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

Bases de datos
País/Región como asunto
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Res ; 2024 Jul 11.
Artículo en Inglés | MEDLINE | ID: mdl-38992154

RESUMEN

Nutritional management of preterm infants recovering from necrotizing enterocolitis (NEC) or focal intestinal perforation (FIP) is challenging, especially in infants managed surgically. The logistics of how, when, and what to feed are unclear and current nutritional practices are primarily based on physiological principles and consensus opinion in individual units, rather than high-quality evidence. The aim of this narrative review is to summarize the literature on nutritional management after NEC or FIP in preterm infants: when to restart enteral nutrition, type of enteral nutrition to use, and how to advance nutrition. We also discuss treatment of micronutrient deficiencies, cholestasis, replacement of stoma losses, and optimal time of stoma closure. In conclusion, there are in sufficient high-quality studies available to provide evidence-based recommendations on the best nutritional practice after NEC or FIP in preterm infants. A local or national consensus based early nutrition guideline agreed upon by a multidisciplinary team including pediatric surgeons, pediatricians/neonatologists, nurses, and nutritionists is recommended. Further studies are urgently needed. IMPACT: There is no good quality evidence or nutritional standard across neonatal units treating infants after medical or surgical NEC or FIP. With this review we hope to start providing some consistency across patients and between providers treating patients with NEC and FIP. Mother's own milk is recommended when restarting enteral nutrition after NEC or FIP. In the absence of high-quality evidence, a consensus based early nutrition guideline agreed upon by a multidisciplinary team is recommended. Nutritional research projects are urgently needed in NEC and FIP patients.

2.
Cytokine ; 167: 156210, 2023 07.
Artículo en Inglés | MEDLINE | ID: mdl-37130487

RESUMEN

BACKGROUND: The induction of maternal inflammation in mice leads to fetal injury that is believed to be IL-6 dependent. The fetal inflammatory response, defined by elevated fetal or amniotic fluid IL-6, has been described as a potential mechanism for subsequent fetal injury. The role of maternal IL-6 production and signaling in the fetal IL-6 response is currently unclear. METHODS: Genetic and anti-IL-6 antibody strategies were used to systematically block the maternal IL-6 response during inflammation. Chorioamnionitis was induced using intraperitoneal injection of lipopolysaccharide (LPS) at mid gestation (E14.5) and late gestation (E18.5). This model was used in pregnant C57Bl/6 dams, IL6-/- dams, C57Bl/6 dams treated with anti-IL-6 (blocks both classical and trans-signaling) or anti-gp130 antibodies (blocks trans-signaling only) and IL6+/- dams. Six hours following LPS injection, maternal serum, placental tissue, amniotic fluid and fetal tissue or serum were collected. A bead-based multiplex assay was used to evaluate levels of IL-6, KC, IL-1ß, TNF, IL-10, IL-22, IFN-γ, IL-13 and IL-17A. RESULTS: Chorioamnionitis in C57Bl/6 dams was characterized by elevated maternal serum levels of IL-6, KC and IL-22 with litter loss during mid gestation. The fetal response to maternal inflammation in C57Bl/6 mice was primarily characterized by elevated levels of IL-6, KC and IL-22 in the placenta, amniotic fluid and fetus during both mid and late gestation. A global IL-6 knockout (IL6-/-) eradicated the maternal, placental, amniotic fluid and fetal IL-6 response to LPS during mid and late gestation and improved litter survival, while minimally influencing the KC or IL-22 responses. Blocking maternal classical IL-6 signaling in C57Bl/6 dams at the time of LPS exposure diminished the maternal, placental, amniotic fluid and fetal IL-6 response during mid and late gestation, while blocking maternal IL-6 trans-signaling only affected fetal IL-6 expression. To evaluate whether maternal IL-6 was crossing the placenta and reaching the fetus, IL-6+/- dams were utilized in the chorioamnionitis model. IL-6+/- dams mounted a systemic inflammatory response following injection with LPS, characterized by elevated IL-6, KC and IL-22. IL-6-/- pups born to IL6+/- dams had decreased amniotic fluid levels of IL-6 and undetectable levels of fetal IL-6 compared to IL-6+/+ littermate controls. CONCLUSION: The fetal response to systemic maternal inflammation is dependent upon maternal IL-6 signaling, but maternal IL-6 is not crossing the placenta and reaching the fetus at detectable levels.


Asunto(s)
Corioamnionitis , Enfermedades Fetales , Animales , Femenino , Ratones , Embarazo , Líquido Amniótico/metabolismo , Corioamnionitis/metabolismo , Modelos Animales de Enfermedad , Enfermedades Fetales/metabolismo , Inflamación/metabolismo , Interleucina-6 , Lipopolisacáridos , Placenta/metabolismo
3.
Pediatr Res ; 91(3): 590-597, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34021272

RESUMEN

BACKGROUND: Necrotizing enterocolitis (NEC) is a devastating intestinal disease of premature infants, with significant mortality and long-term morbidity among survivors. Multiple NEC definitions exist, but no formal head-to-head evaluation has been performed. We hypothesized that contemporary definitions would perform better in evaluation metrics than Bell's and range features would be more frequently identified as important than yes/no features. METHODS: Two hundred and nineteen patients from the University of Iowa hospital with NEC, intestinal perforation, or NEC concern were identified from a 10-year retrospective cohort. NEC presence was confirmed by a blinded investigator. Evaluation metrics were calculated using statistics and six supervised machine learning classifiers for current NEC definitions. Feature importance evaluation was performed on each decision tree classifier. RESULTS: Newer definitions outperformed Bell's staging using both standard statistics and most machine learning classifiers. The decision tree classifier had the highest overall machine learning scores, which resulted in Non-Bell definitions having high sensitivity (0.826, INC) and specificity (0.969, ST), while Modified Bell (IIA+) had reasonable sensitivity (0.783), but poor specificity (0.531). Feature importance evaluation identified nine criteria as important for diagnosis. CONCLUSIONS: This preliminary study suggests that Non-Bell NEC definitions may be better at diagnosing NEC and calls for further examination of definitions and important criteria. IMPACT: This article is the first formal head-to-head evaluation of current available definitions of NEC. Non-Bell NEC definitions may be more effective in identifying NEC based on findings from traditional measures of diagnostic performance and machine learning techniques. Nine features were identified as important for diagnosis from the definitions evaluated within the decision tree when performing supervised classification machine learning. This article serves as a preliminary study to formally evaluate the definitions of NEC utilized and should be expounded upon with a larger and more diverse patient cohort.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades Fetales , Enfermedades del Recién Nacido , Enfermedades del Prematuro , Enterocolitis Necrotizante/diagnóstico , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/diagnóstico , Estudios Retrospectivos
4.
Am J Physiol Gastrointest Liver Physiol ; 321(6): G668-G681, 2021 12 01.
Artículo en Inglés | MEDLINE | ID: mdl-34643097

RESUMEN

MicroRNA-mediated regulation is critical for the proper development and function of the small intestinal (SI) epithelium. However, it is not known which microRNAs are expressed in each of the cell types of the SI epithelium. To bridge this important knowledge gap, we performed comprehensive microRNA profiling in all major cell types of the mouse SI epithelium. We used flow cytometry and fluorescence-activated cell sorting with multiple reporter mouse models to isolate intestinal stem cells, enterocytes, goblet cells, Paneth cells, enteroendocrine cells, tuft cells, and secretory progenitors. We then subjected these cell populations to small RNA-sequencing. The resulting atlas revealed highly enriched microRNA markers for almost every major cell type (https://sethupathy-lab.shinyapps.io/SI_miRNA/). Several of these lineage-enriched microRNAs (LEMs) were observed to be embedded in annotated host genes. We used chromatin-run-on sequencing to determine which of these LEMs are likely cotranscribed with their host genes. We then performed single-cell RNA-sequencing to define the cell type specificity of the host genes and embedded LEMs. We observed that the two most enriched microRNAs in secretory progenitors are miR-1224 and miR-672, the latter of which we found is deleted in hominin species. Finally, using several in vivo models, we established that miR-152 is a Paneth cell-specific microRNA.NEW & NOTEWORTHY In this study, first, microRNA atlas (and searchable web server) across all major small intestinal epithelial cell types is presented. We have demonstrated microRNAs that uniquely mark several lineages, including enteroendocrine and tuft. Identification of a key marker of mouse secretory progenitor cells, miR-672, which we show is deleted in humans. We have used several in vivo models to establish miR-152 as a specific marker of Paneth cells, which are highly understudied in terms of microRNAs.


Asunto(s)
Linaje de la Célula , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Mucosa Intestinal/metabolismo , Intestino Delgado/metabolismo , MicroARNs/genética , Transcriptoma , Animales , Biomarcadores/metabolismo , Separación Celular , Células Cultivadas , Biología Computacional , Perros , Femenino , Citometría de Flujo , Mucosa Intestinal/citología , Intestino Delgado/citología , Masculino , Ratones Endogámicos C57BL , Ratones Transgénicos , MicroARNs/metabolismo , Organoides , RNA-Seq , Análisis de la Célula Individual
5.
Pediatr Res ; 88(Suppl 1): 30-34, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855510

RESUMEN

Necrotizing enterocolitis (NEC) remains among the most common and devastating diseases in neonates. Despite advances in neonatal clinical care, specific treatment strategies and diagnostic modalities remain lacking. As a result, morbidity and mortality remain high. Improved understanding of the pathogenesis of NEC has the potential for improved therapeutics. Some of the areas of research leading to promising discoveries include inhibition of Toll-like receptor signaling, modulation of vascular endothelial growth factor signal pathways, defining metabolomic alterations in NEC to discover potential biomarkers, probing for genetic predispositions to NEC susceptibility, determining mechanistic relations between anemia and NEC, and microflora modulation through the use of probiotics. All of these areas may represent novel promising approaches to the prevention and treatment of NEC. This review will focus on these current and possible therapeutic perspectives.


Asunto(s)
Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/terapia , Probióticos/uso terapéutico , Anemia/complicaciones , Animales , Investigación Biomédica , Enterocolitis Necrotizante/metabolismo , Predisposición Genética a la Enfermedad , Humanos , Lactante , Recien Nacido Extremadamente Prematuro , Recién Nacido , Recien Nacido Prematuro , Enfermedades del Prematuro/patología , Inflamación , Ratones , Leche Humana , Estrés Oxidativo , Transducción de Señal , Receptores Toll-Like , Resultado del Tratamiento , Factor A de Crecimiento Endotelial Vascular/metabolismo
6.
Pediatr Res ; 88(Suppl 1): 10-15, 2020 08.
Artículo en Inglés | MEDLINE | ID: mdl-32855506

RESUMEN

Necrotizing enterocolitis (NEC) is a leading cause of morbidity and mortality in hospitalized infants. First classified through Bell staging in 1978, a number of additional definitions of NEC have been proposed in the subsequent decades. In this review, we summarize eight current definitions of NEC, and explore similarities and differences in clinical signs and radiographic features included within these definitions, as well as their limitations. We highlight the importance of a global consensus on defining NEC to improve NEC research and outcomes, incorporating input from participants at an international NEC conference. We also highlight the important role of patient-families in helping to redefine NEC.


Asunto(s)
Enterocolitis Necrotizante/diagnóstico , Enfermedades del Recién Nacido/diagnóstico , Enfermedades del Prematuro/diagnóstico , Centers for Disease Control and Prevention, U.S. , Consenso , Enterocolitis Necrotizante/clasificación , Femenino , Humanos , Lactante , Recién Nacido , Enfermedades del Recién Nacido/clasificación , Recien Nacido Prematuro , Enfermedades del Prematuro/clasificación , Masculino , Neonatología/normas , Riesgo , Factores de Riesgo , Reino Unido , Estados Unidos
7.
Pediatr Res ; 88(1): 66-76, 2020 07.
Artículo en Inglés | MEDLINE | ID: mdl-31242501

RESUMEN

BACKGROUND: Preterm infants are susceptible to unique pathology due to their immaturity. Mouse models are commonly used to study immature intestinal disease, including necrotizing enterocolitis (NEC). Current NEC models are performed at a variety of ages, but data directly comparing intestinal developmental stage equivalency between mice and humans are lacking. METHODS: Small intestines were harvested from C57BL/6 mice at 3-4 days intervals from birth to P28 (n = 8 at each age). Preterm human small intestine samples representing 17-23 weeks of completed gestation were obtained from the University of Pittsburgh Health Sciences Tissue Bank, and at term gestation during reanastamoses after resection for NEC (n = 4-7 at each age). Quantification of intestinal epithelial cell types and messenger RNA for marker genes were evaluated on both species. RESULTS: Overall, murine and human developmental trends over time are markedly similar. Murine intestine prior to P10 is most similar to human fetal intestine prior to viability. Murine intestine at P14 is most similar to human intestine at 22-23 weeks completed gestation, and P28 murine intestine is most similar to human term intestine. CONCLUSION: Use of C57BL/6J mice to model the human immature intestine is reasonable, but the age of mouse chosen is a critical factor in model development.


Asunto(s)
Epitelio/crecimiento & desarrollo , Regulación del Desarrollo de la Expresión Génica , Intestinos/crecimiento & desarrollo , Animales , Enterocolitis Necrotizante/metabolismo , Epitelio/patología , Receptores ErbB/metabolismo , Perfilación de la Expresión Génica , Homeostasis , Humanos , Enfermedades Intestinales/metabolismo , Intestinos/patología , Ratones , Ratones Endogámicos C57BL
8.
Proc Natl Acad Sci U S A ; 113(19): E2627-35, 2016 May 10.
Artículo en Inglés | MEDLINE | ID: mdl-27114524

RESUMEN

Interleukin (IL)-18 is an important effector of innate and adaptive immunity, but its expression must also be tightly regulated because it can potentiate lethal systemic inflammation and death. Healthy and septic human neonates demonstrate elevated serum concentrations of IL-18 compared with adults. Thus, we determined the contribution of IL-18 to lethality and its mechanism in a murine model of neonatal sepsis. We find that IL-18-null neonatal mice are highly protected from polymicrobial sepsis, whereas replenishing IL-18 increased lethality to sepsis or endotoxemia. Increased lethality depended on IL-1 receptor 1 (IL-1R1) signaling but not adaptive immunity. In genome-wide analyses of blood mRNA from septic human neonates, expression of the IL-17 receptor emerged as a critical regulatory node. Indeed, IL-18 administration in sepsis increased IL-17A production by murine intestinal γδT cells as well as Ly6G(+) myeloid cells, and blocking IL-17A reduced IL-18-potentiated mortality to both neonatal sepsis and endotoxemia. We conclude that IL-17A is a previously unrecognized effector of IL-18-mediated injury in neonatal sepsis and that disruption of the deleterious and tissue-destructive IL-18/IL-1/IL-17A axis represents a novel therapeutic approach to improve outcomes for human neonates with sepsis.


Asunto(s)
Interleucina-17/antagonistas & inhibidores , Interleucina-17/inmunología , Interleucina-18/inmunología , Sepsis Neonatal/inmunología , Sepsis Neonatal/terapia , Tasa de Supervivencia , Animales , Animales Recién Nacidos , Anticuerpos Monoclonales/uso terapéutico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Terapia Molecular Dirigida/métodos , Sepsis Neonatal/patología , Resultado del Tratamiento
10.
Am J Pathol ; 184(10): 2768-78, 2014 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-25216938

RESUMEN

Necrotizing enterocolitis (NEC) affects up to 10% of premature infants, has a mortality of 30%, and can leave surviving patients with significant morbidity. Neuregulin-4 (NRG4) is an ErbB4-specific ligand that promotes epithelial cell survival. Thus, this pathway could be protective in diseases such as NEC, in which epithelial cell death is a major pathologic feature. We sought to determine whether NRG4-ErbB4 signaling is protective in experimental NEC. NRG4 was used i) in the newborn rat formula feeding/hypoxia model; ii) in a recently developed model in which 14- to 16-day-old mice are injected with dithizone to induce Paneth cell loss, followed by Klebsiella pneumoniae infection to induce intestinal injury; and iii) in bacterially infected IEC-6 cells in vitro. NRG4 reduced NEC incidence and severity in the formula feed/hypoxia rat model. It also reduced Paneth cell ablation-induced NEC and prevented dithizone-induced Paneth cell loss in mice. In vitro, cultured ErbB4(-/-) ileal epithelial enteroids had reduced Paneth cell markers and were highly sensitive to inflammatory cytokines. Furthermore, NRG4 blocked, through a Src-dependent pathway, Cronobacter muytjensii-induced IEC-6 cell apoptosis. The potential clinical relevance of these findings was demonstrated by the observation that NRG4 and its receptor ErbB4 are present in human breast milk and developing human intestine, respectively. Thus, NRG4-ErbB4 signaling may be a novel pathway for therapeutic intervention or prevention in NEC.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Neurregulinas/metabolismo , Receptor ErbB-4/metabolismo , Transducción de Señal , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/metabolismo , Células Epiteliales/metabolismo , Femenino , Humanos , Íleon/metabolismo , Intestinos/patología , Ratones , Células de Paneth/metabolismo , Ratas , Ratas Sprague-Dawley
11.
Mediators Inflamm ; 2014: 852378, 2014.
Artículo en Inglés | MEDLINE | ID: mdl-25242872

RESUMEN

BACKGROUND: Premature infants are commonly subject to intestinal inflammation. Since the human small intestine does not reach maturity until term gestation, premature infants have a unique challenge, as either acute or chronic inflammation may alter the normal development of the intestinal tract. Tumor necrosis factor (TNF) has been shown to acutely alter goblet cell numbers and villus length in adult mice. In this study we tested the effects of TNF on villus architecture and epithelial cells at different stages of development of the immature small intestine. METHODS: To examine the effects of TNF-induced inflammation, we injected acute, brief, or chronic exposures of TNF in neonatal and juvenile mice. RESULTS: TNF induced significant villus blunting through a TNF receptor-1 (TNFR1) mediated mechanism, leading to loss of villus area. This response to TNFR1 signaling was altered during intestinal development, despite constant TNFR1 protein expression. Acute TNF-mediated signaling also significantly decreased Paneth cells. CONCLUSIONS: Taken together, the morphologic changes caused by TNF provide insight as to the effects of inflammation on the developing intestinal tract. Additionally, they suggest a mechanism which, coupled with an immature immune system, may help to explain the unique susceptibility of the immature intestine to inflammatory diseases such as NEC.


Asunto(s)
Intestino Delgado/efectos de los fármacos , Intestino Delgado/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Factores de Necrosis Tumoral/farmacología , Animales , Apoptosis/efectos de los fármacos , Western Blotting , Humanos , Ratones , Ratones Noqueados , Reacción en Cadena de la Polimerasa , Receptores Tipo I de Factores de Necrosis Tumoral/genética , Receptores Tipo II del Factor de Necrosis Tumoral/genética
12.
J Perinatol ; 2024 Jun 06.
Artículo en Inglés | MEDLINE | ID: mdl-38844520

RESUMEN

Necrotizing enterocolitis (NEC) continues to be a leading cause of morbidity and mortality in preterm infants. As modern medicine significantly improves the survival of extremely premature infants, the persistence of NEC underscores our limited understanding of its pathogenesis. Due to early delivery, a preterm infant's exposure to amniotic fluid (AF) is abruptly truncated. Replete with bioactive molecules, AF plays an important role in fetal intestinal maturation and preparation for contact with the environment, thus its absence during development of the intestine may contribute to increased susceptibility to NEC. Human milk (HM), particularly during the initial phases of lactation, is a cornerstone of neonatal intestinal defense. The concentrations and activities of several bioactive factors in HM parallel those of AF, suggesting continuity of protection. In this review, we discuss the predominant overlapping bioactive components of HM and AF, with an emphasis on those associated with intestinal growth or reduction of NEC.

13.
Front Vet Sci ; 11: 1275293, 2024.
Artículo en Inglés | MEDLINE | ID: mdl-38318150

RESUMEN

Introduction: Paneth cells are critically important to intestinal health, including protecting intestinal stem cells, shaping the intestinal microbiome, and regulating host immunity. Understanding Paneth cell biology in the immature intestine is often modeled in rodents with little information in larger mammals such as sheep. Previous studies have only established the distribution pattern of Paneth cells in healthy adult sheep. Our study aimed to examine the ontogeny, quantification, and localization of Paneth cells in fetal and newborn lambs at different gestational ages and with perinatal transient asphyxia. We hypothesized that ovine Paneth cell distribution at birth resembles the pattern seen in humans (highest concentrations in the ileum) and that ovine Paneth cell density is gestation-dependent. Methods: Intestinal samples were obtained from 126-127 (preterm, with and without perinatal transient asphyxia) and 140-141 (term) days gestation sheep. Samples were quantified per crypt in at least 100 crypts per animal and confirmed as Paneth cells through in immunohistochemistry. Results: Paneth cells had significantly higher density in the ileum compared to the jejunum and were absent in the colon. Discussion: Exposure to perinatal transient asphyxia acutely decreased Paneth cell numbers. These novel data support the possibility of utilizing ovine models for understanding Paneth cell biology in the fetus and neonate.

14.
iScience ; 27(6): 109993, 2024 Jun 21.
Artículo en Inglés | MEDLINE | ID: mdl-38846005

RESUMEN

Necrotizing enterocolitis (NEC) is a leading cause of preterm infant morbidity and mortality. Treatment for NEC is limited and non-targeted, which makes new treatment and prevention strategies critical. Host defense peptides (HDPs) are essential components of the innate immune system and have multifactorial mechanisms in host defense. LL-37 and hBD2 are two HDPs that have been shown in prior literature to protect from neonatal sepsis-induced mortality or adult inflammatory bowel disease, respectively. Therefore, this article sought to understand if these two HDPs could influence NEC severity in murine preclinical models. NEC was induced in P14-16 C57Bl/6 mice and HDPs were provided as a pretreatment or treatment. Both LL-37 and hBD2 resulted in decreased NEC injury scores as a treatment and hBD2 as a pretreatment. Our data suggest LL-37 functions through antimicrobial properties, while hBD2 functions through decreases in inflammation and improvement of intestinal barrier integrity.

17.
Front Pediatr ; 11: 1182597, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37303753

RESUMEN

Necrotizing Enterocolitis (NEC) is one of the leading causes of gastrointestinal emergency in preterm infants. Although NEC was formally described in the 1960's, there is still difficulty in diagnosis and ultimately treatment for NEC due in part to the multifactorial nature of the disease. Artificial intelligence (AI) and machine learning (ML) techniques have been applied by healthcare researchers over the past 30 years to better understand various diseases. Specifically, NEC researchers have used AI and ML to predict NEC diagnosis, NEC prognosis, discover biomarkers, and evaluate treatment strategies. In this review, we discuss AI and ML techniques, the current literature that has applied AI and ML to NEC, and some of the limitations in the field.

18.
Front Pediatr ; 11: 1161342, 2023.
Artículo en Inglés | MEDLINE | ID: mdl-37082706

RESUMEN

NEC remains one of the most common causes of mortality and morbidity in preterm infants. Animal models of necrotizing enterocolitis (NEC) have been crucial in improving our understanding of this devastating disease and identifying biochemical pathways with therapeutic potential. The pathogenesis of NEC remains incompletely understood, with no specific entity that unifies all infants that develop NEC. Therefore, investigators rely on animal models to manipulate variables and provide a means to test interventions, making them valuable tools to enhance our understanding and prevent and treat NEC. The advancements in molecular analytic tools, genetic manipulation, and imaging modalities and the emergence of scientific collaborations have given rise to unique perspectives and disease correlates, creating novel pathways of investigation. A critical review and understanding of the current phenotypic considerations of the highly relevant animal models of NEC are crucial to developing novel therapeutic and preventative strategies for NEC.

19.
Nutrients ; 14(9)2022 Apr 24.
Artículo en Inglés | MEDLINE | ID: mdl-35565748

RESUMEN

Necrotizing enterocolitis (NEC), an inflammatory disease of the intestine, is a common gastrointestinal emergency among preterm infants. Intestinal barrier dysfunction, hyperactivation of the premature immune system, and dysbiosis are thought to play major roles in the disease. Human milk (HM) is protective, but the mechanisms underpinning formula feeding as a risk factor in the development of NEC are incompletely understood. Hyaluronic acid 35 kDa (HA35), a bioactive glycosaminoglycan of HM, accelerates intestinal development in murine pups during homeostasis. In addition, HA35 prevents inflammation-induced tissue damage in pups subjected to murine NEC, incorporating Paneth cell dysfunction and dysbiosis. We hypothesized HA35 treatment would reduce histological injury and mortality in a secondary mouse model of NEC incorporating formula feeding. NEC-like injury was induced in 14-day mice by dithizone-induced disruption of Paneth cells and oral gavage of rodent milk substitute. Mortality and histological injury, serum and tissue cytokine levels, stool bacterial sequencing, and bulk RNA-Seq comparisons were analyzed. HA35 significantly reduced the severity of illness in this model, with a trend toward reduced mortality, while RNA-Seq analysis demonstrated HA35 upregulated genes associated with goblet cell function and innate immunity. Activation of these critical protective and reparative mechanisms of the small intestine likely play a role in the reduced pathology and enhanced survival trends of HA-treated pups subjected to intestinal inflammation in this secondary model of NEC, providing potentially interesting translational targets for the human preterm disease.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Animales , Animales Recién Nacidos , Modelos Animales de Enfermedad , Disbiosis , Enterocolitis Necrotizante/microbiología , Humanos , Ácido Hialurónico/farmacología , Recién Nacido , Recien Nacido Prematuro , Inflamación , Ratones
20.
Nutrients ; 14(3)2022 Jan 24.
Artículo en Inglés | MEDLINE | ID: mdl-35276854

RESUMEN

Necrotizing enterocolitis (NEC) is a disease mainly of preterm infants with a 30-50% mortality rate and long-term morbidities for survivors. Treatment strategies are limited and have not improved in decades, prompting research into prevention strategies, particularly with probiotics. Recent work with the probiotic B. infantis EVC001 suggests that this organism may generate a more appropriate microbiome for preterm infants who generally have inappropriate gut colonization and inflammation, both risk factors for NEC. Experimental NEC involving Paneth cell disruption in combination with bacterial dysbiosis or formula feeding was induced in P14-16 C57Bl/6 mice with or without gavaged B. infantis. Following completion of the model, serum, small intestinal tissue, the cecum, and colon were harvested to examine inflammatory cytokines, injury, and the microbiome, respectively. EVC001 treatment significantly decreased NEC in a bacterial dysbiosis dependent model, but this decrease was model-dependent. In the NEC model dependent on formula feeding, no difference in injury was observed, but trending to significant differences was observed in serum cytokines. EVC001 also improved wound closure at six and twelve hours compared to the sham control in intestinal epithelial monolayers. These findings suggest that B. infantis EVC001 can prevent experimental NEC through anti-inflammatory and epithelial barrier restoration properties.


Asunto(s)
Enterocolitis Necrotizante , Enfermedades del Recién Nacido , Animales , Bifidobacterium longum subspecies infantis , Enterocolitis Necrotizante/microbiología , Enterocolitis Necrotizante/prevención & control , Humanos , Recién Nacido , Recien Nacido Prematuro , Ratones , Ratones Endogámicos C57BL
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA