Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 22
Filtrar
Más filtros

Banco de datos
Tipo del documento
Intervalo de año de publicación
1.
Pediatr Res ; 2024 Feb 28.
Artículo en Inglés | MEDLINE | ID: mdl-38418592

RESUMEN

BACKGROUND: Recombinant human IGF-1/binding protein-3 (rhIGF-1/BP3) is currently being tested in phase II clinical trials in premature infants to prevent bronchopulmonary dysplasia, but its impact on the neonatal intestine remains unclear. The aim of this study was to determine whether rhIGF-1/BP3 protects against necrotizing enterocolitis (NEC) in mice and to investigate the mechanisms involved. METHODS: Neonatal mice were dam fed or injected intraperitoneally with rhIGF-1/BP3 (or vehicle) and submitted to an experimental NEC model. Serum IGF-1 was assessed by ELISA and intestinal vascular endothelial growth factor (VEGF) and VEGF receptor 2 (VEGFR2) expression by Western blot. Intestinal endothelial cell proliferation, and enterocyte proliferation and migration were examined by immunofluorescence. Pup survival and histological intestinal injury were determined. RESULTS: In pups exposed to experimental NEC, serum IBP3-bound IGF-1 level was decreased. Exogenous rhIGF-1/BP3 preserved VEGF and VEGFR2 protein expression, decreased vascular permeability, and preserved endothelial cell proliferation in the small intestine. Furthermore, rhIGF-1/BP3 promoted enterocyte proliferation and migration, which effects were attenuated by inhibiting VEGFR2 signaling, decreased enterocyte apoptosis and decreased systemic and intestinal inflammation. rhIGF-1/BP3 improved survival and reduced the incidence of severe intestinal injury in experimental NEC. CONCLUSIONS: Exogenous rhIGF-1/BP3 protects neonatal mice against experimental NEC via multiple mechanisms. IMPACT: Exogenous rhIGF-1/BP3 preserves intestinal microvascular development and integrity, promotes enterocyte proliferation and migration, decreases local and systemic inflammation, and protects neonatal mice against NEC. The article adds pre-clinical evidence of a protective role for rhIGF-1/BP3 on the premature gut. It provides evidence supporting the use of rhIGF1/BP3 in premature neonates to protect against NEC.

2.
J Pediatr ; 259: 113478, 2023 08.
Artículo en Inglés | MEDLINE | ID: mdl-37182664

RESUMEN

OBJECTIVE: To test the hypothesis that nailfold capillaroscopy can noninvasively detect dysregulated retinal angiogenesis and predict retinopathy of prematurity (ROP) in infants born premature before its development. METHODS: In a cohort of 32 infants born <33 weeks of gestation, 1386 nailfold capillary network images of the 3 middle fingers of each hand were taken during the first month of life. From these, 25 infants had paired data taken 2 weeks apart during the first month of life. Images were analyzed for metrics of peripheral microvascular density using a machine learning-based segmentation approach and a previously validated microvascular quantification platform (REAVER vascular analysis). Results were correlated with subsequent development of ROP based on a published consensus ROP severity scale. RESULTS: In total, 18 of 32 (56%) (entire cohort) and 13 of 25 (52%) (2-time point subgroup) developed ROP. Peripheral vascular density decreased significantly during the first month of life. In the paired time point analysis, vessel length density, a key metric of peripheral vascular density, was significantly greater at both time points among infants who later developed ROP (15 563 and 11 996 µm/mm2, respectively) compared with infants who did not (12 252 and 8845 µm/mm2, respectively) (P < .001, both time points). A vessel length density cutoff of >15 100 at T1 or at T2 correctly detected 3 of 3 infants requiring ROP therapy. In a mixed-effects linear regression model, peripheral vascular density metrics were significantly correlated with ROP severity. CONCLUSIONS: Nailfold microvascular density assessed during the first month of life is a promising, noninvasive biomarker to identify premature infants at highest risk for ROP before detection on eye exam.


Asunto(s)
Retinopatía de la Prematuridad , Recién Nacido , Lactante , Humanos , Retinopatía de la Prematuridad/diagnóstico , Retinopatía de la Prematuridad/terapia , Angioscopía Microscópica , Recien Nacido Prematuro , Retina , Edad Gestacional , Factores de Riesgo
3.
Am J Pathol ; 189(3): 604-618, 2019 03.
Artículo en Inglés | MEDLINE | ID: mdl-30593820

RESUMEN

Necrotizing enterocolitis (NEC) is a devastating disease affecting premature infants with intestinal inflammation and necrosis. The neonatal intestinal inflammatory response is rich in macrophages, and blood monocyte count is low in human NEC. We previously found that NF-κB mediates the intestinal injury in experimental NEC. However, the role of NF-κB in myeloid cells during NEC remains unclear. Herein, inhibitor of kappaB kinase ß (IKKß), a critical kinase mediating NF-κB activation, was deleted in lysozyme M (Lysm)-expressing cells, which were found to be Cd11b+Ly6c+ monocytes but not Cd11b+Ly6c- macrophages in the dam-fed neonatal mouse intestine. NEC induced differentiation of monocytes into intestinal macrophages and up-regulation of monocyte recruitment genes (eg, L-selectin) in the macrophage compartment in wild-type mice, but not in pups with IKKß deletion in Lysm+ cells. Thus, NF-κB is required for NEC-induced monocyte activation, recruitment, and differentiation in neonatal intestines. Furthermore, pups with Lysm-IKKß deletion had improved survival and decreased incidence of severe NEC compared with littermate controls. Decreased NEC severity was not associated with an improved intestinal barrier. In contrast, NEC was unabated in mice with IKKß deletion in intestinal epithelial cells. Together, these data suggest that recruitment of Ly6c+ monocytes into the intestine, NF-κB activation in these cells, and differentiation of Ly6c+ monocytes into macrophages are critical cellular and molecular events in NEC development to promote disease.


Asunto(s)
Antígenos Ly/metabolismo , Enterocolitis Necrotizante/metabolismo , Células Epiteliales/metabolismo , Macrófagos/metabolismo , Monocitos/metabolismo , FN-kappa B/metabolismo , Animales , Antígenos Ly/genética , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/patología , Células Epiteliales/patología , Eliminación de Gen , Quinasa I-kappa B/genética , Quinasa I-kappa B/metabolismo , Selectina L/genética , Selectina L/metabolismo , Macrófagos/patología , Ratones , Ratones Transgénicos , FN-kappa B/antagonistas & inhibidores , FN-kappa B/genética , Regulación hacia Arriba
4.
Am J Physiol Gastrointest Liver Physiol ; 317(1): G57-G66, 2019 07 01.
Artículo en Inglés | MEDLINE | ID: mdl-31125264

RESUMEN

Prenatal inflammation is a risk factor for necrotizing enterocolitis (NEC), and it increases intestinal injury in a rat NEC model. We previously showed that maldevelopment of the intestinal microvasculature and lack of vascular endothelial growth factor (VEGF) receptor 2 (VEGFR2) signaling play a role in experimental NEC. However, whether prenatal inflammation affects the intestinal microvasculature remains unknown. In this study, mouse dams were injected intraperitoneally with lipopolysaccharide (LPS) or saline at embryonic day 17. Neonatal intestinal microvasculature density, endothelial cell proliferation, and intestinal VEGF-A and VEGFR2 proteins were assessed in vivo. Maternal and fetal serum TNF concentrations were measured by ELISA. The impact of TNF on the neonatal intestinal microvasculature was examined in vitro and in vivo, and we determined whether prenatal LPS injection exacerbates experimental NEC via TNF. Here we found that prenatal LPS injection significantly decreased intestinal microvascular density, endothelial cell proliferation, and VEGF and VEGFR2 protein expression in neonatal mice. Prenatal LPS injection increased maternal and fetal serum levels of TNF. TNF decreased VEGFR2 protein in vitro in neonatal endothelial cells. Postnatal TNF administration in vivo decreased intestinal microvasculature density, endothelial cell proliferation, and VEGF and VEGFR2 protein expression and increased the incidence of severe NEC. These effects were ameliorated by stabilizing hypoxia-inducible factor-1α, the master regulator of VEGF. Furthermore, prenatal LPS injection significantly increased the incidence of severe NEC in our model, and the effect was dependent on endogenous TNF. Our study suggests that prenatal inflammation increases the susceptibility to NEC, downregulates intestinal VEGFR2 signaling, and affects perinatal intestinal microvascular development via a TNF mechanism. NEW & NOTEWORTHY This report provides new evidence that maternal inflammation decreases neonatal intestinal VEGF receptor 2 signaling and endothelial cell proliferation, impairs intestinal microvascular development, and predisposes neonatal mouse pups to necrotizing enterocolitis (NEC) through inflammatory cytokines such as TNF. Our data suggest that alteration of intestinal microvascular development may be a key mechanism by which premature infants exposed to prenatal inflammation are at risk for NEC and preserving the VEGF/VEGF receptor 2 signaling pathway may help prevent NEC development.


Asunto(s)
Enterocolitis Necrotizante/metabolismo , Inflamación/metabolismo , Intestino Delgado/irrigación sanguínea , Microvasos/metabolismo , Neovascularización Fisiológica , Efectos Tardíos de la Exposición Prenatal , Factor de Necrosis Tumoral alfa/metabolismo , Animales , Permeabilidad Capilar , Proliferación Celular , Células Cultivadas , Modelos Animales de Enfermedad , Células Endoteliales/metabolismo , Células Endoteliales/patología , Enterocolitis Necrotizante/etiología , Enterocolitis Necrotizante/patología , Enterocolitis Necrotizante/fisiopatología , Femenino , Edad Gestacional , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Inflamación/complicaciones , Inflamación/patología , Inflamación/fisiopatología , Lipopolisacáridos , Ratones Endogámicos C57BL , Microvasos/patología , Microvasos/fisiopatología , Embarazo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
5.
Pediatr Res ; 83(2): 545-553, 2018 02.
Artículo en Inglés | MEDLINE | ID: mdl-29068435

RESUMEN

BackgroundNecrotizing enterocolitis (NEC) is a devastating neonatal disease characterized by intestinal necrosis. Hypoxia-inducible factor-1α (HIF-1α) has a critical role in cellular oxygen homeostasis. Here, we hypothesized that prolyl hydroxylase (PHD) inhibition, which stabilizes HIF-1α, protects against NEC by promoting intestinal endothelial cell proliferation and improving intestinal microvascular integrity via vascular endothelial growth factor (VEGF) signaling.MethodsTo assess the role of PHD inhibition in a neonatal mouse NEC model, we administered dimethyloxalylglycine (DMOG) or vehicle to pups before or during the NEC protocol, and determined mortality and incidence of severe intestinal injury. We assessed intestinal VEGF by western blot analysis and quantified endothelial cell and epithelial cell proliferation following immunofluorescence.ResultsDMOG decreased mortality and incidence of severe NEC, increased intestinal VEGF expression, and increased intestinal villus endothelial and epithelial cell proliferation in experimental NEC. Inhibiting VEGFR2 signaling eliminated DMOG's protective effect on intestinal injury severity, survival, and endothelial cell proliferation while sparing DMOG's protective effect on intestinal epithelial cell proliferation.ConclusionDMOG upregulates intestinal VEGF, promotes endothelial cell proliferation, and protects against intestinal injury and mortality in experimental NEC in a VEGFR2 dependent manner. DMOG's protective effect on the neonatal intestinal mucosa may be mediated via VEGFR2 dependent improvement of the intestinal microvasculature.


Asunto(s)
Aminoácidos Dicarboxílicos/farmacología , Enterocolitis Necrotizante/patología , Intestinos/patología , Microcirculación , Animales , Animales Recién Nacidos , Proliferación Celular , Modelos Animales de Enfermedad , Células Endoteliales/citología , Subunidad alfa del Factor 1 Inducible por Hipoxia/metabolismo , Intestinos/irrigación sanguínea , Intestinos/lesiones , Ratones , Ratones Endogámicos C57BL , Prolil Hidroxilasas/metabolismo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
6.
Am J Pathol ; 186(7): 1837-1846, 2016 07.
Artículo en Inglés | MEDLINE | ID: mdl-27157990

RESUMEN

Intestinal adaptation to small-bowel resection (SBR) after necrotizing enterocolitis expands absorptive surface areas and promotes enteral autonomy. Survivin increases proliferation and blunts apoptosis. The current study examines survivin in intestinal epithelial cells after ileocecal resection. Wild-type and epithelial Pik3r1 (p85α)-deficient mice underwent sham surgery or 30% resection. RNA and protein were isolated from small bowel to determine levels of ß-catenin target gene expression, activated caspase-3, survivin, p85α, and Trp53. Healthy and post-resection human infant small-bowel sections were analyzed for survivin, Ki-67, and TP53 by immunohistochemistry. Five days after ileocecal resection, epithelial levels of survivin increased relative to sham-operated on mice, which correlated with reduced cleaved caspase-3, p85α, and Trp53. At baseline, p85α-deficient intestinal epithelial cells had less Trp53 and more survivin, and relative responses to resection were blunted compared with wild-type. In infant small bowel, survivin in transit amplifying cells increased 71% after SBR. Resection increased proliferation and decreased numbers of TP53-positive epithelial cells. Data suggest that ileocecal resection reduces p85α, which lowers TP53 activation and releases survivin promoter repression. The subsequent increase in survivin among transit amplifying cells promotes epithelial cell proliferation and lengthens crypts. These findings suggest that SBR reduces p85α and TP53, which increases survivin and intestinal epithelial cell expansion during therapeutic adaptation in patients with short bowel syndrome.


Asunto(s)
Adaptación Fisiológica/fisiología , Proteínas Inhibidoras de la Apoptosis/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Síndrome del Intestino Corto/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Western Blotting , Fosfatidilinositol 3-Quinasa Clase Ia , Procedimientos Quirúrgicos del Sistema Digestivo/efectos adversos , Modelos Animales de Enfermedad , Enterocolitis Necrotizante/cirugía , Proteínas de la Matriz Extracelular/metabolismo , Regulación de la Expresión Génica , Humanos , Inmunohistoquímica , Lactante , Recién Nacido , Proteínas Inhibidoras de la Apoptosis/biosíntesis , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Proteínas Represoras/biosíntesis , Síndrome del Intestino Corto/etiología , Survivin
7.
Am J Physiol Gastrointest Liver Physiol ; 310(9): G716-25, 2016 05 01.
Artículo en Inglés | MEDLINE | ID: mdl-26950855

RESUMEN

The pathogenesis of necrotizing enterocolitis (NEC), a common gastrointestinal disease affecting premature infants, remains poorly understood. We previously found that intestinal VEGF-A expression is decreased in human NEC samples and in a neonatal mouse NEC model prior to detectable histological injury. Therefore, we hypothesized that lack of VEGF receptor 2 (VEGFR2) signaling facilitates neonatal intestinal injury by impairing intestinal microvasculature development. Here, we found that intestinal VEGF-A and its receptor, VEGFR2, were highly expressed at the end of fetal life and significantly decreased after birth in mice. Furthermore, selective inhibition of VEGFR2 kinase activity and exposure to a neonatal NEC protocol significantly decreased the density of the intestinal microvascular network, which was further reduced when both interventions were provided together. Furthermore, VEGFR2 inhibition resulted in greater mortality and incidence of severe injury in pups submitted to the NEC model. The percentage of lamina propria endothelial cells was decreased during NEC induction, and further decreased when VEGFR2 signaling was inhibited. This was associated with decreased endothelial cell proliferation rather than apoptosis. In conclusion, we found that VEGF-A and VEGFR2 proteins are highly expressed in the intestine before birth, and are significantly downregulated in the immediate neonatal period. Furthermore, VEGFR2 signaling is necessary to maintain the integrity of the intestinal mucosal microvasculature during the postnatal period and lack of VEGFR2 signaling predisposes to NEC in neonatal mice.


Asunto(s)
Enterocolitis Necrotizante/metabolismo , Mucosa Intestinal/metabolismo , Microvasos/patología , Receptor 2 de Factores de Crecimiento Endotelial Vascular/genética , Animales , Células Cultivadas , Enterocolitis Necrotizante/genética , Enterocolitis Necrotizante/patología , Mucosa Intestinal/irrigación sanguínea , Mucosa Intestinal/crecimiento & desarrollo , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Microvasos/crecimiento & desarrollo , Transducción de Señal , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/metabolismo
8.
Proc Natl Acad Sci U S A ; 108 Suppl 1: 4623-30, 2011 Mar 15.
Artículo en Inglés | MEDLINE | ID: mdl-21282652

RESUMEN

Imbalance in the regulatory immune mechanisms that control intestinal cellular and bacterial homeostasis may lead to induction of the detrimental inflammatory signals characterized in humans as inflammatory bowel disease. Induction of proinflammatory cytokines (i.e., IL-12) induced by dendritic cells (DCs) expressing pattern recognition receptors may skew naive T cells to T helper 1 polarization, which is strongly implicated in mucosal autoimmunity. Recent studies show the ability of probiotic microbes to treat and prevent numerous intestinal disorders, including Clostridium difficile-induced colitis. To study the molecular mechanisms involved in the induction and repression of intestinal inflammation, the phosphoglycerol transferase gene that plays a key role in lipoteichoic acid (LTA) biosynthesis in Lactobacillus acidophilus NCFM (NCK56) was deleted. The data show that the L. acidophilus LTA-negative in LTA (NCK2025) not only down-regulated IL-12 and TNFα but also significantly enhanced IL-10 in DCs and controlled the regulation of costimulatory DC functions, resulting in their inability to induce CD4(+) T-cell activation. Moreover, treatment of mice with NCK2025 compared with NCK56 significantly mitigated dextran sulfate sodium and CD4(+)CD45RB(high)T cell-induced colitis and effectively ameliorated dextran sulfate sodium-established colitis through a mechanism that involves IL-10 and CD4(+)FoxP3(+) T regulatory cells to dampen exaggerated mucosal inflammation. Directed alteration of cell surface components of L. acidophilus NCFM establishes a potential strategy for the treatment of inflammatory intestinal disorders.


Asunto(s)
Autoinmunidad/inmunología , Colitis/inmunología , Colitis/microbiología , Regulación de la Expresión Génica/inmunología , Lactobacillus acidophilus/metabolismo , Lipopolisacáridos/deficiencia , Animales , Linfocitos T CD4-Positivos/inmunología , Colitis/inducido químicamente , Cartilla de ADN/genética , Sulfato de Dextran/toxicidad , Citometría de Flujo , Técnica del Anticuerpo Fluorescente , Eliminación de Gen , Proteínas de Homeodominio/genética , Interleucina-10/genética , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Factor 88 de Diferenciación Mieloide/genética , Reacción en Cadena de la Polimerasa , Ácidos Teicoicos , Transferasas (Grupos de Otros Fosfatos Sustitutos)/genética
9.
Am J Pathol ; 181(4): 1306-15, 2012 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-22863952

RESUMEN

Chronic ulcerative colitis (CUC) is characterized by increased intestinal epithelial cell (IEC) apoptosis associated with elevated tumor necrosis factor (TNF), inducible nitric oxide synthase (iNOS), and p53. We previously showed that p53 is increased in crypt IECs in human colitis and is needed for IEC apoptosis in chronic dextran sulfate sodium-colitis. Herein, we examined the roles of TNF and iNOS in regulating p53-induced IEC apoptosis in CUC. The IEC TUNEL staining, caspases 3, 8, and 9, and p53 protein levels, induced by anti-CD3 monoclonal antibody (mAb) activation of T cells, were markedly reduced in TNF receptor 1 and 2 gene knockout mice. Induction of IEC apoptosis correlated with increased p53, which was attenuated in iNOS(-/-) mice. IEC p53 levels and apoptosis were reduced in IL-10(-/-) colitic mice treated with neutralizing TNF mAb and the iNOS inhibitor, aminoguanidine, further suggesting that TNF and iNOS are upstream of p53 during colitis-induced IEC apoptosis. IEC apoptosis and p53 levels were assessed in control versus untreated or anti-TNF-treated CUC patients with equivalent levels of inflammation. Data indicated that IEC apoptosis and p53 levels were clearly higher in untreated CUC but markedly reduced in patients treated with anti-TNF mAb. Therefore, TNF-induced iNOS activates a p53-dependent pathway of IEC apoptosis in CUC. The inhibition of IEC apoptosis may be an important mechanism for mucosal healing in anti-TNF-treated CUC patients.


Asunto(s)
Apoptosis , Células Epiteliales/metabolismo , Células Epiteliales/patología , Enfermedades Inflamatorias del Intestino/metabolismo , Enfermedades Inflamatorias del Intestino/patología , Factor de Necrosis Tumoral alfa/metabolismo , Proteína p53 Supresora de Tumor/metabolismo , Animales , Colitis/patología , Enterocitos/enzimología , Enterocitos/patología , Células Epiteliales/inmunología , Humanos , Enfermedades Inflamatorias del Intestino/inmunología , Ratones , Ratones Endogámicos C57BL , Óxido Nítrico Sintasa de Tipo II/metabolismo , Receptores Tipo I de Factores de Necrosis Tumoral/deficiencia , Receptores Tipo I de Factores de Necrosis Tumoral/metabolismo , Receptores Tipo II del Factor de Necrosis Tumoral/deficiencia , Receptores Tipo II del Factor de Necrosis Tumoral/metabolismo , Transducción de Señal , Linfocitos T/inmunología
10.
Gastroenterology ; 141(3): 1036-45, 2011 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-21699775

RESUMEN

BACKGROUND & AIMS: Inflammatory bowel disease (IBD) is associated with increased apoptosis of intestinal epithelial cells (IECs). Mutations in the tumor suppressor p53 appear during early stages of progression from colitis to cancer. We investigated the role of p53 and its target, p53-upregulated modulator of apoptosis (PUMA), in inflammation-induced apoptosis of IECs. METHODS: Apoptosis was induced in mouse models of mucosal inflammation. Responses of IECs to acute, T-cell activation were assessed in wild-type, p53⁻/⁻, Bid⁻/⁻, Bim⁻/⁻, Bax3⁻/⁻, Bak⁻/⁻, PUMA⁻/⁻, and Noxa⁻/⁻ mice. Responses of IECs to acute and chronic colitis were measured in mice following 1 or 3 cycles of dextran sulfate sodium (DSS), respectively. Apoptosis was assessed by TUNEL staining and measuring activity of caspases 3 and 9; levels of p53 and PUMA were assessed in colon tissue from patients with and without ulcerative colitis. RESULTS: Apoptosis of IECs occurred in the lower crypts of colitic tissue from humans and mice. Colitis induction with anti-CD3 or 3 cycles of DSS increased apoptosis and protein levels of p53 and PUMA in colonic crypt IECs. In p53⁻/⁻ and PUMA⁻/⁻ mice, apoptosis of IECs was significantly reduced but inflammation was not. Levels of p53 and PUMA were increased in inflamed mucosal tissues of mice with colitis and in patients with UC, compared with controls. Induction of PUMA in IECs of p53⁻/⁻ mice indicated that PUMA-mediated apoptosis was independent of p53. CONCLUSIONS: In mice and humans, colon inflammation induces apoptosis of IECs via p53-dependent and - independent mechanisms; PUMA also activates an intrinsic apoptosis pathway associated with colitis.


Asunto(s)
Proteínas Reguladoras de la Apoptosis/fisiología , Apoptosis/fisiología , Colitis/patología , Colitis/fisiopatología , Mucosa Intestinal/patología , Proteínas Proto-Oncogénicas/fisiología , Proteína p53 Supresora de Tumor/fisiología , Proteínas Supresoras de Tumor/fisiología , Animales , Proteínas Reguladoras de la Apoptosis/genética , Estudios de Casos y Controles , Caspasa 3/metabolismo , Caspasa 9/metabolismo , Colitis/inducido químicamente , Colon/metabolismo , Colon/patología , Colon/fisiopatología , Sulfato de Dextran/efectos adversos , Modelos Animales de Enfermedad , Humanos , Mucosa Intestinal/metabolismo , Mucosa Intestinal/fisiopatología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Proteínas Proto-Oncogénicas/genética , Transducción de Señal/fisiología , Linfocitos T/patología , Linfocitos T/fisiología , Proteína p53 Supresora de Tumor/genética , Proteínas Supresoras de Tumor/genética
11.
Newborn (Clarksville) ; 1(1): 32-43, 2022.
Artículo en Inglés | MEDLINE | ID: mdl-35846894

RESUMEN

Necrotizing enterocolitis (NEC) is a major cause of morbidity and mortality in premature infants. NEC is characterized by intestinal tissue inflammation and necrosis. The intestinal barrier is altered in NEC, which potentially contributes to its pathogenesis by promoting intestinal bacterial translocation and stimulating the inflammatory response. In premature infants, many components of the intestinal barrier are immature. This article reviews the different components of the intestinal barrier and how their immaturity contributes to intestinal barrier dysfunction and NEC.

12.
Commun Biol ; 5(1): 320, 2022 04 06.
Artículo en Inglés | MEDLINE | ID: mdl-35388142

RESUMEN

Necrotizing enterocolitis (NEC) is a deadly bowel necrotic disease of premature infants. Low levels of plasma IGF-1 predispose premature infants to NEC. While increasing evidence suggests that defective perinatal intestinal microvascular development plays a role in NEC, the involved mechanism remains incompletely understood. We report here that serum and intestinal IGF-1 are developmentally regulated during the perinatal period in mice and decrease during experimental NEC. Neonatal intestinal macrophages produce IGF-1 and promote endothelial cell sprouting in vitro via IGF-1 signaling. In vivo, in the neonatal intestine, macrophage-derived IGF-1 promotes VEGF expression and endothelial cell proliferation and protects against experimental NEC. Exogenous IGF-1 preserves intestinal microvascular density and protects against experimental NEC. In human NEC tissues, villous endothelial cell proliferation and IGF-1- producing macrophages are decreased compared to controls. Together, our results suggest that defective IGF-1-production by neonatal macrophages impairs neonatal intestinal microvascular development and predisposes the intestine to necrotizing enterocolitis.


Asunto(s)
Enterocolitis Necrotizante , Animales , Enterocolitis Necrotizante/metabolismo , Enterocolitis Necrotizante/prevención & control , Femenino , Humanos , Recién Nacido , Factor I del Crecimiento Similar a la Insulina/metabolismo , Intestinos , Macrófagos/metabolismo , Ratones , Embarazo , Transducción de Señal
13.
Infect Immun ; 79(5): 1863-72, 2011 May.
Artículo en Inglés | MEDLINE | ID: mdl-21343355

RESUMEN

Citrobacter rodentium infection of mice induces cell-mediated immune responses associated with crypt hyperplasia and epithelial ß-catenin signaling. Recent data suggest that phosphatidylinositol-3-kinase (PI3K)/Akt signaling cooperates with Wnt to activate ß-catenin in intestinal stem and progenitor cells through phosphorylation at Ser552 (P-ß-catenin(552)). Our aim was to determine whether epithelial PI3K/Akt activation is required for ß-catenin signaling and host defense against C. rodentium. C57BL/6 mice were infected with C. rodentium and treated with dimethyl sulfoxide (DMSO) (vehicle control) or with the PI3K inhibitor LY294002 or wortmannin. The effects of infection on PI3K activation and ß-catenin signaling were analyzed by immunohistochemistry. The effects of PI3K inhibition on host defense were analyzed by the quantification of splenic and colon bacterial clearance, and adaptive immune responses were measured by real-time PCR (RT-PCR) and enzyme-linked immunosorbent assay (ELISA). Increased numbers of P-ß-catenin(552)-stained epithelial cells were found throughout expanded crypts in C. rodentium colitis. We show that the inhibition of PI3K signaling attenuates epithelial Akt activation, the Ser552 phosphorylation and activation of ß-catenin, and epithelial cell proliferative responses during C. rodentium infection. PI3K inhibition impairs bacterial clearance despite having no impact on mucosal cytokine (gamma interferon [IFN-γ], tumor necrosis factor [TNF], interleukin-17 [IL-17], and IL-1ß) or chemokine (CXCL1, CXCL5, CXCL9, and CXCL10) induction. The results suggest that the host defense against C. rodentium requires epithelial PI3K activation to induce Akt-mediated ß-catenin signaling and the clearance of C. rodentium independent of adaptive immune responses.


Asunto(s)
Citrobacter rodentium/inmunología , Infecciones por Enterobacteriaceae/inmunología , Infecciones por Enterobacteriaceae/metabolismo , Células Epiteliales/metabolismo , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal/inmunología , beta Catenina/metabolismo , Animales , Colitis/inmunología , Colitis/metabolismo , Colitis/patología , Infecciones por Enterobacteriaceae/patología , Activación Enzimática/efectos de los fármacos , Activación Enzimática/inmunología , Inhibidores Enzimáticos/farmacología , Ensayo de Inmunoadsorción Enzimática , Células Epiteliales/inmunología , Células Epiteliales/patología , Técnica del Anticuerpo Fluorescente , Inmunohistoquímica , Ratones , Ratones Endogámicos C57BL , Proteínas Proto-Oncogénicas c-akt/inmunología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Transducción de Señal/efectos de los fármacos
14.
Gastroenterology ; 138(2): 595-605, 605.e1-3, 2010 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-19879273

RESUMEN

BACKGROUND & AIMS: Mesalamine is a mainstay therapeutic agent in chronic ulcerative colitis (CUC) in which condition it reverses crypt architectural changes and reduces colitis-associated cancer (CAC). The present study addressed the possibility that mesalamine reduces beta-catenin-associated progenitor cell activation, Akt-phosphorylated beta-catenin(Ser552) (P-beta-catenin), and colitis-induced dysplasia (CID). METHODS: Effects of mesalamine on P-beta-catenin staining and function were assessed by immunohistochemistry and quantitative reverse-transcription polymerase chain reaction (qRT-PCR) in biopsy specimens of CUC in mild or "refractory" severe mucosal inflammation. Effects of mesalamine on epithelial proliferation and activation of Akt and beta-catenin were assessed in interleukin (IL)-10(-/-) colitis and CID by immunohistochemistry and Western blotting. Dysplasia was assessed by counting the number and lengths of lesions per colon. RESULTS: Data from IL-10(-/-) and human colitis samples show that mesalamine reduced Akt activation and P-beta-catenin levels in the middle and upper crypt. Reductions in P-beta-catenin in CUC biopsy specimens with severe inflammation suggested that mesalamine reduced P-beta-catenin levels in tissue refractory to mesalamine's anti-inflammatory effects. In IL-10(-/-) mice, mesalamine reduced CID concordant with inhibition of crypt Akt and beta-catenin signaling. CONCLUSIONS: The results are consistent with the model that mesalamine contributes to chemoprevention in CAC by reducing beta-catenin signaling within intestinal progenitors.


Asunto(s)
Antiinflamatorios no Esteroideos/farmacología , Colitis Ulcerosa/metabolismo , Colon/metabolismo , Células Epiteliales/metabolismo , Mesalamina/farmacología , beta Catenina/metabolismo , Animales , Biopsia , Proliferación Celular/efectos de los fármacos , Colitis Ulcerosa/patología , Colon/efectos de los fármacos , Colon/patología , Modelos Animales de Enfermedad , Progresión de la Enfermedad , Células Epiteliales/efectos de los fármacos , Células Epiteliales/patología , Humanos , Interleucina-10/genética , Interleucina-10/metabolismo , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados
15.
Gastroenterology ; 139(3): 869-81, 881.e1-9, 2010 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-20580720

RESUMEN

BACKGROUND & AIMS: Mechanisms responsible for crypt architectural distortion in chronic ulcerative colitis (CUC) are not well understood. Data indicate that serine/threonine protein kinase Akt (Akt) signaling cooperates with Wingless (Wnt) to activate beta-catenin in intestinal stem and progenitor cells through phosphorylation at Ser552 (P-beta-catenin(552)). We investigated whether phosphoinositide 3-kinase (PI3K) is required for Akt-mediated activation of beta-catenin during intestinal inflammation. METHODS: The class IA subunit of PI3K was conditionally deleted from intestinal epithelial cells in mice named I-pik3r1KO. Acute inflammation was induced in mice and intestines were analyzed by biochemical and histologic methods. The effects of chemically blocking PI3K in colitic interleukin-10(-/-) mice were examined. Biopsy samples from patients were examined. RESULTS: Compared with wild-type, I-pik3r1KO mice had reduced T-cell-mediated Akt and beta-catenin signaling in intestinal stem and progenitor cells and limited crypt epithelial proliferation. Biochemical analyses indicated that PI3K-Akt signaling increased nuclear total beta-catenin and P-beta-catenin(552) levels and reduced N-terminal beta-catenin phosphorylation, which is associated with degradation. PI3K inhibition in interleukin-10(-/-) mice impaired colitis-induced epithelial Akt and beta-catenin activation, reduced progenitor cell expansion, and prevented dysplasia. Human samples had increased numbers of progenitor cells with P-beta-catenin(552) throughout expanded crypts and increased messenger RNA expression of beta-catenin target genes in CUC, colitis-associated cancer, tubular adenomas, and sporadic colorectal cancer, compared with control samples. CONCLUSIONS: PI3K-Akt signaling cooperates with Wnt to increase beta-catenin signaling during inflammation. PI3K-induced and Akt-mediated beta-catenin signaling are required for progenitor cell activation during the progression from CUC to CAC; these factors might be used as biomarkers of dysplastic transformation in the colon.


Asunto(s)
Colitis/enzimología , Colon/enzimología , Mucosa Intestinal/enzimología , Fosfatidilinositol 3-Quinasas/metabolismo , Transducción de Señal , Células Madre/enzimología , beta Catenina/metabolismo , Animales , Biopsia , Proliferación Celular , Colitis/complicaciones , Colitis/genética , Colitis/inmunología , Colitis/patología , Colon/efectos de los fármacos , Colon/inmunología , Colon/patología , Neoplasias del Colon/enzimología , Neoplasias del Colon/patología , Colonoscopía , Modelos Animales de Enfermedad , Humanos , Interleucina-10/deficiencia , Interleucina-10/genética , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/inmunología , Mucosa Intestinal/patología , Ratones , Ratones Endogámicos C57BL , Ratones Noqueados , Fosfatidilinositol 3-Quinasas/deficiencia , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación , Inhibidores de Proteínas Quinasas/farmacología , Proteínas Proto-Oncogénicas c-akt/metabolismo , Transducción de Señal/efectos de los fármacos , Células Madre/efectos de los fármacos , Células Madre/inmunología , Células Madre/patología , Linfocitos T/enzimología , Linfocitos T/inmunología , Factores de Tiempo , Proteínas Wnt/metabolismo
16.
Neonatology ; 107(3): 191-8, 2015.
Artículo en Inglés | MEDLINE | ID: mdl-25659996

RESUMEN

BACKGROUND: Decreased intestinal perfusion may contribute to the development of necrotizing enterocolitis (NEC). Vascular endothelial growth factor (VEGF) is an angiogenic protein necessary for the development and maintenance of capillary networks. Whether VEGF is dysregulated in NEC remains unknown. OBJECTIVES: The objective of this study was to determine whether intestinal VEGF expression is altered in a neonatal mouse model of NEC and in human NEC patients. METHODS: We first assessed changes of intestinal VEGF mRNA and protein in a neonatal mouse NEC model before significant injury occurs. We then examined whether exposure to formula feeding, bacterial inoculation, cold stress and/or intermittent hypoxia affected intestinal VEGF expression. Last, we visualized VEGF protein in intestinal tissues of murine and human NEC and control cases by immunohistochemistry. RESULTS: Intestinal VEGF protein and mRNA were significantly decreased in pups exposed to the NEC protocol compared to controls. Hypoxia, cold stress and commensal bacteria, when administered together, significantly downregulated intestinal VEGF expression, while they had no significant effect when given alone. VEGF was localized to a few single intestinal epithelial cells and some cells of the lamina propria and myenteric plexus. VEGF staining was decreased in murine and human NEC intestines when compared to control tissues. CONCLUSION: Intestinal VEGF protein is reduced in human and experimental NEC. Decreased VEGF production might contribute to NEC pathogenesis.


Asunto(s)
Enterocolitis Necrotizante/genética , Mucosa Intestinal/patología , Factor A de Crecimiento Endotelial Vascular/metabolismo , Animales , Animales Recién Nacidos , Respuesta al Choque por Frío , Modelos Animales de Enfermedad , Regulación hacia Abajo , Humanos , Hipoxia/fisiopatología , Inmunohistoquímica , Lactante , Recién Nacido , Mucosa Intestinal/metabolismo , Ratones , Ratones Endogámicos C57BL , ARN Mensajero/genética , Factor A de Crecimiento Endotelial Vascular/genética
17.
Curr HIV Res ; 2(4): 395-401, 2004 Oct.
Artículo en Inglés | MEDLINE | ID: mdl-15544459

RESUMEN

Much attention has been focused on CD4+ memory/primed T cells and monocytes/macrophages as the predominant targets of HIV infection. Considerable evidence, however, indicates that cell types that are classically not permissive to HIV infection, can be a potent source of productive virus replication in response to immune modulation. This immune modulation can be in the form of cytokine stimulation pre- or post-infection or by activating these "non-conventional" targets. In this review, we will provide evidence for immune modulation of HIV infection with special emphasis on immune modulation of HIV replication in classically non-HIV permissive primary cells of both lymphoid and non-lymphoid origins. We will also examine the impact of HIV infection of these non-conventional targets as they pertain to both HIV immuno- and neuro-pathogenesis.


Asunto(s)
Astrocitos/virología , Citocinas/inmunología , Infecciones por VIH/fisiopatología , VIH-1/patogenicidad , Replicación Viral/inmunología , Complejo SIDA Demencia/inmunología , Complejo SIDA Demencia/fisiopatología , Complejo SIDA Demencia/virología , Adulto , Línea Celular , Niño , Preescolar , Infecciones por VIH/inmunología , Infecciones por VIH/virología , VIH-1/fisiología , Humanos , Linfocitos/virología , Replicación Viral/fisiología
18.
Inflamm Bowel Dis ; 19(10): 2051-60, 2013 Sep.
Artículo en Inglés | MEDLINE | ID: mdl-23867870

RESUMEN

BACKGROUND: Mesalamine, 5-aminosalicylic acid (5-ASA), is a potent antioxidant and is known to enhance peroxisome proliferator-activated receptor γ activity in the intestine. Our previous studies suggested reduced Phosphoinositide 3-Kinase (PI3K)/ß-catenin signaling as a mechanism for 5-ASA chemoprevention in chronic ulcerative colitis (CUC). We now hypothesize that 5-ASA mediates changes in intestinal epithelial cell (IEC) reactive oxygen species during colitis to affect phosphatase and tensin homolog (PTEN), PI3K, and ß-catenin signaling. METHODS: Here, we examined effects of 5-ASA on oxidant-induced cell signaling pathways in HT-29 cells, IECs from mice, and biopsy tissue from control and CUC patients. Samples were selected to control for inflammation between untreated and 5-ASA-treated CUC patients. RESULTS: Direct evaluation of IEC in H2O2-stimulated whole colonic crypts indicated that 5-ASA reduces reactive oxygen species levels in lower crypt IECs where long-lived progenitor cells reside. Analysis of biopsies from patient samples revealed that 5-ASA increases expression of the antioxidant catalase in CUC patients. Also, 5-ASA increased nuclear peroxisome proliferator-activated receptor γ protein and target gene expression. Data showed 5-ASA-induced peroxisome proliferator-activated receptor γ DNA binding to the PTEN promoter (chromatin immunoprecipitation) and reduced both phosphorylated and oxidized (inactive) PTEN protein levels. Analysis of patient samples revealed 5-ASA that also reduced levels of active phosphorylated Akt in inflamed colitis tissue. Reduced PI3K/Akt signaling and expression of ß-catenin target genes in 5-ASA-treated CUC patients additionally suggests enhanced PTEN activity as well. CONCLUSIONS: Therefore, 5-ASA reduces CUC-induced reactive oxygen species in colonic progenitor cells and enhances PTEN activity, thus attenuating PI3K/Akt signaling. These data suggest that the antioxidant properties of 5-ASA may be the predominant mechanism for 5-ASA chemoprevention.


Asunto(s)
Antiinflamatorios no Esteroideos/uso terapéutico , Antioxidantes/uso terapéutico , Colitis Ulcerosa/complicaciones , Neoplasias del Colon/tratamiento farmacológico , Mesalamina/uso terapéutico , Fosfatidilinositol 3-Quinasas/metabolismo , Células Madre/efectos de los fármacos , Adolescente , Animales , Western Blotting , Proliferación Celular/efectos de los fármacos , Inmunoprecipitación de Cromatina , Enfermedad Crónica , Colitis Ulcerosa/tratamiento farmacológico , Colitis Ulcerosa/metabolismo , Neoplasias del Colon/etiología , Neoplasias del Colon/metabolismo , Citometría de Flujo , Humanos , Peróxido de Hidrógeno/farmacología , Mucosa Intestinal/citología , Mucosa Intestinal/efectos de los fármacos , Mucosa Intestinal/metabolismo , Captura por Microdisección con Láser , Ratones , Ratones Endogámicos C57BL , Oxidantes/farmacología , PPAR gamma/genética , PPAR gamma/metabolismo , Fosfohidrolasa PTEN/genética , Fosfohidrolasa PTEN/metabolismo , Fosfatidilinositol 3-Quinasas/genética , Inhibidores de las Quinasa Fosfoinosítidos-3 , Fosforilación/efectos de los fármacos , Proteínas Proto-Oncogénicas c-akt/genética , Proteínas Proto-Oncogénicas c-akt/metabolismo , ARN Mensajero/genética , Reacción en Cadena en Tiempo Real de la Polimerasa , Reacción en Cadena de la Polimerasa de Transcriptasa Inversa , Células Madre/metabolismo , Células Madre/patología , beta Catenina/genética , beta Catenina/metabolismo
19.
Inflamm Bowel Dis ; 18(2): 323-32, 2012 Feb.
Artículo en Inglés | MEDLINE | ID: mdl-22009715

RESUMEN

BACKGROUND: Activated effector T cells contribute to tissue injury observed in inflammatory bowel disease. T cells are recruited to effector sites after activation in peripheral lymph nodes directs expression of tissue-specific homing receptors. One such mechanism for effector T cell recruitment employs activation-induced fucosylation of P-selectin glycoprotein ligand (PSGL)-1 that mediates binding to endothelial P-selectin. Here we examine the differential role of PSGL-1 in recruiting effector T-cell subsets in colitis. METHODS: C57BL/6 wildtype and PSGL-1 mice received 2.5% dextran sodium sulfate (DSS) for 6 days and were euthanized 7 and 14 days after the initiation of DSS. Disease activity was monitored throughout. Histologic colitis scores, colonic CD4+ accumulation, and cytokine production were assessed at days 7 and 14. Recruitment of T-helper (Th) subsets was assessed by enumerating adoptively transferred Th1 or Th17 CD4+ cells 2 days after transfer to DSS-treated mice. RESULTS: DSS colitis increases CD4+ T cells in colonic tissue and induces Th1 (interferon gamma [IFN-γ], tumor necrosis factor [TNF]) and Th17 (interleukin [IL]-17, IL-22) cytokines. Loss of PSGL-1 attenuates DSS colitis, decreases colonic CD4+ T cell numbers, and reduces both Th1 and Th17 cytokine production. Colitis increases recruitment of Th1 (19-fold) and Th17 (2.5-fold) cells. PSGL-1 deficiency in transferred T cells abrogates colonic recruitment of Th1 cells in DSS colitis, whereas Th17 recruitment is unaffected. CONCLUSIONS: PSGL-1 selectively controls Th1 recruitment in colitis. Whereas Th17 recruitment is independent of PSGL-1, generation of colonic Th17 cytokine requires initial Th1 recruitment. Therefore, attenuating PSGL-1 binding may prevent colonic recruitment of disease-causing Th1 cells that promote local Th17 generation.


Asunto(s)
Movimiento Celular/fisiología , Colitis/inmunología , Glicoproteínas de Membrana/fisiología , Células TH1/inmunología , Células Th17/inmunología , Animales , Recuento de Células , Colitis/inducido químicamente , Citocinas/metabolismo , Sulfato de Dextran/toxicidad , Ensayo de Inmunoadsorción Enzimática , Citometría de Flujo , Ratones , Ratones Endogámicos C57BL , Reacción en Cadena en Tiempo Real de la Polimerasa , Índice de Severidad de la Enfermedad
20.
Virology ; 350(2): 443-52, 2006 Jul 05.
Artículo en Inglés | MEDLINE | ID: mdl-16542695

RESUMEN

Interleukin (IL)-7 plays several roles critical to T cell maturation, survival, and homeostasis. Because of these functions, IL-7 is under investigation as an immune-modulator for therapeutic use in lymphopenic clinical conditions, including HIV. We reported that naive T cells, typically not permissive to HIV, can be productively infected when pre-treated with IL-7. We evaluated the mechanism by which IL-7-mediates this effect. IL-7 potently up-regulated the transcriptional factor NFAT, but had no effect on NFkappaB. Blocking NFAT activity using a number of reagents, such as Cyclosporin A, FK-506, or the NFAT-specific inhibitor known as VIVIT peptide, all markedly reduced IL-7-mediated induction of HIV replication in naive T cells. Additional neutralization of cytokines present in IL-7-treated cultures and/or those that have NFAT-binding sequences within their promotors indicated that IL-10, IL-4, and most significantly IFNgamma, all contribute to IL-7-induction of HIV productive replication in naive T cells. These data clarify the mechanism by which IL-7 can overcome the block to HIV productive infection in naive T cells, despite their quiescent cell status. These findings are relevant to the treatment of HIV disease and understanding HIV pathogenesis in the naive CD4+ T cell compartment, especially in light of the vigorous pursuit of IL-7 as an in vivo immune modulator.


Asunto(s)
VIH/fisiología , Interleucina-7/farmacología , Activación de Linfocitos , Factores de Transcripción NFATC/metabolismo , Linfocitos T/virología , Replicación Viral/efectos de los fármacos , Antígenos CD/análisis , Linfocitos T CD4-Positivos/inmunología , Linfocitos T CD4-Positivos/virología , VIH/efectos de los fármacos , Humanos , Valores de Referencia , Linfocitos T/inmunología , Replicación Viral/inmunología
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA