Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 8 de 8
Filtrar
Más filtros










Base de datos
Intervalo de año de publicación
1.
Pediatr Res ; 91(1): 101-106, 2022 01.
Artículo en Inglés | MEDLINE | ID: mdl-34561550

RESUMEN

BACKGROUND: Stem cell therapy has been proven to rescue intestinal injury and stimulate intestinal regeneration in necrotizing enterocolitis (NEC). Specifically, stem cells derived from amniotic fluid (AFSCs) and mesenchymal stem cells (MSCs) derived from bone marrow have shown promising results in the treatment of experimental NEC. This study aims to examine the effects of AFSCs and MSCs on the prevention of intestinal injury during experimental NEC. METHODS: Supernatants from AFSC and MSC cultures were collected to perform proteomic analysis. Prior to NEC induction, mice received intraperitoneal injections of phosphate-buffered saline (PBS), 2 × 106 AFSCs, or 2 × 106 MSCs. RESULTS: We found that AFSCs grew faster than MSCs. Proteomic analysis indicated that AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. Administering AFSCs before NEC induction decreased NEC severity and mucosal inflammation. Intestinal proliferation and endogenous stem cell activation were increased after AFSC administration. However, administering MSCs before NEC induction had no beneficial effects. CONCLUSIONS: This study demonstrated that AFSCs and MSCs have different protein release profiles. AFSCs can potentially be used as a preventative strategy for neonates at risk of NEC, while MSCs cannot be used. IMPACT: AFSCs and MSCs have distinct protein secretory profiles, and AFSCs are primarily involved in cell development and growth, while MSCs are involved in immune regulation. AFSCs are unique in transiently enhancing healthy intestinal epithelial cell growth, which offers protection against the development of experimental NEC. The prevention of NEC via the administration of AFSCs should be evaluated in infants at great risk of developing NEC or in infants with early signs of NEC.


Asunto(s)
Líquido Amniótico/citología , Trasplante de Células Madre , Animales , Enterocolitis Necrotizante , Humanos , Recién Nacido , Ratones
2.
Mol Nutr Food Res ; 66(4): e2100893, 2022 02.
Artículo en Inglés | MEDLINE | ID: mdl-34921749

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency affecting preterm infants. Breastmilk protects against NEC, partly due to human milk oligosaccharides (HMOs). HMO compositions are highly diverse, and it is unclear if anti-NEC properties are specific to carbohydrate motifs. Here, this study compares intestinal epithelial transcriptomes of five synthetic HMOs (sHMOs) and examines structure-function relationships of HMOs on intestinal signaling. METHODS AND RESULTS: This study interrogates the transcriptome of Caco-2Bbe1 cells in response to five synthetic HMOs (sHMOs) using RNA sequencing: 2'-fucosyllactose (2'-FL), 3-fucosyllactose (3FL), 6'-siallyllactose (6'-SL), lacto-N-tetraose (LNT), lacto-N-neotetraose (LNnT). Protection against intestinal barrier dysfunction and inflammation occurred in an HMO-dependent manner. Each sHMO exerts a unique set of host transcriptome changes and modulated unique signaling pathways. There is clustering between HMOs bearing similar side chains, with little overlap in gene regulation which is shared by all sHMOs. Interestingly, most sHMOs protect pups against NEC, exerting divergent mechanisms on intestinal cell morphology and inflammation. CONCLUSIONS: These results demonstrate that while structurally distinct HMOs impact intestinal physiology, their mechanisms of action differ. This finding establishes the first structure-function relationship of HMOs in the context of intestinal cell signaling responses and offers a functional framework by which to screen and design HMO-like compounds.


Asunto(s)
Enterocolitis Necrotizante , Leche Humana , Animales , Células CACO-2 , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/prevención & control , Humanos , Lactante , Recién Nacido , Recien Nacido Prematuro , Ratones , Leche Humana/química , Oligosacáridos/química , Relación Estructura-Actividad , Transcriptoma
3.
PLoS One ; 16(12): e0260522, 2021.
Artículo en Inglés | MEDLINE | ID: mdl-34855833

RESUMEN

PURPOSE: Necrotizing enterocolitis (NEC) is one of the most distressing gastrointestinal emergencies affecting neonates. Amniotic fluid stem cells (AFSC) improve intestinal injury and survival in experimental NEC but are difficult to administer. In this study, we evaluated whether conditioned medium (CM) derived from human AFSC have protective effects. METHODS: Three groups of C57BL/6 mice were studied: (i) breast-fed mice as control; (ii) experimental NEC mice receiving PBS; and (iii) experimental NEC mice receiving CM. NEC was induced between post-natal days P5 through P9 via: (A) gavage feeding of hyperosmolar formula four-time a day; (B) 10 minutes hypoxia prior to feeds; and (C) lipopolysaccharide administration on P6 and P7. Intra-peritoneal injections of either PBS or CM were given on P6 and P7. All mice were sacrificed on P9 and terminal ileum were harvested for analyses. RESULTS: CM treatment increased survival and reduced intestinal damage, decreased mucosal inflammation (IL-6; TNF-α), neutrophil infiltration (MPO), and apoptosis (CC3), and also restored angiogenesis (VEGF) in the ileum. Additionally, CM treated mice had increased levels of epithelial proliferation (Ki67) and stem cell activity (Olfm4; Lgr5) compared to NEC+PBS mice, showing restored intestinal regeneration and recovery during NEC induction. CM proteomic analysis of CM content identified peptides that regulated immune and stem cell activity. CONCLUSIONS: CM derived from human AFSC administered in experimental NEC exhibited various benefits including reduced intestinal injury and inflammation, increased enterocyte proliferation, and restored intestinal stem cell activity. This study provides the scientific basis for the use of CM derived from AFSC in neonates with NEC.


Asunto(s)
Enterocolitis Necrotizante , Líquido Amniótico , Humanos , Recién Nacido , Proteómica
4.
Mol Nutr Food Res ; 64(21): e2000519, 2020 11.
Artículo en Inglés | MEDLINE | ID: mdl-32926533

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a devastating gastrointestinal emergency and currently the leading cause of mortality in preterm infants. Recent studies show that human milk oligosaccharides (HMOs) reduce the frequency and incidence of NEC; however, the molecular mechanisms for their protection are largely unexplored. METHODS AND RESULTS: To address this gap, a genome-wide profiling of the intestinal epithelial transcriptome in response to HMOs using RNA-sequencing is performed. It is found that HMOs alter the host transcriptome in 225 unique target genes pertaining to cell proliferation and differentiation, including upregulation of stem cell differentiation marker HMGCS2. To validate these results, differentiation in Caco-2Bbe1 (Caco-2) intestinal cells is verified by Alcian Blue staining and transepithelial electrical resistance (TER) recordings. Furthermore, an in vivo model of NEC is also employed whereby neonatal pups are gavage fed HMOs. Interestingly, HMOs-fed pups show enhanced cell MUC2 differentiation and HMGCS2 expression. CONCLUSIONS: These findings demonstrate HMOs protect against NEC in part by altering the differentiation of the crypt-villus axis. In addition, this study suggests that pooled HMOs directly induce a series of biological processes, which provide mechanistic insights to how HMOs protect the host intestine.


Asunto(s)
Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Oligosacáridos/farmacología , Animales , Células CACO-2 , Ciclo Celular/efectos de los fármacos , Ciclo Celular/genética , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Perros , Enterocolitis Necrotizante/genética , Femenino , Perfilación de la Expresión Génica , Humanos , Hidroximetilglutaril-CoA Sintasa/metabolismo , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Células de Riñón Canino Madin Darby , Masculino , Ratones Endogámicos C57BL , Receptores Activados del Proliferador del Peroxisoma/metabolismo , Transducción de Señal/efectos de los fármacos , Transducción de Señal/genética , Serina-Treonina Quinasas TOR/metabolismo
5.
Cell Death Dis ; 11(9): 750, 2020 09 14.
Artículo en Inglés | MEDLINE | ID: mdl-32929076

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating intestinal disease primarily affecting preterm neonates and causing high morbidity, high mortality, and huge costs for the family and society. The treatment and the outcome of the disease have not changed in recent decades. Emerging evidence has shown that stimulating the Wnt/ß-catenin pathway and enhancing intestinal regeneration are beneficial in experimental NEC, and that they could potentially be used as a novel treatment. Amniotic fluid stem cells (AFSC) and AFSC-derived extracellular vesicles (EV) can be used to improve intestinal injury in experimental NEC. However, the mechanisms by which they affect the Wnt/ß-catenin pathway and intestinal regeneration are unknown. In our current study, we demonstrated that AFSC and EV attenuate NEC intestinal injury by activating the Wnt signaling pathway. AFSC and EV stimulate intestinal recovery from NEC by increasing cellular proliferation, reducing inflammation and ultimately regenerating a normal intestinal epithelium. EV administration has a rescuing effect on intestinal injury when given during NEC induction; however, it failed to prevent injury when given prior to NEC induction. AFSC-derived EV administration is thus a potential emergent novel treatment strategy for NEC.


Asunto(s)
Enterocolitis Necrotizante/genética , Vesículas Extracelulares/metabolismo , Intestinos/lesiones , Vía de Señalización Wnt/genética , Animales , Modelos Animales de Enfermedad , Humanos , Ratones , Ratas
6.
Cell Death Dis ; 10(10): 743, 2019 10 03.
Artículo en Inglés | MEDLINE | ID: mdl-31582728

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by acute intestinal injury. Intestinal stem cell (ISC) renewal is required for gut regeneration in response to acute injury. The Wnt/ß-catenin pathway is essential for intestinal renewal and ISC maintenance. We found that ISC expression, Wnt activity and intestinal regeneration were all decreased in both mice with experimental NEC and in infants with acute active NEC. Moreover, intestinal organoids derived from NEC-injured intestine of both mice and humans failed to maintain proliferation and presented more differentiation. Administration of Wnt7b reversed these changes and promoted growth of intestinal organoids. Additionally, administration of exogenous Wnt7b rescued intestinal injury, restored ISC, and reestablished intestinal epithelial homeostasis in mice with NEC. Our findings demonstrate that during NEC, Wnt/ß-catenin signaling is decreased, ISC activity is impaired, and intestinal regeneration is defective. Administration of Wnt resulted in the maintenance of intestinal epithelial homeostasis and avoidance of NEC intestinal injury.


Asunto(s)
Enterocolitis Necrotizante/fisiopatología , Intestinos/fisiopatología , Regeneración/fisiología , Vía de Señalización Wnt , Animales , Diferenciación Celular/efectos de los fármacos , Proliferación Celular/efectos de los fármacos , Enterocolitis Necrotizante/genética , Células Epiteliales/efectos de los fármacos , Células Epiteliales/metabolismo , Regulación de la Expresión Génica/efectos de los fármacos , Humanos , Intestinos/efectos de los fármacos , Intestinos/patología , Ratones Endogámicos C57BL , Modelos Biológicos , Organoides/efectos de los fármacos , Organoides/metabolismo , Proteínas Proto-Oncogénicas/administración & dosificación , Proteínas Proto-Oncogénicas/farmacología , Regeneración/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/metabolismo , Análisis de Supervivencia , Proteínas Wnt/administración & dosificación , Proteínas Wnt/farmacología , Vía de Señalización Wnt/efectos de los fármacos , Vía de Señalización Wnt/genética
7.
Mol Nutr Food Res ; 63(3): e1800658, 2019 02.
Artículo en Inglés | MEDLINE | ID: mdl-30407734

RESUMEN

SCOPE: Necrotizing enterocolitis (NEC) is a leading cause of morbidity and death in preterm infants, occurring more often in formula-fed than breastfed infants. Studies in both rats and humans show that human milk oligosaccharides (HMOs) lower the incidence of NEC, but the mechanism underlying such protection is currently unclear. METHODS AND RESULTS: By extracting HMOs from pooled human breastmilk, the impact of HMOs on the intestinal mucin levels in a murine model of NEC are investigated. To confirm the results, the findings are validated by exposing human intestinal epithelial cells and intestinal organoids to HMOs and evaluated for mucin expression. HMO-gavage to pups increases Muc2 levels and decreases intestinal permeability to macromolecular dextran. HMO-treated cells have increased Muc2 expression, decreased bacterial attachment and dextran permeability during challenge by enteric pathogens. To identify the mediators involved in HMO induction of mucins, it is demonstrated that HMOs directly induce the expression of chaperone proteins including protein disulfide isomerase (PDI). Suppression of PDI activity removes the protective effects of HMOs on barrier function in vitro as well as NEC protection in vivo. CONCLUSIONS: Taken together, the results provide insights to the possible mechanisms by which HMOs protect the neonatal intestine through upregulation of mucins.


Asunto(s)
Enterocolitis Necrotizante/prevención & control , Leche Humana/química , Mucina 2/genética , Oligosacáridos/farmacología , Animales , Animales Recién Nacidos , Células CACO-2 , Estrés del Retículo Endoplásmico/efectos de los fármacos , Enterocolitis Necrotizante/metabolismo , Células Caliciformes/efectos de los fármacos , Humanos , Mucosa Intestinal/efectos de los fármacos , Ratones , Ratones Endogámicos C57BL , Mucina 2/análisis , Proteína Disulfuro Isomerasas/fisiología
8.
Pediatr Surg Int ; 35(1): 3-7, 2019 Jan.
Artículo en Inglés | MEDLINE | ID: mdl-30382376

RESUMEN

BACKGROUND: Adult intestinal organoids have been used to study ex vivo intestinal injury in adulthood. However, the neonatal intestinal epithelium has many unique features that are different from adult mature intestine. Establishing a neonatal ex vivo organoid model is essential to study the epithelial physiology in early postnatal development and to investigate derangements associated with disease processes during the neonatal period like necrotizing enterocolitis (NEC). METHODS: Fresh and frozen terminal ileum was harvested from mice pups on postnatal day 9. Crypts were isolated and organoids were cultured. Organoids were exposed to hypoxia and lipopolysaccharide (LPS) for 48 h to induce epithelial injury. Inflammatory cytokines and tight junction proteins were evaluated. RESULTS: Robust intestinal organoids can be formed from both fresh and frozen intestinal tissue of neonatal mice pups. Hypoxia and LPS administration induced intestinal inflammation and disrupted tight junctions in these neonatal intestinal organoids. CONCLUSIONS: We have established a novel method to grow organoids from neonatal intestine. We demonstrated that these organoids respond to the injury occurring during neonatal intestinal diseases such as NEC by increasing the organoid inflammation and by disrupting the organoid barrier function. Organoids provide an ex vivo platform to study intestinal physiology and pathology during the neonatal period.


Asunto(s)
Enterocolitis Necrotizante/patología , Íleon/patología , Organoides/patología , Animales , Animales Recién Nacidos , Citocinas/metabolismo , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/metabolismo , Íleon/metabolismo , Mucosa Intestinal/metabolismo , Mucosa Intestinal/patología , Lipopolisacáridos/metabolismo , Ratones , Ratones Endogámicos C57BL , Organoides/metabolismo
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA
...