fibrosis. To investigate the role of IFN-γ, we developed novel IL-13(-/-) /IFN-γ(-/-) double cytokine-deficient mice and examined disease progression in models of type 2-driven fibrosis. As predicted, we showed that fibrosis in the lung and liver are both highly dependent on IL-13. We also observed increased IFN-γ production and inflammatory activity in the tissues of IL-13-deficient mice. Surprisingly, however, an even greater reduction in fibrosis was observed in IL-13/IFN-γ double deficient mice, most notably in the livers of mice chronically infected with Schistosoma mansoni. The increased protection was associated with marked decreases in Tgfb1, Mmp12, and Timp1 mRNA expression in the tissues; reduced inflammation; and decreased expression of important pro-inflammatory mediators such as TNF-α. Experiments conducted with neutralizing monoclonal antibodies to IL-13 and IFN-γ validated the findings with the genetically deficient mice. Together, these studies demonstrate that the reduction in fibrosis observed when IL-13 signalling is suppressed is not dependent on increased IFN-γ activity. Instead, by reducing compensatory increases in type 1-associated inflammation, therapeutic strategies that block IFN-γ and IL-13 activity simultaneously can confer greater protection from progressive fibrosis than IL-13 blockade alone. Published 2016. This article is a U.S. Government work and is in the public domain in the USA.


Asunto(s)
Interferón gamma/genética , Interleucina-13/genética , Cirrosis Hepática/prevención & control , Fibrosis Pulmonar/prevención & control , Schistosoma mansoni/inmunología , Esquistosomiasis mansoni/prevención & control , Animales , Anticuerpos Neutralizantes , Femenino , Granuloma , Humanos , Inflamación , Interferón gamma/metabolismo , Interleucina-13/metabolismo , Hígado/metabolismo , Hígado/patología , Cirrosis Hepática/inmunología , Cirrosis Hepática/patología , Pulmón/metabolismo , Pulmón/patología , Masculino , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fibrosis Pulmonar/inmunología , Fibrosis Pulmonar/patología , Esquistosomiasis mansoni/parasitología , Esquistosomiasis mansoni/patología , Transducción de Señal , Factor de Necrosis Tumoral alfa/genética , Factor de Necrosis Tumoral alfa/metabolismo
20.
Infect Immun ; 84(4): 1032-1044, 2016 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-26810038

RESUMEN

Nematode infection upregulates interleukin-4 (IL-4) and IL-13 and induces STAT6-dependent changes in gut function that promote worm clearance. IL-4 and IL-13 activate the type 2 IL-4 receptor (IL-4R), which contains the IL-13Rα1 and IL-4Rα chains. We used mice deficient in IL-13Rα1 (IL-13Rα1(-/-)) to examine the contribution of IL-13 acting at the type 2 IL-4R to immune and functional responses to primary (Hb1) and secondary (Hb2) infections with the gastrointestinal nematode parasite Heligmosomoides bakeri There were differences between strains in the IL-4 and IL-13 expression responses to Hb1 but not Hb2 infection. Following Hb2 infection, deficient mice had impaired worm expulsion and higher worm fecundity despite normal production of Th2-derived cytokines. The upregulation of IL-25 and IL-13Rα2 in Hb1- and Hb2-infected wild-type (WT) mice was absent in IL-13Rα1(-/-)mice. Goblet cell numbers and resistin-like molecule beta (RELM-ß) expression were attenuated significantly in IL-13Rα1(-/-)mice following Hb2 infections. IL-13Rα1 contributes to the development of alternatively activated macrophages, but the type 1 IL-4R is also important. Hb1 infection had no effects on smooth muscle function or epithelial permeability in either strain, while the enhanced mucosal permeability and changes in smooth muscle function and morphology observed in response to Hb2 infection in WT mice were absent in IL-13Rα1(-/-)mice. Notably, the contribution of claudin-2, which has been linked to IL-13, does not mediate the increased mucosal permeability following Hb2 infection. These results show that activation of IL-13Rα1 is critical for key aspects of the immune and functional responses to Hb2 infection that facilitate expulsion.


Asunto(s)
Heligmosomatoidea , Subunidad alfa1 del Receptor de Interleucina-13/metabolismo , Parasitosis Intestinales/metabolismo , Infecciones por Strongylida/inmunología , Animales , Femenino , Subunidad alfa1 del Receptor de Interleucina-13/genética , Parasitosis Intestinales/inmunología , Mucosa Intestinal/metabolismo , Intestinos/citología , Ratones , Ratones Endogámicos BALB C , Ratones Noqueados , Infecciones por Strongylida/parasitología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA