Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 8 de 8
Filter
Add more filters










Database
Language
Publication year range
1.
Adv Nutr ; 11(2): 200-215, 2020 03 01.
Article in English | MEDLINE | ID: mdl-31386148

ABSTRACT

While conventional nutrition research has yielded biomarkers such as doubly labeled water for energy metabolism and 24-h urinary nitrogen for protein intake, a critical need exists for additional, equally robust biomarkers that allow for objective assessment of specific food intake and dietary exposure. Recent advances in high-throughput MS combined with improved metabolomics techniques and bioinformatic tools provide new opportunities for dietary biomarker development. In September 2018, the NIH organized a 2-d workshop to engage nutrition and omics researchers and explore the potential of multiomics approaches in nutritional biomarker research. The current Perspective summarizes key gaps and challenges identified, as well as the recommendations from the workshop that could serve as a guide for scientists interested in dietary biomarkers research. Topics addressed included study designs for biomarker development, analytical and bioinformatic considerations, and integration of dietary biomarkers with other omics techniques. Several clear needs were identified, including larger controlled feeding studies, testing a variety of foods and dietary patterns across diverse populations, improved reporting standards to support study replication, more chemical standards covering a broader range of food constituents and human metabolites, standardized approaches for biomarker validation, comprehensive and accessible food composition databases, a common ontology for dietary biomarker literature, and methodologic work on statistical procedures for intake biomarker discovery. Multidisciplinary research teams with appropriate expertise are critical to moving forward the field of dietary biomarkers and producing robust, reproducible biomarkers that can be used in public health and clinical research.


Subject(s)
Biomarkers/analysis , Diet , Metabolomics/methods , Biomarkers/blood , Biomarkers/urine , Food , Genomics , Humans , Metagenomics , Nutritional Physiological Phenomena/genetics , Nutritional Sciences/methods , Nutritional Status , Reproducibility of Results
2.
Clin Pharmacol Drug Dev ; 6(2): 176-185, 2017 Mar.
Article in English | MEDLINE | ID: mdl-28263462

ABSTRACT

Human metabolic disease opens a new view to understanding the contribution of the intestinal microbiome to drug metabolism and drug-induced toxicity in gut-liver function. The gut microbiome, a key determinant of intestinal inflammation, also plays a direct role in chronic inflammation and liver disease. Gut bacterial communities directly metabolize certain drugs, reducing their bioavailability and influencing individual variation in drug response. In addition, some microbiome-produced compounds may affect drug pharmacokinetics and pharmacodynamics via altered expression of metabolizing enzymes and drug transporters or genes coding for drug target proteins, drug response phenotypes, and disease states. Molecular-based high-throughput technologies are providing novel insight about host-gut microbiome interactions, homeostasis, and xenobiotic effects associated with wide variation in efficacy or toxicity in humans. It is envisioned that future approaches to treating and preventing liver disease will benefit from in-depth studies of the liver-microbiome axis. Thus, the microbiome shares a fundamental role in human physiology with various organ systems, and its importance must be considered in the rapid evolution of precision medicine. A new emerging perspective of understanding the effect of the gut microbiome on human response to drugs would be indispensable for developing efficacious, safe, and cost-effective precision therapies.


Subject(s)
Intestinal Mucosa/metabolism , Liver/metabolism , Microbiota , Animals , Drug-Related Side Effects and Adverse Reactions , Humans , Pharmaceutical Preparations/metabolism , Precision Medicine
3.
J Nutr ; 145(5): 1123S-1130S, 2015 May.
Article in English | MEDLINE | ID: mdl-25833886

ABSTRACT

Homo sapiens harbor trillions of microbes, whose microbial metagenome (collective genome of a microbial community) using omic validation interrogation tools is estimated to be at least 100-fold that of human cells, which comprise 23,000 genes. This article highlights some of the current progress and open questions in nutrition-related areas of microbiome research. It also underscores the metabolic capabilities of microbial fermentation on nutritional substrates that require further mechanistic understanding and systems biology approaches of studying functional interactions between diet composition, gut microbiota, and host metabolism. Questions surrounding bacterial fermentation and degradation of dietary constituents (particularly by Firmicutes and Bacteroidetes) and deciphering how microbial encoding of enzymes and derived metabolites affect recovery of dietary energy by the host are more complex than previously thought. Moreover, it is essential to understand to what extent the intestinal microbiota is subject to dietary control and to integrate these data with functional metabolic signatures and biomarkers. Many lines of research have demonstrated the significant role of the gut microbiota in human physiology and disease. Probiotic and prebiotic products are proliferating in the market in response to consumer demand, and the science and technology around these products are progressing rapidly. With high-throughput molecular technologies driving the science, studying the bidirectional interactions of host-microbial cometabolism, epithelial cell maturation, shaping of innate immune development, normal vs. dysfunctional nutrient absorption and processing, and the complex signaling pathways involved is now possible. Substantiating the safety and mechanisms of action of probiotic/prebiotic formulations is critical. Beneficial modulation of the human microbiota by using these nutritional and biotherapeutic strategies holds considerable promise as next-generation drugs, vaccinomics, and metabolic agents and in novel food discovery.


Subject(s)
Biomedical Research , Diet/adverse effects , Global Health , Intestinal Mucosa/microbiology , Metabolomics , Microbiology , Microbiota , Animals , Biomedical Research/trends , Congresses as Topic , Humans , Intestinal Mucosa/metabolism , Metabolomics/trends , Microbiology/trends , Prebiotics/adverse effects , Probiotics/adverse effects
4.
Dig Dis Sci ; 49(4): 579-89, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15185861

ABSTRACT

The aim of the present study was to examine the effects of oral supplementation of newborn Balb/c mice with bifidobacteria (B. infantis, B. bifidum) and iron-free apo-lactoferrin (bovine, human) on gut endotoxin concentration and mucosal immunity. Endotoxin concentration was measured in ileocecal filtrates at 7, 14, 21, and 28 days postdelivery by a quantitative limulus amebocyte lysate test. While endotoxin levels in bifidobacteria-fed mice showed a steady rise over time, they were consistently lower than that observed in control animals. Results of lactoferrin supplementation varied depending on the specific time point, but overall by day 28, all treatment groups showed lower intestinal endotoxin concentrations compared to saline fed animals. Neither bifidobacteria nor lactoferrin stimulated an increase in B or T cells, or in cytokine production (IL-6, TNF-alpha, INF-gamma), in Peyer's patches as measured by flow cytometry. Bifidobacteria and lactoferrin were well tolerated as dietary supplements and showed promising potential to reduce gut endotoxin levels.


Subject(s)
Bifidobacterium , Endotoxins/metabolism , Immunity, Mucosal/physiology , Intestines/drug effects , Intestines/immunology , Lactoferrin/pharmacology , Administration, Oral , Analysis of Variance , Animals , Biomarkers , Culture Techniques , Endotoxins/analysis , Enzyme-Linked Immunosorbent Assay , Female , Flow Cytometry , Interferon-gamma/analysis , Interleukin-6/analysis , Intestinal Mucosa/immunology , Intestinal Mucosa/microbiology , Limulus Test , Mice , Mice, Inbred BALB C , Pregnancy , Pregnancy, Animal , Probability , Reference Values , Risk Factors , Sensitivity and Specificity , Tumor Necrosis Factor-alpha/analysis
5.
Clin Pediatr (Phila) ; 42(7): 603-11, 2003 Sep.
Article in English | MEDLINE | ID: mdl-14552519

ABSTRACT

Vaccination compliance rates were calculated for 1995 to 2001 for enrolled patients, based on the Centers for Disease Control and Prevention guidelines and age-appropriate vaccine schedules. The results reported here indicate computerized tracking with the Doctor's Pediatric Immunization Program (Dr. PIP) maintained vaccine compliance rates (> 90%) in healthy and immunocompromised children at 2 months and 12 months of age. Instituting the computerized system has yielded nearly optimal results in both indigenous inner-city clinics. Despite the efficient progress made by automated tracking, the results for specific vaccine strategies (Varicella) and target groups (human immunodeficiency virus, high-risk indigent populations) may require on-going and intensive educational efforts to achieve optimization levels.


Subject(s)
Medical Records Systems, Computerized/statistics & numerical data , Patient Compliance , Vaccination , Adolescent , Adult , Child , Child, Preschool , Hospitals, Urban , Humans , Infant
6.
Dig Dis Sci ; 48(7): 1324-32, 2003 Jul.
Article in English | MEDLINE | ID: mdl-12870790

ABSTRACT

A series of in vitro experiments was performed to test the ability of bovine and human lactoferrin to influence the growth of the gram-positive probiotic bacteria, Bifidobacterium bifidum, Bifidobacterium infantis, and Lactobacillus acidophilus, as well as the gram-negative enteric bacteria, E. coli O157:H7 and Salmonella typhimurium. None of the lactoferrin preparations stimulated the growth of the tested strains. However, iron-free apo-lactoferrin (bovine and human) and 66% iron-saturated bovine lactoferrin dramatically slowed the growth of E. coli O157:H7 in single culture experiments, while 98% iron-saturated preparations had no effect. In coculture experiments of B. infantis and E. coli, the iron-limited preparations of lactoferrin also slowed the growth of the latter without inhibiting the bifidobacteria. These results suggest that lactoferrin in iron-limited forms may have the potential to be combined with probiotic bacteria in biotherapeutic products, which could help balance human gut microflora and limit the overgrowth of certain enteric microorganisms.


Subject(s)
Bifidobacterium/drug effects , Escherichia coli/drug effects , Lactobacillus acidophilus/drug effects , Lactoferrin/pharmacology , Salmonella typhimurium/drug effects , Animals , Bifidobacterium/growth & development , Cattle , Escherichia coli/growth & development , Humans , Iron Compounds/pharmacology , Lactobacillus acidophilus/growth & development , Salmonella typhimurium/growth & development
7.
Pediatr Res ; 51(6): 750-5, 2002 Jun.
Article in English | MEDLINE | ID: mdl-12032272

ABSTRACT

Bifidobacterium species (B. bifidum and B. infantis), with or without prebiotic compounds (arabino-galactan, short-chain fructo-oligosaccharide, iso-malto-dextrins), were orally fed to Balb/c pups (n = 192) to evaluate their potential synergistic effects on modulating the course of rhesus rotavirus (RRV) infection, as well as their ability to mediate the associated mucosal and humoral immune responses. Rotavirus-specific IgA and IgG in serum, rotavirus antigen, and specific IgA in feces were measured by ELISA. Mucosal total IgA and IgG levels were determined in Peyer's patches by flow cytometry. Significantly delayed onset (p = 0.001) and early resolution (p < 0.001) of diarrhea were observed in bifidobacteria-treated, RRV-infected mice compared with RRV-infected control mice. Supplementation with prebiotic compounds did not shorten the clinical diarrhea course more than that observed with bifidobacteria treatment alone. Rotavirus-specific IgA in feces was 16-fold elevated on d 5 postinfection in bifidobacteria-treated, RRV-infected mice compared with the RRV-infected alone group. In addition, the level of rotavirus-specific IgA in serum was four-fold higher in bifidobacteria-treated, RRV-infected litters versus mice challenged with RRV alone on 28 and 42 d postinfection. No enhancement of the immune response was found in RRV-infected mice that were treated with both bifidobacteria and prebiotic compounds over those treated with bifidobacteria only. The findings suggest that bifidobacteria may act as an adjuvant by modulating early mucosal and strong humoral rotavirus-specific immune responses, and mitigate severity of rotavirus-induced diarrhea.


Subject(s)
Bifidobacterium/immunology , Galactans/pharmacology , Oligosaccharides/pharmacology , Rotavirus Infections/drug therapy , Rotavirus Infections/immunology , Rotavirus/immunology , Animals , Antibodies, Viral/blood , Antigens, Viral/analysis , Dextrins/pharmacology , Drug Synergism , Enzyme-Linked Immunosorbent Assay , Feces/virology , Female , Flow Cytometry , Fructose/pharmacology , Immunoglobulin A/blood , Immunoglobulin G/blood , Isomaltose/pharmacology , Mice , Mice, Inbred BALB C , Pregnancy
8.
Am J Hum Biol ; 10(2): 211-219, 1998.
Article in English | MEDLINE | ID: mdl-28561447

ABSTRACT

Bacterial translocation and enteral feeding are factors implicated in neonatal necrotizing enterocolitis (NEC) in the preterm infant. A cohort of 60 preterm low birth-weight (LBW) infants (600-1,600 g at birth) consecutively admitted to the neonatal intensive care unit (NICU; N = 183) were prospectively followed to evaluate the role of bacterial endotoxins (lipopolysaccharides) and enteral feeding in the development of NEC. Stage I NEC was identified in 14/60 (23%) infants. In all, 15% (9/60) of infants followed, which represented roughly 5% of higher risk, LBW infants admitted to the NICU, progressed to Stage II or III NEC disease. Infants not enterally fed (nothing by mouth [NPO]) were at greatest risk of developing NEC. No infant who was breast milk fed progressed to Stage II or III NEC. The protective effect of breast milk was most evident when compared with the combined group of NPO or formula-feeding infants per person-week at risk (RR = .15, P < .04). Toxin-producing bacteria and endotoxin levels in stool filtrates predicted early and advanced stages of NEC disease. Cytokine concentrations (interleukin-6 [IL-6]) in stool appeared of limited value in reflecting mucosally limited disease in the gastrointestinal tract. Overgrowth of toxin-producing bacteria and their toxin products may adversely affect gut barrier function; monitoring endotoxin concentrations in stool filtrates may be most clinically useful in NPO and formula-fed infants identified at risk of developing NEC. Am. J. Hum. Biol. 10:211-219, 1998. © 1998 Wiley-Liss, Inc.

SELECTION OF CITATIONS
SEARCH DETAIL
...