Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 19 de 19
Filtrer
Plus de filtres










Base de données
Gamme d'année
1.
Biologicals ; 86: 101756, 2024 May.
Article de Anglais | MEDLINE | ID: mdl-38479213

RÉSUMÉ

An international hybrid meeting held 21-22 June 2023 in Ottawa, Canada brought together regulators, scientists, and industry experts to discuss a set of principles and best practices in the development and implementation of standards. Although the use of international standards (ISs) and international units (IUs) has been an essential part of ensuring human and animal vaccine quality in the past decades, the types and uses of standards have expanded with technological advances in manufacture and testing of vaccines. The needs of stakeholders are evolving in response to the ever-increasing complexity, diversity, and number of vaccine products as well as increasing efforts to replace animal-based potency tests with in vitro assays that measure relevant quality attributes. As such, there must be a concomitant evolution in the design and implementation of both international and in-house standards. Concomitantly, greater harmonization of regulatory expectations must be achieved through collaboration with standard-setting organizations, national control laboratories and manufacturers. Stakeholders provided perspectives on challenges and several recommendations emerged as essential to advancing agreed upon objectives.


Sujet(s)
Contrôle de qualité , Vaccins , Humains , Vaccins/normes , Animaux , Canada , Normes de référence
2.
Bioanalysis ; 13(6): 415-463, 2021 Mar.
Article de Anglais | MEDLINE | ID: mdl-33533276

RÉSUMÉ

The 14th edition of the Workshop on Recent Issues in Bioanalysis (14th WRIB) was held virtually on June 15-29, 2020 with an attendance of over 1000 representatives from pharmaceutical/biopharmaceutical companies, biotechnology companies, contract research organizations, and regulatory agencies worldwide. The 14th WRIB included three Main Workshops, seven Specialized Workshops that together spanned 11 days in order to allow exhaustive and thorough coverage of all major issues in bioanalysis, biomarkers, immunogenicity, gene therapy and vaccine. Moreover, a comprehensive vaccine assays track; an enhanced cytometry track and updated Industry/Regulators consensus on BMV of biotherapeutics by LCMS were special features in 2020. As in previous years, this year's WRIB continued to gather a wide diversity of international industry opinion leaders and regulatory authority experts working on both small and large molecules to facilitate sharing and discussions focused on improving quality, increasing regulatory compliance and achieving scientific excellence on bioanalytical issues. This 2020 White Paper encompasses recommendations emerging from the extensive discussions held during the workshop and is aimed to provide the Global Bioanalytical Community with key information and practical solutions on topics and issues addressed, in an effort to enable advances in scientific excellence, improved quality and better regulatory compliance. Due to its length, the 2020 edition of this comprehensive White Paper has been divided into three parts for editorial reasons. This publication (Part 3) covers the recommendations on Vaccine, Gene/Cell Therapy, NAb Harmonization and Immunogenicity). Part 1 (Innovation in Small Molecules, Hybrid LBA/LCMS & Regulated Bioanalysis), Part 2A (BAV, PK LBA, Flow Cytometry Validation and Cytometry Innovation) and Part 2B (Regulatory Input) are published in volume 13 of Bioanalysis, issues 4 and 5 (2020), respectively.


Sujet(s)
Thérapie cellulaire et tissulaire , Cytométrie en flux , Thérapie génétique , Réaction de polymérisation en chaine en temps réel , Vaccins/analyse , Humains , Contrôle de qualité , Récepteurs chimériques pour l'antigène/analyse , États-Unis , Food and Drug Administration (USA)
3.
Am J Physiol Gastrointest Liver Physiol ; 304(10): G864-75, 2013 May 15.
Article de Anglais | MEDLINE | ID: mdl-23518680

RÉSUMÉ

Preterm neonates are susceptible to gastrointestinal disorders such as necrotizing enterocolitis (NEC). Maternal milk and colostrum protects against NEC via growth promoting, immunomodulatory, and antimicrobial factors. The fetal enteral diet amniotic fluid (AF), contains similar components, and we hypothesized that postnatal AF administration reduces inflammatory responses and NEC in preterm neonates. Preterm pigs (92% gestation) were delivered by caesarean section and fed parental nutrition (2 days) followed by enteral (2 days) porcine colostrum (COLOS, n = 7), infant formula (FORM, n = 13), or AF supplied before and after introduction of formula (AF, n = 10) in experiment 1, and supplied only during the enteral feeding period in experiment 2 (FORM, n = 16; AF, n = 14). The NEC score was reduced in both AF and COLOS pigs, relative to FORM, when AF was provided prior to full enteral feeding (9.9 and 7.7 compared with 17.3, P < 0.05). There was no effect of AF when provided only during enteral feeding. AF pigs showed decreased bacterial abundance in colon and intestinal inflammation-related genes (e.g., TNF-α, IL-1α, IL-6, NOS) were downregulated, relative to FORM pigs with NEC. Anti-inflammatory properties of AF were supported by delayed maturation and decreased TNF-α production in murine dendritic cells, as well as increased proliferation and migration, and downregulation of IL-6 expression in intestinal cells (IEC-6, IPEC-J2). Like colostrum, AF may reduce NEC development in preterm neonates by suppressing the proinflammatory responses to enteral formula feeding and gut colonization when provided before the onset of NEC.


Sujet(s)
Liquide amniotique/physiologie , Colostrum/physiologie , Entérocolite nécrosante/thérapie , Gastroentérite/thérapie , Animaux , Animaux nouveau-nés , Cytokines/métabolisme , Cellules dendritiques/métabolisme , Nutrition entérale , Entérocolite nécrosante/microbiologie , Entérocolite nécrosante/anatomopathologie , Entérocytes/métabolisme , Femelle , Gastroentérite/microbiologie , Gastroentérite/anatomopathologie , Humains , Nouveau-né , Prématuré , Absorption intestinale , Intestins/microbiologie , Analyse sur microréseau , Nutrition parentérale totale , Perméabilité , Grossesse , ARN/biosynthèse , ARN/génétique , Réaction de polymérisation en chaine en temps réel , Suidae
4.
J Theor Biol ; 321: 83-99, 2013 Mar 21.
Article de Anglais | MEDLINE | ID: mdl-23238281

RÉSUMÉ

Necrotizing enterocolitis (NEC) is a severe disease of the gastrointestinal tract in premature infants, characterized by a disrupted intestinal epithelium and an exaggerated pro-inflammatory response. Since the activation of Toll-like receptor-4 (TLR4) blocks cell migration and proliferation and contributes to an uncontrolled inflammatory response within the intestine, this receptor has been identified as a key contributor to the development of NEC. Toll-like receptor-9 (TLR9) has been shown to sense bacterial genome components (CpG DNA) and to play an anti-inflammatory role in NEC. We present in vitro results demonstrating direct inhibition of TLR4 activation by CpG DNA, and we develop a mathematical model of bacteria-immune interactions within the intestine to investigate how such inhibition of TLR4 signaling might alter inflammation, associated bacterial invasion of tissue, and resulting outcomes. The model predicts that TLR9 can inhibit both the beneficial and detrimental effects of TLR4, and thus a proper balance of action by these two receptors is needed to promote intestinal health. The model results are also used to explore three interventions that could potentially prevent the development of NEC: reducing bacteria in the mucus layer, administering probiotic treatment, and blocking TLR4 activation. While the model shows that these interventions would be successful in most cases, the model is also used to identify situations in which the proposed treatments might be harmful.


Sujet(s)
Bactéries/métabolisme , Entérocolite nécrosante/métabolisme , Entérocolite nécrosante/microbiologie , Inflammation/métabolisme , Récepteurs de type Toll/métabolisme , Animaux , Lignée cellulaire , Simulation numérique , Ilots CpG , ADN/métabolisme , Cellules épithéliales/cytologie , Humains , Intestins/microbiologie , Modèles biologiques
5.
Gastroenterology ; 143(3): 708-718.e5, 2012 Sep.
Article de Anglais | MEDLINE | ID: mdl-22796522

RÉSUMÉ

BACKGROUND & AIMS: Little is known about factors that regulate intestinal epithelial differentiation; microbial recognition receptors such as Toll-like receptor (TLR)4 might be involved. We investigated whether intestinal TLR4 regulates epithelial differentiation and is involved in development of necrotizing enterocolitis (NEC) of the immature intestine. METHODS: Mice with conditional disruption of TLR4 in the intestinal epithelium and TLR4 knockout (TLR4(-/-)) mice were generated by breeding TLR4(loxp/loxp) mice with villin-cre and Ella-cre, respectively. Enterocytes that did not express or overexpressed TLR4 were created by lentiviral or adenoviral transduction. Intestinal organoids were cultured on tissue matrices. Bile acids were measured by colorimetric assays, and microbial composition was determined by 16S pyrosequencing. NEC was induced in 7- to 10-day-old mice by induction of hypoxia twice daily for 4 days. RESULTS: TLR4(-/-) mice and mice with enterocyte-specific deletion of TLR4 were protected from NEC; epithelial differentiation into goblet cells was increased via suppressed Notch signaling in the small intestinal epithelium. TLR4 also regulates differentiation of goblet cells in intestinal organoid and enterocyte cell cultures; differentiation was increased on deletion of TLR4 and restored when TLR4 was expressed ectopically. TLR4 signaling via Notch was increased in intestinal tissue samples from patients with NEC, and numbers of goblet cells were reduced. 16S pyrosequencing revealed that wild-type and TLR4-deficient mice had similar microbial profiles; increased numbers of goblet cells were observed in mice given antibiotics. TLR4 deficiency reduced levels of luminal bile acids in vivo, and addition of bile acids to TLR4-deficient cell cultures prevented differentiation of goblet cells. CONCLUSIONS: TLR4 signaling and Notch are increased in intestinal tissues of patients with NEC and required for induction of NEC in mice. TLR4 prevents goblet cell differentiation, independently of the microbiota. Bile acids might initiate goblet cell development.


Sujet(s)
Différenciation cellulaire , Entérocolite nécrosante/métabolisme , Cellules caliciformes/métabolisme , Intestin grêle/métabolisme , Récepteur de type Toll-4/métabolisme , Animaux , Animaux nouveau-nés , Acides et sels biliaires/métabolisme , Lignée cellulaire , Modèles animaux de maladie humaine , Entérocolite nécrosante/étiologie , Entérocolite nécrosante/génétique , Entérocolite nécrosante/microbiologie , Entérocolite nécrosante/anatomopathologie , Entérocolite nécrosante/prévention et contrôle , Cellules caliciformes/microbiologie , Cellules caliciformes/anatomopathologie , Humains , Hypoxie/complications , Préparation pour nourrissons , Nouveau-né , Intestin grêle/microbiologie , Intestin grêle/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Organoïdes , Interférence par ARN , Rats , Récepteurs Notch/métabolisme , Transduction du signal , Techniques de culture de tissus , Récepteur de type Toll-4/déficit , Récepteur de type Toll-4/génétique , Transfection
6.
Proc Natl Acad Sci U S A ; 109(28): 11330-5, 2012 Jul 10.
Article de Anglais | MEDLINE | ID: mdl-22733781

RÉSUMÉ

The fetal intestinal mucosa is characterized by elevated Toll-like receptor 4 (TLR4) expression, which can lead to the development of necrotizing enterocolitis (NEC)--a devastating inflammatory disease of the premature intestine--upon exposure to microbes. To define endogenous strategies that could reduce TLR4 signaling, we hypothesized that amniotic fluid can inhibit TLR4 signaling within the fetal intestine and attenuate experimental NEC, and we sought to determine the mechanisms involved. We show here that microinjection of amniotic fluid into the fetal (embryonic day 18.5) gastrointestinal tract reduced LPS-mediated signaling within the fetal intestinal mucosa. Amniotic fluid is abundant in EGF, which we show is required for its inhibitory effects on TLR4 signaling via peroxisome proliferator-activated receptor, because inhibition of EGF receptor (EGFR) with cetuximab or EGF-depleted amniotic fluid blocked the inhibitory effects of amniotic fluid on TLR4, whereas amniotic fluid did not prevent TLR4 signaling in EGFR- or peroxisome proliferator-activated receptor γ-deficient enterocytes or in mice deficient in intestinal epithelial EGFR, and purified EGF attenuated the exaggerated intestinal mucosal TLR4 signaling in wild-type mice. Moreover, amniotic fluid-mediated TLR4 inhibition reduced the severity of NEC in mice through EGFR activation. Strikingly, NEC development in both mice and humans was associated with reduced EGFR expression that was restored upon the administration of amniotic fluid in mice or recovery from NEC in humans, suggesting that a lack of amniotic fluid-mediated EGFR signaling could predispose to NEC. These findings may explain the unique susceptibility of premature infants to the development of NEC and offer therapeutic approaches to this devastating disease.


Sujet(s)
Liquide amniotique/métabolisme , Régulation de l'expression des gènes au cours du développement , Muqueuse intestinale/métabolisme , Récepteur de type Toll-4/métabolisme , Animaux , Lignée cellulaire , Entérocolite nécrosante/métabolisme , Entérocytes/métabolisme , Récepteurs ErbB/métabolisme , Humains , Nouveau-né , Muqueuse intestinale/embryologie , Intestins/embryologie , Souris , Microscopie confocale/méthodes , Transduction du signal , Facteurs temps
7.
J Immunol ; 188(9): 4543-57, 2012 May 01.
Article de Anglais | MEDLINE | ID: mdl-22461698

RÉSUMÉ

Necrotizing enterocolitis (NEC) is the leading cause of gastrointestinal-related mortality in premature infants, and it develops under conditions of exaggerated TLR4 signaling in the newborn intestinal epithelium. Because NEC does not develop spontaneously, despite the presence of seemingly tonic stimulation of intestinal TLR4, we hypothesized that mechanisms must exist to constrain TLR4 signaling that become diminished during NEC pathogenesis and focused on the intracellular stress response protein and chaperone heat shock protein-70 (Hsp70). We demonstrate that the induction of intracellular Hsp70 in enterocytes dramatically reduced TLR4 signaling, as assessed by LPS-induced NF-κB translocation, cytokine expression, and apoptosis. These findings were confirmed in vivo, using mice that either globally lacked Hsp70 or overexpressed Hsp70 within the intestinal epithelium. TLR4 activation itself significantly increased Hsp70 expression in enterocytes, which provided a mechanism of autoinhibition of TLR4 signaling in enterocytes. In seeking to define the mechanisms involved, intracellular Hsp70-mediated inhibition of TLR4 signaling required both its substrate-binding EEVD domain and association with the cochaperone CHIP, resulting in ubiquitination and proteasomal degradation of TLR4. The expression of Hsp70 in the intestinal epithelium was significantly decreased in murine and human NEC compared with healthy controls, suggesting that loss of Hsp70 protection from TLR4 could lead to NEC. In support of this, intestinal Hsp70 overexpression in mice and pharmacologic upregulation of Hsp70 reversed TLR4-induced cytokines and enterocyte apoptosis, as well as prevented and treated experimental NEC. Thus, a novel TLR4 regulatory pathway exists within the newborn gut involving Hsp70 that may be pharmacologically activated to limit NEC severity.


Sujet(s)
Protéines du choc thermique HSP70/immunologie , Muqueuse intestinale/immunologie , Transduction du signal/immunologie , Récepteur de type Toll-4/immunologie , Animaux , Apoptose/effets des médicaments et des substances chimiques , Apoptose/immunologie , Entérocolite nécrosante/immunologie , Entérocolite nécrosante/métabolisme , Entérocolite nécrosante/anatomopathologie , Femelle , Protéines du choc thermique HSP70/métabolisme , Humains , Nouveau-né , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Lipopolysaccharides/pharmacologie , Mâle , Souris , Facteur de transcription NF-kappa B/immunologie , Facteur de transcription NF-kappa B/métabolisme , Proteasome endopeptidase complex/immunologie , Proteasome endopeptidase complex/métabolisme , Structure tertiaire des protéines , Protéolyse/effets des médicaments et des substances chimiques , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/métabolisme , Ubiquitin-protein ligases/immunologie , Ubiquitin-protein ligases/métabolisme , Ubiquitination/effets des médicaments et des substances chimiques , Ubiquitination/immunologie
8.
Cell Mol Life Sci ; 68(22): 3623-34, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21986983

RÉSUMÉ

The maintenance of mucosal barrier equilibrium in the intestine requires a delicate and dynamic balance between enterocyte loss by apoptosis and the generation of new cells by proliferation from stem cell precursors at the base of the intestinal crypts. When the balance shifts towards either excessive or insufficient apoptosis, a broad range of gastrointestinal diseases can manifest. Recent work from a variety of laboratories has provided evidence in support of a role for receptors of the innate immune system, including Toll-like receptors 2, 4, and 9 as well as the intracellular pathogen recognition receptor NOD2/CARD15, in the initiation of enterocyte apoptosis. The subsequent induction of enterocyte apoptosis in response to the activation of these innate immune receptors plays a key role in the development of various intestinal diseases, including necrotizing enterocolitis, Crohn's disease, ulcerative colitis, and intestinal cancer. This review will detail the regulatory pathways that govern enterocyte apoptosis, and will explore the role of the innate immune system in the induction of enterocyte apoptosis in gastrointestinal disease.


Sujet(s)
Apoptose/immunologie , Entérocytes/physiologie , Maladies gastro-intestinales/anatomopathologie , Immunité innée/immunologie , Inflammation/anatomopathologie , Muqueuse intestinale/immunologie , Animaux , Entérocytes/cytologie , Maladies gastro-intestinales/immunologie , Humains , Inflammation/immunologie , Muqueuse intestinale/cytologie , Muqueuse intestinale/anatomopathologie , Facteur de transcription NF-kappa B/immunologie , Protéines adaptatrices de signalisation NOD/immunologie , Transduction du signal/immunologie , Récepteurs de type Toll/immunologie
9.
Am J Physiol Gastrointest Liver Physiol ; 301(3): G435-45, 2011 Sep.
Article de Anglais | MEDLINE | ID: mdl-21700903

RÉSUMÉ

Necrotizing enterocolitis (NEC) in preterm infants develops very rapidly from a mild intolerance to enteral feeding into intestinal mucosal hemorrhage, inflammation, and necrosis. We hypothesized that immediate feeding-induced gut responses precede later clinical NEC symptoms in preterm pigs. Fifty-six preterm pigs were fed total parenteral nutrition (TPN) for 48 h followed by enteral feeding for 0, 8, 17, or 34 h with either colostrum (Colos, n = 20) or formula (Form, n = 31). Macroscopic NEC lesions were detected in Form pigs throughout the enteral feeding period (20/31, 65%), whereas most Colos pigs remained protected (1/20, 5%). Just 8 h of formula feeding induced histopathological lesions, as evidenced by capillary stasis and necrosis, epithelial degeneration, edema, and mucosal hemorrhage. These immediate formula-induced changes were paralleled by decreased digestive enzyme activities (lactase and dipeptidylpeptidase IV), increased nutrient fermentation, and altered expression of innate immune defense genes such as interleukins (IL-1α, IL-6, IL-18), nitric oxide synthetase, tight junction proteins (claudins), Toll-like receptors (TLR-4), and TNF-α. In contrast, the first hours of colostrum feeding induced no histopathological lesions, increased maltase activity, and induced changes in gene expressions related to tissue development. Total bacterial density was high after 2 days of parenteral feeding and was not significantly affected by diet (colostrum, formula) or length of enteral feeding (8-34 h), except that a few bacterial groups (Clostridium, Enterococcus, Streptococcus species) increased with time. We conclude that a switch from parenteral to enteral nutrition rapidly induces diet-dependent histopathological, functional, and proinflammatory insults to the immature intestine. Great care is required when introducing enteral feeds to TPN-fed preterm infants, particularly when using formula, because early feeding-induced insults may predispose to NEC lesions that are difficult to revert by later dietary or medical interventions.


Sujet(s)
Nutrition entérale , Entérocolite nécrosante/anatomopathologie , Nutrition parentérale totale , Animaux , Colostrum , Entérocolite nécrosante/étiologie , Humains , Préparation pour nourrissons/pharmacologie , Nouveau-né , Prématuré , Suidae
10.
J Nutr Biochem ; 22(6): 511-21, 2011 Jun.
Article de Anglais | MEDLINE | ID: mdl-21193301

RÉSUMÉ

The gastrointestinal inflammatory disorder, necrotizing enterocolitis (NEC), is among the most serious diseases for preterm neonates. Nutritional, microbiological and immunological dysfunctions all play a role in disease progression but the relationship among these determinants is not understood. The preterm gut is very sensitive to enteral feeding which may either promote gut adaptation and health, or induce gut dysfunction, bacterial overgrowth and inflammation. Uncontrolled inflammatory reactions may be initiated by maldigestion and impaired mucosal protection, leading to bacterial overgrowth and excessive nutrient fermentation. Tumor necrosis factor alpha, toll-like receptors and heat-shock proteins are identified among the immunological components of the early mucosal dysfunction. It remains difficult, however, to distinguish the early initiators of NEC from the later consequences of the disease pathology. To elucidate the mechanisms and identify clinical interventions, animal models showing spontaneous NEC development after preterm birth coupled with different forms of feeding may help. In this review, we summarize the literature and some recent results from studies on preterm pigs on the nutritional, microbial and immunological interactions during the early feeding-induced mucosal dysfunction and later NEC development. We show that introduction of suboptimal enteral formula diets, coupled with parenteral nutrition, predispose to disease, while advancing amounts of mother's milk from birth (particularly colostrum) protects against disease. Hence, the transition from parenteral to enteral nutrition shortly after birth plays a pivotal role to secure gut growth, digestive maturation and an appropriate response to bacterial colonization in the sensitive gut of preterm neonates.


Sujet(s)
Entérocolite nécrosante/étiologie , Tube digestif/immunologie , Tube digestif/microbiologie , Phénomènes physiologiques nutritionnels chez le nourrisson , Maladies du prématuré/étiologie , Animaux , Animaux nouveau-nés , Entérocolite nécrosante/immunologie , Entérocolite nécrosante/microbiologie , Tube digestif/croissance et développement , Protéines du choc thermique/métabolisme , Humains , Système immunitaire/immunologie , Nouveau-né , Prématuré , Maladies du prématuré/immunologie , Maladies du prématuré/microbiologie , Muqueuse intestinale/immunologie , Muqueuse intestinale/microbiologie , Intestin grêle/métabolisme , Métagénome/physiologie
11.
Neonatology ; 99(4): 280-8, 2011.
Article de Anglais | MEDLINE | ID: mdl-21135563

RÉSUMÉ

BACKGROUND: In newborns, colonizing bacteria and enteral nutrition are important for early gut development and immunity. However, in preterm newborns, bacterial colonization, coupled with enteral feeding, can lead to marked intestinal inflammation and disease such as necrotizing enterocolitis (NEC). We hypothesized that the initial bacterial colonization of the gut affects the intestinal proteome independently of enteral feeding. OBJECTIVE: To identify the intestinal proteins affected by the first colonizing bacteria by comparing the intestinal proteome in formula-fed preterm pigs reared under germ free (GF) or conventional conditions. METHODS: Gel-based proteomics of the small intestine to detect proteins that may play a part in the response of the immature intestine to bacterial colonization after birth. RESULTS: Fourteen proteins involved in stress response and detoxification (e.g. heat-shock proteins, peroxiredoxin 1), tissue metabolism and apoptosis (e.g. annexin 2), and some signal transduction pathways were differentially expressed between GF and conventionally reared pigs. CONCLUSION: The premature intestine is highly responsive to initial bacterial colonization and the specific bacteria-related proteome changes may contribute to the stress response that makes the immature intestine sensitive to the pro-inflammatory effects of enteral feeding.


Sujet(s)
Bactéries/croissance et développement , Entérocolite nécrosante/métabolisme , Muqueuse intestinale/métabolisme , Naissance prématurée/médecine vétérinaire , Protéome/métabolisme , Animaux , Animaux nouveau-nés , Charge bactérienne/physiologie , Prédisposition aux maladies , Électrophorèse bidimensionnelle sur gel , Entérocolite nécrosante/étiologie , Entérocolite nécrosante/microbiologie , Entérocolite nécrosante/médecine vétérinaire , Femelle , Intestins/microbiologie , Grossesse , Protéome/analyse , Suidae , Maladies des porcs/étiologie , Maladies des porcs/métabolisme , Maladies des porcs/microbiologie
12.
Pediatr Res ; 69(3): 183-8, 2011 Mar.
Article de Anglais | MEDLINE | ID: mdl-21135755

RÉSUMÉ

Necrotizing enterocolitis (NEC) is the leading cause of death from gastrointestinal disease in the preterm infant. The dismal results of current treatment for NEC highlight the urgent need for greater understanding of the pathogenesis of this disease, and the importance of discovering novel, molecular-specific therapies for it. Current dogma indicates that NEC development reflects an abnormal response by the premature infant to the microbial flora that colonizes the gastrointestinal tract, although the mechanisms that mediate these abnormal bacterial-enterocyte interactions and the reasons for the particularly increased susceptibility of the premature infant to the development of NEC remain incompletely explained. Recent evidence has shed light on an emerging role for the Toll-like receptors (TLRs) of the innate immune system as central players in the pathways that signal in response to enteric bacteria resulting in the development of NEC. We now review recent advances in the field of NEC and identify several exciting potential avenues for novel treatments by focusing on abnormal TLR4 signaling in the premature intestine in the pathogenesis of NEC. In so doing, we seek to offer new hope to the patients and their families who are affected by this devastating disorder.


Sujet(s)
Entérocolite nécrosante/thérapie , Récepteurs de type Toll/métabolisme , Animaux , Entérocolite nécrosante/immunologie , Entérocolite nécrosante/microbiologie , Entérocolite nécrosante/anatomopathologie , Entérocytes/immunologie , Entérocytes/métabolisme , Entérocytes/microbiologie , Tube digestif/métabolisme , Tube digestif/microbiologie , Tube digestif/anatomopathologie , Humains , Immunité innée , Nouveau-né/immunologie , Prématuré/immunologie , Maladies du prématuré/immunologie , Maladies du prématuré/microbiologie , Maladies du prématuré/anatomopathologie , Maladies du prématuré/thérapie , Muqueuse intestinale/métabolisme , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Transduction du signal/physiologie
13.
J Nutr ; 141(2): 223-30, 2011 Feb.
Article de Anglais | MEDLINE | ID: mdl-21178092

RÉSUMÉ

Preterm birth and necrotizing enterocolitis (NEC) is associated with inappropriate gut colonization and immunity, which may be improved by probiotic bacteria. Using a preterm pig model of NEC, we investigated the effects of probiotics on intestinal structure, function, microbiology, and immunology in the immediate postnatal period. Just after birth, caesarean-delivered preterm pigs were inoculated with Lactobacillus paracasei, Bifidobacteria animalis, and Streptococcus thermophilus (total 2.4 × 10(10)/d) either as live (ProLive, n = 14) or gamma-irradiated dead bacteria (ProDead, n = 12) and compared with controls (n = 14). All pigs received parenteral nutrition for 2 d followed by enteral formula feeding until tissue collection on d 5. Compared with control pigs, intestinal weight was lower and NEC incidence was higher in both groups given probiotics (64-67 vs. 14%; P<0.01). Hexose absorption, brush border enzyme activities, and gut barrier function were lower in the ProDead group compared with the other groups (P < 0.05), whereas live probiotics induced higher expression of the proinflammatory cytokines IL-1α and IL-6 (P < 0.05). Probiotics minimally affected gut colonization, except that live probiotics induced a higher density of B. animalis and lower bacterial diversity in the distal intestinal mucosa and lower SCFA concentrations in the colon (P < 0.05). The detrimental effects of probiotic bacteria in this study may relate to the specific strain and dose combination and may have involved the very immature gut immune system and low NEC incidence in the control group. It remains to be determined whether similar adverse responses to probiotics occur in preterm infants.


Sujet(s)
Côlon/microbiologie , Entérocolite nécrosante/microbiologie , Muqueuse intestinale/microbiologie , Intestin grêle/microbiologie , Probiotiques/effets indésirables , Animaux , Bifidobacterium , Entérocolite nécrosante/métabolisme , Entérocolite nécrosante/anatomopathologie , Incidence , Absorption intestinale , Muqueuse intestinale/métabolisme , Muqueuse intestinale/anatomopathologie , Intestin grêle/métabolisme , Intestin grêle/anatomopathologie , Lactobacillus , Modèles animaux , Soutien nutritionnel , Taille d'organe , Streptococcus thermophilus , Suidae
14.
Gastroenterology ; 139(3): 904-17, 917.e1-6, 2010 Sep.
Article de Anglais | MEDLINE | ID: mdl-20580721

RÉSUMÉ

BACKGROUND & AIMS: Factors that regulate enterocyte apoptosis in necrotizing enterocolitis (NEC) remain incompletely understood, although Toll-like receptor-4 (TLR4) signaling in enterocytes plays a major role. Nucleotide-binding oligomerization domain-2 (NOD2) is an immune receptor that regulates other branches of the immune system, although its effects on TLR4 in enterocytes and its role in NEC remain unknown. We now hypothesize that activation of NOD2 in the newborn intestine inhibits TLR4, and that failure of NOD2 signaling leads to NEC through increased TLR4-mediated enterocyte apoptosis. METHODS: The effects of NOD2 on enterocyte TLR4 signaling and intestinal injury and repair were assessed in enterocytes lacking TLR4 or NOD2, in mice with intestinal-specific wild-type or dominant-negative TLR4 or NOD2, and in mice with NEC. A protein array was performed on NOD2-activated enterocytes to identify novel effector molecules involved. RESULTS: TLR4 activation caused apoptosis in newborn but not adult small intestine or colon, and its intestinal expression was influenced by NOD2. NOD2 activation inhibited TLR4 in enterocytes, but not macrophages, and reversed the effects of TLR4 on intestinal mucosal injury and repair. Protection from TLR4-induced enterocyte apoptosis by NOD2 required a novel pathway linking NOD2 with the apoptosis mediator second mitochondria-derived activator of caspase/direct inhibitor of apoptosis-binding protein with low PI (SMAC-DIABLO), both in vitro and in vivo. Strikingly, activation of NOD2 reduced SMAC-DIABLO expression, attenuated the extent of enterocyte apoptosis, and reduced the severity of NEC. CONCLUSIONS: These findings reveal a novel inhibitory interaction between TLR4 and NOD2 signaling in enterocytes leading to the regulation of enterocyte apoptosis and suggest a therapeutic role for NOD2 in the protection of intestinal diseases such as NEC.


Sujet(s)
Endotoxémie/métabolisme , Entérocolite nécrosante/métabolisme , Muqueuse intestinale/métabolisme , Protéine adaptatrice de signalisation NOD2/métabolisme , Transduction du signal , Récepteur de type Toll-4/métabolisme , Acétylmuramyl alanyl isoglutamine/pharmacologie , Facteurs âges , Animaux , Animaux nouveau-nés , Apoptose , Protéines régulatrices de l'apoptose , Protéines de transport/métabolisme , Lignée cellulaire , Mouvement cellulaire , Modèles animaux de maladie humaine , Endotoxémie/génétique , Endotoxémie/anatomopathologie , Entérocolite nécrosante/génétique , Entérocolite nécrosante/anatomopathologie , Entérocolite nécrosante/prévention et contrôle , Entérocytes/métabolisme , Humains , Muqueuse intestinale/effets des médicaments et des substances chimiques , Muqueuse intestinale/anatomopathologie , Souris , Souris de lignée C57BL , Souris knockout , Protéines mitochondriales/métabolisme , Facteur de transcription NF-kappa B/métabolisme , Protéine adaptatrice de signalisation NOD2/agonistes , Protéine adaptatrice de signalisation NOD2/déficit , Protéine adaptatrice de signalisation NOD2/génétique , Analyse par réseau de protéines , Rats , Indice de gravité de la maladie , Transduction du signal/effets des médicaments et des substances chimiques , Récepteur de type Toll-4/génétique , Transduction génétique
15.
Am J Physiol Gastrointest Liver Physiol ; 297(6): G1115-25, 2009 Dec.
Article de Anglais | MEDLINE | ID: mdl-19808655

RÉSUMÉ

Necrotizing enterocolitis (NEC) remains the most severe gastrointestinal disorder in preterm infants. It is associated with the initiation of enteral nutrition and may be related to immature carbohydrate digestive capacity. We tested the hypothesis that a formula containing maltodextrin vs. a formula containing lactose as the principal source of carbohydrate would predispose preterm pigs to a higher NEC incidence. Cesarean-derived preterm pigs were given total parenteral nutrition for 48 h followed by total enteral nutrition with a lactose-based (n = 11) or maltodextrin-based (n = 11) formula for 36 h. A higher incidence (91% vs. 27%) and severity (score of 3.3 vs. 1.8) of NEC were observed in the maltodextrin than in the lactose group. This higher incidence of NEC in the maltodextrin group was associated with significantly lower activities of lactase, maltase, and aminopeptidase; reduced villus height; transiently reduced in vivo aldohexose uptake; and reduced ex vivo aldohexose uptake capacity in the middle region of the small intestine. Bacterial diversity was low for both diets, but alterations in bacterial composition and luminal concentrations of short-chain fatty acids were observed in the maltodextrin group. In a second study, we quantified net portal absorption of aldohexoses (glucose and galactose) during acute jejunal infusion of a maltodextrin- or a lactose-based formula (n = 8) into preterm pigs. We found lower net portal aldohexose absorption (4% vs. 42%) and greater intestinal recovery of undigested carbohydrate (68% vs. 27%) in pigs acutely perfused with the maltodextrin-based formula than those perfused with the lactose-based formula. The higher digestibility of the lactose than the maltodextrin in the formulas can be attributed to a 5- to 20-fold higher hydrolytic activity of tissue-specific lactase than maltases. We conclude that carbohydrate maldigestion is sufficient to increase the incidence and severity of NEC in preterm pigs.


Sujet(s)
Phénomènes physiologiques nutritionnels chez l'animal , Digestion , Entérocolite nécrosante/physiopathologie , Intestins/physiopathologie , Aminopeptidases/métabolisme , Animaux , Animaux nouveau-nés , Césarienne , Modèles animaux de maladie humaine , Nutrition entérale , Entérocolite nécrosante/induit chimiquement , Entérocolite nécrosante/enzymologie , Entérocolite nécrosante/microbiologie , Galactose/métabolisme , Glucose/métabolisme , Humains , Hydrolyse , Préparation pour nourrissons/administration et posologie , Nouveau-né , Absorption intestinale , Intestins/enzymologie , Intestins/croissance et développement , Intestins/microbiologie , Lactase/métabolisme , Lactose/administration et posologie , Nutrition parentérale , Polyosides/administration et posologie , Naissance prématurée , Indice de gravité de la maladie , Suidae , Facteurs temps , alpha-Glucosidase/métabolisme
16.
J Nutr ; 138(8): 1437-44, 2008 Aug.
Article de Anglais | MEDLINE | ID: mdl-18641188

RÉSUMÉ

Following preterm birth, bacterial colonization and enteral formula feeding predispose neonates to gut dysfunction and necrotizing enterocolitis (NEC), a serious gastrointestinal inflammatory disease. We hypothesized that administration of probiotics would beneficially influence early bacterial colonization, thereby reducing the susceptibility to formula-induced gut atrophy, dysfunction, and NEC. Caesarean-delivered preterm pigs were provided total parenteral nutrition (1.5 d) followed by enteral feeding (2 d) with porcine colostrum (COLOS; n = 5), formula (FORM; n = 9), or formula with probiotics (FORM-P; Bifidobacterium animalis and Lactobacillus: L. acidophilus, L. casei, L. pentosus, L. plantarum; n = 13). Clinical NEC scores were reduced (P < 0.05) in FORM-P (2.0 +/- 0.2) and COLOS groups (1.7 +/- 0.5) compared with FORM pigs (3.4 +/- 0.6). Lower NEC scores were associated with elevated intestinal weight, mucosa proportion, villus height, RNA integrity, and brush border aminopeptidase A and N activities, and lower gastric organic acid concentration in the FORM-P and COLOS groups (P < 0.05). Diversity of the mucosa-associated bacteria in the distal small intestine was similar among formula-fed pigs, yet the abundance of specific bacterial groups differed between FORM-P and FORM pigs. FORM-P pigs had lower colonization density of a potential pathogen, Clostridium perfringens, and had commensal Lactobacillus bacteria more closely associated with enterocytes along the villus-crypt axis relative to FORM pigs. These results suggest that probiotic administration immediately after birth promotes the colonization of a beneficial commensal microbiota capable of limiting the formula-induced mucosal atrophy, dysfunction, and pathogen load in preterm neonates, thereby reducing the incidence and severity of NEC.


Sujet(s)
Entérocolite nécrosante/prévention et contrôle , Maladies gastro-intestinales/prévention et contrôle , Intestins/microbiologie , Probiotiques/usage thérapeutique , Animaux , Animaux nouveau-nés , Bifidobacterium/physiologie , Cytokines/génétique , Cytokines/métabolisme , Régime alimentaire/effets indésirables , Modèles animaux de maladie humaine , Calendrier d'administration des médicaments , Entérocolite nécrosante/traitement médicamenteux , Femelle , Maladies gastro-intestinales/traitement médicamenteux , Intestins/cytologie , Intestins/effets des médicaments et des substances chimiques , Intestins/enzymologie , Lactobacillus/physiologie , Mâle , Grossesse , Naissance prématurée , Probiotiques/administration et posologie , Répartition aléatoire , Suidae
17.
PLoS One ; 2(7): e677, 2007 Jul 25.
Article de Anglais | MEDLINE | ID: mdl-17653288

RÉSUMÉ

BACKGROUND: CCL25/TECK and CCL28/MEC are CC chemokines primarily expressed in thymic dendritic cells and mucosal epithelial cells. Their receptors, CCR9 and CCR10, are mainly expressed on T and B lymphocytes. In human, mouse, pig and sheep CCL25 and CCL28 play an important role in the segregation and the compartmentalization of the mucosal immune system. As evidenced by early comparisons of germ-free and conventional animals, the intestinal bacterial microflora has a marked effect on host intestinal immune functions. However, little is known about the impact of bacterial colonization on constitutive and induced chemokine expressions as well as on the generation of anti-inflammatory mechanisms. METHODOLOGY/PRINCIPAL FINDINGS: Therefore, we decided to focus by qPCR on the mRNA expression of two main gut chemokines, CCL25 and CCL28, their receptors CCR9 and CCR10, the Tregs marker Foxp3 and anti-inflammatory cytokines TGF-beta and IL-10 following colonization with different bacterial species within the small intestine. To accomplish this we used an original germ-free neonatal pig model and monoassociated pigs with a representative Gram-negative (Escherichia coli) or Gram-positive (Lactobacillus fermentum) commensal bacteria commonly isolated from the neonatal pig intestine. Our results show a consistent and marked effect of microbial colonization on the mRNA expression of intestinal chemokines, chemokine receptors, Foxp3 and TGF-beta. Moreover, as evidenced by in vitro experiments using two different cell lines, the pattern of regulation of CCL25 and CCL28 expression in the gut appears complex and suggests an additional role for in vivo factors. CONCLUSIONS/SIGNIFICANCE: Taken together, the results highlight the key role of bacterial microflora in the development of a functional intestinal immune system in an elegant and relevant model for human immune system development.


Sujet(s)
Chimiokines CC/génétique , Récepteurs CCR10/génétique , Récepteurs CCR/génétique , Animaux , Chimiokines/génétique , Escherichia coli/génétique , Facteurs de transcription Forkhead/génétique , Régulation de l'expression des gènes , Humains , Intestin grêle/physiologie , Souris , ARN messager/génétique , Ovis , Suidae , Lymphocytes T régulateurs/physiologie , Transcription génétique
18.
J Proteome Res ; 6(7): 2596-604, 2007 Jul.
Article de Anglais | MEDLINE | ID: mdl-17542629

RÉSUMÉ

The gastrointestinal tract harbors a complex community of bacteria, of which many may be beneficial. Studies of germ-free animal models have shown that the gastrointestinal microbiota not only assists in making nutrients available for the host but also contributes to intestinal health and development. We studied small intestinal protein expression patterns in gnotobiotic pigs maintained germ-free, or monoassociated with either Lactobacillus fermentum or non-pathogenic Escherichia coli. A common reference design in combination with labeling with stable isobaric tags allowed the individual comparison of 12 animals. Our results showed that bacterial colonization differentially affected mechanisms such as proteolysis, epithelial proliferation, and lipid metabolism, which is in good agreement with previous studies of other germ-free animal models. We have also found that E. coli has a profound effect on actin remodeling and intestinal proliferation, which may be related to stimulated migration and turnover of enterocytes. Regulations related to L. fermentum colonization involved individual markers for immunoregulatory mechanisms.


Sujet(s)
Escherichia coli/physiologie , Muqueuse intestinale/métabolisme , Intestins/microbiologie , Limosilactobacillus fermentum/physiologie , Protéome/métabolisme , Actines/métabolisme , Animaux , Prolifération cellulaire , Protéines/analyse , Protéines/métabolisme , Protéome/analyse , Sus scrofa/métabolisme , Sus scrofa/microbiologie
19.
Gastroenterology ; 130(6): 1776-92, 2006 May.
Article de Anglais | MEDLINE | ID: mdl-16697741

RÉSUMÉ

BACKGROUND & AIMS: Preterm birth and formula feeding are key risk factors associated with necrotizing enterocolitis (NEC) in infants, but little is known about intestinal conditions that predispose to disease. Thus, structural, functional, and microbiologic indices were used to investigate the etiology of spontaneous NEC development in preterm pigs. METHODS: Piglets were delivered by cesarean section at 92% gestation, reared in infant incubators, and fed infant formula or colostrum every 3 hours (n = 120) until tissue collection at 1-2 days of age. RESULTS: Clinical and histopathologic signs of NEC were observed in 57% of pigs fed FORMULA (26/46) and in 5% of pigs fed COLOSTRUM (2/38) (P < .05). Relative to COLOSTRUM, both healthy and sick FORMULA pigs had reduced intestinal villous heights, enzyme activities, nutrient absorption, and antioxidant levels and higher inducible nitric oxide synthetase activity (P < .05). In healthy pigs, mucosal microbial diversity remained low and diet independent. NEC pigs showed bacterial overgrowth, and a high mucosal density of Clostridium perfringens was detected in some but not all pigs. Germ-free conditions and antiserum against Clostridium perfringens toxin prevented intestinal dysfunction and NEC in formula-fed pigs, whereas the gut trophic factors, epidermal growth factor, and glucagon-like peptide 2 had limited effects. CONCLUSIONS: A subclinical, formula-induced mucosal atrophy and dysfunction predispose to NEC and bacterial overgrowth. The adverse feeding effects are colonization dependent and may be reduced by factors in colostrum that include antibodies against aggressive toxins such as those of Clostridium perfringens.


Sujet(s)
Entérocolite nécrosante/épidémiologie , Entérocolite nécrosante/prévention et contrôle , Préparation pour nourrissons/administration et posologie , Intestin grêle/métabolisme , Gestation animale , Animaux , Animaux nouveau-nés , Séquence nucléotidique , Ponction-biopsie à l'aiguille , Causalité , Numération de colonies microbiennes , Colostrum , ADN bactérien/analyse , Entérocolite nécrosante/anatomopathologie , Femelle , Tube digestif/microbiologie , Immunohistochimie , Absorption intestinale/physiologie , Muqueuse intestinale/microbiologie , Muqueuse intestinale/anatomopathologie , Intestin grêle/embryologie , Intestin grêle/anatomopathologie , Données de séquences moléculaires , Réaction de polymérisation en chaîne/méthodes , Grossesse , Naissance prématurée , Probabilité , Facteurs de risque , Sensibilité et spécificité , Suidae
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE
...