Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 7 de 7
Filter
Add more filters










Database
Language
Publication year range
1.
J Pediatr Gastroenterol Nutr ; 71(3): 393-400, 2020 09.
Article in English | MEDLINE | ID: mdl-32826717

ABSTRACT

OBJECTIVE: Determine the minimum dosage of alanyl-glutamine (Ala-Gln) required to improve gut integrity and growth in children at risk of environmental enteropathy (EE). METHODS: This was a double-blinded randomized placebo-controlled dose-response trial. We enrolled 140 children residing in a low-income community in Fortaleza, Brazil. Participants were 2 to 60 months old and had weight-for-age (WAZ), height-for-age (HAZ), or weight-for-height (WHZ) z-scores less than -1. We randomized children to 10 days of nutritional supplementation: Ala-Gln at 3 g/day, Ala-Gln at 6 g/day, Ala-Gln at 12 g/day, or an isonitrogenous dose of glycine (Gly) placebo at 12.5 g/day. Our primary outcome was urinary lactulose-mannitol excretion testing. Secondary outcomes were anthropometry, fecal markers of inflammation, urine metabolic profiles, and malabsorption (spot fecal energy). RESULTS: Of 140 children, 103 completed 120 days of follow-up (24% dropout). In the group receiving the highest dose of Ala-Gln, we detected a modest improvement in urinary lactulose excretion from 0.19% on day 1 to 0.17% on day 10 (P = 0.05). We observed significant but transient improvements in WHZ at day 10 in 2 Ala-Gln groups, and in WHZ and WAZ in all Ala-Gln groups at day 30. We detected no effects on fecal inflammatory markers, diarrheal morbidity, or urine metabolic profiles; but did observe modest reductions in fecal energy and fecal lactoferrin in participants receiving Ala-Gln. CONCLUSIONS: Intermediate dose Ala-Gln promotes short-term improvement in gut integrity and ponderal growth in children at risk of EE. Lower doses produced improvements in ponderal growth in the absence of enhanced gut integrity.


Subject(s)
Dipeptides , Nutritional Status , Brazil , Child , Child, Preschool , Glutamine , Humans , Infant , Inflammation
2.
Pediatr Infect Dis J ; 36(12): 1177-1185, 2017 Dec.
Article in English | MEDLINE | ID: mdl-28230705

ABSTRACT

Malnutrition results in serious consequences for growth and cognitive development in children. We studied select child and maternal biologic factors, socioeconomic factors, enteric pathogenic burden and gut function biomarkers in 402 children 6-24 months of age in Northeastern Brazil. In this prospective case-control study, not being fed colostrum [odds ratio (OR): 3.29, 95% confidence interval (CI): 1.73-6.26], maternal age ≥18 years (OR: 1.88, 95% CI: 1.10-3.22) and no electric fan (OR: 2.46, 95% CI: 1.22-4.96) or bicycle (OR: 1.80, 95% CI: 1.10-2.95) in the household were positively associated, and higher birth weight (OR: 0.27, 95% CI: 0.19-0.38), larger head circumference (OR: 0.74, 95% CI: 0.66-0.82) and shortness of breath in the last 2 weeks (OR: 0.49, 95% CI: 0.27-0.90) were negatively associated with malnutrition. Subclinical enteric pathogen infections were common, and enteroaggregative Escherichia coli infections were more prevalent in malnourished children (P = 0.045). Biomarkers such as the lactulose-mannitol test, myeloperoxidase, neopterin and calprotectin were highly elevated in both malnourished and nourished children. Nourished children had a better systemic immune response than the malnourished children, as detected by elevated serum amyloid A-1 and soluble cluster of differentiation protein 14 biomarkers (P < 0.001). Serum amyloid A-1 and soluble cluster of differentiation protein 14 were also associated with better nutritional Z scores. Neonatal, maternal and socioeconomic factors were associated with malnutrition in children. There was a substantial subclinical enteric pathogen burden, particularly with enteroaggregative E. coli, in malnourished children.


Subject(s)
Child Nutrition Disorders/epidemiology , Child Nutrition Disorders/physiopathology , Malnutrition/epidemiology , Malnutrition/physiopathology , Biomarkers/blood , Biomarkers/metabolism , Brazil/epidemiology , Case-Control Studies , Child Nutrition Disorders/metabolism , Child Nutrition Disorders/microbiology , Child, Preschool , Enteropathogenic Escherichia coli , Escherichia coli Infections , Fatty Acid-Binding Proteins/blood , Humans , Infant , Inflammation , Malnutrition/metabolism , Malnutrition/microbiology , Prospective Studies , Serum Amyloid A Protein/analysis
3.
PLoS One ; 11(9): e0158772, 2016.
Article in English | MEDLINE | ID: mdl-27690129

ABSTRACT

Critical to the design and assessment of interventions for enteropathy and its developmental consequences in children living in impoverished conditions are non-invasive biomarkers that can detect intestinal damage and predict its effects on growth and development. We therefore assessed fecal, urinary and systemic biomarkers of enteropathy and growth predictors in 375 6-26 month-old children with varying degrees of malnutrition (stunting or wasting) in Northeast Brazil. 301 of these children returned for followup anthropometry after 2-6m. Biomarkers that correlated with stunting included plasma IgA anti-LPS and anti-FliC, zonulin (if >12m old), and intestinal FABP (I-FABP, suggesting prior barrier disruption); and with citrulline, tryptophan and with lower serum amyloid A (SAA) (suggesting impaired defenses). In contrast, subsequent growth was predicted in those with higher fecal MPO or A1AT and also by higher L/M, plasma LPS, I-FABP and SAA (showing intestinal barrier disruption and inflammation). Better growth was predicted in girls with higher plasma citrulline and in boys with higher plasma tryptophan. Interactions were also seen with fecal MPO and neopterin in predicting subsequent growth impairment. Biomarkers clustered into markers of 1) functional intestinal barrier disruption and translocation, 2) structural intestinal barrier disruption and inflammation and 3) systemic inflammation. Principle components pathway analyses also showed that L/M with %L, I-FABP and MPO associate with impaired growth, while also (like MPO) associating with a systemic inflammation cluster of kynurenine, LBP, sCD14, SAA and K/T. Systemic evidence of LPS translocation associated with stunting, while markers of barrier disruption or repair (A1AT and Reg1 with low zonulin) associated with fecal MPO and neopterin. We conclude that key noninvasive biomarkers of intestinal barrier disruption, LPS translocation and of intestinal and systemic inflammation can help elucidate how we recognize, understand, and assess effective interventions for enteropathy and its growth and developmental consequences in children in impoverished settings.

4.
Am J Physiol Gastrointest Liver Physiol ; 308(10): G831-9, 2015 May 15.
Article in English | MEDLINE | ID: mdl-25792564

ABSTRACT

L-glutamine (Gln) is a key metabolic fuel for intestinal epithelial cell proliferation and survival and may be conditionally essential for gut homeostasis during catabolic states. We show that L-alanyl-L-glutamine (Ala-Gln), a stable Gln dipeptide, protects mice against jejunal crypt depletion in the setting of dietary protein and fat deficiency. Separately, we show that murine crypt cultures (enteroids) derived from the jejunum require Gln or Ala-Gln for maximal expansion. Once expanded, enteroids deprived of Gln display a gradual atrophy of cryptlike domains, with decreased epithelial proliferation, but stable proportions of Paneth and goblet cell differentiation, at 24 h. Replenishment of enteroid medium with Gln selectively activates mammalian target of rapamycin (mTOR) signaling pathways, rescues proliferation, and promotes crypt regeneration. Gln deprivation beyond 48 h leads to destabilization of enteroids but persistence of EGFP-Lgr5-positive intestinal stem cells with the capacity to regenerate enteroids upon Gln rescue. Collectively, these findings indicate that Gln deprivation induces a reversible quiescence of intestinal stem cells and provides new insights into nutritional regulation of intestinal epithelial homeostasis.


Subject(s)
Dipeptides/metabolism , Glutamine/metabolism , Intestinal Mucosa/cytology , Intestinal Mucosa/metabolism , Stem Cells/cytology , Stem Cells/metabolism , TOR Serine-Threonine Kinases/metabolism , Animals , Cell Differentiation/physiology , Cells, Cultured , Epithelial Cells , Female , Male , Mice , Mice, Inbred C57BL , Signal Transduction/physiology
5.
Vaccine ; 32(1): 48-53, 2013 Dec 17.
Article in English | MEDLINE | ID: mdl-24200975

ABSTRACT

BACKGROUND: Conflicting evidence links malnutrition to the reduced efficacy of rotavirus vaccines in developing countries, where diarrhea and undernutrition remain leading causes of child deaths. Here, we adapted mouse models of rotavirus vaccination (rhesus rotavirus, RRV), rotavirus infection (EDIM), and protein-energy malnutrition (PEM) to test the hypothesis that undernutrition reduces rotavirus vaccine immunogenicity and efficacy. METHODS: We randomized wild type Balb/C dams with 3-day-old pups to a control diet (CD) or an isocaloric, multideficient regional basic diet (RBD) that produces PEM. At 3 weeks of age, we weaned CD and RBD pups to their dams' diet and subrandomized weanlings to receive a single dose of either live oral rotavirus vaccine (RRV) or PBS. At 6 weeks of age, we orally challenged all groups with murine rotavirus (EDIM). Serum and stool specimens were collected before and after RRV and EDIM administration to measure viral shedding and antibody responses by ELISA. RESULTS: RBD pups and weanlings exhibited significant failure to thrive compared to age-matched CD mice (P<.0001). RRV vaccination induced higher levels of serum anti-RV IgA responses in RBD vs. CD mice (P<.0001). Vaccination protected CD and RBD mice equally against EDIM infection, as measured by viral shedding. In unvaccinated RBD mice, EDIM shedding peaked 1 day earlier (P<.05), however we detected no effects of undernutrition on viral clearance nor of infection on bodyweight. EDIM infection provoked higher anti-RV serum IgA levels in RBD vs. CD mice, regardless of vaccination (P<.0001). Last, RRV vaccination mitigated stool IgA responses to EDIM more in CD vs. RBD mice (P<.0001). CONCLUSIONS: Despite modulated IgA responses to vaccination and infection, undernutrition does not impair rotavirus vaccine efficacy nor exacerbate infection in this mouse model of protein-energy malnutrition. Alternative models are needed to elucidate host-pathogen factors undermining rotavirus vaccine effectiveness in high-risk global settings.


Subject(s)
Immunoglobulin A/immunology , Protein-Energy Malnutrition/immunology , Rotavirus Infections/immunology , Rotavirus Infections/prevention & control , Rotavirus Vaccines/immunology , Rotavirus/immunology , Animals , Antibodies, Viral/blood , Antibodies, Viral/immunology , Diet , Disease Models, Animal , Immunization , Immunoglobulin G/blood , Immunoglobulin G/immunology , Mice
6.
Inflammopharmacology ; 17(4): 221-8, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19609650

ABSTRACT

The effect of lupeol, a natural pentacyclic triterpene on ethanol-induced gastric damage in mice was evaluated. The gastroprotection was assessed by determination of changes in mean gastric lesion area, quantification of mucosal non-protein sulfhydryls (NP-SH), and characterized using drugs that influence the endogenous prostaglandins, alpha(2)-adrenoceptors, nitric oxide, K(ATP)-channels, and intracellular calcium. Orally administered lupeol (3, 10, and 30 mg/kg) significantly and dose-dependently attenuated the ethanol-induced gastric damage by 39-69%, whereas the positive control N-acetylcysteine (NAC, 300 mg/kg, i.p.) afforded 32% protection. Both lupeol and NAC restored the NP-SH depleted by ethanol but the lupeol effect was only marginal. Lupeol gastroprotection was attenuated by indomethacin and L-NAME, the respective COX and NO-synthase inhibitors and was weakly sensitive to alpha(2)-adrenergic antagonist yohimbine and K(ATP)-channel blocker glibenclamide, but more profoundly to calcium blocker verapamil. These pharmacological effects of lupeol may synergistically contribute to alleviating the ethanol-associated gastric damage, which is multifactorial.


Subject(s)
Anti-Inflammatory Agents/pharmacology , Ethanol/toxicity , Gastric Mucosa/drug effects , Triterpenes/pharmacology , Acetylcysteine/pharmacology , Animals , Anti-Inflammatory Agents/administration & dosage , Calcium/metabolism , Central Nervous System Depressants/toxicity , Dose-Response Relationship, Drug , Gastric Mucosa/pathology , KATP Channels/drug effects , KATP Channels/metabolism , Male , Mice , Nitric Oxide/metabolism , Pentacyclic Triterpenes , Prostaglandins/metabolism , Rats , Rats, Wistar , Receptors, Adrenergic, alpha-2/drug effects , Receptors, Adrenergic, alpha-2/metabolism , Sulfhydryl Compounds/metabolism , Triterpenes/administration & dosage
7.
Planta Med ; 73(13): 1372-6, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17918041

ABSTRACT

In search of novel gastroprotective agents, mangiferin, a naturally occurring glucosylxanthone from Mangifera indica L. (Anacardiaceae), was evaluated in mice on gastric injury induced by ethanol and indomethacin. The effects of mangiferin on gastric mucosal damage were assessed by determination of changes in mean gastric lesion area or ulcer score in mice and on gastric secretory volume and total acidity in 4-h pylorus-ligated rats. Mangiferin (3, 10 and 30 mg/kg, P. O.) significantly attenuated the gastric damage induced by ethanol by 30, 35, and 63 %, and of indomethacin by 22, 23 and 57 %, respectively. N-Acetylcysteine (750 mg/kg, I. P.) and lansoprazole (30 mg/kg, P. O.) used as positive controls in these ulcerogenic models resulted in 50 % and 76 % suppression of gastric injury, respectively. In 4-h pylorus-ligated rats, intraduodenally applied mangiferin (30 mg/kg) caused significant diminutions in gastric secretory volume and total acidity. In addition, like N-acetylcysteine, a donor of sulfhydryls, mangiferin effectively prevented the ethanol-associated depletion of gastric mucosal non-protein sulfhydryl content in mice, suggesting an antioxidant action. These findings provide evidence that mangiferin affords gastroprotection against gastric injury induced by ethanol and indomethacin most possibly through the antisecretory and antioxidant mechanisms of action.


Subject(s)
Anti-Ulcer Agents/pharmacology , Mangifera , Phytotherapy , Stomach Ulcer/prevention & control , Xanthones/pharmacology , Animals , Anti-Ulcer Agents/administration & dosage , Anti-Ulcer Agents/therapeutic use , Dose-Response Relationship, Drug , Ethanol , Gastric Acid , Hydrogen-Ion Concentration , Indomethacin , Ligation , Male , Mice , Plant Bark , Plant Extracts/administration & dosage , Plant Extracts/pharmacology , Plant Extracts/therapeutic use , Rats , Rats, Wistar , Stomach Ulcer/chemically induced , Xanthones/administration & dosage , Xanthones/therapeutic use
SELECTION OF CITATIONS
SEARCH DETAIL
...