Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 208
Filtrar
1.
Front Immunol ; 15: 1434281, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39104529

RESUMEN

Necrotizing enterocolitis (NEC) is one of the most devasting diseases affecting preterm neonates. However, despite a lot of research, NEC's pathogenesis remains unclear. It is known that the pathogenesis is a multifactorial process, including (1) a pathological microbiome with abnormal bacterial colonization, (2) an immature immune system, (3) enteral feeding, (3) an impairment of microcirculation, and (4) possibly ischemia-reperfusion damage to the intestine. Overall, the immaturity of the mucosal barrier and the increased expression of Toll-like receptor 4 (TLR4) within the intestinal epithelium result in an intestinal hyperinflammation reaction. Concurrently, a deficiency in counter-regulatory mediators can be seen. The sum of these processes can ultimately result in intestinal necrosis leading to very high mortality rates of the affected neonates. In the last decade no substantial advances in the treatment of NEC have been made. Thus, NEC animal models as well as in vitro models have been employed to better understand NEC's pathogenesis on a cellular and molecular level. This review will highlight the different models currently in use to study immunological aspects of NEC.


Asunto(s)
Modelos Animales de Enfermedad , Enterocolitis Necrotizante , Enterocolitis Necrotizante/inmunología , Humanos , Animales , Recién Nacido , Mucosa Intestinal/inmunología , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Microbioma Gastrointestinal/inmunología , Receptor Toll-Like 4/metabolismo , Receptor Toll-Like 4/inmunología , Recien Nacido Prematuro/inmunología
2.
Adv Exp Med Biol ; 1449: 43-57, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39060730

RESUMEN

Intestinal bacteria, also known as gut microbiota, are a rich ecology of microorganisms found in the human digestive tract. Extensive study has highlighted their critical relevance in preserving human health. New research has revealed that bacterial viability is not invariably necessary to induce health benefits. Postbiotics (defined soluble substances produced as a byproduct of the metabolic processes of living microbes) have thus emerged as an important topic of research. They contribute to shaping the gut microbiota, exert immune-modulation activity, and improve the integrity of the gut barrier.Alterations in preterm gut colonization associated with intestinal barrier immaturity and the increased reactivity of the intestinal mucosa to colonizing bacteria have been implicated in the pathogenesis of necrotizing enterocolitis. Postbiotics have shown promising outcomes in reducing the risk of developing NEC, lowering inflammation, encouraging the development of good bacteria, and strengthening the intestinal barrier. This is an important advancement in newborn care and highlights the potential of postbiotics to avoid severe intestinal disorders.


Asunto(s)
Enterocolitis Necrotizante , Microbioma Gastrointestinal , Enterocolitis Necrotizante/prevención & control , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/inmunología , Humanos , Recién Nacido , Mucosa Intestinal/microbiología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Probióticos/uso terapéutico , Animales , Recien Nacido Prematuro , Intestinos/microbiología
3.
Int Immunopharmacol ; 139: 112590, 2024 Sep 30.
Artículo en Inglés | MEDLINE | ID: mdl-38996778

RESUMEN

BACKGROUND: Neonatal necrotizing enterocolitis (NEC) is one of the most prevalent and severe intestinal emergencies in newborns. The inflammatory activation of macrophages is associated with the intestinal injury of NEC. The neuroimmune regulation mediated by α7 nicotinic acetylcholine receptor (α7nAChR) plays an important role in regulating macrophage activation and inflammation progression, but in NEC remains unclear. This study aims to explore the effect of macrophage α7nAChR on NEC. METHODS: Mice NEC model were conducted with high-osmolarity formula feeding, hypoxia, and cold stimulation. The α7nAChR agonist PNU-282987 and mTOR inhibitor rapamycin were treated by intraperitoneal injections in mice. The expression and distribution of macrophages, α7nAChR, and phospho-mammalian target of rapamycin (p-mTOR) in the intestines of NEC patients and mice was assessed using immunohistochemistry, immunofluorescence, and flow cytometry. The expression of NLRP3, activated caspase-1 and IL-1ß in mice intestines was detected by flow cytometry, western blot or ELISA. In vitro, the mouse RAW264.7 macrophage cell line was also cultured followed by various treatments. Expression of p-mTOR, NLRP3, activated caspase-1, and IL-1ß in macrophages was determined. RESULTS: Macrophages accumulated in the intestines and the expression of α7nAChR in the mucosal and submucosal layers of the intestines was increased in both the NEC patients and mice. The p-mTOR and CD68 were increased and co-localized in intestines of NEC patients. In vitro, α7nAChR agonist PNU-282987 significantly reduced the increase of NLRP3, activated caspase-1, and IL-1ß in macrophages. PNU-282987 also significantly reduced the increase of p-mTOR. The effect was blocked by AMPK inhibitor compound C. The expression of NLRP3, activated caspase-1, and IL-1ß was inhibited after mTOR inhibitor rapamycin treatment. In NEC model mice, PNU-282987 reduced the expression of p-mTOR, NLRP3, activated caspase-1, and IL-1ß in the intestine. Meanwhile, rapamycin significantly attenuated NLRP3 activation and the release of IL-1ß. Moreover, the proportion of intestinal macrophages and intestinal injury decreased after PNU-282987 treatment. CONCLUSION: Macrophage α7nAChR activation mitigates NLRP3 inflammasome activation by modulating mTOR phosphorylation, and subsequently alleviates intestinal inflammation and injury in NEC.


Asunto(s)
Benzamidas , Compuestos Bicíclicos con Puentes , Enterocolitis Necrotizante , Interleucina-1beta , Macrófagos , Proteína con Dominio Pirina 3 de la Familia NLR , Transducción de Señal , Serina-Treonina Quinasas TOR , Receptor Nicotínico de Acetilcolina alfa 7 , Animales , Receptor Nicotínico de Acetilcolina alfa 7/metabolismo , Receptor Nicotínico de Acetilcolina alfa 7/agonistas , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/inmunología , Ratones , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Humanos , Compuestos Bicíclicos con Puentes/farmacología , Compuestos Bicíclicos con Puentes/uso terapéutico , Macrófagos/efectos de los fármacos , Macrófagos/metabolismo , Macrófagos/inmunología , Interleucina-1beta/metabolismo , Benzamidas/farmacología , Benzamidas/uso terapéutico , Recién Nacido , Transducción de Señal/efectos de los fármacos , Células RAW 264.7 , Modelos Animales de Enfermedad , Masculino , Ratones Endogámicos C57BL , Animales Recién Nacidos , Femenino
4.
Front Immunol ; 15: 1403018, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38881893

RESUMEN

Necrotizing enterocolitis (NEC) is a severe gastrointestinal disease primarily affecting premature neonates, marked by poorly understood pro-inflammatory signaling cascades. Recent advancements have shed light on a subset of endogenous molecular patterns, termed chromatin-associated molecular patterns (CAMPs), which belong to the broader category of damage-associated molecular patterns (DAMPs). CAMPs play a crucial role in recognizing pattern recognition receptors and orchestrating inflammatory responses. This review focuses into the realm of CAMPs, highlighting key players such as extracellular cold-inducible RNA-binding protein (eCIRP), high mobility group box 1 (HMGB1), cell-free DNA, neutrophil extracellular traps (NETs), histones, and extracellular RNA. These intrinsic molecules, often perceived as foreign, have the potential to trigger immune signaling pathways, thus contributing to NEC pathogenesis. In this review, we unravel the current understanding of the involvement of CAMPs in both preclinical and clinical NEC scenarios. We also focus on elucidating the downstream signaling pathways activated by these molecular patterns, providing insights into the mechanisms that drive inflammation in NEC. Moreover, we scrutinize the landscape of targeted therapeutic approaches, aiming to mitigate the impact of tissue damage in NEC. This in-depth exploration offers a comprehensive overview of the role of CAMPs in NEC, bridging the gap between preclinical and clinical insights.


Asunto(s)
Alarminas , Cromatina , Enterocolitis Necrotizante , Humanos , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/inmunología , Alarminas/metabolismo , Alarminas/inmunología , Cromatina/metabolismo , Animales , Transducción de Señal , Recién Nacido , Proteína HMGB1/metabolismo
5.
Cell Mol Immunol ; 21(6): 575-588, 2024 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-38632385

RESUMEN

Neonates are susceptible to inflammatory disorders such as necrotizing enterocolitis (NEC) due to their immature immune system. The timely appearance of regulatory immune cells in early life contributes to the control of inflammation in neonates, yet the underlying mechanisms of which remain poorly understood. In this study, we identified a subset of neonatal monocytes characterized by high levels of neuropilin-1 (Nrp1), termed Nrp1high monocytes. Compared with their Nrp1low counterparts, Nrp1high monocytes displayed potent immunosuppressive activity. Nrp1 deficiency in myeloid cells aggravated the severity of NEC, whereas adoptive transfer of Nrp1high monocytes led to remission of NEC. Mechanistic studies showed that Nrp1, by binding to its ligand Sema4a, induced intracellular p38-MAPK/mTOR signaling and activated the transcription factor KLF4. KLF4 transactivated Nos2 and enhanced the production of nitric oxide (NO), a key mediator of immunosuppression in monocytes. These findings reveal an important immunosuppressive axis in neonatal monocytes and provide a potential therapeutic strategy for treating inflammatory disorders in neonates.


Asunto(s)
Animales Recién Nacidos , Enterocolitis Necrotizante , Inflamación , Factor 4 Similar a Kruppel , Factores de Transcripción de Tipo Kruppel , Monocitos , Neuropilina-1 , Monocitos/metabolismo , Monocitos/inmunología , Animales , Neuropilina-1/metabolismo , Neuropilina-1/genética , Inflamación/patología , Inflamación/inmunología , Humanos , Factores de Transcripción de Tipo Kruppel/metabolismo , Factores de Transcripción de Tipo Kruppel/genética , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/prevención & control , Ratones , Óxido Nítrico Sintasa de Tipo II/metabolismo , Óxido Nítrico/metabolismo , Transducción de Señal , Serina-Treonina Quinasas TOR/metabolismo , Ratones Endogámicos C57BL , Recién Nacido , Proteínas Quinasas p38 Activadas por Mitógenos/metabolismo , Ratones Noqueados
6.
Int Immunopharmacol ; 133: 112058, 2024 May 30.
Artículo en Inglés | MEDLINE | ID: mdl-38613883

RESUMEN

Fetal growth restriction (FGR) is a major cause of premature and low-weight births, which increases the risk of necrotizing enterocolitis (NEC); however, the association remains unclear. We report a close correlation between placental polymorphonuclear myeloid-derived suppressor cells (PMN-MDSCs) and NEC. Newborns with previous FGR exhibited intestinal inflammation and more severe NEC symptoms than healthy newborns. Placental PMN-MDSCs are vital regulators of fetal development and neonatal gut inflammation. Placental single-cell transcriptomics revealed that PMN-MDSCs populations and olfactomedin-4 gene (Olfm4) expression levels were significantly increased in PMN-MDSCs in later pregnancy compared to those in early pregnancy and non-pregnant females. Female mice lacking Olfm4 in myeloid cells mated with wild-type males showed FGR during pregnancy, with a decreased placental PMN-MDSCs population and expression of growth-promoting factors (GPFs) from placental PMN-MDSCs. Galectin-3 (Gal-3) stimulated the OLFM4-mediated secretion of GPFs by placental PMN-MDSCs. Moreover, GPF regulation via OLFM4 in placental PMN-MDSCs was mediated via hypoxia inducible factor-1α (HIF-1α). Notably, the offspring of mothers lacking Olfm4 exhibited intestinal inflammation and were susceptible to NEC. Additionally, OLFM4 expression decreased in placental PMN-MDSCs from pregnancies with FGR and was negatively correlated with neonatal morbidity. These results revealed that placental PMN-MDSCs contributed to fetal development and ameliorate newborn intestinal inflammation.


Asunto(s)
Retardo del Crecimiento Fetal , Células Supresoras de Origen Mieloide , Placenta , Animales , Femenino , Embarazo , Humanos , Placenta/inmunología , Placenta/metabolismo , Recién Nacido , Células Supresoras de Origen Mieloide/inmunología , Células Supresoras de Origen Mieloide/metabolismo , Retardo del Crecimiento Fetal/inmunología , Ratones , Ratones Noqueados , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/metabolismo , Factor Estimulante de Colonias de Granulocitos/metabolismo , Factor Estimulante de Colonias de Granulocitos/genética , Ratones Endogámicos C57BL , Masculino , Galectinas/metabolismo , Galectinas/genética , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Subunidad alfa del Factor 1 Inducible por Hipoxia/genética , Intestinos/inmunología , Intestinos/patología
7.
Mucosal Immunol ; 17(4): 599-617, 2024 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-38555026

RESUMEN

Proper development of mucosal immunity is critical for human health. Over the past decade, it has become evident that in humans, this process begins in utero. However, there are limited data on the unique features and functions of fetal mucosal immune cells. To address this gap, we integrated several single-cell ribonucleic acid sequencing datasets of the human small intestine (SI) to create an SI transcriptional atlas throughout the human life span, ranging from the first trimester to adulthood, with a focus on immune cells. Fetal SI displayed a complex immune landscape comprising innate and adaptive immune cells that exhibited distinct transcriptional programs from postnatal samples, especially compared with pediatric and adult samples. We identified shifts in myeloid populations across gestation and progression of memory T-cell states throughout the human lifespan. In particular, there was a marked shift of memory T cells from those with stem-like properties in the fetal samples to fully differentiated cells with a high expression of activation and effector function genes in adult samples, with neonatal samples containing both features. Finally, we demonstrate that the SI developmental atlas can be used to elucidate improper trajectories linked to mucosal diseases by implicating developmental abnormalities underlying necrotizing enterocolitis, a severe intestinal complication of prematurity. Collectively, our data provide valuable resources and important insights into intestinal immunity that will facilitate regenerative medicine and disease understanding.


Asunto(s)
Feto , Intestino Delgado , Análisis de la Célula Individual , Humanos , Intestino Delgado/inmunología , Intestino Delgado/metabolismo , Feto/inmunología , Adulto , Niño , Células T de Memoria/inmunología , Células T de Memoria/metabolismo , Adolescente , Femenino , Preescolar , Inmunidad Mucosa , Lactante , Recién Nacido , Mucosa Intestinal/inmunología , Mucosa Intestinal/metabolismo , Transcriptoma , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/genética , Inmunidad Innata , Inmunidad Adaptativa , Perfilación de la Expresión Génica , Embarazo , Diferenciación Celular , Memoria Inmunológica
8.
J Pediatr Gastroenterol Nutr ; 75(1): 97-103, 2022 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-35442233

RESUMEN

OBJECTIVES: Human milk reduces the incidence of necrotizing enterocolitis (NEC). Prior studies have demonstrated that exogenous surfactant protein-A (SP-A) modulates intestinal inflammation, reduces NEC-like pathology in SP-A-deficient (SPAKO) pups, and may contribute to breast milk's immunomodulatory potential. We hypothesize that SP-A is present in milk and impacts inflammatory responses in the terminal ileum of neonatal mice. METHODS: Human milk was collected at postpartum days 1-3 and 28. Mouse milk was collected at postpartum days 1-10. SP-A was detected in milk through immunoprecipitation and western blot analysis. The impact of murine wild-type (WT) milk on SPAKO pup ileum was evaluated in a model of intestinal inflammation via cross-rearing experiments. Terminal ileum was evaluated for inflammatory cytokine and toll-like receptor 4 (TLR4) mRNA expression via quantitative real-time RT-PCR. RESULTS: SP-A was detected in human milk and wild type (WT) mouse milk, but not in SPAKO mouse milk. Expression of TLR4, interleukin (IL)-1ß, IL-6, and tumor necrosis factor (TNF)-α was decreased in SPAKO pups reared with WT dams compared to SPAKO pups reared with SPAKO dams, with a peak effect at day of life 14. When inflammation was induced using a lipopolysaccharide-induced model of inflammation, expression of TLR4, IL-1ß, IL-6, CXCL-1, and TNF-α was significantly lower in SPAKO pups reared with WT dams compared to SPAKO pups reared with SPAKO dams. CONCLUSIONS: SP-A is present in human and murine milk and plays a role in lowering inflammation in murine pup terminal ileum. Both baseline inflammation and induced inflammatory responses are reduced via exposure to SP-A in milk with the effect amplified in inflammatory conditions.


Asunto(s)
Enterocolitis Necrotizante , Leche Humana , Proteína A Asociada a Surfactante Pulmonar , Receptor Toll-Like 4 , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/inmunología , Femenino , Humanos , Agentes Inmunomoduladores/farmacología , Recién Nacido , Inflamación/inmunología , Inflamación/metabolismo , Interleucina-6 , Ratones , Leche Humana/efectos de los fármacos , Leche Humana/inmunología , Proteína A Asociada a Surfactante Pulmonar/genética , Proteína A Asociada a Surfactante Pulmonar/inmunología , Tensoactivos , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/metabolismo , Factor de Necrosis Tumoral alfa/metabolismo
9.
Biomed Res Int ; 2021: 1877075, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34712727

RESUMEN

The objective of this study was to explore the therapeutic effects of berberine on necrotic enteritis (NE) in broilers caused by Clostridium perfringens. A total of 240 1-day-old Arbor Acres chicks were divided into four groups, as negative controls (NC), positive controls (PC), berberine- (BER-) treated, or lincomycin- (LMY-) treated groups. Broilers were challenged with C. perfringens at 15-21 days of age, followed by BER or LMY supplied in drinking water for 7 days. Experimental results showed that C. perfringens infection significantly decreased growth performance and increased intestinal necrosis index and the number of C. perfringens present to 6.45 Log10CFU/g (P < 0.001). Proinflammatory cytokines in the ileum were significantly increased, but the expression of ileal tight junction proteins occludin and claudin-1 was significantly reduced. Both BER and LMY ameliorated some of these observations. Compared with the PC group, the number of C. perfringens in the cecum was significantly decreased following treatment (P < 0.001), and growth performance and small intestine morphology were similar to those of the NC group (P > 0.05). IL-1ß, IL-6, and TNF-α levels as well as occludin and claudin-1 expression were also significantly improved (P < 0.05). BER has the potential to replace antibiotics for NE caused by C. perfringens.


Asunto(s)
Berberina/farmacología , Pollos/inmunología , Pollos/microbiología , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/microbiología , Intestinos/microbiología , Intestinos/patología , Animales , Ciego/efectos de los fármacos , Ciego/microbiología , Clostridium perfringens/efectos de los fármacos , Citocinas/metabolismo , Dieta , Enterocolitis Necrotizante/genética , Femenino , Regulación de la Expresión Génica/efectos de los fármacos , Íleon/metabolismo , Intestinos/inmunología , Lincomicina/farmacología , ARN Mensajero/genética , ARN Mensajero/metabolismo , Proteínas de Uniones Estrechas/genética , Proteínas de Uniones Estrechas/metabolismo
10.
Mediators Inflamm ; 2021: 6259381, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34675753

RESUMEN

OBJECTIVE: To determine the role of sodium butyrate in intestinal inflammation via regulation of high-mobility group box-1 (HMGB1), we analyzed the potential mechanism in necrotizing enterocolitis (NEC) in a neonatal mouse model. METHODS: A NEC model was created with hypoxia and cold exposure and artificial overfeeding. C57BL/6 neonatal mice were randomized into three groups: the control, untreated NEC, and sodium butyrate (150 mM)-pretreated NEC groups. Pathological variations in ileocecal intestinal tissue were observed by HE staining and scored in a double-blind manner. The mRNA expression levels of HMGB1, Toll-like receptor 4 (TLR4), nuclear factor-κB (NF-κB), and inflammatory cytokines in intestinal tissues were determined by quantitative real-time PCR. The protein levels of HMGB1 and associated cytokines in intestinal tissues were evaluated using ELISA. The relative protein expression levels of TLR4 and NF-κB in intestinal tissues were quantified by western blot. RESULTS: Sodium butyrate administration improved the body weight and survival rate of NEC mice; relieved intestinal pathological injury; reduced the intestinal expression of HMGB1, TLR4, NF-κB, interleukin- (IL-) 1ß, IL-6, IL-8, and TNF-α; and increased the intestinal expression of IL-10 (P < 0.05). Treatment with butyrate decreased the proportion of opportunistic Clostridium_sensu_stricto_1 and Enterococcus and increased the proportion of beneficial Firmicutes and Lactobacillus in the NEC model. CONCLUSIONS: Sodium butyrate intervention relieves intestinal inflammation and partially corrects the disrupted intestinal flora in mice with NEC.


Asunto(s)
Ácido Butírico/uso terapéutico , Enterocolitis Necrotizante/tratamiento farmacológico , Animales , Ácido Butírico/farmacología , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/patología , Femenino , Proteína HMGB1/genética , Proteína HMGB1/fisiología , Intestinos/microbiología , Intestinos/patología , Masculino , Ratones , Ratones Endogámicos C57BL , FN-kappa B/genética , FN-kappa B/fisiología , Distribución Aleatoria , Receptor Toll-Like 4/genética , Receptor Toll-Like 4/fisiología
11.
BMC Microbiol ; 21(1): 225, 2021 08 06.
Artículo en Inglés | MEDLINE | ID: mdl-34362295

RESUMEN

BACKGROUND: Necrotising enterocolitis (NEC) is a devastating bowel disease, primarily affecting premature infants, with a poorly understood aetiology. Prior studies have found associations in different cases with an overabundance of particular elements of the faecal microbiota (in particular Enterobacteriaceae or Clostridium perfringens), but there has been no explanation for the different results found in different cohorts. Immunological studies have indicated that stimulation of the TLR4 receptor is involved in development of NEC, with TLR4 signalling being antagonised by the activated TLR9 receptor. We speculated that differential stimulation of these two components of the signalling pathway by different microbiota might explain the dichotomous findings of microbiota-centered NEC studies. Here we used shotgun metagenomic sequencing and qPCR to characterise the faecal microbiota community of infants prior to NEC onset and in a set of matched controls. Bayesian regression was used to segregate cases from control samples using both microbial and clinical data. RESULTS: We found that the infants suffering from NEC fell into two groups based on their microbiota; one with low levels of CpG DNA in bacterial genomes and the other with high abundances of organisms expressing LPS. The identification of these characteristic communities was reproduced using an external metagenomic validation dataset. We propose that these two patterns represent the stimulation of a common pathway at extremes; the LPS-enriched microbiome suggesting overstimulation of TLR4, whilst a microbial community with low levels of CpG DNA suggests reduction of the counterbalance to TLR4 overstimulation. CONCLUSIONS: The identified microbial community patterns support the concept of NEC resulting from TLR-mediated pathways. Identification of these signals suggests characteristics of the gastrointestinal microbial community to be avoided to prevent NEC. Potential pre- or pro-biotic treatments may be designed to optimise TLR signalling.


Asunto(s)
Enterocolitis Necrotizante/microbiología , Células Epiteliales/inmunología , Microbioma Gastrointestinal/genética , Enfermedades del Prematuro/microbiología , Receptor Toll-Like 4/inmunología , Teorema de Bayes , ADN Bacteriano/genética , Enterocolitis Necrotizante/inmunología , Células Epiteliales/microbiología , Heces/microbiología , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/inmunología , Metagenómica , ARN Ribosómico 16S/genética , Receptor Toll-Like 4/genética
13.
Cell Rep Med ; 2(6): 100320, 2021 06 15.
Artículo en Inglés | MEDLINE | ID: mdl-34195684

RESUMEN

Necrotizing enterocolitis (NEC) is a deadly intestinal inflammatory disorder that primarily affects premature infants and lacks adequate therapeutics. Interleukin (IL)-22 plays a critical role in gut barrier maintenance, promoting epithelial regeneration, and controlling intestinal inflammation in adult animal models. However, the importance of IL-22 signaling in neonates during NEC remains unknown. We investigated the role of IL-22 in the neonatal intestine under homeostatic and inflammatory conditions by using a mouse model of NEC. Our data reveal that Il22 expression in neonatal murine intestine is negligible until weaning, and both human and murine neonates lack IL-22 production during NEC. Mice deficient in IL-22 or lacking the IL-22 receptor in the intestine display a similar susceptibility to NEC, consistent with the lack of endogenous IL-22 during development. Strikingly, treatment with recombinant IL-22 during NEC substantially reduces inflammation and enhances epithelial regeneration. These findings may provide a new therapeutic strategy to attenuate NEC.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Interleucinas/genética , Mucosa Intestinal/inmunología , Proteínas Recombinantes/farmacología , Regeneración/inmunología , Animales , Animales Recién Nacidos , Quimiocina CXCL1/genética , Quimiocina CXCL1/inmunología , Quimiocina CXCL2/genética , Quimiocina CXCL2/inmunología , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/patología , Microbioma Gastrointestinal/inmunología , Regulación del Desarrollo de la Expresión Génica , Humanos , Recién Nacido , Enfermedades del Recién Nacido/inmunología , Enfermedades del Recién Nacido/microbiología , Enfermedades del Recién Nacido/patología , Recien Nacido Prematuro , Interleucina-1beta/genética , Interleucina-1beta/inmunología , Interleucinas/inmunología , Mucosa Intestinal/metabolismo , Mucosa Intestinal/microbiología , Ratones , Ratones Noqueados , Isoformas de Proteínas/genética , Isoformas de Proteínas/inmunología , Receptores de Interleucina/genética , Receptores de Interleucina/inmunología , Regeneración/genética , Transducción de Señal , Destete , Interleucina-22
14.
Gut Microbes ; 13(1): 1951113, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34264803

RESUMEN

Premature birth, especially if born before week 32 of gestation, is associated with increased risk of neonatal morbidity and mortality. Prophylactic use of probiotics has been suggested to protect preterm infants via supporting a healthy gut microbiota (GM) development, but the suggested strains and doses vary between studies. In this study, we profiled the GM of 5, 10 and 30-day fecal samples from two cohorts of preterm neonates (born <30 weeks of gestation) recruited in the same neonatal intensive care unit. One cohort (n = 165) was recruited from September 2006 to January 2009 before probiotics were introduced in the clinic. The second cohort (n = 87) was recruited from May 2010 to October 2011 after introducing Lacticaseibacillus rhamnosus GG and Bifidobacterium animalis ssp. lactis BB-12 supplementation policy. Through V3-V4 region 16S rRNA gene amplicon sequencing, a distinct increase of L. rhamnosus and B. animalis was found in the fecal samples of neonates supplemented with probiotics. During the first 30 days of life, the preterm GM went through similarly patterned progression of bacterial populations. Staphylococcus and Weissella dominated in early samples, but was gradually overtaken by Veillonella, Enterococcus and Enterobacteriaceae. Probiotic supplementation was associated with pronounced reduction of Weissella, Veillonella spp. and the opportunistic pathogen Klebsiella. Potential nosocomial pathogens Citrobacter and Chryseobacterium species also gradually phased out. In conclusion, probiotic supplementation to preterm neonates affected gut colonization by certain bacteria, but did not change the overall longitudinal bacterial progression in the neonatal period.Abbreviations: GM: Gut microbiota; ASV: Amplicon sequence variant; NEC: Necrotizing enterocolitis; DOL: Days of life; NICU: Neonatal intensive care unit; ESPGHAN: European Society for Pediatric Gastroenterology, Hepatology and Nutrition; Db-RDA: Distance-based redundancy analysis; PERMANOVA: Permutational multivariate analysis of variance; ANCOM: Analysis of compositions of microbiomes; LGG: Lacticaseibacillus (former Lactobacillus) rhamnosus GG; BB-12: Bifidobacterium animalis ssp. lactis BB-12; DGGE: Denaturing Gradient Gel Electrophoresis.


Asunto(s)
Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/inmunología , Microbioma Gastrointestinal/efectos de los fármacos , Recien Nacido Prematuro/crecimiento & desarrollo , Probióticos/farmacología , Probióticos/uso terapéutico , Bifidobacterium animalis/aislamiento & purificación , Estudios de Cohortes , Dinamarca , Heces/microbiología , Femenino , Microbioma Gastrointestinal/inmunología , Humanos , Recién Nacido , Enfermedades del Recién Nacido/tratamiento farmacológico , Enfermedades del Recién Nacido/inmunología , Recien Nacido Prematuro/inmunología , Lactobacillus/aislamiento & purificación , Masculino
15.
Int Immunopharmacol ; 96: 107787, 2021 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-34162151

RESUMEN

Despite the fact that melatonin regulates the expression of long noncoding RNAs (lncRNAs) under different physiological and pathological conditions, it has not been confirmed whether melatonin-induced lncRNAs regulate the differentiation of Treg and Th17 cells. Herein, we show that the expression of LINC01512 is significantly down-regulated and correlates with imbalanced Treg/Th17 ratios in necrotising enterocolitis (NEC) tissues. Through gain- and loss-of-function approaches, we found that LINC01512 promotes the differentiation of Treg cells but interferes with that of Th17 cells. Mechanistically, LINC01512 promotes SIRT1 in Treg and Th17 cells, and subsequently enhances the differentiation of Treg cells and inhibits that of Th17 cells. Furthermore, we demonstrate that melatonin up-regulates the transcription of LINC01512 via the AMPK signalling pathway and that the blockade of AMPK represses LINC01512 expression in Treg and Th17 cells. Overall, our results confirm that SIRT1-regulated differentiation of Treg/Th17 cells is actually modulated by melatonin-induced LINC0512. Moreover, manipulation of the AMPK/LINC01512/SIRT1 axis via melatonin may be a novel therapeutic approach to reduce inflammation.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Melatonina/farmacología , ARN Largo no Codificante , Sirtuina 1/inmunología , Linfocitos T Reguladores/efectos de los fármacos , Células Th17/efectos de los fármacos , Diferenciación Celular , Humanos , Linfocitos T Reguladores/inmunología , Células Th17/inmunología
16.
Front Immunol ; 12: 650709, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33968047

RESUMEN

Prior to birth, the neonate has limited exposure to pathogens. The transition from the intra-uterine to the postnatal environment initiates a series of complex interactions between the newborn host and a variety of potential pathogens that persist over the first few weeks of life. This transition is particularly complex in the case of the premature and very low birth weight infant, who may be susceptible to many disorders as a result of an immature and underdeveloped immune system. Chief amongst these disorders is necrotizing enterocolitis (NEC), an acute inflammatory disorder that leads to necrosis of the intestine, and which can affect multiple systems and have the potential to result in long term effects if the infant is to survive. Here, we examine what is known about the interplay of the immune system with the maternal uterine environment, microbes, nutritional and other factors in the pathogenesis of neonatal pathologies such as NEC, while also taking into consideration the effects on the long-term health of affected children.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Microbioma Gastrointestinal/inmunología , Enfermedades del Recién Nacido/inmunología , Efectos Tardíos de la Exposición Prenatal/inmunología , Femenino , Humanos , Recién Nacido , Recien Nacido Prematuro/inmunología , Recién Nacido de muy Bajo Peso/inmunología , Embarazo
17.
Biomed Res Int ; 2021: 5568724, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-33880370

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is one of the most serious gastrointestinal disease-causing high morbidity and mortality in premature infants. However, the underlying mechanism of the pathogenesis of NEC is still not fully understood. METHODS: RNA sequencing of intestinal specimens from 9 NEC and 5 controls was employed to quantify the gene expression levels. RNA sequencing was employed to quantify the gene expression levels. DESeq2 tool was used to identify the differentially expressed genes. The biological function, pathways, transcription factors, and immune cells dysregulated in NEC were characterized by gene set enrichment analysis. RESULTS: In the present study, we analyzed RNA sequencing data of NECs and controls and revealed that immune-related pathways were highly activated, while some cellular responses to external stimuli-related pathways were inactivated in NEC. Moreover, B cells, macrophages M1, and plasma cells were identified as the major cell types involved in NEC. Furthermore, we also found that inflammation-related transcription factor genes, such as STAT1, STAT2, and IRF2, were significantly activated in NEC, further suggesting that these TFs might play critical roles in NEC pathogenesis. In addition, NEC samples exhibited heterogeneity to some extent. Interestingly, two subgroups in the NEC samples were identified by hierarchical clustering analysis. Notably, B cells, T cells, Th1, and Tregs involved in adaptive immune were predicted to highly infiltrate into subgroup I, while subgroup II was significantly infiltrated by neutrophils. The heterogeneity of immune cells in NEC indicated that both innate and adaptive immunes might induce NEC-related inflammatory response. CONCLUSIONS: In summary, we systematically analyzed inflammation-related genes, signaling pathways, and immune cells to characterize the NEC pathogenesis and samples, which greatly improved our understanding of the roles of inflammatory responses in NEC.


Asunto(s)
Biología Computacional , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/inmunología , Recien Nacido Prematuro/inmunología , Inflamación/genética , Transducción de Señal/genética , Estudios de Casos y Controles , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Redes Reguladoras de Genes , Humanos , Recién Nacido , Factores de Transcripción/metabolismo
18.
J Neuroinflammation ; 18(1): 66, 2021 Mar 06.
Artículo en Inglés | MEDLINE | ID: mdl-33676524

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is an inflammatory gastrointestinal disease in premature neonates with high mortality and morbidity, while the underlining mechanism of intestinal injury and profound neurological dysfunction remains unclear. Here, we aimed to investigate the involvement of NLPR3 inflammasome activation in NEC-related enterocolitis and neuroinflammation, especially long-term cognitive impairment, meanwhile, explore the protective effect of NLRP3 inhibitor MCC950 on NEC in mice. METHODS: NLRP3 inflammasome activation in the intestine and brain was assessed in the NEC mouse model, and NLRP3 inhibitor MCC950 was administrated during the development of NEC. Survival rate, histopathological injury of the intestine and brain, and expression of mature IL-1ß and other pro-inflammatory cytokines were analyzed. Long-term cognitive impairment was evaluated by behavioral test. RESULTS: The expression of NLRP3 and mature IL-1ß in the intestine and brain was greatly upregulated in NEC mice compared to the controls. MCC950 treatment efficiently improved NEC survival rate, reduced intestinal and brain inflammation, and ameliorated the severity of pathological damage in both organs. Additionally, in vivo blockage of NLRP3 inflammasome with MCC950 in early life of NEC pups potently protected against NEC-associated long-term cognitive impairment. CONCLUSIONS: Our findings suggest that NLRP3 inflammasome activation participates in NEC-induced intestinal and brain injury, and early intervention with NLRP3 inhibitor may provide beneficial therapeutic effect on NEC infants.


Asunto(s)
Disfunción Cognitiva/inmunología , Enterocolitis Necrotizante/inmunología , Inflamasomas/inmunología , Inflamación/inmunología , Proteína con Dominio Pirina 3 de la Familia NLR/inmunología , Animales , Disfunción Cognitiva/metabolismo , Disfunción Cognitiva/patología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Furanos/farmacología , Indenos/farmacología , Inflamasomas/metabolismo , Inflamación/metabolismo , Inflamación/patología , Ratones , Ratones Endogámicos C57BL , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Sulfonamidas/farmacología
19.
Nat Commun ; 12(1): 1042, 2021 02 15.
Artículo en Inglés | MEDLINE | ID: mdl-33589625

RESUMEN

Necrotizing enterocolitis (NEC) is a disease of premature infants characterized by acute intestinal necrosis. Current dogma suggests that NEC develops in response to post-natal dietary and bacterial factors, and so a potential role for in utero factors in NEC remains unexplored. We now show that during pregnancy, administration of a diet rich in the aryl hydrocarbon receptor (AHR) ligand indole-3-carbinole (I3C), or of breast milk, activates AHR and prevents NEC in newborn mice by reducing Toll-like receptor 4 (TLR4) signaling in the newborn gut. Protection from NEC requires activation of AHR in the intestinal epithelium which is reduced in mouse and human NEC, and is independent of leukocyte activation. Finally, we identify an AHR ligand ("A18") that limits TLR4 signaling in mouse and human intestine, and prevents NEC in mice when administered during pregnancy. In summary, AHR signaling is critical in NEC development, and maternally-delivered, AHR-based therapies may alleviate NEC.


Asunto(s)
Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/genética , Enterocolitis Necrotizante/genética , Indoles/administración & dosificación , Leche Humana/fisiología , Receptores de Hidrocarburo de Aril/genética , Receptor Toll-Like 4/genética , Animales , Animales Recién Nacidos , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/agonistas , Factores de Transcripción con Motivo Hélice-Asa-Hélice Básico/inmunología , Citocromo P-450 CYP1A1/genética , Citocromo P-450 CYP1A1/inmunología , Dieta/métodos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/inmunología , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Femenino , Regulación de la Expresión Génica , Humanos , Recién Nacido , Recien Nacido Prematuro , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ligandos , Exposición Materna , Ratones , Embarazo , Receptores de Hidrocarburo de Aril/agonistas , Receptores de Hidrocarburo de Aril/inmunología , Transducción de Señal , Porcinos , Receptor Toll-Like 4/inmunología
20.
Inflamm Res ; 70(3): 343-358, 2021 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-33564933

RESUMEN

BACKGROUND: Activation of intestinal macrophages is implicated in the pathogenesis of neonatal necrotizing enterocolitis (NEC), yet its precise mechanisms remain unclear. OBJECTIVE: The purpose of this study is to investigate the role of macrophages and TNF-α via an inflammatory MicroRNA in NEC. MATERIALS AND METHODS: Immunofluorescence (IF) staining of CD68, iNOS, and Arg-1 was employed to identify phenotypes of macrophage in the intestines of NEC infants and NEC mice. Expression of TNF-α, c-kit, and miR-222 was evaluated by qRT-PCR, Western blot, and immunochemical staining from the tissue samples. RESULTS: Large number of M1 macrophage infiltration was found in the NEC intestines. Expression of CD68, iNOS, and TNF-α were significantly increased, while c-kit was decreased distinctly in the NEC group. In the early phase of NEC mouse model, inhibition of M1 macrophages reduced the incidence of NEC and intestinal inflammation. We found that TNF-α upregulated the expression of miRNA-222 and inhibited the expression of c-kit. Conversely, such decrease of c-kit expression could be reversed by miR-222 antagonists. Furtherly, dual-luciferase assay confirmed that c-kit can be inhibited by miR-222 directly. CONCLUSION: Macrophages activation in NEC intestine results in an increased inflammatory response and TNF-α production, accompanied with miR-222 upregulation and c-kit suppression. Modulations of M1 macrophages, TNF-α or miR-222 may be potential therapeutic targets for NEC treatment.


Asunto(s)
Enterocolitis Necrotizante/inmunología , Macrófagos/inmunología , MicroARNs , Proteínas Proto-Oncogénicas c-kit/inmunología , Factor de Necrosis Tumoral alfa/inmunología , Animales , Enterocolitis Necrotizante/patología , Femenino , Humanos , Recién Nacido , Intestino Delgado/inmunología , Intestino Delgado/patología , Activación de Macrófagos , Masculino , Ratones Endogámicos C57BL , Regulación hacia Arriba
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA