Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 100
Filtrar
2.
J Perinatol ; 2023 Dec 27.
Artigo em Inglês | MEDLINE | ID: mdl-38151596

RESUMO

OBJECTIVE: Neonatal Opioid Withdrawal Syndrome (NOWS) has been associated with the development of necrotizing enterocolitis (NEC) in term and late-preterm neonates. In this study, we used stool gene expression to determine if an increase in baseline inflammation in the intestine of infants with NOWS is associated with these findings. STUDY DESIGN: Stool samples were prospectively collected between days 1-3 and days 4-9 after delivery for opioid-exposed ( n = 9) or non-exposed neonates (n = 8). Stool gene expression for TLR4 and HMGB1 was determined via real-time PCR. RESULTS: TLR4 expression was higher in the stool of the non-exposed group in both time periods, between days 1-3 (P < 0.0001) and days 4-9 (P < 0.05) after delivery. No significant difference in HMGB1 expression was found at either time point (P > 0.05). CONCLUSION: These findings point to an important interplay between opioid exposure and/or NOWS and the inflammatory milieu of the neonatal intestine.

4.
J Vis Exp ; (197)2023 07 28.
Artigo em Inglês | MEDLINE | ID: mdl-37590536

RESUMO

Necrotizing enterocolitis (NEC) is a severe and potentially fatal intestinal disease that has been difficult to study due to its complex pathogenesis, which remains incompletely understood. The pathophysiology of NEC includes disruption of intestinal tight junctions, increased gut barrier permeability, epithelial cell death, microbial dysbiosis, and dysregulated inflammation. Traditional tools to study NEC include animal models, cell lines, and human or mouse intestinal organoids. While studies using those model systems have improved the field's understanding of disease pathophysiology, their ability to recapitulate the complexity of human NEC is limited. An improved in vitro model of NEC using microfluidic technology, named NEC-on-a-chip, has now been developed. The NEC-on-a-chip model consists of a microfluidic device seeded with intestinal enteroids derived from a preterm neonate, co-cultured with human endothelial cells and the microbiome from an infant with severe NEC. This model is a valuable tool for mechanistic studies into the pathophysiology of NEC and a new resource for drug discovery testing for neonatal intestinal diseases. In this manuscript, a detailed description of the NEC-on-a-chip model will be provided.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Microbiota , Animais , Lactente , Camundongos , Humanos , Recém-Nascido , Disbiose , Células Endoteliais , Microfluídica
5.
Semin Pediatr Surg ; 32(3): 151306, 2023 Jun.
Artigo em Inglês | MEDLINE | ID: mdl-37276783

RESUMO

Maternal breast milk is the penultimate nutritional source for term and preterm neonates. Its composition is highly complex and includes multiple factors that enhance the development of nearly every neonatal organ system leading to both short- and long-term health benefits. Intensive research is focused on identifying breast milk components that enhance infant health. However, this research is complicated by the significant impact of maternal factors and the processing of pumped breast milk on bioactive ingredients. Optimizing enteral nutrition is particularly important for preterm neonates who miss the transplacental acquisition of nutrients in the third trimester of pregnancy and are at risk for illnesses associated with gut barrier dysfunction, including sepsis and necrotizing enterocolitis. In this review, we will discuss the health benefits of breast milk and its bioactive components.


Assuntos
Leite Humano , Sepse , Lactente , Gravidez , Feminino , Recém-Nascido , Humanos , Recém-Nascido Prematuro , Fórmulas Infantis , Nutrição Enteral
6.
Front Pediatr ; 11: 1188050, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37334221

RESUMO

Necrotizing enterocolitis (NEC) is an intestinal disease that primarily impacts preterm infants. The pathophysiology of NEC involves a complex interplay of factors that result in a deleterious immune response, injury to the intestinal mucosa, and in its most severe form, irreversible intestinal necrosis. Treatments for NEC remain limited, but one of the most effective preventative strategies for NEC is the provision of breast milk feeds. In this review, we discuss mechanisms by which bioactive nutrients in breast milk impact neonatal intestinal physiology and the development of NEC. We also review experimental models of NEC that have been used to study the role of breast milk components in disease pathophysiology. These models are necessary to accelerate mechanistic research and improve outcomes for neonates with NEC.

7.
Front Pediatr ; 11: 1184940, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-37325361

RESUMO

Introduction: Necrotizing enterocolitis (NEC) is a potentially fatal intestinal disease primarily affecting preterm infants. Early diagnosis of neonates with NEC is crucial to improving outcomes; however, traditional diagnostic tools remain inadequate. Biomarkers represent an opportunity to improve the speed and accuracy of diagnosis, but they are not routinely used in clinical practice. Methods: In this study, we utilized an aptamer-based proteomic discovery assay to identify new serum biomarkers of NEC. We compared levels of serum proteins in neonates with and without NEC and identified ten differentially expressed serum proteins between these groups. Results: We detected two proteins, C-C motif chemokine ligand 16 (CCL16) and immunoglobulin heavy constant alpha 1 and 2 heterodimer (IGHA1 IGHA2), that were significantly increased during NEC and eight that were significantly decreased. Generation of receiver operating characteristic (ROC) curves revealed that alpha-fetoprotein (AUC = 0.926), glucagon (AUC = 0.860), and IGHA1 IGHA2 (AUC = 0.826) were the proteins that best differentiated patients with and without NEC. Discussion: These findings indicate that further investigation into these serum proteins as a biomarker for NEC is warranted. In the future, laboratory tests incorporating these differentially expressed proteins may improve the ability of clinicians to diagnose infants with NEC rapidly and accurately.

8.
J Immunol Res ; 2023: 5356646, 2023.
Artigo em Inglês | MEDLINE | ID: mdl-36959922

RESUMO

Specific biomarkers of intestinal injury associated with necrotizing enterocolitis (NEC) are needed to diagnose and monitor intestinal mucosal injury and recovery. This study aims to develop and test a modified enzyme-linked immunosorbent assay (ELISA) protocol to detect the total keratin 8 (K8) in the stool of newborns with NEC and investigate the clinical value of fecal K8 as a marker of intestinal injury specifically associated with NEC. We collected fecal samples from five newborns with NEC and five gestational age-matched premature neonates without NEC at the Lucile Packard Children's Hospital Stanford and Washington University School of Medicine, respectively. Fecal K8 levels were measured using a modified ELISA protocol and Western blot, and fecal calprotectin was measured using a commercial ELISA kit. Clinical data, including gestational age, birth weight, Bell stage for NEC, feeding strategies, total white blood cell (WBC) count, and other pertinent clinical variables, were collected and analyzed. Fecal K8 levels were significantly higher in the pre-NEC group (1-2 days before diagnosis of NEC) and NEC group than those in the non-NEC group (p = 0.013, p = 0.041). Moreover, fecal K8 was relatively higher at the onset of NEC and declined after the resolution of the disease (p = 0.019). Results with similar trends to fecal K8 were also seen in fecal calprotectin (p = 0.046), but not seen in total WBC count (p = 0.182). In conclusion, a modified ELISA protocol for the total K8 protein was successfully developed for the detection of fecal K8 in the clinical setting of premature newborns with NEC. Fecal K8 is noted to be significantly increased in premature newborns with NEC and may, therefore, serve as a noninvasive and specific marker for intestinal epithelial injury associated with NEC.


Assuntos
Enterocolite Necrosante , Recém-Nascido Prematuro , Humanos , Recém-Nascido , Enterocolite Necrosante/diagnóstico , Fezes , Recém-Nascido Prematuro/metabolismo , Queratina-8/metabolismo , Complexo Antígeno L1 Leucocitário
9.
JCI Insight ; 8(8)2023 04 24.
Artigo em Inglês | MEDLINE | ID: mdl-36881475

RESUMO

Necrotizing enterocolitis (NEC) is a deadly gastrointestinal disease of premature infants that is associated with an exaggerated inflammatory response, dysbiosis of the gut microbiome, decreased epithelial cell proliferation, and gut barrier disruption. We describe an in vitro model of the human neonatal small intestinal epithelium (Neonatal-Intestine-on-a-Chip) that mimics key features of intestinal physiology. This model utilizes intestinal enteroids grown from surgically harvested intestinal tissue from premature infants and cocultured with human intestinal microvascular endothelial cells within a microfluidic device. We used our Neonatal-Intestine-on-a-Chip to recapitulate NEC pathophysiology by adding infant-derived microbiota. This model, named NEC-on-a-Chip, simulates the predominant features of NEC, including significant upregulation of proinflammatory cytokines, decreased intestinal epithelial cell markers, reduced epithelial proliferation, and disrupted epithelial barrier integrity. NEC-on-a-Chip provides an improved preclinical model of NEC that facilitates comprehensive analysis of the pathophysiology of NEC using precious clinical samples. This model is an advance toward a personalized medicine approach to test new therapeutics for this devastating disease.


Assuntos
Células Endoteliais , Enterocolite Necrosante , Lactente , Recém-Nascido , Humanos , Recém-Nascido Prematuro , Mucosa Intestinal , Dispositivos Lab-On-A-Chip
10.
J Perinatol ; 43(1): 103-107, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36097287

RESUMO

Fortification of human milk is the standard of care for very low birth weight (VLBW) infants and is required to support adequate postnatal growth and development. Achieving adequate growth velocity and preventing growth faltering is critical for the developing neonatal brain and optimizing long-term neurodevelopmental outcomes. Mother's milk is the gold standard nutrition to feed preterm infants, however, it does not provide the nutrients needed to support the growth of VLBW infants. After the decision is made to use mother's milk (if available) or alternatively, donor human milk, many dilemmas exist with regards to additional treatment decisions surrounding the type of fortification to use, when to fortify, and the duration of fortification. In this article, we will review the differences in mother's milk compared to donor milk, the different types of human milk fortifiers, the optimal timing of fortification, and discuss when to discontinue human milk fortification.


Assuntos
Recém-Nascido Prematuro , Leite Humano , Lactente , Recém-Nascido , Humanos , Alimentos Fortificados , Recém-Nascido de muito Baixo Peso , Estado Nutricional , Fenômenos Fisiológicos da Nutrição do Lactente
11.
J Perinatol ; 43(1): 114-119, 2023 01.
Artigo em Inglês | MEDLINE | ID: mdl-36127395

RESUMO

Intestinal failure in neonatal and pediatric populations can be debilitating for patients and difficult to manage for clinicians. Management strategies include referral to an intestinal rehabilitation center, small volume trophic feeds to stimulate the intestine with cautious advancement of enteral nutrition using a standardized and evidence-based feeding protocol, and supplemental parenteral nutrition to optimize an infant's growth and nutrition. In this review, we discuss the causes of intestinal failure, parenteral nutrition strategies, enteral feeding initiation and advancement protocols, as well as the challenges in feeding an infant with intestinal failure.


Assuntos
Recém-Nascido Prematuro , Insuficiência Intestinal , Recém-Nascido , Criança , Lactente , Humanos , Neonatologistas , Nutrição Parenteral/métodos , Nutrição Enteral/métodos
12.
Semin Perinatol ; 47(1): 151690, 2023 02.
Artigo em Inglês | MEDLINE | ID: mdl-36581527

RESUMO

Human breast milk is the optimal nutrition for all infants and is comprised of many bioactive and immunomodulatory components. The components in human milk, such as probiotics, human milk oligosaccharides (HMOs), extracellular vesicles, peptides, immunoglobulins, growth factors, cytokines, and vitamins, play a critical role in guiding neonatal development beyond somatic growth. In this review, we will describe the bioactive factors in human milk and discuss how these factors shape neonatal immunity, the intestinal microbiome, intestinal development, and more from the inside out.


Assuntos
Microbioma Gastrointestinal , Leite Humano , Lactente , Recém-Nascido , Feminino , Criança , Humanos , Desenvolvimento Infantil , Oligossacarídeos , Estado Nutricional , Aleitamento Materno
13.
Mucosal Immunol ; 15(6): 1181-1187, 2022 06.
Artigo em Inglês | MEDLINE | ID: mdl-36380094

RESUMO

Rapid development of the fetal and neonatal intestine is required to meet the growth requirements of early life and form a protective barrier against external insults encountered by the intestinal mucosa. The fetus receives nutrition via the placenta and is protected from harmful pathogens in utero, which leads to intestinal development in a relatively quiescent environment. Upon delivery, the intestinal mucosa is suddenly tasked with providing host defense and meeting nutritional demands. To serve these functions, an array of specialized epithelial cells develop from intestinal stem cells starting in utero and continuing postnatally. Intestinal disease results when these homeostatic processes are interrupted. For preterm neonates, the most common pathology resulting from epithelial barrier dysfunction is necrotizing enterocolitis (NEC). In this review, we discuss the normal development and function of the intestinal epithelium in early life as well as how disruption of these processes can lead to NEC.


Assuntos
Enterocolite Necrosante , Mucosa Intestinal , Recém-Nascido , Humanos , Mucosa Intestinal/patologia , Enterocolite Necrosante/patologia , Intestinos/patologia , Células Epiteliais/patologia
14.
Clin Epigenetics ; 14(1): 49, 2022 04 11.
Artigo em Inglês | MEDLINE | ID: mdl-35410447

RESUMO

OBJECTIVE: Necrotizing enterocolitis (NEC) is the most common and lethal gastrointestinal disease affecting preterm infants. NEC develops suddenly and is characterized by gut barrier destruction, an inflammatory response, intestinal necrosis and multi-system organ failure. There is currently no method for early NEC detection, and the pathogenesis of NEC remains unclear. DESIGN: To further understand the molecular mechanisms that support NEC, we used solution phase hybridization and next-generation DNA sequencing of bisulfite converted DNA to perform targeted genome-wide analysis of DNA methylation at high read depth. RESULTS: We found that ileal samples from surgical NEC infants (n = 5) exist in a broadly hypermethylated state relative to their non-NEC counterparts (n = 9). These trends were not uniform, with hypermethylation being most consistently observed outside CpG islands and promoters. We further identified several biologically interesting gene promoters that displayed differential methylation in NEC and a number of biological pathways that appear dysregulated in NEC. We also found that DNA methylation patterns identified in ileal NEC tissue were correlated with those found and published previously in stool samples from NEC-affected infants. CONCLUSION: We confirmed that surgical NEC is associated with broad DNA hypermethylation in the ileum, and this may be detectable in stool samples of affected individuals. Thus, an epigenomic liquid biopsy of stool may have significant potential as a biomarker with respect to the diagnostic/predictive detection of NEC. Our findings, along with recent similar observations in colon, suggest that epigenomic dysregulation is a significant feature of surgical NEC. These findings motivate future studies which will involve the longitudinal screening of samples obtained prior to the onset of NEC. Our long-term goal is the development of novel screening, diagnostic and phenotyping methods for NEC.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Ilhas de CpG , Metilação de DNA , Enterocolite Necrosante/diagnóstico , Enterocolite Necrosante/genética , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Intestino Delgado/patologia
15.
Neonatology ; 119(3): 334-344, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-35313308

RESUMO

INTRODUCTION: The neonatal sequential organ failure assessment (nSOFA) score is a tool for calculating mortality risk of infants in the neonatal intensive care unit. The utility of the nSOFA in determining the risk of mortality or the association with surgical intervention among infants with necrotizing enterocolitis (NEC) has not been investigated. METHODS: We performed a retrospective, cohort study of preterm (<37 weeks) infants with NEC Bell's stage ≥ IIA at six hospitals from 2008 to 2020. An nSOFA score (range 0-15) was assigned to each patient at nine time points from 48 h before or after clinical illness was suspected. RESULTS: Of the 259 infants, nSOFA scores for infants who died (n = 39) or had the composite outcome of surgery or death (n = 114) were significantly higher (p < 0.05) early in the NEC course compared to nSOFA scores for infants who survived medical NEC. Twelve hours after evaluation, the area under the receiver operating characteristic curve was 0.87 (95% confidence interval [CI], 0.80-0.93) to discriminate for mortality and 0.84 (95% CI, 0.79-0.90) for surgery or death (p < 0.001). A maximum nSOFA score of ≥4 at -6, 0, 6, or 12 h following evaluation was associated with a 20-fold increase in mortality and 19-fold increase in surgery or death compared with a score of <4 (p < 0.001). CONCLUSION: In this multicenter cohort, the nSOFA score was able to discriminate well for death as well as surgery or death among infants with NEC. The nSOFA is a clinical research tool that may be used in infants with NEC to improve classification by objective quantification of organ dysfunction.


Assuntos
Enterocolite Necrosante , Doenças do Recém-Nascido , Estudos de Coortes , Enterocolite Necrosante/complicações , Enterocolite Necrosante/diagnóstico , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Escores de Disfunção Orgânica , Estudos Retrospectivos
16.
J Biomed Inform ; 128: 104031, 2022 04.
Artigo em Inglês | MEDLINE | ID: mdl-35183765

RESUMO

Preterm birth affects more than 10% of all births worldwide. Such infants are much more prone to Growth Faltering (GF), an issue that has been unsolved despite the implementation of numerous interventions aimed at optimizing preterm infant nutrition. To improve the ability for early prediction of GF risk for preterm infants we collected a comprehensive, large, and unique clinical and microbiome dataset from 3 different sites in the US and the UK. We use and extend machine learning methods for GF prediction from clinical data. We next extend graphical models to integrate time series clinical and microbiome data. A model that integrates clinical and microbiome data improves on the ability to predict GF when compared to models using clinical data only. Information on a small subset of the taxa is enough to help improve model accuracy and to predict interventions that can improve outcome. We show that a hierarchical classifier that only uses a subset of the taxa for a subset of the infants is both the most accurate and cost-effective method for GF prediction. Further analysis of the best classifiers enables the prediction of interventions that can improve outcome.


Assuntos
Microbiota , Nascimento Prematuro , Humanos , Lactente , Recém-Nascido , Recém-Nascido Prematuro , Aprendizado de Máquina
17.
Immunity ; 55(2): 237-253.e8, 2022 02 08.
Artigo em Inglês | MEDLINE | ID: mdl-35081371

RESUMO

The Th17 cell-lineage-defining cytokine IL-17A contributes to host defense and inflammatory disease by coordinating multicellular immune responses. The IL-17 receptor (IL-17RA) is expressed by diverse intestinal cell types, and therapies targeting IL-17A induce adverse intestinal events, suggesting additional tissue-specific functions. Here, we used multiple conditional deletion models to identify a role for IL-17A in secretory epithelial cell differentiation in the gut. Paneth, tuft, goblet, and enteroendocrine cell numbers were dependent on IL-17A-mediated induction of the transcription factor ATOH1 in Lgr5+ intestinal epithelial stem cells. Although dispensable at steady state, IL-17RA signaling in ATOH1+ cells was required to regenerate secretory cells following injury. Finally, IL-17A stimulation of human-derived intestinal organoids that were locked into a cystic immature state induced ATOH1 expression and rescued secretory cell differentiation. Our data suggest that the cross talk between immune cells and stem cells regulates secretory cell lineage commitment and the integrity of the mucosa.


Assuntos
Fatores de Transcrição Hélice-Alça-Hélice Básicos/metabolismo , Mucosa Intestinal/citologia , Receptores Acoplados a Proteínas G/metabolismo , Receptores de Interleucina-17/metabolismo , Células-Tronco/metabolismo , Animais , Comunicação Celular , Diferenciação Celular/efeitos dos fármacos , Linhagem da Célula/efeitos dos fármacos , Colite/induzido quimicamente , Colite/metabolismo , Colite/patologia , Sulfato de Dextrana/efeitos adversos , Humanos , Interleucina-17/metabolismo , Interleucina-17/farmacologia , Mucosa Intestinal/metabolismo , Intestinos/efeitos dos fármacos , Intestinos/metabolismo , Intestinos/patologia , Camundongos , Camundongos Knockout , NF-kappa B/metabolismo , Receptores de Interleucina-17/deficiência , Fatores de Transcrição SOX9/metabolismo , Transdução de Sinais , Células-Tronco/citologia
18.
Commun Biol ; 5(1): 47, 2022 01 12.
Artigo em Inglês | MEDLINE | ID: mdl-35022507

RESUMO

Lysosomal membrane permeabilization (LMP) and cathepsin release typifies lysosome-dependent cell death (LDCD). However, LMP occurs in most regulated cell death programs suggesting LDCD is not an independent cell death pathway, but is conscripted to facilitate the final cellular demise by other cell death routines. Previously, we demonstrated that Caenorhabditis elegans (C. elegans) null for a cysteine protease inhibitor, srp-6, undergo a specific LDCD pathway characterized by LMP and cathepsin-dependent cytoplasmic proteolysis. We designated this cell death routine, lysoptosis, to distinguish it from other pathways employing LMP. In this study, mouse and human epithelial cells lacking srp-6 homologues, mSerpinb3a and SERPINB3, respectively, demonstrated a lysoptosis phenotype distinct from other cell death pathways. Like in C. elegans, this pathway depended on LMP and released cathepsins, predominantly cathepsin L. These studies suggested that lysoptosis is an evolutionarily-conserved eukaryotic LDCD that predominates in the absence of neutralizing endogenous inhibitors.


Assuntos
Antígenos de Neoplasias/genética , Morte Celular , Células Epiteliais/fisiologia , Serpinas/genética , Animais , Antígenos de Neoplasias/metabolismo , Linhagem Celular Tumoral , Humanos , Camundongos , Serpinas/metabolismo
19.
Front Pediatr ; 10: 1107404, 2022.
Artigo em Inglês | MEDLINE | ID: mdl-36714655

RESUMO

Necrotizing enterocolitis (NEC) is a devastating, multifactorial disease mainly affecting the intestine of premature infants. Recent discoveries have significantly enhanced our understanding of risk factors, as well as, cellular and genetic mechanisms of this complex disease. Despite these advancements, no essential, single risk factor, nor the mechanism by which each risk factor affects NEC has been elucidated. Nonetheless, recent research indicates that maternal factors, antibiotic exposure, feeding, hypoxia, and altered gut microbiota pose a threat to the underdeveloped immunity of preterm infants. Here we review predisposing factors, status of unwarranted immune responses, and microbial pathogenesis in NEC based on currently available scientific evidence. We additionally discuss novel techniques and models used to study NEC and how this research translates from the bench to the bedside into potential treatment strategies.

SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...