Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 306
Filtrar
1.
J Cell Mol Med ; 28(14): e18534, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-39031467

RESUMEN

Intestinal dysbiosis is believed to play a role in the development of necrotizing enterocolitis (NEC). The efficacy of JNK-inhibitory peptide (CPJIP) in treating NEC was assessed. Treatment with CPJIP led to a notable reduction in p-JNK expression in IEC-6 cells and NEC mice. Following LPS stimulation, the expression of RNA and protein of claudin-1, claudin-3, claudin-4 and occludin was significantly decreased, with this decrease being reversed by CPJIP administration, except for claudin-3, which remained consistent in NEC mice. Moreover, the expression levels of the inflammatory factors TNF-α, IL-1ß and IL-6 were markedly elevated, a phenomenon that was effectively mitigated by the addition of CPJIP in both IEC-6 cells and NEC mice. CPJIP administration resulted in improved survival rates, ameliorated microscopic intestinal mucosal injury, and increased the total length of the intestines and colon in NEC mice. Additionally, CPJIP treatment led to a reduction in serum concentrations of FD-4, D-lactate and DAO. Furthermore, our results revealed that CPJIP effectively inhibited intestinal cell apoptosis and promoted cell proliferation in the intestine. This study represents the first documentation of CPJIP's ability to enhance the expression of tight junction components, suppress inflammatory responses, and rescue intestinal cell fate by inhibiting JNK activation, ultimately mitigating intestinal severity. These findings suggest that CPJIP has the potential to serve as a promising candidate for the treatment of NEC.


Asunto(s)
Apoptosis , Enterocolitis Necrotizante , Inflamación , Mucosa Intestinal , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Animales , Ratones , Mucosa Intestinal/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/patología , Inflamación/metabolismo , Inflamación/tratamiento farmacológico , Inflamación/patología , Apoptosis/efectos de los fármacos , Péptidos/farmacología , Modelos Animales de Enfermedad , Proliferación Celular/efectos de los fármacos , Ratones Endogámicos C57BL , Línea Celular , Ratas , Proteínas Quinasas JNK Activadas por Mitógenos/metabolismo , Lipopolisacáridos , Funcion de la Barrera Intestinal
2.
Cells ; 13(14)2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-39056761

RESUMEN

Necrotizing enterocolitis (NEC) is a complex, multifactorial gastrointestinal disorder predominantly affecting preterm infants. The pathogenesis of this condition involves a complex interplay between intestinal barrier dysfunction, microbial dysbiosis, and an altered immune response. This study investigates the potential role of endogenous hyaluronan (HA) in both the early phases of intestinal development and in the context of NEC-like intestinal injury. We treated neonatal CD-1 mouse pups with PEP1, a peptide inhibiting HA receptor interactions, from postnatal days 8 to 12. We evaluated postnatal intestinal developmental indicators, such as villi length, crypt depth, epithelial cell proliferation, crypt fission, and differentiation of goblet and Paneth cells, in PEP1-treated animals compared with those treated with scrambled peptide. PEP1 treatment significantly impaired intestinal development, as evidenced by reductions in villi length, crypt depth, and epithelial cell proliferation, along with a decrease in crypt fission activity. These deficits in PEP1-treated animals correlated with increased susceptibility to NEC-like injuries, including higher mortality rates, and worsened histological intestinal injury. These findings highlight the role of endogenous HA in supporting intestinal development and protecting against NEC.


Asunto(s)
Enterocolitis Necrotizante , Homeostasis , Ácido Hialurónico , Intestinos , Animales , Ácido Hialurónico/farmacología , Ácido Hialurónico/metabolismo , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/tratamiento farmacológico , Ratones , Homeostasis/efectos de los fármacos , Intestinos/patología , Intestinos/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Animales Recién Nacidos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de los fármacos , Modelos Animales de Enfermedad
3.
Int Immunopharmacol ; 138: 112567, 2024 Sep 10.
Artículo en Inglés | MEDLINE | ID: mdl-38950458

RESUMEN

BACKGROUND: Imbalanced intestinal microbiota and damage to the intestinal barrier contribute to the development of necrotizing enterocolitis (NEC). Autoinducer-2 (AI-2) plays a crucial role in repairing intestinal damage and reducing inflammation. OBJECTIVE: This study aimed to investigate the impact of AI-2 on the expression of intestinal zonula occludens-1 (ZO-1) and occludin proteins in NEC. We evaluated its effects in vivo using NEC mice and in vitro using lipopolysaccharide (LPS)-stimulated intestinal cells. METHODS: Pathological changes in the intestines of neonatal mice were assessed using histological staining and scoring. Cell proliferation was measured using the cell counting kit-8 (CCK-8) assay to determine the optimal conditions for LPS and AI-2 interventions. Real-time quantitative polymerase chain reaction (RT-qPCR) was used to analyze the mRNA levels of matrix metalloproteinase-3 (MMP3), protease activated receptor-2 (PAR2), interleukin-1ß (IL-1ß), and IL-6. Protein levels of MMP3, PAR2, ZO-1, and occludin were evaluated using western blot, immunohistochemistry, or immunofluorescence. RESULTS: AI-2 alleviated NEC-induced intestinal damage (P < 0.05) and enhanced the proliferation of damaged IEC-6 cells (P < 0.05). AI-2 intervention reduced the mRNA and protein expressions of MMP3 and PAR2 in intestinal tissue and cells (P < 0.05). Additionally, it increased the protein levels of ZO-1 and occludin (P < 0.05), while reducing IL-1ß and IL-6 mRNA expression (P < 0.05). CONCLUSION: AI-2 intervention enhances the expression of tight junction proteins (ZO-1 and occludin), mitigates intestinal damage in NEC neonatal mice and IEC-6 cells, potentially by modulating PAR2 and MMP3 signaling. AI-2 holds promise as a protective intervention for NEC. AI-2 plays a crucial role in repairing intestinal damage and reducing inflammation.


Asunto(s)
Enterocolitis Necrotizante , Metaloproteinasa 3 de la Matriz , Receptor PAR-2 , Transducción de Señal , Animales , Humanos , Ratones , Animales Recién Nacidos , Línea Celular , Proliferación Celular/efectos de los fármacos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Homoserina/análogos & derivados , Homoserina/farmacología , Mucosa Intestinal/patología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/inmunología , Intestinos/patología , Intestinos/efectos de los fármacos , Lactonas/farmacología , Lipopolisacáridos , Metaloproteinasa 3 de la Matriz/metabolismo , Metaloproteinasa 3 de la Matriz/genética , Ratones Endogámicos C57BL , Ocludina/metabolismo , Ocludina/genética , Receptor PAR-2/metabolismo , Receptor PAR-2/genética , Transducción de Señal/efectos de los fármacos , Proteína de la Zonula Occludens-1/metabolismo , Proteína de la Zonula Occludens-1/genética
4.
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
5.
Gut Microbes ; 16(1): 2379566, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-39013030

RESUMEN

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in premature infants with no specific treatments available. We aimed to identify the molecular mechanisms underlying NEC and investigate the therapeutic effects of Bacteroides fragilis on NEC. Clinical samples of infant feces, bile acid-targeted metabolomics, pathological staining, bioinformatics analysis, NEC rat model, and co-immunoprecipitation were used to explore the pathogenesis of NEC. Taxonomic characterization of the bile salt hydrolase (bsh) gene, enzyme activity assays, 16S rRNA sequencing, and organoids were used to explore the therapeutic effects of B. fragilis on NEC-related intestinal damage. Clinical samples, NEC rat models, and in vitro experiments revealed that total bile acid increased in the blood but decreased in feces. Moreover, the levels of FXR and other bile acid metabolism-related genes were abnormal, resulting in disordered bile acid metabolism in NEC. Taurochenodeoxycholic acid accelerated NEC pathogenesis and taurodeoxycholate alleviated NEC. B. fragilis displayed bsh genes and enzyme activity and alleviated intestinal damage by restoring gut microbiota dysbiosis and bile acid metabolism abnormalities by inhibiting the FXR-NLRP3 signaling pathway. Our results provide valuable insights into the therapeutic role of B. fragilis in NEC. Administering B. fragilis may substantially alleviate intestinal damage in NEC.


Asunto(s)
Amidohidrolasas , Bacteroides fragilis , Ácidos y Sales Biliares , Enterocolitis Necrotizante , Microbioma Gastrointestinal , Proteína con Dominio Pirina 3 de la Familia NLR , Receptores Citoplasmáticos y Nucleares , Transducción de Señal , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/tratamiento farmacológico , Animales , Proteína con Dominio Pirina 3 de la Familia NLR/metabolismo , Proteína con Dominio Pirina 3 de la Familia NLR/genética , Bacteroides fragilis/metabolismo , Bacteroides fragilis/genética , Transducción de Señal/efectos de los fármacos , Ácidos y Sales Biliares/metabolismo , Ratas , Receptores Citoplasmáticos y Nucleares/metabolismo , Receptores Citoplasmáticos y Nucleares/genética , Microbioma Gastrointestinal/efectos de los fármacos , Amidohidrolasas/metabolismo , Amidohidrolasas/genética , Humanos , Ratas Sprague-Dawley , Recién Nacido , Modelos Animales de Enfermedad , Masculino , Femenino , Probióticos/administración & dosificación , Probióticos/farmacología , Recien Nacido Prematuro , Disbiosis/microbiología
6.
BMJ Case Rep ; 17(6)2024 Jun 05.
Artículo en Inglés | MEDLINE | ID: mdl-38839401

RESUMEN

Retinopathy of prematurity (ROP) and necrotising enterocolitis (NEC) are complications of prematurity. Despite being quite different in terms of incidence, pathogenesis and consequences, both share a pathogenic role of aberrant vascularisation: increased in ROP, deficient for NEC. Current therapy for ROP includes the use of anti-vascular endothelial growth factor (anti-VEGF) agents, which are able to interrupt retinal hypervascularity. Despite being delivered intravitreously, anti-VEGF used in ROP can be absorbed into circulation and exert systemic effects. We present here a case of an ex-27 weeks gestational age infant, presenting multiple NEC risk factors, treated at 2 months of age with low-dose ranibizumab, who developed a large bowel NEC episode in the first week after treatment. We believe that this further report of an association between anti-VEGF agents and NEC could be interesting for the identification of children at risk of severe adverse events and stimulating further research on the topic.


Asunto(s)
Inhibidores de la Angiogénesis , Enterocolitis Necrotizante , Inyecciones Intravítreas , Ranibizumab , Retinopatía de la Prematuridad , Humanos , Retinopatía de la Prematuridad/tratamiento farmacológico , Enterocolitis Necrotizante/tratamiento farmacológico , Ranibizumab/administración & dosificación , Ranibizumab/uso terapéutico , Recién Nacido , Inhibidores de la Angiogénesis/administración & dosificación , Inhibidores de la Angiogénesis/efectos adversos , Masculino , Recien Nacido Prematuro , Femenino , Factor A de Crecimiento Endotelial Vascular/antagonistas & inhibidores
7.
Biomed Pharmacother ; 176: 116876, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38850657

RESUMEN

Necrotizing enterocolitis (NEC) is one of the most common and serious intestinal illnesses in newborns and seriously affects their long-term prognosis and survival. Butyrate is a short-chain fatty acid that can relieve intestinal inflammation, but its mechanism of action is unclear. Results from an in vivo neonatal rat model has shown that butyrate caused an improved recovery from NEC. These protective effects were associated with the metabolite of hesperetin, as determined by metabolomics and molecular biological analysis. Furthermore, transcriptomics combined with inhibitor assays were used to investigate the mechanism of action of hesperetin in an in vitro NEC model (IEC-6 cells exposed to LPS) to further investigate the mechanism by which butyrate attenuates NEC. The transcriptomics analysis showed that the PI3K-Akt signaling pathway was involved in the anti-NEC effect of hesperitin. Subsequently, the results using an inhibitor of PI3K (LY294002) indicated that the suppression could be explained by the hesperetin-induced expression of tight junction (TJ) proteins by potentially blocking the PI3K-Akt signaling pathway. In summary, the present study demonstrated that butyrate could improve recovery from NEC with a hesperetin metabolite, causing potential inhibition of the phosphorylation of the PI3K-Akt signaling pathway, resulting in the increased expression of TJ proteins. These findings reveal a potential new therapeutic pathway for the treatment of NEC.


Asunto(s)
Enterocolitis Necrotizante , Hesperidina , Fosfatidilinositol 3-Quinasas , Proteínas Proto-Oncogénicas c-akt , Ratas Sprague-Dawley , Transducción de Señal , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Hesperidina/farmacología , Animales , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Fosfatidilinositol 3-Quinasas/metabolismo , Ratas , Animales Recién Nacidos , Modelos Animales de Enfermedad , Butiratos/farmacología , Línea Celular
8.
J Pediatr Surg ; 59(7): 1266-1270, 2024 Jul.
Artículo en Inglés | MEDLINE | ID: mdl-38561306

RESUMEN

BACKGROUND: We sought to evaluate postoperative antibiotic practices in a large population of patients with necrotizing enterocolitis (NEC) and determine whether any regimens were associated with better outcomes. METHODS: The Pediatric Health Information Systems (PHIS) database was queried to identify patients who underwent an intestinal resection for acute NEC between July, 2016 and June, 2021. Data regarding post-resection antibiotic therapy, cutaneous or intraabdominal infection, and fungal or antibiotic-resistant infection were collected. RESULTS: 130 infants at 38 children's hospitals met inclusion criteria. Postoperative antibiotics were administered for a median of 13 days. The most frequently used antibiotics were vancomycin and piperacillin/tazobactam. Antibiotic duration greater than five days was not associated with a lower incidence of infection. No antibiotic was associated with a lower incidence of any of the complications assessed, although ampicillin was associated with more infections, overall. The incidence of fungal infection and treatment with a parenteral anti-fungal medication was greater with vancomycin. No antibiotic combination was used enough to be assessed. CONCLUSIONS: Administration of antibiotics for more than five days after resection for NEC was not associated with better infectious outcomes and no single antibiotic demonstrated superior efficacy. Consistent with prior studies, fungal infections were more frequent with vancomycin. TYPE OF STUDY: Retrospective database study, level 3B. LEVEL OF EVIDENCE: II.


Asunto(s)
Antibacterianos , Enterocolitis Necrotizante , Humanos , Enterocolitis Necrotizante/cirugía , Enterocolitis Necrotizante/tratamiento farmacológico , Antibacterianos/uso terapéutico , Antibacterianos/administración & dosificación , Recién Nacido , Masculino , Estudios Retrospectivos , Femenino , Lactante , Resultado del Tratamiento , Vancomicina/uso terapéutico , Vancomicina/administración & dosificación , Complicaciones Posoperatorias/epidemiología , Complicaciones Posoperatorias/etiología , Incidencia , Combinación Piperacilina y Tazobactam/uso terapéutico , Combinación Piperacilina y Tazobactam/administración & dosificación
9.
Sci Rep ; 14(1): 7665, 2024 04 01.
Artículo en Inglés | MEDLINE | ID: mdl-38561398

RESUMEN

The integrity of the intestinal mucus barrier is crucial for human health, as it serves as the body's first line of defense against pathogens. However, postnatal development of the mucus barrier and interactions between maturity and its ability to adapt to external challenges in neonatal infants remain unclear. In this study, we unveil a distinct developmental trajectory of the mucus barrier in preterm piglets, leading to enhanced mucus microstructure and reduced mucus diffusivity compared to term piglets. Notably, we found that necrotizing enterocolitis (NEC) is associated with increased mucus diffusivity of our large pathogen model compound, establishing a direct link between the NEC condition and the mucus barrier. Furthermore, we observed that addition of sodium decanoate had varying effects on mucus diffusivity depending on maturity and health state of the piglets. These findings demonstrate that regulatory mechanisms governing the neonatal mucosal barrier are highly complex and are influenced by age, maturity, and health conditions. Therefore, our results highlight the need for specific therapeutic strategies tailored to each neonatal period to ensure optimal gut health.


Asunto(s)
Ácidos Decanoicos , Enterocolitis Necrotizante , Moco , Recién Nacido , Animales , Humanos , Porcinos , Inflamación , Suplementos Dietéticos , Enterocolitis Necrotizante/tratamiento farmacológico , Mucosa Intestinal
10.
Int Immunopharmacol ; 128: 111360, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38176339

RESUMEN

Bile acids (BAs) have increasingly been implicated in the onset and progression of necrotizing enterocolitis (NEC); multiple findings have demonstrated their ability to induce damage to the intestinal epithelium, thereby exacerbating disease severity. Although we previously showed that melatonin was able to treat NEC by correcting the Treg/Th17 imbalance, the modulatory effect of melatonin on BAs remains unclear. In this study, we conducted transcriptome analysis on intestinal tissues from patients with NEC and validated these findings. Subsequently, we treated mice with melatonin alone or in combination with an agonist/inhibitor of Sirtuin 1 (SIRT1) to assess faecal and serum BA levels, the expression levels of BA transporters and regulators, and the extent of intestinal injury. Our transcriptome results indicated dysregulation of BA metabolism and abnormal expression of BA transporters in patients with NEC, which were also observed in our NEC mouse model. Furthermore, exogenous BAs were found to aggravate NEC severity in mice. Notably, melatonin effectively restored the aberrant expression of BA transporters, such as apical membrane sodium-dependent bile acid transporters (ASBT), ileal bile acid-binding protein (IBABP), and organic solute transporter-alpha (OST-α), by upregulating SIRT1 expression while reducing farnesoid X receptor (FXR) acetylation, consequently leading to decreased serum and faecal BA levels and mitigated NEC severity. Thus, we propose a potential mechanism through which melatonin reduces BA levels via the SIRT1/FXR signalling axis in an NEC mouse model. Collectively, these results highlight that melatonin holds promise for reducing BA levels and represents a promising therapeutic strategy for treating NEC.


Asunto(s)
Enterocolitis Necrotizante , Melatonina , Animales , Humanos , Ratones , Ácidos y Sales Biliares/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Intestinos , Hígado , Melatonina/farmacología , Melatonina/uso terapéutico , Sirtuina 1/metabolismo
11.
Life Sci ; 339: 122420, 2024 Feb 15.
Artículo en Inglés | MEDLINE | ID: mdl-38218534

RESUMEN

Human milk is the most valuable source of nutrition for infants. The structure and function of human milk oligosaccharides (HMOs), which are key components of human milk, have long been attracting particular research interest. Several recent studies have found HMOs to be efficacious in the prevention and treatment of necrotizing enterocolitis (NEC). Additionally, they could be developed in the future as non-invasive predictive markers for NEC. Based on previous findings and the well-defined functions of HMOs, we summarize potential protective mechanisms of HMOs against neonatal NEC, which include: modulating signal receptor function, promoting intestinal epithelial cell proliferation, reducing apoptosis, restoring intestinal blood perfusion, regulating microbial prosperity, and alleviating intestinal inflammation. HMOs supplementation has been demonstrated to be protective against NEC in both animal studies and clinical observations. This calls for mass production and use of HMOs in infant formula, necessitating more research into the safety of industrially produced HMOs and the appropriate dosage in infant formula.


Asunto(s)
Enterocolitis Necrotizante , Leche Humana , Lactante , Animales , Recién Nacido , Humanos , Leche Humana/química , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/prevención & control , Intestinos , Proliferación Celular , Oligosacáridos/farmacología , Oligosacáridos/uso terapéutico , Oligosacáridos/análisis
12.
J Am Coll Surg ; 238(1): 82-98, 2024 Jan 01.
Artículo en Inglés | MEDLINE | ID: mdl-37870229

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is a devastating condition where inflammatory changes and necrosis in the gut results in activation of brain microglia and subsequent neurodevelopmental impairment. Chondroitin sulfate (CS) is a glycosaminoglycan in human breast milk that is absent in conventional formulas. We hypothesized that oral formula supplementation with CS during a murine model of experimental NEC would not only attenuate intestinal injury, but also brain injury. STUDY DESIGN: NEC was induced in mouse pups on postnatal days (PNDs) 5 to 8. Three conditions were studied: (1) breastfed controls, (2) NEC, and (3) NEC+enteral CS (formula+200 mg/kg/d of CS). Pups were euthanized on PND 9 or reunited with dams by the evening of PND 8. Intestinal segments were H&E stained, and immunohistochemistry was performed on brain tissue for Iba-1 to assess for microglial morphology and cortical changes. Neurodevelopmental assays were performed on mice reunited with foster dams on PND 9. Single-cell RNA-sequencing analysis was performed on human intestinal epithelial cells exposed to (1) nothing, (2) hydrogen peroxide (H 2 O 2 ) alone, or (3) H 2 O 2 + CS to look at the differential gene expression between groups. Groups were compared with ANOVA or Kruskal-Wallis tests as appropriate with p < 0.05 considered significant. RESULTS: Compared with NEC, mice treated with oral CS showed improved clinical outcomes, decreased intestinal injury, and attenuated microglial activation and deleterious cortical change. Mice with CS performed better on early neurodevelopmental assays when compared with NEC alone. Single-cell analysis of HIEC-6 cells demonstrated that CS treatment down regulated several inflammatory pathways including nuclear factor κB-suggesting an explanation for the improved Th17 intestinal cytokine profile. CONCLUSIONS: Oral CS supplementation improved both physiological, clinical, and developmental outcomes. These data suggest that CS is a safe compound for formula supplementation for the prevention of NEC.


Asunto(s)
Lesiones Encefálicas , Enterocolitis Necrotizante , Femenino , Animales , Ratones , Recién Nacido , Humanos , Sulfatos de Condroitina/metabolismo , Sulfatos de Condroitina/uso terapéutico , Enterocolitis Necrotizante/tratamiento farmacológico , Lesiones Encefálicas/metabolismo , Suplementos Dietéticos , Modelos Animales de Enfermedad , Mucosa Intestinal
13.
J Perinatol ; 44(4): 587-593, 2024 Apr.
Artículo en Inglés | MEDLINE | ID: mdl-37863983

RESUMEN

OBJECTIVE: Evaluate the impact of a multidisciplinary guideline standardizing antibiotic duration and enteral feeding practices following medical necrotizing enterocolitis (mNEC). STUDY DESIGN: For preterm infants with Bell Stage 2 A mNEC and negative blood culture, antibiotic treatment was standardized to 7 days. Trophic feeds of unfortified human milk began 72 h after resolution of pneumatosis. Feeds were advanced by 20 cc/kg/day starting on the last day of antibiotics. Primary outcomes were antibiotic days and days to full feeds, defined as 120 cc/kg/day of enteral nutrition. Secondary outcomes included central line days and length of stay (LOS). RESULTS: Antibiotic duration decreased 23%. Time to start trophic feeds and time to full feeds decreased 33 and 16% respectively. Central line use dropped (98 to 72% of infants) and central line days were reduced by 59%. CONCLUSION: Implementation of a mNEC QI package reduced antibiotic duration, time to full feeds, central line use and CL days.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Recién Nacido , Humanos , Recien Nacido Prematuro , Enterocolitis Necrotizante/tratamiento farmacológico , Mejoramiento de la Calidad , Nutrición Enteral , Antibacterianos/uso terapéutico , Recién Nacido de muy Bajo Peso
14.
J Pediatr Surg ; 58(12): 2391-2398, 2023 Dec.
Artículo en Inglés | MEDLINE | ID: mdl-37684170

RESUMEN

BACKGROUND: Hydrogen sulfide (H2S) has been shown to improve outcomes in a murine model of necrotizing enterocolitis (NEC). There is evidence in humans that H2S relies on endothelial nitric oxide synthase (eNOS) to exert its protective effects, potentially through the persulfidation of eNOS at the Cysteine 443 residue. We obtained a novel mouse strain with a mutation at this residue (eNOSC440G) and hypothesized that this locus would be critical for GYY4137 (an H2S donor) to exert its protective effects. METHODS: Necrotizing enterocolitis was induced in 5-day old wild type (WT) and eNOSC440G mice using intermittent exposure to hypoxia and hypothermia in addition to gavage formula feeds. On postnatal day 9, mice were humanely euthanized. Data collected included daily weights, clinical sickness scores, histologic lung injury, intestinal injury (macroscopically and histologically), and intestinal perfusion. During the NEC model, pups received daily intraperitoneal injections of either GYY4137 (50 mg/kg) or PBS (vehicle). Data were tested for normality and compared using t-test or Mann-Whitney, and a p-value <0.05 was considered significant. RESULTS: In WT mice, the administration of GYY4137 significantly improved clinical sickness scores, attenuated intestinal and lung injury, and improved mesenteric perfusion compared to vehicle (p < 0.05). In eNOSC440G mice, the treatment and vehicle groups had similar clinical sickness scores, intestinal and lung injury scores, and intestinal perfusion. CONCLUSIONS: GYY4137 administration improves clinical outcomes, attenuates intestinal and lung injury, and improves perfusion in a murine model of necrotizing enterocolitis. The beneficial effects of GYY4137 are dependent on the Cys440 residue of eNOS.


Asunto(s)
Enterocolitis Necrotizante , Sulfuro de Hidrógeno , Enfermedades del Recién Nacido , Lesión Pulmonar , Humanos , Recién Nacido , Animales , Ratones , Óxido Nítrico Sintasa de Tipo III , Sulfuro de Hidrógeno/farmacología , Enterocolitis Necrotizante/tratamiento farmacológico , Modelos Animales de Enfermedad , Óxido Nítrico
15.
Physiol Rep ; 11(17): e15819, 2023 09.
Artículo en Inglés | MEDLINE | ID: mdl-37697223

RESUMEN

Necrotizing enterocolitis (NEC) continues to be a devastating disease in preterm neonates and has a paucity of medical management options. Chondroitin sulfate (CS) is a naturally occurring glycosaminoglycan (GAG) in human breast milk (HM) and has been shown to reduce inflammation. We hypothesized that supplementation with CS in an experimental NEC model would alter microbial diversity, favorably alter the cytokine profile, and (like other sulfur compounds) improve outcomes in experimental NEC via the eNOS pathway. NEC was induced in 5-day-old pups. Six groups were studied (n = 9-15/group): (1) WT breastfed and (2) Formula fed controls, (3) WT NEC, (4) WT NEC + CS, (5) eNOS KO (knockout) NEC, and (6) eNOS KO NEC + CS. Pups were monitored for clinical sickness score and weights. On postnatal day 9, the pups were killed. Stool was collected from rectum and microbiome analysis was done with 16 s rRNA sequencing. Intestinal segments were examined histologically using a well-established injury scoring system and segments were homogenized and analyzed for cytokine profile. Data were analyzed using GraphPad Prism with p < 0.05 considered significant. CS supplementation in formula improved experimental NEC outcomes when compared to NEC alone. CS supplementation resulted in similar improvement in NEC in both the WT and eNOS KO mice. CS supplementation did not result in microbial changes when compared to NEC alone. Our data suggest that although CS supplementation improved outcomes in NEC, this protection is not conferred via the eNOS pathway or alteration of microbial diversity. CS therapy in NEC does improve the intestinal cytokine profile and further experiments will explore the mechanistic role of CS in altering immune pathways in this disease.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades Fetales , Femenino , Recién Nacido , Humanos , Animales , Ratones , Sulfatos de Condroitina/uso terapéutico , Enterocolitis Necrotizante/tratamiento farmacológico , Modelos Animales de Enfermedad , Suplementos Dietéticos , Citocinas
16.
Sci Rep ; 13(1): 13395, 2023 08 17.
Artículo en Inglés | MEDLINE | ID: mdl-37591866

RESUMEN

Prostaglandin E2 (PGE2) is implicated in intestinal inflammation and intestinal blood flow regulation with a paradoxical effect on the pathogenesis of necrotizing enterocolitis (NEC), which is not yet well understood. In the current study, we found that PGE2, EP4, and COX-2 varied at different distances from the most damaged area in the terminal ileum obtained from human infants with NEC. PGE2 administration alleviated the phenotype of experimental NEC and the intestinal microvascular features in experimental NEC, but this phenomenon was inhibited by eNOS depletion, suggesting that PGE2 promoted intestinal microcirculatory perfusion through eNOS. Furthermore, PGE2 administration increased the VEGF content in MIMECs under TNFα stress and promoted MIMEC proliferation. This response to PGE2 was involved in eNOS phosphorylation and nitric oxide (NO) production and was blocked by the EP4 antagonist in vitro, suggesting that targeting the PGE2-EP4-eNOS axis might be a potential clinical and therapeutic strategy for NEC treatment. The study is reported in accordance with ARRIVE guidelines ( https://arriveguidelines.org ).


Asunto(s)
Dinoprostona , Enterocolitis Necrotizante , Lactante , Humanos , Recién Nacido , Microcirculación , Intestinos , Ciclooxigenasa 2 , Enterocolitis Necrotizante/tratamiento farmacológico
17.
J Pediatr Surg ; 58(10): 1982-1989, 2023 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-37479571

RESUMEN

BACKGROUND: Antibiotic overutilization in the neonatal intensive care unit (NICU) has many adverse effects, and necrotizing enterocolitis (NEC) is one of the most common indications for antibiotics in premature infants. Evidence for a preferred antibiotic regimen for NEC is lacking. This project aims to reduce piperacillin-tazobactam use and overall antibiotic duration in neonates with NEC through the implementation of an antibiotic stewardship pathway based on the modified Bell stage classification system. METHODS: A multidisciplinary team consisting of neonatology, pharmacy, infectious disease, and surgery developed an antibiotic protocol for the management of NEC based on Bell stage. Recommendations included 48 h of ampicillin/gentamicin (AG) for stage I, 5-10 days of AG for stage II, the addition of metronidazole for stage IIIA, and 7-14 days of piperacillin-tazobactam (PT) for stage IIIB. We evaluated overall antibiotic and PT exposure, progression to surgical NEC, NEC recurrence, antibiotic resistance, bacteremia/fungemia, and mortality 1 year pre- and post-protocol implementation. RESULTS: 27 patients pre-intervention and 44 post-intervention were analyzed. Antibiotic exposure was reduced from a median 119.19 to 80.65 days of therapy (DOT) per 1000 patient days (p = 0.11). PT exposure decreased after protocol implementation (median 68.78 vs. 7.97 DOT per 1000 patient days, p = 0.002). There were no significant differences in morbidity or mortality outcomes. CONCLUSIONS: Antibiotic stewardship strategies can be implemented in the NICU without compromising outcomes in patients with NEC. Bell stage stratification appears to be an effective method for antibiotic selection. Further studies are needed in a larger population to optimize regimens and ensure safety. TYPE OF STUDY: Retrospective comparative study. LEVEL OF EVIDENCE: Level III.


Asunto(s)
Programas de Optimización del Uso de los Antimicrobianos , Enterocolitis Necrotizante , Enfermedades Fetales , Enfermedades del Recién Nacido , Lactante , Femenino , Recién Nacido , Humanos , Enterocolitis Necrotizante/tratamiento farmacológico , Mejoramiento de la Calidad , Estudios Retrospectivos , Recien Nacido Prematuro , Antibacterianos/uso terapéutico , Ampicilina/uso terapéutico , Combinación Piperacilina y Tazobactam/uso terapéutico
18.
Tissue Cell ; 83: 102128, 2023 Aug.
Artículo en Inglés | MEDLINE | ID: mdl-37413858

RESUMEN

AIM: This study aims to explore the possible effect of Astragaloside IV (AS-IV) on necrotizing enterocolitis (NEC) neonatal rat models and verify the possible implication of TNF-like ligand 1 A (TL1A) and NF-κB signal pathway. METHODS: NEC neonatal rat models were established through formula feeding, cold/asphyxia stress and Lipopolysaccharide (LPS) gavage method. The appearance, activity and skin as well as the pathological status of rats subjected to NEC modeling were assessed. The intestinal tissues were observed after H&E staining. The expression of oxidative stress biomarkers (SOD, MDA and GSH-Px) and inflammatory cytokines (TNF-α, IL-1ß and IL-6) were detected by ELISA and qRT-PCR. Western blotting and immunohistochemistry were applied to detect expressions of TL1A and NF-κB signal pathway-related proteins. Cell apoptosis was assessed by TUNEL. RESULTS: NEC neonatal rat models were established successfully, in which TL1A was highly expressed and NF-κB signal pathway was activated, while TL1A and NF-κB signal pathway can be suppressed by AS-IV treatment in NEC rats. Meanwhile, inflammatory response in intestinal tissues was increased in NEC rat models and AS-IV can attenuate inflammatory response in NEC rats through inhibiting TL1A and NF-κb signal pathway. CONCLUSION: AS-IV can inhibit TL1A expression and NF-κb signal pathway to attenuate the inflammatory response in NEC neonatal rat models.


Asunto(s)
Enterocolitis Necrotizante , FN-kappa B , Ratas , Animales , FN-kappa B/metabolismo , Animales Recién Nacidos , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/patología , Ligandos , Ratas Sprague-Dawley , Transducción de Señal , Inflamación/patología , Modelos Animales de Enfermedad
19.
Semin Pediatr Surg ; 32(3): 151308, 2023 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-37295297

RESUMEN

Antibiotic therapy remains a cornerstone of treatment of both medical and surgical presentations of necrotizing enterocolitis (NEC). However, guidelines regarding the administration of antibiotics for the treatment of NEC are lacking and practices vary amongst clinicians. Although the pathogenesis of NEC is unknown, there is consensus that the infant gastrointestinal microbiome contributes to the disease. The presumed connection between dysbiosis and NEC has prompted some to study whether early prophylactic enteral antibiotics can prevent NEC. Yet others have taken an opposing approach, studying whether perinatal antibiotic exposure increases the risk of NEC by inducing a state of dysbiosis. This narrative review summarizes what is known about antibiotics and their association with the infant microbiome and NEC, current antibiotic prescribing practices for infants with medical and surgical NEC, as well as potential strategies to further optimize the use of antibiotics in this population of infants.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades Fetales , Microbioma Gastrointestinal , Enfermedades del Recién Nacido , Lactante , Embarazo , Femenino , Recién Nacido , Humanos , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/etiología , Disbiosis/complicaciones , Disbiosis/tratamiento farmacológico , Antibacterianos/uso terapéutico
20.
Gut Microbes ; 15(1): 2221758, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37358104

RESUMEN

Microbiome-targeting therapies have received great attention as approaches to prevent disease in infants born preterm, but their safety and efficacy remain uncertain. Here we summarize the existing literature, focusing on recent meta-analyses and systematic reviews that evaluate the performance of probiotics, prebiotics, and/or synbiotics in clinical trials and studies, emphasizing interventions for which the primary or secondary outcomes were prevention of necrotizing enterocolitis, late-onset sepsis, feeding intolerance, and/or reduction in hospitalization length or all-cause mortality. Current evidence suggests that probiotics and prebiotics are largely safe but conclusions regarding their effectiveness in the neonatal intensive care unit have been mixed. To address this ambiguity, we evaluated publications that collectively support benefits of probiotics with moderate to high certainty evidence in a recent comprehensive network meta-analysis, highlighting limitations in these trials that make it difficult to support with confidence the routine, universal administration of probiotics to preterm infants.


Asunto(s)
Enterocolitis Necrotizante , Microbioma Gastrointestinal , Microbiota , Probióticos , Humanos , Recién Nacido , Enterocolitis Necrotizante/tratamiento farmacológico , Enterocolitis Necrotizante/prevención & control , Recien Nacido Prematuro , Unidades de Cuidado Intensivo Neonatal , Prebióticos , Probióticos/efectos adversos
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA