Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 65
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Nat Immunol ; 17(11): 1300-1311, 2016 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-27668799

RESUMEN

Mucosal-associated invariant T cells (MAIT cells) detect microbial vitamin B2 derivatives presented by the antigen-presenting molecule MR1. Here we defined three developmental stages and checkpoints for the MAIT cell lineage in humans and mice. Stage 1 and stage 2 MAIT cells predominated in thymus, while stage 3 cells progressively increased in abundance extrathymically. Transition through each checkpoint was regulated by MR1, whereas the final checkpoint that generated mature functional MAIT cells was controlled by multiple factors, including the transcription factor PLZF and microbial colonization. Furthermore, stage 3 MAIT cell populations were expanded in mice deficient in the antigen-presenting molecule CD1d, suggestive of a niche shared by MAIT cells and natural killer T cells (NKT cells). Accordingly, this study maps the developmental pathway and checkpoints that control the generation of functional MAIT cells.


Asunto(s)
Diferenciación Celular/inmunología , Células T Invariantes Asociadas a Mucosa/citología , Células T Invariantes Asociadas a Mucosa/fisiología , Timo/inmunología , Timo/metabolismo , Animales , Antígenos CD1d/genética , Biomarcadores , Diferenciación Celular/genética , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Humanos , Inmunofenotipificación , Células Progenitoras Linfoides/inmunología , Células Progenitoras Linfoides/metabolismo , Masculino , Ratones , Ratones Noqueados , MicroARNs/genética
2.
Nat Immunol ; 16(4): 354-65, 2015 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-25729923

RESUMEN

Interleukin 37 (IL-37) and IL-1R8 (SIGIRR or TIR8) are anti-inflammatory orphan members of the IL-1 ligand family and IL-1 receptor family, respectively. Here we demonstrate formation and function of the endogenous ligand-receptor complex IL-37-IL-1R8-IL-18Rα. The tripartite complex assembled rapidly on the surface of peripheral blood mononuclear cells upon stimulation with lipopolysaccharide. Silencing of IL-1R8 or IL-18Rα impaired the anti-inflammatory activity of IL-37. Whereas mice with transgenic expression of IL-37 (IL-37tg mice) with intact IL-1R8 were protected from endotoxemia, IL-1R8-deficient IL-37tg mice were not. Proteomic and transcriptomic investigations revealed that IL-37 used IL-1R8 to harness the anti-inflammatory properties of the signaling molecules Mer, PTEN, STAT3 and p62(dok) and to inhibit the kinases Fyn and TAK1 and the transcription factor NF-κB, as well as mitogen-activated protein kinases. Furthermore, IL-37-IL-1R8 exerted a pseudo-starvational effect on the metabolic checkpoint kinase mTOR. IL-37 thus bound to IL-18Rα and exploited IL-1R8 to activate a multifaceted intracellular anti-inflammatory program.


Asunto(s)
Subunidad alfa del Receptor de Interleucina-18/inmunología , Interleucina-1/inmunología , Leucocitos Mononucleares/inmunología , Receptores de Interleucina-1/inmunología , Transducción de Señal/inmunología , Animales , Línea Celular , Regulación de la Expresión Génica , Humanos , Inmunidad Innata , Inflamación/genética , Inflamación/inmunología , Inflamación/patología , Interleucina-1/genética , Subunidad alfa del Receptor de Interleucina-18/antagonistas & inhibidores , Subunidad alfa del Receptor de Interleucina-18/genética , Leucocitos Mononucleares/efectos de los fármacos , Leucocitos Mononucleares/patología , Lipopolisacáridos/farmacología , Quinasas Quinasa Quinasa PAM/genética , Quinasas Quinasa Quinasa PAM/inmunología , Ratones , Ratones Transgénicos , FN-kappa B/genética , FN-kappa B/inmunología , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/inmunología , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Proto-Oncogénicas/inmunología , Proteínas Proto-Oncogénicas c-fyn/genética , Proteínas Proto-Oncogénicas c-fyn/inmunología , ARN Interferente Pequeño/genética , ARN Interferente Pequeño/inmunología , Proteínas de Unión al ARN/genética , Proteínas de Unión al ARN/inmunología , Proteínas Tirosina Quinasas Receptoras/genética , Proteínas Tirosina Quinasas Receptoras/inmunología , Receptores de Interleucina-1/antagonistas & inhibidores , Receptores de Interleucina-1/deficiencia , Receptores de Interleucina-1/genética , Factor de Transcripción STAT3/genética , Factor de Transcripción STAT3/inmunología , Serina-Treonina Quinasas TOR/genética , Serina-Treonina Quinasas TOR/inmunología , Tirosina Quinasa c-Mer
3.
J Neuroinflammation ; 20(1): 241, 2023 Oct 20.
Artículo en Inglés | MEDLINE | ID: mdl-37864272

RESUMEN

BACKGROUND: Perinatal infection/inflammation is associated with a high risk for neurological injury and neurodevelopmental impairment after birth. Despite a growing preclinical evidence base, anti-inflammatory interventions have not been established in clinical practice, partly because of the range of potential targets. We therefore systematically reviewed preclinical studies of immunomodulation to improve neurological outcomes in the perinatal brain and assessed their therapeutic potential. METHODS: We reviewed relevant studies published from January 2012 to July 2023 using PubMed, Medline (OvidSP) and EMBASE databases. Studies were assessed for risk of bias using the SYRCLE risk of bias assessment tool (PROSPERO; registration number CRD42023395690). RESULTS: Forty preclinical publications using 12 models of perinatal neuroinflammation were identified and divided into 59 individual studies. Twenty-seven anti-inflammatory agents in 19 categories were investigated. Forty-five (76%) of 59 studies reported neuroprotection, from all 19 categories of therapeutics. Notably, 10/10 (100%) studies investigating anti-interleukin (IL)-1 therapies reported improved outcome, whereas half of the studies using corticosteroids (5/10; 50%) reported no improvement or worse outcomes with treatment. Most studies (49/59, 83%) did not control core body temperature (a known potential confounder), and 25 of 59 studies (42%) did not report the sex of subjects. Many studies did not clearly state whether they controlled for potential study bias. CONCLUSION: Anti-inflammatory therapies are promising candidates for treatment or even prevention of perinatal brain injury. Our analysis highlights key knowledge gaps and opportunities to improve preclinical study design that must be addressed to support clinical translation.


Asunto(s)
Antiinflamatorios , Neuroprotección , Embarazo , Animales , Femenino , Humanos , Encéfalo
4.
Int J Mol Sci ; 24(3)2023 Feb 01.
Artículo en Inglés | MEDLINE | ID: mdl-36769133

RESUMEN

Preterm birth is a major contributor to neonatal morbidity and mortality. Complications of prematurity such as bronchopulmonary dysplasia (BPD, affecting the lung), pulmonary hypertension associated with BPD (BPD-PH, heart), white matter injury (WMI, brain), retinopathy of prematurity (ROP, eyes), necrotizing enterocolitis (NEC, gut) and sepsis are among the major causes of long-term morbidity in infants born prematurely. Though the origins are multifactorial, inflammation and in particular the imbalance of pro- and anti-inflammatory mediators is now recognized as a key driver of the pathophysiology underlying these illnesses. Here, we review the involvement of the interleukin (IL)-1 family in perinatal inflammation and its clinical implications, with a focus on the potential of these cytokines as therapeutic targets for the development of safe and effective treatments for early life inflammatory diseases.


Asunto(s)
Displasia Broncopulmonar , Enfermedades del Recién Nacido , Nacimiento Prematuro , Retinopatía de la Prematuridad , Lactante , Embarazo , Femenino , Recién Nacido , Humanos , Interleucina-1 , Recien Nacido Prematuro , Antiinflamatorios/uso terapéutico , Displasia Broncopulmonar/etiología , Displasia Broncopulmonar/tratamiento farmacológico , Enfermedades del Recién Nacido/tratamiento farmacológico , Inflamación/complicaciones , Inflamación/tratamiento farmacológico , Retinopatía de la Prematuridad/tratamiento farmacológico
5.
J Physiol ; 600(19): 4361-4373, 2022 10.
Artículo en Inglés | MEDLINE | ID: mdl-36068678

RESUMEN

The cardiovascular impact of dexamethasone (Dex) is not well understood. Most data are obtained from a 6 week, high-dose regimen, and are limited to findings of hypertension and cardiac hypertrophy. The present study ascertained the impact of low-dose Dex on cardiac indices when administered to extremely preterm infants for lung disease. A pre-post intervention prospective echocardiographic (Echo) study was undertaken, with cardiac assessments performed before and within 24 h after completion of first course of therapy (10 day regimen, cumulative 0.89 mg kg-1 ). Thirty infants with a gestational age of 24.6 ± 1.1 weeks and birthweight of 612 ± 125 g, respectively, were studied. The age at Dex administration was 20 ± 9 days. Fractional inspired oxygen decreased from 0.7 ± 0.23 to 0.35 ± 0.14 (P < 0.001). Patent ductus arteriosus was noted in 20 infants at Echo1. At Echo2, the ductal diameter decreased from 2.16 ± 0.8 to 1.1 ± 0.8 mm (P = 0.0003), with complete closure in 7/20 (35%). A reduction in left pulmonary artery end-diastolic velocity was noted (17 ± 12 to 9 ± 10 cm s-1 , P < 0.001). Pulmonary vascular resistance decreased (increased time to peak velocity/right ventricular ejection time, 0.2 ± 0.03 to 0.23  ± 0.03, P = 0.0001) and right ventricular systolic performance improved (tricuspid annular plane systolic excursion, 4.9 ± 0.8 to 5.5 ± 0.9 mm, P = 0.02). No significant changes in fractional shortening and left ventricular mass were noted. A significant increase in blood pressure was noted. As a percentage of pre-treatment baseline, the mean increase for systolic blood pressure was 20.3% (95% confidence interval = 14-26) on day 2 (P = 0.008). Low-dose Dex influenced cardiovascular parameters related to pulmonary circulation. KEY POINTS: Corticosteroid therapy is frequently used in preterm infants who are dependent on ventilator support. Echocardiographic studies in infants administered a 6 week course of steroids have noted left ventricular hypertrophy, outlet obstruction and hypertension, but no information is available on right heart indices. The cardiopulmonary effects of the current, significantly lesser cumulative dose (10 day regimen, commonly described as 'DART') have not been evaluated. The present study noted a significant influence on ductal and pulmonary circulation indices. Left heart architecture and function was maintained, whereas a significant but transient increase in blood pressure was noted.


Asunto(s)
Hipertensión , Enfermedades Pulmonares , Corticoesteroides , Dexametasona/uso terapéutico , Ecocardiografía , Ventrículos Cardíacos/diagnóstico por imagen , Humanos , Lactante , Recien Nacido Extremadamente Prematuro , Recién Nacido , Oxígeno , Estudios Prospectivos
6.
Nat Immunol ; 11(11): 1014-22, 2010 Nov.
Artículo en Inglés | MEDLINE | ID: mdl-20935647

RESUMEN

The function of interleukin 37 (IL-37; formerly IL-1 family member 7) has remained elusive. Expression of IL-37 in macrophages or epithelial cells almost completely suppressed production of pro-inflammatory cytokines, whereas the abundance of these cytokines increased with silencing of endogenous IL-37 in human blood cells. Anti-inflammatory cytokines were unaffected. Mice with transgenic expression of IL-37 were protected from lipopolysaccharide-induced shock, and showed markedly improved lung and kidney function and reduced liver damage after treatment with lipopolysaccharide. Transgenic mice had lower concentrations of circulating and tissue cytokines (72-95% less) than wild-type mice and showed less dendritic cell activation. IL-37 interacted intracellularly with Smad3 and IL-37-expressing cells and transgenic mice showed less cytokine suppression when endogenous Smad3 was depleted. IL-37 thus emerges as a natural suppressor of innate inflammatory and immune responses.


Asunto(s)
Inmunidad Innata/inmunología , Interleucina-1/inmunología , Animales , Línea Celular , Técnicas de Silenciamiento del Gen , Humanos , Interleucina-1/genética , Ratones , Ratones Transgénicos , Transducción de Señal , Proteína smad3/inmunología
7.
Paediatr Respir Rev ; 44: 19-30, 2022 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-36503648

RESUMEN

Bronchopulmonary dysplasia (BPD) is the most common respiratory sequela of prematurity, and infants born with fetal growth restriction (FGR) are disproportionately represented in BPD statistics, as factors which affect somatic growth may also affect pulmonary growth. Effects of in-utero hypoxia underlying FGR on lung parenchymal architecture predisposing to BPD are well documented, but the pulmonary vascular constructs are not well appreciated. Disruption of angiogenesis during critical periods of lung growth impairs alveolarization, contributing to BPD pathogenesis. Pulmonary artery thickness/stiffness has been noted in FGR in the initial postnatal weeks, and also in well-grown infants with established BPD. The lack of waveform cushioning by the major arteries exposes the pulmonary resistance vessels to higher pulsatile stress, thereby accelerating microvascular disease. Reactive oxygen species, increased sympathetic activity and endothelial dysfunction are common mediators in FGR and BPD; each putative targets for prevention and/or therapeutics using interleukin (IL)-1 receptor antagonist (IL-1Ra), melatonin or inhibition of renin-angiotensin-aldosterone system. While BPD is the archetypal respiratory disease of infancy, effects of FGR on pulmonary function are long-term, extending well into childhood. This narrative links FGR in very/extremely preterm infants with BPD through the vascular affliction as a mechanistic and potentially, therapeutic pathway. Our objectives were to depict the burden of disease for FGR and BPD amongst preterm infants, portray vascular involvement in the placenta in FGR and BPD cohorts, provide high resolution vascular ultrasound information in both cohorts with a view to address therapeutic relevance, and lastly, link this information with paediatric age-group lung diseases.


Asunto(s)
Displasia Broncopulmonar , Enfermedades del Recién Nacido , Lactante , Embarazo , Femenino , Recién Nacido , Humanos , Niño , Retardo del Crecimiento Fetal , Pulmón , Recien Nacido Extremadamente Prematuro
8.
Nucleic Acids Res ; 48(13): 7052-7065, 2020 07 27.
Artículo en Inglés | MEDLINE | ID: mdl-32544249

RESUMEN

Oligonucleotide-based therapeutics have become a reality, and are set to transform management of many diseases. Nevertheless, the modulatory activities of these molecules on immune responses remain incompletely defined. Here, we show that gene targeting 2'-O-methyl (2'OMe) gapmer antisense oligonucleotides (ASOs) can have opposing activities on Toll-Like Receptors 7 and 8 (TLR7/8), leading to divergent suppression of TLR7 and activation of TLR8, in a sequence-dependent manner. Surprisingly, TLR8 potentiation by the gapmer ASOs was blunted by locked nucleic acid (LNA) and 2'-methoxyethyl (2'MOE) modifications. Through a screen of 192 2'OMe ASOs and sequence mutants, we characterized the structural and sequence determinants of these activities. Importantly, we identified core motifs preventing the immunosuppressive activities of 2'OMe ASOs on TLR7. Based on these observations, we designed oligonucleotides strongly potentiating TLR8 sensing of Resiquimod, which preserve TLR7 function, and promote strong activation of phagocytes and immune cells. We also provide proof-of-principle data that gene-targeting ASOs can be selected to synergize with TLR8 agonists currently under investigation as immunotherapies, and show that rational ASO selection can be used to prevent unintended immune suppression of TLR7. Taken together, our work characterizes the immumodulatory effects of ASOs to advance their therapeutic development.


Asunto(s)
Oligodesoxirribonucleótidos Antisentido/farmacología , Receptor Toll-Like 7/metabolismo , Receptor Toll-Like 8/metabolismo , Células Cultivadas , Humanos , Imidazoles/metabolismo , Leucocitos Mononucleares , Oligonucleótidos/metabolismo , Receptor Toll-Like 7/agonistas , Receptor Toll-Like 8/agonistas
9.
J Neuroinflammation ; 18(1): 189, 2021 Aug 31.
Artículo en Inglés | MEDLINE | ID: mdl-34465372

RESUMEN

BACKGROUND: Increased systemic and tissue levels of interleukin (IL)-1ß are associated with greater risk of impaired neurodevelopment after birth. In this study, we tested the hypothesis that systemic IL-1 receptor antagonist (Ra) administration would attenuate brain inflammation and injury in near-term fetal sheep exposed to lipopolysaccharide (LPS). METHODS: Chronically instrumented near-term fetal sheep at 0.85 of gestation were randomly assigned to saline infusion (control, n = 9), repeated LPS infusions (0 h = 300 ng, 24 h = 600 ng, 48 h = 1200 ng, n = 8) or repeated LPS plus IL-1Ra infusions (13 mg/kg infused over 4 h) started 1 h after each LPS infusion (n = 9). Sheep were euthanized 4 days after starting infusions for histology. RESULTS: LPS infusions increased circulating cytokines and were associated with electroencephalogram (EEG) suppression with transiently reduced mean arterial blood pressure, and increased carotid artery perfusion and fetal heart rate (P < 0.05 vs. control for all). In the periventricular and intragyral white matter, LPS-exposure increased IL-1ß immunoreactivity, numbers of caspase 3+ cells and microglia, reduced astrocyte and olig-2+ oligodendrocyte survival but did not change numbers of mature CC1+ oligodendrocytes, myelin expression or numbers of neurons in the cortex and subcortical regions. IL-1Ra infusions reduced circulating cytokines and improved recovery of EEG activity and carotid artery perfusion. Histologically, IL-1Ra reduced microgliosis, IL-1ß expression and caspase-3+ cells, and improved olig-2+ oligodendrocyte survival. CONCLUSION: IL-1Ra improved EEG activity and markedly attenuated systemic inflammation, microgliosis and oligodendrocyte loss following LPS exposure in near-term fetal sheep. Further studies examining the long-term effects on brain maturation are now needed.


Asunto(s)
Encéfalo/efectos de los fármacos , Encefalitis/tratamiento farmacológico , Proteína Antagonista del Receptor de Interleucina 1/farmacología , Lipopolisacáridos/farmacología , Oligodendroglía/efectos de los fármacos , Sustancia Blanca/efectos de los fármacos , Animales , Encéfalo/metabolismo , Encéfalo/patología , Encefalitis/metabolismo , Encefalitis/patología , Femenino , Proteína Antagonista del Receptor de Interleucina 1/uso terapéutico , Oligodendroglía/metabolismo , Oligodendroglía/patología , Embarazo , Ovinos , Sustancia Blanca/metabolismo , Sustancia Blanca/patología
10.
J Cell Mol Med ; 21(6): 1128-1138, 2017 06.
Artículo en Inglés | MEDLINE | ID: mdl-27957795

RESUMEN

Bronchopulmonary dysplasia (BPD) is a severe lung disease of preterm infants, which is characterized by fewer, enlarged alveoli and increased inflammation. BPD has grave consequences for affected infants, but no effective and safe therapy exists. We previously showed that prophylactic treatment with interleukin-1 receptor antagonist (IL-1Ra) prevents murine BPD induced by perinatal inflammation and hyperoxia. Here, we used the same BPD model to assess whether an alternative anti-inflammatory agent, protein C (PC), is as effective as IL-1Ra against BPD. We also tested whether delayed administration or a higher dose of IL-1Ra affects its ability to ameliorate BPD and investigated aspects of drug safety. Pups were reared in room air (21% O2 ) or hyperoxia (65% or 85% O2 ) and received daily injections with vehicle, 1200 IU/kg PC, 10 mg/kg IL-1Ra (early or late onset) or 100 mg/kg IL-1Ra. After 3 or 28 days, lung and brain histology were assessed and pulmonary cytokines were analysed using ELISA and cytokine arrays. We found that PC only moderately reduced the severe impact of BPD on lung structure (e.g. 18% increased alveolar number by PC versus 34% by IL-1Ra); however, PC significantly reduced IL-1ß, IL-1Ra, IL-6 and macrophage inflammatory protein (MIP)-2 by up to 89%. IL-1Ra at 10 mg/kg prevented BPD more effectively than 100 mg/kg IL-1Ra, but only if treatment commenced at day 1 of life. We conclude that prophylactic low-dose IL-1Ra and PC ameliorate BPD and have potential as the first remedy for one of the most devastating diseases preterm babies face.


Asunto(s)
Displasia Broncopulmonar/tratamiento farmacológico , Inflamación/tratamiento farmacológico , Proteína Antagonista del Receptor de Interleucina 1/administración & dosificación , Proteína C/administración & dosificación , Animales , Animales Recién Nacidos , Antiinflamatorios/administración & dosificación , Antiinflamatorios/efectos adversos , Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/patología , Modelos Animales de Enfermedad , Femenino , Humanos , Lactante , Recién Nacido , Inflamación/complicaciones , Inflamación/patología , Proteína Antagonista del Receptor de Interleucina 1/efectos adversos , Pulmón/efectos de los fármacos , Pulmón/patología , Ratones , Embarazo , Proteína C/efectos adversos , Alveolos Pulmonares/efectos de los fármacos , Alveolos Pulmonares/patología
11.
Hum Mol Genet ; 24(2): 436-49, 2015 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-25209981

RESUMEN

Harlequin ichthyosis (HI) is a severe skin disease which leads to neonatal death in ∼50% of cases. It is the result of mutations in ABCA12, a protein that transports lipids required to establish the protective skin barrier needed after birth. To better understand the life-threatening newborn HI phenotype, we analysed the developing epidermis for consequences of lipid dysregulation in mouse models. We observed a pro-inflammatory signature which was characterized by chemokine upregulation in embryonic skin which is distinct from that seen in other types of ichthyosis. Inflammation also persisted in grafted HI skin. To examine the contribution of inflammation to disease development, we overexpressed interleukin-37b to globally suppress fetal inflammation, observing considerable improvements in keratinocyte differentiation. These studies highlight inflammation as an unexpected contributor to HI disease development in utero, and suggest that inhibiting inflammation may reduce disease severity.


Asunto(s)
Ictiosis Lamelar/embriología , Ictiosis Lamelar/inmunología , Animales , Diferenciación Celular , Quimiocinas/genética , Quimiocinas/inmunología , Modelos Animales de Enfermedad , Epidermis/embriología , Epidermis/inmunología , Femenino , Humanos , Ictiosis Lamelar/genética , Ictiosis Lamelar/fisiopatología , Interleucina-1/genética , Interleucina-1/inmunología , Queratinocitos/citología , Masculino , Ratones , Ratones Noqueados , Fenotipo , Piel/embriología , Piel/inmunología
12.
Proc Natl Acad Sci U S A ; 111(42): 15178-83, 2014 Oct 21.
Artículo en Inglés | MEDLINE | ID: mdl-25294929

RESUMEN

IL-1 family member IL-37 limits innate inflammation in models of colitis and LPS-induced shock, but a role in adaptive immunity remains unknown. Here, we studied mice expressing human IL-37b isoform (IL-37tg) subjected to skin contact hypersensitivity (CHS) to dinitrofluorobenzene. CHS challenge to the hapten was significantly decreased in IL-37tg mice compared with wild-type (WT) mice (-61%; P < 0.001 at 48 h). Skin dendritic cells (DCs) were present and migrated to lymph nodes after antigen uptake in IL-37tg mice. When hapten-sensitized DCs were adoptively transferred to WT mice, antigen challenge was greatly impaired in mice receiving DCs from IL-37tg mice compared with those receiving DCs from WT mice (-60%; P < 0.01 at 48 h). In DCs isolated from IL-37tg mice, LPS-induced increase of MHC II and costimulatory molecule CD40 was reduced by 51 and 31%, respectively. In these DCs, release of IL-1ß, IL-6, and IL-12 was reduced whereas IL-10 secretion increased (37%). Consistent with these findings, DCs from IL-37tg mice exhibited a lower ability to stimulate syngeneic and allogeneic naive T cells as well as antigen-specific T cells and displayed enhanced induction of T regulatory (Treg) cells (86%; P < 0.001) in vitro. Histological analysis of CHS skin in mice receiving hapten-sensitized DCs from IL-37tg mice revealed a marked reduction in CD8(+) T cells (-74%) but an increase in Treg cells (2.6-fold). Together, these findings reveal that DCs expressing IL-37 are tolerogenic, thereby impairing activation of effector T-cell responses and inducing Treg cells. IL-37 thus emerges as an inhibitor of adaptive immunity.


Asunto(s)
Inmunidad Adaptativa , Células Dendríticas/citología , Interleucina-1/metabolismo , Animales , Linfocitos T CD8-positivos/citología , Movimiento Celular , Quimiotaxis , Citocinas/metabolismo , Dermatitis por Contacto/inmunología , Dinitrofluorobenceno/química , Citometría de Flujo , Haptenos/química , Humanos , Inmunidad Innata , Ratones , Ratones Endogámicos C57BL , Ratones Transgénicos , Fagocitosis , Piel/metabolismo , Linfocitos T Reguladores/citología
13.
Proc Natl Acad Sci U S A ; 111(7): 2650-5, 2014 Feb 18.
Artículo en Inglés | MEDLINE | ID: mdl-24481253

RESUMEN

IL-37 is a fundamental inhibitor of innate immunity. Human IL-37 has a caspase-1 cleavage site and translocates to the nucleus upon LPS stimulation. Here, we investigated whether caspase-1 processing affects IL-37-mediated suppression of LPS-induced cytokines and the release from cells by analyzing a caspase-1 cleavage site mutant IL-37 (IL-37D20A). Nuclear translocation of IL-37D20A is significantly impaired compared with WT IL-37 in transfected cells. LPS-induced IL-6 was decreased in cells expressing WT IL-37 but not IL-37D20A. The function of IL-37 in transfected bone marrow-derived macrophages is nucleotide-binding oligomerization domain-like receptor family, pyrin domain containing 3 (NLRP3) inflammasome-dependent, because IL-37 transfection in apoptosis-associated speck-like protein containing a carboxyl-terminal caspase recruitment domain- and NLRP3-deficient cells does not reduce levels of IL-6 and IL-1ß upon LPS stimulation. IL-37-expressing macrophages release both precursor and mature IL-37, but only the externalization of mature IL-37 was dependent on ATP. Precursor and mature IL-37 was also secreted from human dendritic cells and peripheral blood mononuclear cells. To determine whether IL-37 is active in the extracellular compartment, we pretreated IL-37 transgenic mice with IL-37-neutralizing antibodies before LPS challenge. In IL-37-expressing mice, neutralizing IL-37 antibodies reversed the suppression of LPS-induced serum IL-6. In contrast, the addition of neutralizing antibody did not reverse suppression of LPS-induced IL-6 in mouse macrophages transfected with IL-37. Although caspase-1 is required for nuclear translocation of intracellular IL-37 and for secretion of mature IL-37, the release of the IL-37 precursor is independent of caspase-1 activation. IL-37 now emerges as a dual-function cytokine with intra- and extracellular properties for suppressing innate inflammation.


Asunto(s)
Anticuerpos Neutralizantes/inmunología , Caspasa 1/metabolismo , Núcleo Celular/metabolismo , Inmunidad Innata/inmunología , Interleucina-1/metabolismo , Transporte Activo de Núcleo Celular/inmunología , Animales , Western Blotting , Caspasa 1/genética , Línea Celular , Escherichia coli , Técnica del Anticuerpo Fluorescente , Humanos , Interleucina-6/sangre , Lipopolisacáridos , Ratones , Ratones Transgénicos , Microscopía Confocal , Mutagénesis Sitio-Dirigida
14.
Am J Respir Cell Mol Biol ; 55(6): 858-868, 2016 12.
Artículo en Inglés | MEDLINE | ID: mdl-27482635

RESUMEN

Bronchopulmonary dysplasia (BPD) is a chronic disease of extreme prematurity that has serious long-term consequences including increased asthma risk. We earlier identified IL-1 receptor antagonist (IL-1Ra) as a potent inhibitor of murine BPD induced by combining perinatal inflammation (intraperitoneal LPS to pregnant dams) and exposure of pups to hyperoxia (fraction of inspired oxygen = 0.65). In this study, we determined whether airway remodeling and hyperresponsiveness similar to asthma are evident in this model, and whether IL-1Ra is protective. During 28-day exposure to air or hyperoxia, pups received vehicle or 10 mg/kg IL-1Ra by daily subcutaneous injection. Lungs were then prepared for histology and morphometry of alveoli and airways, or for real-time PCR, or inflated with agarose to prepare precision-cut lung slices to visualize ex vivo intrapulmonary airway contraction and relaxation by phase-contrast microscopy. In pups reared under normoxic conditions, IL-1Ra treatment did not affect alveolar or airway structure or airway responses. Pups reared in hyperoxia developed a severe BPD-like lung disease, with fewer, larger alveoli, increased subepithelial collagen, and increased expression of α-smooth muscle actin and cyclin D1. After hyperoxia, methacholine elicited contraction with similar potency but with an increased maximum reduction in lumen area (air, 44%; hyperoxia, 89%), whereas dilator responses to salbutamol were maintained. IL-1Ra treatment prevented hyperoxia-induced alveolar disruption and airway fibrosis but, surprisingly, not the increase in methacholine-induced airway contraction. The current study is the first to demonstrate ex vivo airway hyperreactivity caused by systemic maternal inflammation and postnatal hyperoxia, and it reveals further preclinical mechanistic insights into IL-1Ra as a treatment targeting key pathophysiological features of BPD.


Asunto(s)
Remodelación de las Vías Aéreas (Respiratorias) , Hiperreactividad Bronquial/complicaciones , Hiperreactividad Bronquial/metabolismo , Displasia Broncopulmonar/complicaciones , Displasia Broncopulmonar/metabolismo , Proteína Antagonista del Receptor de Interleucina 1/metabolismo , Remodelación de las Vías Aéreas (Respiratorias)/efectos de los fármacos , Albuterol/farmacología , Animales , Hiperreactividad Bronquial/patología , Hiperreactividad Bronquial/fisiopatología , Displasia Broncopulmonar/patología , Displasia Broncopulmonar/fisiopatología , Modelos Animales de Enfermedad , Femenino , Hiperoxia/complicaciones , Hiperoxia/metabolismo , Hiperoxia/patología , Hiperoxia/fisiopatología , Pulmón/efectos de los fármacos , Pulmón/metabolismo , Pulmón/patología , Pulmón/fisiopatología , Ratones Endogámicos C57BL , Contracción Muscular/efectos de los fármacos , Relajación Muscular/efectos de los fármacos , Embarazo , Alveolos Pulmonares/patología
15.
Am J Physiol Lung Cell Mol Physiol ; 311(6): L1076-L1081, 2016 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-27760764

RESUMEN

Bronchopulmonary dysplasia (BPD) is often complicated by pulmonary hypertension (PH). We investigated three biomarkers potentially suitable as screening markers for extremely preterm infants at risk of BPD-associated PH. In this prospective observational cohort study conducted in a tertiary neonatal intensive care unit, 83 preterm infants with BPD born <28-wk gestation and still inpatients at 36-wk corrected age received an echocardiogram and blood tests of B-type natriuretic peptide (BNP), troponin I, and YKL-40. Infants were analyzed according to echocardiographic evidence of tricuspid regurgitation (TR). Thirty infants had evidence of TR on echocardiogram at 36-wk corrected age. Infants with or without TR had similar baseline demographics: mean ± SD gestational age 261 ± 12 vs. 261 ± 11 wk and birth weight 830 ± 206 vs. 815 ± 187 g, respectively. There was no difference in duration of respiratory support. The right ventricular systolic pressure of infants with evidence of TR was 40 ± 16 mmHg. BNP was the only biomarker that proved to be significantly higher in infants with evidence of TR: median (interquartile range) serum level 54.5 (35-105) vs. 41.5 (30-59) pg/ml, P = 0.043. Subgroup analysis of infants with severe BPD requiring discharge on home oxygen or BPD-related mortality revealed similar results. There was no difference between groups for troponin I and YKL-40. In conclusion, increased serum levels of BNP were associated with evidence of TR at 36-wk corrected gestational age in extremely preterm infants, suggesting a potential role as a screening biomarker for BPD-associated PH.


Asunto(s)
Displasia Broncopulmonar/sangre , Proteína 1 Similar a Quitinasa-3/sangre , Hipertensión Pulmonar/sangre , Recien Nacido Extremadamente Prematuro/sangre , Péptido Natriurético Encefálico/sangre , Troponina I/sangre , Biomarcadores/sangre , Displasia Broncopulmonar/complicaciones , Demografía , Femenino , Humanos , Hipertensión Pulmonar/complicaciones , Masculino , Ventilación Pulmonar , Factores de Riesgo
16.
Expert Rev Mol Med ; 18: e12, 2016 06 24.
Artículo en Inglés | MEDLINE | ID: mdl-27341512

RESUMEN

Necrotising enterocolitis (NEC) is an uncommon, but devastating intestinal inflammatory disease that predominantly affects preterm infants. NEC is sometimes dubbed the spectre of neonatal intensive care units, as its onset is insidiously non-specific, and once the disease manifests, the damage inflicted on the baby's intestine is already disastrous. Subsequent sepsis and multi-organ failure entail a mortality of up to 65%. Development of effective treatments for NEC has stagnated, largely because of our lack of understanding of NEC pathogenesis. It is clear, however, that NEC is driven by a profoundly dysregulated immune system. NEC is associated with local increases in pro-inflammatory mediators, e.g. Toll-like receptor (TLR) 4, nuclear factor-κB, tumour necrosis factor, platelet-activating factor (PAF), interleukin (IL)-18, interferon-gamma, IL-6, IL-8 and IL-1ß. Deficiencies in counter-regulatory mechanisms, including IL-1 receptor antagonist (IL-1Ra), TLR9, PAF-acetylhydrolase, transforming growth factor beta (TGF-ß)1&2, IL-10 and regulatory T cells likely facilitate a pro-inflammatory milieu in the NEC-afflicted intestine. There is insufficient evidence to conclude a predominance of an adaptive Th1-, Th2- or Th17-response in the disease. Our understanding of the accompanying regulation of systemic immunity remains poor; however, IL-1Ra, IL-6, IL-8 and TGF-ß1 show promise as biomarkers. Here, we chart the emerging immunological landscape that underpins NEC by reviewing the involvement and potential clinical implications of innate and adaptive immune mediators and their regulation in NEC.


Asunto(s)
Susceptibilidad a Enfermedades/inmunología , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/metabolismo , Factores de Edad , Animales , Biomarcadores , Ensayos Clínicos como Asunto , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Susceptibilidad a Enfermedades/metabolismo , Enterocolitis Necrotizante/diagnóstico , Enterocolitis Necrotizante/epidemiología , Humanos , Sistema Inmunológico/citología , Sistema Inmunológico/inmunología , Sistema Inmunológico/metabolismo , Inmunidad , Factores Inmunológicos/metabolismo , Evaluación del Resultado de la Atención al Paciente , Fenotipo , Receptores Inmunológicos/metabolismo , Receptores de Reconocimiento de Patrones/metabolismo , Factores de Riesgo , Índice de Severidad de la Enfermedad , Transducción de Señal
17.
J Immunol ; 192(2): 589-602, 2014 Jan 15.
Artículo en Inglés | MEDLINE | ID: mdl-24337385

RESUMEN

IL-32 is a multifaceted cytokine with a role in infections, autoimmune diseases, and cancer, and it exerts diverse functions, including aggravation of inflammation and inhibition of virus propagation. We previously identified IL-32 as a critical regulator of endothelial cell (EC) functions, and we now reveal that IL-32 also possesses angiogenic properties. The hyperproliferative ECs of human pulmonary arterial hypertension and glioblastoma multiforme exhibited a markedly increased abundance of IL-32, and, significantly, the cytokine colocalized with integrin αVß3. Vascular endothelial growth factor (VEGF) receptor blockade, which resulted in EC hyperproliferation, increased IL-32 three-fold. Small interfering RNA-mediated silencing of IL-32 negated the 58% proliferation of ECs that occurred within 24 h in scrambled-transfected controls. Reduction of IL-32 neither affected apoptosis (insignificant changes in Bak-1, Bcl-2, Bcl-xL, lactate dehydrogenase, annexin V, and propidium iodide) nor VEGF or TGF-ß levels, but siIL-32-transfected adult and neonatal ECs produced up to 61% less NO, IL-8, and matrix metalloproteinase-9, and up to 3-fold more activin A and endostatin. In coculture-based angiogenesis assays, IL-32γ dose-dependently increased tube formation up to 3-fold; an αVß3 inhibitor prevented this activity and reduced IL-32γ-induced IL-8 by 85%. In matrigel plugs loaded with IL-32γ, VEGF, or vehicle and injected into live mice, we observed the anticipated VEGF-induced increase in neocapillarization (8-fold versus vehicle), but unexpectedly, IL-32γ was equally angiogenic. A second signal such as IFN-γ was required to render cells responsive to exogenous IL-32γ; importantly, this was confirmed using a completely synthetic preparation of IL-32γ. In summary, we add angiogenic properties that are mediated by integrin αVß3 but VEGF-independent to the portfolio of IL-32, implicating a role for this versatile cytokine in pulmonary arterial hypertension and neoplastic diseases.


Asunto(s)
Interleucinas/metabolismo , Neovascularización Patológica/metabolismo , Neovascularización Patológica/patología , Activinas/metabolismo , Animales , Apoptosis/fisiología , Células Cultivadas , Endostatinas/metabolismo , Hipertensión Pulmonar Primaria Familiar , Glioblastoma/embriología , Glioblastoma/patología , Células Endoteliales de la Vena Umbilical Humana , Humanos , Hipertensión Pulmonar/metabolismo , Hipertensión Pulmonar/patología , Integrina alfaVbeta3/metabolismo , Interferón gamma/metabolismo , Interleucina-8/metabolismo , Metaloproteinasa 9 de la Matriz/metabolismo , Ratones , Óxidos de Nitrógeno/metabolismo , Receptores de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores , Receptores de Factores de Crecimiento Endotelial Vascular/metabolismo , Factor de Crecimiento Transformador beta1/metabolismo , Factor A de Crecimiento Endotelial Vascular/metabolismo
18.
Proc Natl Acad Sci U S A ; 110(35): 14384-9, 2013 Aug 27.
Artículo en Inglés | MEDLINE | ID: mdl-23946428

RESUMEN

Bronchopulmonary dysplasia (BPD) is a common lung disease of premature infants, with devastating short- and long-term consequences. The pathogenesis of BPD is multifactorial, but all triggers cause pulmonary inflammation. No therapy exists; therefore, we investigated whether the anti-inflammatory interleukin-1 receptor antagonist (IL-1Ra) prevents murine BPD. We precipitated BPD by perinatal inflammation (lipopolysaccharide injection to pregnant dams) and rearing pups in hyperoxia (65% or 85% O2). Pups were treated daily with IL-1Ra or vehicle for up to 28 d. Vehicle-injected animals in both levels of hyperoxia developed a severe BPD-like lung disease (alveolar number and gas exchange area decreased by up to 60%, alveolar size increased up to fourfold). IL-1Ra prevented this structural disintegration at 65%, but not 85% O2. Hyperoxia depleted pulmonary immune cells by 67%; however, extant macrophages and dendritic cells were hyperactivated, with CD11b and GR1 (Ly6G/C) highly expressed. IL-1Ra partially rescued the immune cell population in hyperoxia (doubling the viable cells), reduced the percentage that were activated by up to 63%, and abolished the unexpected persistence of IL-1α and IL-1ß on day 28 in hyperoxia/vehicle-treated lungs. On day 3, perinatal inflammation and hyperoxia each triggered a distinct pulmonary immune response, with some proinflammatory mediators increasing up to 20-fold and some amenable to partial or complete reversal with IL-1Ra. In summary, our analysis reveals a pivotal role for IL-1α/ß in murine BPD and an involvement for MIP (macrophage inflammatory protein)-1α and TREM (triggering receptor expressed on myeloid cells)-1. Because it effectively shields newborn mice from BPD, IL-1Ra emerges as a promising treatment for a currently irremediable disease that may potentially brighten the prognosis of the tiny preterm patients.


Asunto(s)
Displasia Broncopulmonar/prevención & control , Hiperoxia/complicaciones , Inflamación/complicaciones , Proteína Antagonista del Receptor de Interleucina 1/fisiología , Animales , Displasia Broncopulmonar/etiología , Modelos Animales de Enfermedad , Femenino , Humanos , Recién Nacido , Lipopolisacáridos/farmacología , Pulmón/efectos de los fármacos , Pulmón/inmunología , Ratones , Ratones Endogámicos C57BL , Embarazo
19.
Proc Natl Acad Sci U S A ; 108(40): 16711-6, 2011 Oct 04.
Artículo en Inglés | MEDLINE | ID: mdl-21873195

RESUMEN

IL-37, a newly described member of the IL-1 family, functions as a fundamental inhibitor of innate inflammation and immunity. In the present study, we examined a role for IL-37 during experimental colitis. A transgenic mouse strain was generated to express human IL-37 (hIL-37tg), and these mice were subjected to dextran sulfate sodium (DSS)-induced colitis. Despite the presence of a CMV promoter to drive expression of IL-37, mRNA transcripts were not present in colons at the resting state. Expression was observed only upon disruption of the epithelial barrier, with a six- to sevenfold increase (P = 0.02) on days 3 and 5 after continuous exposure to DSS. During the development of colitis, clinical disease scores were reduced by 50% (P < 0.001), and histological indices of colitis were one-third less in hIL-37tg mice compared with WT counterparts (P < 0.001). Reduced inflammation was associated with decreased leukocyte recruitment into the colonic lamina propria. In addition, release of IL-1ß and TNFα from ex vivo colonic explant tissue was decreased 5- and 13-fold, respectively, compared with WT (P ≤ 0.005), whereas IL-10 was increased sixfold (P < 0.001). However, IL-10 was not required for the anti-inflammatory effects of IL-37 because IL-10-receptor antibody blockade did not reverse IL-37-mediated protection. Mechanistically, IL-37 originating from hematopoietic cells was sufficient to exert anti-inflammatory effects because WT mice reconstituted with hIL-37tg bone marrow were protected from colitis. Thus, IL-37 emerges as key modulator of intestinal inflammation.


Asunto(s)
Colitis/inmunología , Interleucina-1/metabolismo , Análisis de Varianza , Animales , Colitis/patología , Sulfato de Dextran , Citometría de Flujo , Humanos , Interleucina-1/inmunología , Interleucina-1beta/metabolismo , Ratones , Ratones Transgénicos , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Factor de Necrosis Tumoral alfa/metabolismo
20.
Vaccines (Basel) ; 12(3)2024 Mar 01.
Artículo en Inglés | MEDLINE | ID: mdl-38543895

RESUMEN

Introduction: The World Health Organization (WHO) recommends vaccination against hepatitis B as soon as possible following birth for all infants, regardless of prematurity. Hepatitis B vaccination at birth is clearly justified, represents a crucial step in the global control of perinatally acquired hepatitis B and there are no safety concerns in infants born at term. However, there is limited information on the safety of the hepatitis B vaccine in preterm infants, whose immune responses and morbidity risk differ from those in infants born at term. Objectives: The objectives of this paper are to systematically review the literature regarding the safety and risk of adverse events following immunisation (AEFIs) associated with the administration of the hepatitis B vaccine (monovalent or as part of a combination vaccine) to preterm infants. Methods: We performed a search for relevant papers published between 1 January 2002 and 30 March 2023 in the Ovid MEDLINE, Ovid Embase, Cochrane Central Register of Controlled Trials and CINAHL Plus databases. Two authors independently reviewed and analysed each article to include in the systematic review. Narrative synthesis is presented. Results: Twenty-one relevant papers were identified and included in this systematic review. The vast majority of data pertained to multi-antigen (combination) vaccine preparations and vaccination episodes from 6 weeks of age onwards. We found no publications investigating the timing of the birth dose of the hepatitis B vaccine, and AEFI reporting was exclusively short-term (hours to days following administration). There was substantial variability in the reported rate of AEFIs between studies, ranging from 0% to 96%. Regardless of frequency, AEFIs were mostly minor and included injection site reactions, temperature instability and self-limiting cardiorespiratory events. Six studies reported serious adverse events (SAEs) such as the requirement for escalation of respiratory support. However, these occurred predominantly in high-risk infant populations and were rare (~1%). Using the Grading of Recommendations, Assessment, Development, and Evaluations (GRADE) approach, the certainty of evidence was assessed as very low. Conclusions: Despite substantial variability between the relatively small number of published studies in terms of cohort selection, definitions, vaccine preparations and reporting, hepatitis B-containing vaccines (mostly as combination vaccines) appear to be relatively well tolerated in preterm infants from 6 weeks of age. Research focusing on the safety of hepatitis B vaccine in preterm infants specifically within 7 days of birth is lacking, particularly regarding long-term morbidity risk. Further research in this area is required.

SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA