Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 21
Filtrar
Mais filtros










Base de dados
Intervalo de ano de publicação
1.
Bio Protoc ; 14(10): e4993, 2024 May 20.
Artigo em Inglês | MEDLINE | ID: mdl-38798982

RESUMO

Anemia is a common and serious health problem, nearly universally diagnosed in preterm infants, and is associated with increased morbidity and mortality worldwide. Red blood cell (RBC) transfusion is a lifesaving and mainstay therapy; however, it has critical adverse effects. One consequence is necrotizing enterocolitis (NEC), an inflammatory bowel necrosis disease in preterm infants. The murine model of phlebotomy-induced anemia and RBC transfusion-associated NEC enables a detailed study of the molecular mechanisms underlying these morbidities and the evaluation of potential new therapeutic strategies. This protocol describes a detailed procedure for obtaining murine pups with phlebotomy-induced anemia and delivering an RBC transfusion that develops NEC.

2.
Pediatr Res ; 92(6): 1575-1579, 2022 12.
Artigo em Inglês | MEDLINE | ID: mdl-35322186

RESUMO

BACKGROUND: Anemia is a nearly universal diagnosis in preterm infants, caused by phlebotomy, and exacerbated by the underlying erythropoietic immaturity. Newborn infants are exposed to the unique stressor of fetal-to-neonatal transition, which requires significant adaptation ex utero. Accordingly, the preterm infant's response to anemia may alter the ability to confront underlying illness. This study utilized our preclinical mouse model of phlebotomy-induced anemia (PIA) to comprehensively investigate associated hematological changes. METHODS: C57BL/6 mice were subjected to timed phlebotomy between postnatal days 2--10 to induce severe anemia. Complete blood counts were determined by the Sysmex XT-2000iV analyzer. RESULTS: Anemic pups showed a gradual reduction of RBC and hemoglobin (Hb) and increased reticulocyte (RET) counts and red cell distribution width (RDW), however, with reduced RET-Hb from postnatal day (P) of 4 onwards. Elevated levels of high fluorescent RET and immature reticulocyte fraction (IRF) were noted in anemic mouse pups, but low and medium fluorescent RET were reduced. Also, the reduction of mean corpuscular volume (MCV), mean corpuscular hemoglobin (MCH), and mean corpuscular hemoglobin concentration (MCHC) were noted in anemic pups. No changes were seen in lymphocytes, but monocytes and neutrophils were significantly elevated from P4-P6. CONCLUSIONS: PIA in mouse pups is associated with hematological changes that may be exacerbating factors in neonatal diseases. IMPACT: Anemia is common and often severe in premature infants. Investigation of hematological parameters in settings of preclinical anemia may be an index of therapeutic strategies. Preclinical model evaluating the effects of neonatal anemia on the remainder of complete blood count. Detailed time kinetic phlebotomy-induced anemic mice enable us to study the impact on developmental delays in erythropoiesis and possible strategic intervention. Hematological effects of severe anemia in mice might provide insight on how best to investigate anemia in preterm infants.


Assuntos
Anemia , Flebotomia , Recém-Nascido , Humanos , Animais , Camundongos , Animais Recém-Nascidos , Flebotomia/efeitos adversos , Recém-Nascido Prematuro , Camundongos Endogâmicos C57BL , Anemia/complicações
3.
J Biol Chem ; 298(3): 101621, 2022 03.
Artigo em Inglês | MEDLINE | ID: mdl-35074429

RESUMO

Inhibition of P300 acetyltransferase activity by specific inhibitor C646 has been shown to improve insulin signaling. However, the underlying molecular mechanism of this improvement remains unclear. In this study, we analyzed P300 levels of obese patients and found that they were significantly increased in liver hepatocytes. In addition, large amounts of P300 appeared in the cytoplasm. Inhibition of P300 acetyltransferase activity by C646 drastically increased tyrosine phosphorylation of the insulin receptor protein substrates (IRS1/2) without affecting the tyrosine phosphorylation of the beta subunit of the insulin receptor (IRß) in hepatocytes in the absence of insulin. Since IRS1/2 requires membrane translocation and binding to inositol compounds for normal functions, we also examined the role of acetylation on binding to phosphatidylinositol(4,5)P2 and found that IRS1/2 acetylation by P300 reduced this binding. In contrast, we show that inhibition of IRS1/2 acetylation by C646 facilitates IRS1/2 membrane translocation. Intriguingly, we demonstrate that C646 activates IRß's tyrosine kinase activity and directly promotes IRß interaction with IRS1/2, leading to the tyrosine phosphorylation of IRS1/2 and subsequent activation of insulin signaling even in the absence of insulin. In conclusion, these data reveal the unique effects of C646 in activating insulin signaling in patients with obesity and diabetes.


Assuntos
Benzoatos , Inibidores Enzimáticos , Proteínas Substratos do Receptor de Insulina , Nitrobenzenos , Pirazolonas , Receptor de Insulina , Fatores de Transcrição de p300-CBP , Benzoatos/farmacologia , Inibidores Enzimáticos/farmacologia , Humanos , Insulina/metabolismo , Proteínas Substratos do Receptor de Insulina/genética , Proteínas Substratos do Receptor de Insulina/metabolismo , Nitrobenzenos/farmacologia , Fosforilação , Pirazolonas/farmacologia , Receptor de Insulina/metabolismo , Tirosina/metabolismo , Fatores de Transcrição de p300-CBP/antagonistas & inibidores , Fatores de Transcrição de p300-CBP/metabolismo
4.
ACS Omega ; 6(46): 31312-31327, 2021 Nov 23.
Artigo em Inglês | MEDLINE | ID: mdl-34805715

RESUMO

The emergence of a variety of highly transmissible SARS-CoV-2 variants, the causative agent of COVID-19, with multiple spike mutations poses serious challenges in overcoming the ongoing deadly pandemic. It is, therefore, essential to understand how these variants gain enhanced ability to evade immune responses with a higher rate of spreading infection. To address this question, here we have individually assessed the effects of SARS-CoV-2 variant-specific spike (S) protein receptor-binding domain (RBD) mutations E484K, K417N, L452Q, L452R, N501Y, and T478K that characterize and differentiate several emerging variants. Despite the hundreds of apparently neutral mutations observed in the domains other than the RBD, we have shown that each RBD mutation site is differentially engaged in an interdomain allosteric network involving mutation sites from a distant domain, affecting interactions with the human receptor angiotensin-converting enzyme-2 (ACE2). This allosteric network couples the residues of the N-terminal domain (NTD) and the RBD, which are modulated by the RBD-specific mutations and are capable of propagating mutation-induced perturbations between these domains through a combination of structural changes and effector-dependent modulations of dynamics. One key feature of this network is the inclusion of compensatory mutations segregated into three characteristically different clusters, where each cluster residue site is allosterically coupled with specific RBD mutation sites. Notably, each RBD mutation acted like a positive allosteric modulator; nevertheless, K417N was shown to have the largest effects among all of the mutations on the allostery and thereby holds the highest binding affinity with ACE2. This result will be useful for designing the targeted control measure and therapeutic efforts aiming at allosteric modulators.

5.
Proc Natl Acad Sci U S A ; 117(20): 10958-10969, 2020 05 19.
Artigo em Inglês | MEDLINE | ID: mdl-32366656

RESUMO

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants and an orphan disease with no specific treatment. Most patients with confirmed NEC develop moderate-severe thrombocytopenia requiring one or more platelet transfusions. Here we used our neonatal murine model of NEC-related thrombocytopenia to investigate mechanisms of platelet depletion associated with this disease [K. Namachivayam, K. MohanKumar, L. Garg, B. A. Torres, A. Maheshwari, Pediatr. Res. 81, 817-824 (2017)]. In this model, enteral administration of immunogen trinitrobenzene sulfonate (TNBS) in 10-d-old mouse pups produces an acute necrotizing ileocolitis resembling human NEC within 24 h, and these mice developed thrombocytopenia at 12 to 15 h. We hypothesized that platelet activation and depletion occur during intestinal injury following exposure to bacterial products translocated across the damaged mucosa. Surprisingly, platelet activation began in our model 3 h after TNBS administration, antedating mucosal injury or endotoxinemia. Platelet activation was triggered by thrombin, which, in turn, was activated by tissue factor released from intestinal macrophages. Compared to adults, neonatal platelets showed enhanced sensitivity to thrombin due to higher expression of several downstream signaling mediators and the deficiency of endogenous thrombin antagonists. The expression of tissue factor in intestinal macrophages was also unique to the neonate. Targeted inhibition of thrombin by a nanomedicine-based approach was protective without increasing interstitial hemorrhages in the inflamed bowel or other organs. In support of these data, we detected increased circulating tissue factor and thrombin-antithrombin complexes in patients with NEC. Our findings show that platelet activation is an important pathophysiological event and a potential therapeutic target in NEC.


Assuntos
Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/patologia , Doenças do Recém-Nascido/metabolismo , Trombina/metabolismo , Animais , Animais Recém-Nascidos , Plaquetas/metabolismo , Modelos Animais de Doenças , Humanos , Recém-Nascido , Inflamação/metabolismo , Enteropatias/patologia , Intestinos/lesões , Intestinos/patologia , Macrófagos/metabolismo , Camundongos , Trombocitopenia/metabolismo
6.
Am J Physiol Gastrointest Liver Physiol ; 318(4): G705-G716, 2020 04 01.
Artigo em Inglês | MEDLINE | ID: mdl-32090604

RESUMO

Anemia is a frequent diagnosis in critically ill infants, but the clinical implications of severe anemia in these patients remain unclear. In this study, we examined preweaned mice to investigate the effects of severe anemia during early infancy on gut mucosal permeability. C57BL/6 mice were subjected to timed phlebotomy between postnatal days (P) 2-10 to induce severe anemia (hematocrits 20%-24%), and intestinal permeability was tracked longitudinally between P10 and P20 as intestine-to-plasma translocation of enteral macromolecules and bacterial translocation. Epithelial junctions were evaluated by electron microscopy, polymerase chain reactions, immunohistochemistry, and/or enzyme immunoassays on intestinal tissues, Caco-2 intestinal epithelial-like cells, and colonic organoids. Preweaned mouse pups showed an age-related susceptibility to severe anemia, with increased intestinal permeability to enteral macromolecules (dextran, ovalbumin, ß-lactoglobulin) and luminal bacteria. Electron micrographs showed increased paracellular permeability and ultrastructural abnormalities of the adherens junctions. These findings were explained by the loss of E-cadherin in epithelial cells, which was caused by destabilization of the E-cadherin (Cdh1) mRNA because of microRNA let-7e-5p binding to the 3'-untranslated region. Severe anemia resulted in a disproportionate and persistent increase in intestinal permeability in preweaned mice because of the disruption of epithelial adherens junctions. These changes are mediated via microRNA let-7e-mediated depletion of Cdh1 mRNA.NEW & NOTEWORTHY This research article shows that newborn infants with severe anemia show an age-related susceptibility to developing increased intestinal permeability to ingested macromolecules. This abnormal permeability develops because of abnormalities in intestinal epithelial junctions caused by a deficiency of the molecule E-cadherin in epithelial cells. The deficiency of E-cadherin is caused by destabilization of its mRNA precursor because of increased expression and binding of another molecule, the microRNA let-7e-5p, to the E-cadherin mRNA.


Assuntos
Junções Aderentes/patologia , Anemia Neonatal/patologia , Mucosa Intestinal/patologia , Intestinos/patologia , Junções Aderentes/ultraestrutura , Animais , Animais Recém-Nascidos , Células CACO-2 , Caderinas/genética , Caderinas/metabolismo , Feminino , Regulação da Expressão Gênica/efeitos dos fármacos , Humanos , Masculino , Camundongos , MicroRNAs/antagonistas & inibidores , MicroRNAs/genética , MicroRNAs/metabolismo , Permeabilidade , RNA Mensageiro/genética , RNA Mensageiro/metabolismo
7.
Pediatr Res ; 87(5): 847-852, 2020 04.
Artigo em Inglês | MEDLINE | ID: mdl-31756731

RESUMO

BACKGROUND: Fetal swallowing of human amniotic fluid (hAF) containing trophic factors (TFs) promotes gastrointestinal tract (GIT) development. Preterm birth interrupts hAF swallowing, which may increase the risk of necrotizing enterocolitis (NEC). Postnatally, it is difficult to replicate fetal swallowing of hAF due to volume. We aimed to evaluate whether hAF lyophilization is feasible and its effect on hAF-borne TFs. METHODS: We collected hAF (n = 16) from uncomplicated pregnancies. hAF was divided into three groups: unprocessed control (C), concentration by microfiltration (F), and by dialysis and lyophilization (L). EGF, HGF, GM-CSF, and TGF-α were measured in each group by multiplex assay. Bioavailability of TFs was measured by proliferation and LPS-induced IL-8 production by intestinal epithelial cells FHs74. RESULTS: After dialysis/lyophilization, GM-CSF and TGF-α were preserved with partial loss of EGF and HGF. hAF increased cell proliferation and reduced LPS-induced IL-8 production compared to medium alone. Compared to control, dialysis/lyophilization and filtration of hAF increased FHs74 cell proliferation (p < 0.001) and decreased LPS-induced IL-8 production (p < 0.01). CONCLUSIONS: Lyophilization and filtration of hAF is feasible with partial loss of TFs but maintains and even improves bioavailability of TFs measured by proliferation and LPS-induced IL-8 production by FHs74.


Assuntos
Líquido Amniótico/metabolismo , Enterocolite Necrosante/metabolismo , Liofilização , Trato Gastrointestinal/embriologia , Líquido Amniótico/química , Proliferação de Células , Criopreservação , Deglutição , Feminino , Fator Estimulador de Colônias de Granulócitos e Macrófagos/metabolismo , Humanos , Inflamação , Interleucina-8/metabolismo , Gravidez , Fator de Crescimento Transformador alfa/metabolismo
8.
Nat Commun ; 10(1): 3494, 2019 08 02.
Artigo em Inglês | MEDLINE | ID: mdl-31375667

RESUMO

Necrotizing enterocolitis (NEC) is an idiopathic, inflammatory bowel necrosis of premature infants. Clinical studies have linked NEC with antecedent red blood cell (RBC) transfusions, but the underlying mechanisms are unclear. Here we report a neonatal murine model to investigate this association. C57BL/6 mouse pups rendered anemic by timed phlebotomy and then given RBC transfusions develop NEC-like intestinal injury with prominent necrosis, inflammation, and submucosal edema/separation of the lamina propria in the ileocecal region and colon within 12-24 h. The anemic intestine is infiltrated by inflammatory macrophages, which are activated in situ by RBC transfusions via a Toll-like receptor (TLR)-4-mediated mechanism and cause bowel injury. Chelation of RBC degradation products with haptoglobin, absence of TLR4, macrophage depletion, and inhibition of macrophage activation is protective. Intestinal injury worsens with increasing severity and the duration of anemia prior to transfusion, indicating a need for the re-evaluation of current transfusion guidelines for premature infants.


Assuntos
Anemia/complicações , Enterocolite Necrosante/etiologia , Transfusão de Eritrócitos/efeitos adversos , Doenças do Recém-Nascido/etiologia , Anemia/terapia , Animais , Animais Recém-Nascidos , Ceco/patologia , Colo/patologia , Modelos Animais de Doenças , Enterocolite Necrosante/patologia , Humanos , Íleo/patologia , Recém-Nascido , Doenças do Recém-Nascido/patologia , Recém-Nascido Prematuro , Mucosa Intestinal/patologia , Macrófagos/imunologia , Macrófagos/metabolismo , Camundongos , Receptor 4 Toll-Like/imunologia , Receptor 4 Toll-Like/metabolismo
9.
Pediatr Res ; 81(5): 817-824, 2017 May.
Artigo em Inglês | MEDLINE | ID: mdl-28085792

RESUMO

BACKGROUND: Thrombocytopenia is frequently encountered in infants with necrotizing enterocolitis (NEC). To develop a preclinical model of NEC-related thrombocytopenia, we measured serial platelet counts in 10-d-old (P10) mouse pups with trinitrobenzene sulfonic acid (TNBS)-induced NEC-like injury. We also measured platelet volume indices, immature platelet fraction (IPF), and megakaryocyte number/ploidy in these animals. METHODS: Platelet counts, platelet volume indices, and IPF were measured in control (N = 65) and TNBS-treated pups (N = 104) using an automated hematology analyzer. Bone marrow megakaryocyte number, ploidy and CD41 expression were measured by flow cytometry. These findings were confirmed in a small cohort of P3 mice with NEC-like injury. RESULTS: Murine pups with TNBS-mediated NEC-like injury developed thrombocytopenia at 15-24 h after exposure to TNBS. Intestinal injury was associated with increased platelet volume indices (mean platelet volume, platelet-to-large cell ratio, and platelet distribution width), and IPF, indicating increased thrombopoiesis. These mice also showed increased megakaryocyte number, ploidy, and CD41 expression, indicating increased megakaryocyte differentiation. CONCLUSION: Similar to human NEC, murine NEC-like injury was also associated with decreased platelet counts. There was evidence of increased megakaryocyte differentiation and thrombopoiesis, which favors peripheral consumption of platelets as the likely mechanism of thrombocytopenia in these animals, over decreased platelet production.


Assuntos
Plaquetas , Enterocolite Necrosante/sangue , Megacariócitos , Trombocitopenia/sangue , Trombopoese , Animais , Animais Recém-Nascidos , Biomarcadores/sangue , Plaquetas/metabolismo , Plaquetas/patologia , Modelos Animais de Doenças , Enterocolite Necrosante/induzido quimicamente , Enterocolite Necrosante/patologia , Intestinos/patologia , Volume Plaquetário Médio , Megacariócitos/metabolismo , Megacariócitos/patologia , Camundongos Endogâmicos C57BL , Contagem de Plaquetas , Glicoproteína IIb da Membrana de Plaquetas/sangue , Ploidias , Trombocitopenia/etiologia , Trombocitopenia/patologia , Fatores de Tempo , Ácido Trinitrobenzenossulfônico
10.
Semin Perinatol ; 41(1): 52-60, 2017 02.
Artigo em Inglês | MEDLINE | ID: mdl-27832931

RESUMO

Cytokines and growth factors play diverse roles in the uninflamed fetal/neonatal intestinal mucosa and in the development of inflammatory bowel injury during necrotizing enterocolitis (NEC). During gestational development and the early neonatal period, the fetal/premature intestine is exposed to high levels of many "inflammatory" cytokines and growth factors, first via swallowed amniotic fluid in utero and then, after birth, in colostrum and mother's milk. This article reviews the dual, seemingly counter-intuitive roles of cytokines, where these agents play a "trophic" role and promote maturation of the uninflamed mucosa, but can also cause inflammation and promote intestinal injury during NEC.


Assuntos
Citocinas/metabolismo , Enterocolite Necrosante/fisiopatologia , Fator de Crescimento Semelhante a EGF de Ligação à Heparina/metabolismo , Doenças do Prematuro/fisiopatologia , Inflamação/fisiopatologia , Mucosa Intestinal/fisiopatologia , Suscetibilidade a Doenças/imunologia , Enterocolite Necrosante/imunologia , Enterocolite Necrosante/metabolismo , Regulação da Expressão Gênica no Desenvolvimento/imunologia , Humanos , Imunidade Inata , Fenômenos Fisiológicos da Nutrição do Lactente , Recém-Nascido de Peso Extremamente Baixo ao Nascer , Lactente Extremamente Prematuro , Recém-Nascido , Doenças do Prematuro/imunologia , Doenças do Prematuro/metabolismo , Inflamação/imunologia , Mucosa Intestinal/imunologia
11.
Pediatr Res ; 81(1-1): 99-112, 2017 01.
Artigo em Inglês | MEDLINE | ID: mdl-27656771

RESUMO

BACKGROUND: We have shown previously that enteral administration of 2, 4, 6-trinitrobenzene sulfonic acid in 10-d-old C57BL/6 pups produces an acute necrotizing enterocolitis with histopathological and inflammatory changes similar to human necrotizing enterocolitis (NEC). To determine whether murine neonatal 2, 4, 6-trinitrobenzene sulfonic acid (TNBS)-mediated intestinal injury could be used as a NEC model, we compared gene expression profiles of TNBS-mediated intestinal injury and NEC. METHODS: Whole-genome microarray analysis was performed on proximal colon from control and TNBS-treated pups (n = 8/group). For comparison, we downloaded human microarray data of NEC (n = 5) and surgical control (n = 4) from a public database. Data were analyzed using the software programs Partek Genomics Suite and Ingenuity Pathway Analysis. RESULTS: We detected extensive changes in gene expression in murine TNBS-mediated intestinal injury and human NEC. Using fold-change cut-offs of ±1.5, we identified 4,440 differentially-expressed genes (DEGs) in murine TNBS-mediated injury and 1,377 in NEC. Murine TNBS-mediated injury and NEC produced similar changes in expression of orthologous genes (r = 0.611, P < 0.001), and also activated nearly-identical biological processes and pathways. Lipopolysaccharide was top predicted upstream regulator in both the murine and human datasets. CONCLUSION: Murine neonatal TNBS-mediated enterocolitis and human NEC activate nearly-identical biological processes, signaling pathways, and transcriptional networks.


Assuntos
Enterocolite Necrosante/genética , Redes Reguladoras de Genes/efeitos dos fármacos , Mucosa Intestinal/metabolismo , Intestinos/lesões , Ácido Trinitrobenzenossulfônico/toxicidade , Animais , Animais Recém-Nascidos , Modelos Animais de Doenças , Enterocolite Necrosante/etiologia , Perfilação da Expressão Gênica , Humanos , Intestinos/efeitos dos fármacos , Lipopolissacarídeos/toxicidade , Camundongos , Camundongos Endogâmicos C57BL , Especificidade da Espécie
12.
Pediatr Res ; 79(6): 951-61, 2016 06.
Artigo em Inglês | MEDLINE | ID: mdl-26859364

RESUMO

BACKGROUND: Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. Based on our recent findings of increased Smad7 expression in surgically resected bowel affected by NEC, we hypothesized that NEC macrophages undergo inflammatory activation because increased Smad7 expression renders these cells resistant to normal, gut-specific, transforming growth factor (TGF)-ß-mediated suppression of inflammatory pathways. METHODS: We used surgically resected human NEC tissue, murine models of NEC-like injury, bone marrow-derived and intestinal macrophages, and RAW264.7 cells. Smad7 and IκB kinase-beta (IKK-ß) were measured by quantitative PCR, western blots, and immunohistochemistry. Promoter activation was confirmed in luciferase reporter and chromatin immunoprecipitation assays. RESULTS: NEC macrophages showed increased Smad7 expression, particularly in areas with severe tissue damage and high bacterial load. Lipopolysaccharide-induced Smad7 expression suppressed TGF-ß signaling and augmented nuclear factor-kappa B (NF-κB) activation and cytokine production in macrophages. Smad7-mediated NF-κB activation was likely mediated via increased expression of IKK-ß, which, further increased Smad7 expression in a feed-forward loop. We show that Smad7 induced IKK-ß expression through direct binding to the IKK-ß promoter and its transcriptional activation. CONCLUSION: Smad7 expression in NEC macrophages interrupts TGF-ß signaling and promotes NF-κB-mediated inflammatory signaling in these cells through increased expression of IKK-ß.


Assuntos
Enterocolite Necrosante/metabolismo , Mucosa Intestinal/metabolismo , Macrófagos/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Humanos , Quinase I-kappa B/metabolismo , Inflamação , Lipopolissacarídeos/metabolismo , Camundongos , Camundongos Endogâmicos C57BL , NF-kappa B/metabolismo , Células RAW 264.7 , Transdução de Sinais
13.
PLoS One ; 10(7): e0134003, 2015.
Artigo em Inglês | MEDLINE | ID: mdl-26225425

RESUMO

OBJECTIVE: We have shown previously that preterm infants are at risk of necrotizing enterocolitis (NEC), an inflammatory bowel necrosis typically seen in infants born prior to 32 weeks' gestation, because of the developmental deficiency of transforming growth factor (TGF)-ß2 in the intestine. The present study was designed to investigate all-trans retinoic acid (atRA) as an inducer of TGF-ß2 in intestinal epithelial cells (IECs) and to elucidate the involved signaling mechanisms. METHODS: AtRA effects on intestinal epithelium were investigated using IEC6 cells. TGF-ß2 expression was measured using reverse transcriptase-quantitative polymerase chain reaction (RT-qPCR) and Western blots. Signaling pathways were investigated using Western blots, transiently-transfected/transduced cells, kinase arrays, chromatin immunoprecipitation, and selective small molecule inhibitors. RESULTS: AtRA-treatment of IEC6 cells selectively increased TGF-ß2 mRNA and protein expression in a time- and dose-dependent fashion, and increased the activity of the TGF-ß2 promoter. AtRA effects were mediated via RhoA GTPase, Rho-associated, coiled-coil-containing protein kinase 1 (ROCK1), p38α MAPK, and activating transcription factor (ATF)-2. AtRA increased phospho-ATF2 binding to the TGF-ß2 promoter and increased histone H2B acetylation in the TGF-ß2 nucleosome, which is typically associated with transcriptional activation. CONCLUSIONS: AtRA induces TGF-ß2 expression in IECs via RhoA- and p38α MAPK-mediated activation of the transcription factor ATF2. Further studies are needed to investigate the role of atRA as a protective/therapeutic agent in gut mucosal inflammation.


Assuntos
Fator 2 Ativador da Transcrição/metabolismo , Mucosa Intestinal/metabolismo , Proteína Quinase 14 Ativada por Mitógeno/metabolismo , Fator de Crescimento Transformador beta/biossíntese , Tretinoína/farmacologia , Proteína rhoA de Ligação ao GTP/metabolismo , Humanos , Mucosa Intestinal/citologia , Fosforilação , Proteínas Smad/metabolismo
14.
Am J Physiol Gastrointest Liver Physiol ; 309(3): G171-80, 2015 Aug 01.
Artigo em Inglês | MEDLINE | ID: mdl-26045614

RESUMO

Human milk contains biologically important amounts of transforming growth factor-ß2 isoform (TGF-ß2), which is presumed to protect against inflammatory gut mucosal injury in the neonate. In preclinical models, enterally administered TGF-ß2 can protect against experimental necrotizing enterocolitis, an inflammatory bowel necrosis of premature infants. In this study, we investigated whether TGF-ß bioactivity in human preterm milk could be enhanced for therapeutic purposes by adding recombinant TGF-ß2 (rTGF-ß2) to milk prior to feeding. Milk-borne TGF-ß bioactivity was measured by established luciferase reporter assays. Molecular interactions of TGF-ß2 were investigated by nondenaturing gel electrophoresis and immunoblots, computational molecular modeling, and affinity capillary electrophoresis. Addition of rTGF-ß2 (20-40 nM) to human preterm milk samples failed to increase TGF-ß bioactivity in milk. Milk-borne TGF-ß2 was bound to chondroitin sulfate (CS) containing proteoglycan(s) such as biglycan, which are expressed in high concentrations in milk. Chondroitinase treatment of milk increased the bioactivity of both endogenous and rTGF-ß2, and consequently, enhanced the ability of preterm milk to suppress LPS-induced NF-κB activation in macrophages. These findings provide a mechanism for the normally low bioavailability of milk-borne TGF-ß2 and identify chondroitinase digestion of milk as a potential therapeutic strategy to enhance the anti-inflammatory effects of preterm milk.


Assuntos
Condroitinases e Condroitina Liases/metabolismo , Enterocolite Necrosante , Leite Humano , Fator de Crescimento Transformador beta2/metabolismo , Animais , Anti-Inflamatórios/metabolismo , Disponibilidade Biológica , Linhagem Celular , Proteoglicanas de Sulfatos de Condroitina/metabolismo , Enterocolite Necrosante/metabolismo , Enterocolite Necrosante/prevenção & controle , Humanos , Recém-Nascido , Recém-Nascido Prematuro , Inflamação/metabolismo , Mucosa Intestinal/metabolismo , Ativação de Macrófagos/fisiologia , Camundongos , Leite Humano/enzimologia , Leite Humano/metabolismo , NF-kappa B/metabolismo , Proteínas Recombinantes/metabolismo
15.
Pediatr Res ; 77(4): 500-5, 2015 Apr.
Artigo em Inglês | MEDLINE | ID: mdl-25588190

RESUMO

BACKGROUND: Vascular endothelial growth factor (VEGF), a well-characterized regulator of angiogenesis, has been mechanistically implicated in retinal neovascularization and in the pathogenesis of retinopathy of prematurity. However, the ontogeny of VEGF expression in the human fetal retina is not well known. Because retinal vasculature grows with gestational maturation, we hypothesized that VEGF expression also increases in the midgestation human fetal eye as a function of gestational age. METHODS: To identify changes in VEGF gene expression during normal human development, we measured VEGF mRNA by quantitative PCR and measured VEGF protein by enzyme-linked immunosorbent assay and western blots in 10-24 wk gestation fetal vitreous, retina, and serum. RESULTS: VEGF mRNA expression in the retina increased with gestational age. VEGF isoform A, particularly its VEGF121 splice variant, contributed to this positive correlation. Consistent with these findings, we detected increasing VEGF121 protein concentrations in vitreous humor from fetuses of 10-24 wk gestation, while VEGF concentrations decreased in fetal serum. CONCLUSION: VEGF121 mRNA and protein concentrations increase with increasing gestational age in the developing human retina. We speculate that VEGF plays an important role in normal retinal vascular development, and that preterm delivery affects production of this vascular growth factor.


Assuntos
Regulação da Expressão Gênica no Desenvolvimento , RNA Mensageiro/metabolismo , Retina/embriologia , Neovascularização Retiniana , Fator A de Crescimento do Endotélio Vascular/metabolismo , Corpo Vítreo/embriologia , Actinas/metabolismo , Western Blotting , Ensaio de Imunoadsorção Enzimática , Idade Gestacional , Gliceraldeído-3-Fosfato Desidrogenases/metabolismo , Humanos , Subunidade alfa do Fator 1 Induzível por Hipóxia/metabolismo , Recém-Nascido , Recém-Nascido Prematuro , RNA Ribossômico 18S/metabolismo , Retinopatia da Prematuridade/metabolismo
16.
Am J Physiol Gastrointest Liver Physiol ; 306(5): G361-9, 2014 Mar 01.
Artigo em Inglês | MEDLINE | ID: mdl-24407592

RESUMO

Fetal swallowing of amniotic fluid, which contains numerous cytokines and growth factors, plays a key role in gut mucosal development. Preterm birth interrupts this exposure to amniotic fluid-borne growth factors, possibly contributing to the increased risk of necrotizing enterocolitis (NEC) in premature infants. We hypothesized that supplementation of formula feeds with amniotic fluid can provide amniotic fluid-borne growth factors and prevent experimental NEC in rat pups. We compared NEC-like injury in rat pups fed with infant formula vs. formula supplemented either with 30% amniotic fluid or recombinant hepatocyte growth factor (HGF). Cytokines/growth factors in amniotic fluid were measured by immunoassays. Amniotic fluid and HGF effects on enterocyte migration, proliferation, and survival were measured in cultured IEC6 intestinal epithelial cells. Finally, we used an antibody array to investigate receptor tyrosine kinase (RTK) activation and immunoblots to measure phosphoinositide 3-kinase (PI3K) signaling. Amniotic fluid supplementation in oral feeds protected rat pups against NEC-like injury. HGF was the most abundant growth factor in rat amniotic fluid in our panel of analytes. Amniotic fluid increased cell migration, proliferation, and cell survival in vitro. These effects were reproduced by HGF and blocked by anti-HGF antibody or a PI3K inhibitor. HGF transactivated several RTKs in IEC6 cells, indicating that its effects extended to multiple signaling pathways. Finally, similar to amniotic fluid, recombinant HGF also reduced the frequency and severity of NEC-like injury in rat pups. Amniotic fluid supplementation protects rat pups against experimental NEC, which is mediated, at least in part, by HGF.


Assuntos
Líquido Amniótico/metabolismo , Enterocolite Necrosante/prevenção & controle , Fator de Crescimento de Hepatócito/administração & dosagem , Líquido Amniótico/química , Ração Animal , Animais , Células Cultivadas , Citocinas/metabolismo , Suplementos Nutricionais , Células Epiteliais/efeitos dos fármacos , Feminino , Regulação Enzimológica da Expressão Gênica , Fator de Crescimento de Hepatócito/química , Fator de Crescimento de Hepatócito/metabolismo , Humanos , Lactente , Fórmulas Infantis , Mucosa Intestinal/citologia , Gravidez , Ratos , Ratos Sprague-Dawley , Receptores Proteína Tirosina Quinases/genética , Receptores Proteína Tirosina Quinases/metabolismo
17.
Lab Invest ; 94(2): 150-60, 2014 Feb.
Artigo em Inglês | MEDLINE | ID: mdl-24365747

RESUMO

Neonates and young infants exposed to extracorporeal circulation during extracorporeal membrane oxygenation (ECMO) and cardiopulmonary bypass are at risk of developing a systemic inflammatory response syndrome with multi-organ dysfunction. We used a piglet model of ECMO to investigate the hypothesis that epithelial apoptosis is an early event that precedes villous damage during ECMO-related bowel injury. Healthy 3-week-old piglets were subjected to ECMO for up to 8 h. Epithelial apoptosis was measured in histopathological analysis, nuclear imaging, and terminal deoxynucleotidyl transferase dUTP nick end labeling. Plasma intestinal fatty acid-binding protein (I-FABP) levels were measured by enzyme immunoassay. Intestinal mast cells were isolated by fluorescence-assisted cell sorting. Cleaved caspase-8, caspase-9, phospho-p38 MAPK, and fas ligand expression were investigated by immunohistochemistry, western blots, and reverse transcriptase-quantitative PCR. Piglet ECMO was associated with increased gut epithelial apoptosis. Extensive apoptotic changes were noted on villus tips and in scattered crypt cells after 2 h of ECMO. After 8 h, the villi were denuded and apoptotic changes were evident in a majority of crypt cells. Increased circulating I-FABP levels, a marker of gut epithelial injury, showed that epithelial injury occurred during ECMO. We detected increased cleaved caspase-8, but not cleaved caspase-9, in epithelial cells indicating that the extrinsic apoptotic pathway was active. ECMO was associated with increased fas ligand expression in intestinal mast cells, which was induced through activation of the p38 mitogen-activated protein kinase. We conclude that epithelial apoptosis is an early event that initiates gut mucosal injury in a piglet model of ECMO.


Assuntos
Apoptose/fisiologia , Oxigenação por Membrana Extracorpórea/efeitos adversos , Mucosa Intestinal/lesões , Mucosa Intestinal/fisiopatologia , Animais , Animais Recém-Nascidos , Western Blotting , Caspase 8/metabolismo , Caspase 9/metabolismo , Núcleo Celular/ultraestrutura , Primers do DNA/genética , Ensaio de Imunoadsorção Enzimática , Proteína Ligante Fas/metabolismo , Proteínas de Ligação a Ácido Graxo/sangue , Citometria de Fluxo , Imuno-Histoquímica , Marcação In Situ das Extremidades Cortadas , Mucosa Intestinal/citologia , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Estatísticas não Paramétricas , Suínos , Proteínas Quinases p38 Ativadas por Mitógeno/metabolismo
18.
Am J Physiol Gastrointest Liver Physiol ; 304(12): G1055-65, 2013 Jun 15.
Artigo em Inglês | MEDLINE | ID: mdl-23558011

RESUMO

Human milk contains substantial amounts of transforming growth factor (TGF)-ß, particularly the isoform TGF-ß2. We previously showed in preclinical models that enterally administered TGF-ß2 can protect against necrotizing enterocolitis (NEC), an inflammatory bowel necrosis of premature infants. In this study we hypothesized that premature infants remain at higher risk of NEC than full-term infants, even when they receive their own mother's milk, because preterm human milk contains less bioactive TGF-ß than full-term milk. Our objective was to compare TGF-ß bioactivity in preterm vs. full-term milk and identify factors that activate milk-borne TGF-ß. Mothers who delivered between 23 0/7 and 31 6/7 wk or at ≥37 wk of gestation provided milk samples at serial time points. TGF-ß bioactivity and NF-κB signaling were measured using specific reporter cells and in murine intestinal tissue explants. TGF-ß1, TGF-ß2, TGF-ß3, and various TGF-ß activators were measured by real-time PCR, enzyme immunoassays, or established enzymatic activity assays. Preterm human milk showed minimal TGF-ß bioactivity in the native state but contained a large pool of latent TGF-ß. TGF-ß2 was the predominant isoform of TGF-ß in preterm milk. Using a combination of several in vitro and ex vivo models, we show that neuraminidase is a key regulator of TGF-ß bioactivity in human milk. Finally, we show that addition of bacterial neuraminidase to preterm human milk increased TGF-ß bioactivity. Preterm milk contains large quantities of TGF-ß, but most of it is in an inactive state. Addition of neuraminidase can increase TGF-ß bioactivity in preterm milk and enhance its anti-inflammatory effects.


Assuntos
Leite Humano/metabolismo , Neuraminidase/metabolismo , Fator de Crescimento Transformador beta/metabolismo , Animais , Feminino , Expressão Gênica , Humanos , Lactação/metabolismo , Camundongos , Leite Humano/enzimologia , NF-kappa B/genética , NF-kappa B/metabolismo , Neuraminidase/genética , Nascimento Prematuro/metabolismo , Isoformas de Proteínas/genética , Isoformas de Proteínas/metabolismo , Nascimento a Termo/metabolismo , Fatores de Tempo , Fator de Crescimento Transformador beta/genética
19.
Am J Physiol Gastrointest Liver Physiol ; 304(2): G167-80, 2013 Jan 15.
Artigo em Inglês | MEDLINE | ID: mdl-23154975

RESUMO

Preterm infants may be at risk of necrotizing enterocolitis (NEC) due to deficiency of transforming growth factor-ß 2 (TGF-ß(2)) in the developing intestine. We hypothesized that low epithelial TGF-ß(2) expression in preterm intestine and during NEC results from diminished autocrine induction of TGF-ß(2) in these cells. Premature baboons delivered at 67% gestation were treated per current norms for human preterm infants. NEC was diagnosed by clinical and radiological findings. Inflammatory cytokines, TGF-ß(2), Smad7, Ski, and strawberry notch N (SnoN)/Ski-like oncoprotein (SKIL) was measured using quantitative reverse transcriptase-polymerase chain reaction, immunoblots, and immunohistochemistry. Smad7 effects were examined in transfected IEC6 intestinal epithelial cells in vitro. Findings were validated in archived human tissue samples of NEC. NEC was recorded in seven premature baboons. Consistent with existing human data, premature baboon intestine expressed less TGF-ß(2) than term intestine. TGF-ß(2) expression was regulated in epithelial cells in an autocrine fashion, which was interrupted in the premature intestine and during NEC due to increased expression of Smad7. LPS increased Smad7 binding to the TGF-ß(2) promoter and was associated with dimethylation of the lysine H3K9, a marker of transcriptional silencing, on the nucleosome of TGF-ß(2). Increased Smad7 expression in preterm intestine was correlated with the deficiency of SnoN/SKIL, a repressor of the Smad7 promoter. Smad7 inhibits autocrine expression of TGF-ß(2) in intestinal epithelial cells in the normal premature intestine and during NEC. Increased Smad7 expression in the developing intestine may be due to a developmental deficiency of the SnoN/SKIL oncoprotein.


Assuntos
Comunicação Autócrina , Colo/metabolismo , Enterocolite Necrosante/metabolismo , Mucosa Intestinal/metabolismo , Proteína Smad7/metabolismo , Fator de Crescimento Transformador beta2/metabolismo , Animais , Western Blotting , Linhagem Celular , Colo/patologia , Proteínas de Ligação a DNA/metabolismo , Modelos Animais de Doenças , Regulação para Baixo , Enterocolite Necrosante/genética , Enterocolite Necrosante/patologia , Idade Gestacional , Humanos , Imuno-Histoquímica , Mucosa Intestinal/patologia , Peptídeos e Proteínas de Sinalização Intracelular/metabolismo , Papio anubis , Papio cynocephalus , Nascimento Prematuro , Proteínas Proto-Oncogênicas/metabolismo , Reação em Cadeia da Polimerase Via Transcriptase Reversa , Proteína Smad7/genética , Transfecção , Fator de Crescimento Transformador beta2/genética
20.
Am J Physiol Gastrointest Liver Physiol ; 303(1): G93-102, 2012 Jul.
Artigo em Inglês | MEDLINE | ID: mdl-22538401

RESUMO

Necrotizing enterocolitis (NEC) is an inflammatory bowel necrosis of premature infants. In tissue samples of NEC, we identified numerous macrophages and a few neutrophils but not many lymphocytes. We hypothesized that these pathoanatomic characteristics of NEC represent a common tissue injury response of the gastrointestinal tract to a variety of insults at a specific stage of gut development. To evaluate developmental changes in mucosal inflammatory response, we used trinitrobenzene sulfonic acid (TNBS)-induced inflammation as a nonspecific insult and compared mucosal injury in newborn vs. adult mice. Enterocolitis was induced in 10-day-old pups and adult mice (n = 25 animals per group) by administering TNBS by gavage and enema. Leukocyte populations were enumerated in human NEC and in murine TNBS-enterocolitis using quantitative immunofluorescence. Chemokine expression was measured using quantitative polymerase chain reaction, immunoblots, and immunohistochemistry. Macrophage recruitment was investigated ex vivo using intestinal tissue-conditioned media and bone marrow-derived macrophages in a microchemotaxis assay. Similar to human NEC, TNBS enterocolitis in pups was marked by a macrophage-rich leukocyte infiltrate in affected tissue. In contrast, TNBS-enterocolitis in adult mice was associated with pleomorphic leukocyte infiltrates. Macrophage precursors were recruited to murine neonatal gastrointestinal tract by the chemokine CXCL5, a known chemoattractant for myeloid cells. We also demonstrated increased expression of CXCL5 in surgically resected tissue samples of human NEC, indicating that a similar pathway was active in NEC. We concluded that gut mucosal injury in the murine neonate is marked by a macrophage-rich leukocyte infiltrate, which contrasts with the pleomorphic leukocyte infiltrates in adult mice. In murine neonatal enterocolitis, macrophages were recruited to the inflamed gut mucosa by the chemokine CXCL5, indicating that CXCL5 and its cognate receptor CXCR2 merit further investigation as potential therapeutic targets in NEC.


Assuntos
Mucosa Intestinal/patologia , Macrófagos/fisiologia , Envelhecimento/fisiologia , Animais , Animais Recém-Nascidos , Western Blotting , Quimiocina CXCL5/fisiologia , Quimiotaxia de Leucócito/efeitos dos fármacos , Quimiotaxia de Leucócito/fisiologia , Eletroforese em Gel de Gradiente Desnaturante , Ensaio de Imunoadsorção Enzimática , Humanos , Imuno-Histoquímica , Técnicas In Vitro , Recém-Nascido , Recém-Nascido Prematuro , Inflamação/patologia , Enteropatias/induzido quimicamente , Enteropatias/patologia , Mucosa Intestinal/citologia , Camundongos , Infiltração de Neutrófilos/fisiologia , Reação em Cadeia da Polimerase , Fatores de Risco , Ácido Trinitrobenzenossulfônico
SELEÇÃO DE REFERÊNCIAS
DETALHE DA PESQUISA
...