1
|
Soliman A, Rodriguez-Vera L, Alarcia-Lacalle A, Pippa LF, Subhani S, Lukacova V, Duconge J, de Moraes NV, Vozmediano V. Leveraging Omeprazole PBPK/PD Modeling to Inform Drug-Drug Interactions and Specific Recommendations for Pediatric Labeling. Pharmaceutics 2025; 17:373. [PMID: 40143036 PMCID: PMC11944414 DOI: 10.3390/pharmaceutics17030373] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 03/05/2025] [Accepted: 03/06/2025] [Indexed: 03/28/2025] Open
Abstract
Background/Objectives: Omeprazole is widely used for managing gastrointestinal disorders like GERD, ulcers, and H. pylori infections. However, its use in pediatrics presents challenges due to drug interactions (DDIs), metabolic variability, and safety concerns. Omeprazole's pharmacokinetics (PK), primarily influenced by CYP2C19 metabolism, is affected by ontogenetic changes in enzyme expression, complicating dosing in children. Methods: This study aimed to develop and validate a physiologically based pharmacokinetic (PBPK) model for omeprazole and its metabolites to predict age-related variations in metabolism and response. Results: The PBPK model successfully predicted exposure to parent and metabolites in adults and pediatrics, incorporating competitive and mechanism-based inhibition of CYP2C19 and CYP3A4 by omeprazole and its metabolites. By accounting for age-dependent metabolic pathways, the model enabled priori predictions of omeprazole exposure in different age groups. Linking PK to the pharmacodynamics (PD) model, we described the impact of age-related physiological changes on intragastric pH, the primary outcome for proton pump inhibitors efficacy. Conclusions: The PBPK-PD model allowed for the virtual testing of dosing scenarios, providing an alternative to clinical studies in pediatrics where traditional DDI studies are challenging. This approach offers valuable insights for accurate dosing recommendations in pediatrics, accounting for age-dependent variability in metabolism, and underscores the potential of PBPK modeling in guiding pediatric drug development.
Collapse
Affiliation(s)
- Amira Soliman
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA (L.R.-V.); (N.V.d.M.)
- Department of Pharmacy Practice, Faculty of Pharmacy, Helwan University, Helwan, Cairo 11795, Egypt
| | - Leyanis Rodriguez-Vera
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA (L.R.-V.); (N.V.d.M.)
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA
| | - Ana Alarcia-Lacalle
- Pharmacokinetic, Nanotechnology and Gene Therapy Group (PharmaNanoGene), Faculty of Pharmacy, Centro de Investigación Lascaray Ikergunea, University of the Basque Country UPV/EHU, 01006 Vitoria-Gasteiz, Spain;
- Bioaraba, Microbiology, Infectious Disease, Antimicrobial Agents, and Gene Therapy, 01009 Vitoria-Gasteiz, Spain
| | - Leandro F. Pippa
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA (L.R.-V.); (N.V.d.M.)
| | - Saima Subhani
- Simulation Plus, Inc., Lancaster, CA 93534, USA; (S.S.); (V.L.)
| | - Viera Lukacova
- Simulation Plus, Inc., Lancaster, CA 93534, USA; (S.S.); (V.L.)
| | - Jorge Duconge
- Department of Pharmaceutical Sciences, School of Pharmacy, University of Puerto Rico, Medical Sciences Campus, San Juan, PR 00936, USA;
| | - Natalia V. de Moraes
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA (L.R.-V.); (N.V.d.M.)
| | - Valvanera Vozmediano
- Center for Pharmacometrics and Systems Pharmacology, Department of Pharmaceutics, College of Pharmacy, University of Florida, Orlando, FL 32827, USA (L.R.-V.); (N.V.d.M.)
- Model Informed Development, CTI Laboratories, Covington, KY 41011, USA
| |
Collapse
|
2
|
Van der Veken M, Brouwers J, Parrott N, Augustijns P, Stillhart C. Investigating the effect of whey and casein proteins on drug solubility from a paediatric drug absorption perspective. Int J Pharm X 2024; 8:100290. [PMID: 40104183 PMCID: PMC11915521 DOI: 10.1016/j.ijpx.2024.100290] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 10/02/2024] [Accepted: 10/05/2024] [Indexed: 03/20/2025] Open
Abstract
Considering the predominantly milk-based diet of neonates and infants and their immature gastrointestinal digestion, milk proteins may affect drug behaviour and absorption in this population. Using in vitro models, this study investigated the impact of the representative milk proteins, whey and casein, on the solubility and permeation of the lipophilic model drugs spironolactone, clopidogrel and ritonavir. Drug solubility experiments revealed that the presence of milk proteins increased drug solubility. Next, permeation studies demonstrated that the same milk proteins reduced drug permeation across an artificial membrane. These results highlight the importance of the solubility-permeability interplay and indicate the effect of these proteins may be considered during (paediatric) drug development. Lastly, the findings underscore the importance of considering milk protein-drug interactions to optimize drug delivery strategies during (paediatric) drug development and especially for the youngest and most vulnerable part of this population.
Collapse
Affiliation(s)
- Matthias Van der Veken
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49-Box 921, 3000 Leuven, Belgium
| | - Joachim Brouwers
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49-Box 921, 3000 Leuven, Belgium
| | - Neil Parrott
- Pharmaceutical Sciences, Roche Pharma Research and Early Development, Roche Innovation Centre Basel, 4070 Basel, Switzerland
| | - Patrick Augustijns
- Drug Delivery and Disposition, KU Leuven, Gasthuisberg O&N II, Herestraat 49-Box 921, 3000 Leuven, Belgium
| | - Cordula Stillhart
- Pharmaceutical R&D, F. Hoffmann-La Roche Ltd., 4070 Basel, Switzerland
| |
Collapse
|
3
|
Thomassen G, Abrahamse E, Mischke M, Becker M, Bartke N, Knol J, Renes I. In vitro gastrointestinal lipid handling and bioaccessibility rate of infant formula with large phospholipid-coated lipid droplets are different from those of standard formula and closer to human milk. Food Hydrocoll 2024; 156:110336. [DOI: 10.1016/j.foodhyd.2024.110336] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
4
|
Huang JR, Song JR, Cai WS, Shao ZW, Zhou DY, Song L. Enhancing vitamin D 3 bioaccessibility: Unveiling hydrophobic interactions in soybean protein isolate and vitamin D 3 binding via an infant in vitro digestion model. Food Chem 2024; 451:139507. [PMID: 38696940 DOI: 10.1016/j.foodchem.2024.139507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 03/26/2024] [Accepted: 04/26/2024] [Indexed: 05/04/2024]
Abstract
In the domain of infant nutrition, optimizing the absorption of crucial nutrients such as vitamin D3 (VD3) is paramount. This study harnessed dynamic-high-pressure microfluidization (DHPM) on soybean protein isolate (SPI) to engineer SPI-VD3 nanoparticles for fortifying yogurt. Characterized by notable binding affinity (Ka = 0.166 × 105 L·mol-1) at 80 MPa and significant surface hydrophobicity (H0 = 3494), these nanoparticles demonstrated promising attributes through molecular simulations. During simulated infant digestion, the 80 MPa DHPM-treated nanoparticles showcased an impressive 74.4% VD3 bioaccessibility, delineating the pivotal roles of hydrophobicity, bioaccessibility, and micellization dynamics. Noteworthy was their traversal through the gastrointestinal tract, illuminating bile salts' crucial function in facilitating VD3 re-encapsulation, thereby mitigating crystallization and augmenting absorption. Moreover, DHPM treatment imparted enhancements in nanoparticle integrity and hydrophobic properties, consequently amplifying VD3 bioavailability. This investigation underscores the potential of SPI-VD3 nanoparticles in bolstering VD3 absorption, thereby furnishing invaluable insights for tailored infant nutrition formulations.
Collapse
Affiliation(s)
- Jia-Rong Huang
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Jing-Ru Song
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Wan-Shuang Cai
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Zhen-Wen Shao
- Qingdao Seawit Life Science Co., Ltd., Qingdao 370200, China
| | - Da-Yong Zhou
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China
| | - Liang Song
- SKL of Marine Food Processing & Safety Control, National Engineering Research Center of Seafood, Collaborative Innovation Center of Seafood Deep Processing, Liaoning Province Key Laboratory for Marine Food Science and Technology, School of Food Science and Technology, Dalian Polytechnic University, Dalian 116034, China.
| |
Collapse
|
5
|
Rodriguez MD, León AE, Bustos MC. Co-ingestion of Cereals and Legumes during Infant Complementary Feeding: Starch and Protein in vitro Digestion. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2024; 79:489-496. [PMID: 38642194 DOI: 10.1007/s11130-024-01170-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 03/17/2024] [Indexed: 04/22/2024]
Abstract
This study explores the impact of co-ingesting cereals and legumes on starch and protein during simulated infant in vitro digestion. Various legumes (chickpeas, lentils, peas) were added to cereals (durum wheat, brown rice, white maize), and their effects on starch and protein hydrolysis were analyzed. Substituting 50% of cereal with legumes increased proteins, minerals, and dietary fiber. Infant food with legumes exhibited smoother pasting properties. Legumes in cereal purées led to varying starch hydrolysis trends, with the lowest values in durum wheat with chickpea and all cereal blends with peas. Resistant starch levels exceeding 50% were found in infant food samples. Digested protein hydrolysis increased with legumes in durum wheat, except for peas. Brown rice mixtures decreased significantly compared to the control with chickpeas (61%) and peas (42%), while lentil blends increased by 46%. Legumes generally did not significantly affect starch bioavailability, even with α-amylase inhibitors. Lentil-cereal purées could enhance infant food nutritional value.
Collapse
Affiliation(s)
- Marianela D Rodriguez
- Facultad de Ciencias Agropecuarias, Universidad Nacional de Córdoba (UNC), Córdoba, Córdoba, Argentina
| | - Alberto E León
- Facultad de Ciencias Agropecuarias, Universidad Nacional de Córdoba (UNC), Córdoba, Córdoba, Argentina
- Instituto de Ciencia y tecnología de los Alimentos Córdoba (ICyTAC), Conicet - UNC, Córdoba, Córdoba, Argentina
| | - Mariela C Bustos
- Instituto de Ciencia y tecnología de los Alimentos Córdoba (ICyTAC), Conicet - UNC, Córdoba, Córdoba, Argentina.
| |
Collapse
|
6
|
Li J, Zhu F. Whey protein hydrolysates and infant formulas: Effects on physicochemical and biological properties. Compr Rev Food Sci Food Saf 2024; 23:e13337. [PMID: 38578124 DOI: 10.1111/1541-4337.13337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2023] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 04/06/2024]
Abstract
Whey protein hydrolysates are recognized for their substantial functional and biological properties. Their high digestibility and amino acid composition make them a valuable ingredient to hydrolyzed whey infant formulas, enhancing both product functionality and nutritional values for infant growth. It is important to understand the functional and biological properties of whey protein hydrolysates for their applications in infant formula systems. This review explored preparation methods of whey protein hydrolysates for infant formula-based applications. The effects of whey protein hydrolysate on the physicochemical and biological properties of hydrolyzed whey infant formulas were summarized. The influences of whey protein hydrolysates on the functional and nutritional properties of formulas from manufacturing to infant consumption were discussed. Whey protein hydrolysates are crucial components in the preparation of infant formula, tailored to meet the functional and nutritional demands of the product. The selection of enzyme types and hydrolysis parameters is decisive for obtaining "optimal" whey protein hydrolysates that match the intended characteristics. "Optimal" whey protein hydrolysates offer diverse functionalities, including solubility, emulsification and production stability to hydrolyzed whey infant formulas during manufacturing processes and formulations. They simultaneously promote protein digestibility, infant growth and other potential health benefits, including reduced allergenic potential, as supported by in vitro, in vivo and clinical trials. Overall, the precise selection of enzymes and hydrolysis parameters in the production of whey protein hydrolysates is crucial in achieving the desired characteristics and functional benefits for hydrolyzed whey infant formulas, making them critical in the development of infant nutrition products.
Collapse
Affiliation(s)
- Jiecheng Li
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| | - Fan Zhu
- School of Chemical Sciences, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
7
|
Power ML, Muletz-Wolz CR, Bornbusch SL. Microbiome: Mammalian milk microbiomes: sources of diversity, potential functions, and future research directions. REPRODUCTION AND FERTILITY 2024; 5:e230056. [PMID: 38513351 PMCID: PMC11046322 DOI: 10.1530/raf-23-0056] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 03/18/2024] [Indexed: 03/23/2024] Open
Abstract
Abstract Milk is an ancient, fundamental mammalian adaptation that provides nutrition and biochemical communication to offspring. Microbiomes have been detected in milk of all species studied to date. In this review, we discuss: (a) routes by which microbes may enter milk; (b) evidence for proposed milk microbiome adaptive functions; (c) variation in milk microbiomes across mammals; and (d) future research directions, including suggestions for how to address outstanding questions on the viability and functionality of milk microbiomes. Milk microbes may be sourced from the maternal gastrointestinal tract, oral, skin, and mammary gland microbiomes and from neonatal oral and skin microbiomes. Given the variety of microbial sources, stochastic processes strongly influence milk microbiome assembly, but milk microbiomes appear to be influenced by maternal evolutionary history, diet, environment, and milk nutrients. Milk microbes have been proposed to colonize the neonatal intestinal tract and produce gene and metabolic products that influence physiology, metabolism, and immune system development. Limited epidemiological data indicate that early-life exposure to milk microbes can result in positive, long-term health outcomes. Milk microbiomes can be modified by dietary changes including providing the mother with probiotics and prebiotics. Milk replacers (i.e. infant formula) may benefit from supplementation with probiotics and prebiotics, but data are lacking on probiotics' usefulness, and supplementation should be evidence based. Overall, milk microbiome literature outside of human and model systems is scarce. We highlight the need for mechanistic studies in model species paired with comparative studies across mammals to further our understanding of mammalian milk microbiome evolution. A broader study of milk microbiomes has the potential to inform animal care with relevance to ex situ endangered species. Lay summary Milk is an ancient adaptation that supports the growth and development of mammalian neonates and infants. Beyond its fundamental nutritional function, milk influences all aspects of neonatal development, especially immune function. All kinds of milks so far studied have contained a milk microbiome. In this review, we focus on what is known about the collection of bacterial members found in milk microbiomes. Milk microbiomes include members sourced from maternal and infant microbiomes and they appear to be influenced by maternal evolutionary history, diet, milk nutrients, and environment, as well as by random chance. Once a neonate begins nursing, microbes from milk colonize their gut and produce byproducts that influence their physiology, metabolism, and immune development. Empirical data on milk microbiomes outside of humans and model systems are sparse. Greater study of milk microbiomes across mammals will expand our understanding of mammalian evolution and improve the health of animals under human care.
Collapse
Affiliation(s)
- Michael L Power
- Center for Species Survival, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| | - Carly R Muletz-Wolz
- Center for Conservation Genomics, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| | - Sally L Bornbusch
- Center for Conservation Genomics, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
- Department of Nutrition Science, Smithsonian’s National Zoo and Conservation Biology Institute, Washington, District of Columbia, USA
| |
Collapse
|
8
|
Jiang H, Xu Y, Chen G, Liu T, Yang Y, Mao X. Digestive properties and peptide profiles exhibited significant differences between skim camel milk and bovine milk powder after static in vitro simulated infant gastrointestinal digestion. Food Res Int 2024; 178:113860. [PMID: 38309893 DOI: 10.1016/j.foodres.2023.113860] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2023] [Revised: 12/09/2023] [Accepted: 12/14/2023] [Indexed: 02/05/2024]
Abstract
This study aims to analyze the differences in digestion properties and peptide profiles between the skim camel and bovine milk powder after static in vitro simulated infant gastrointestinal digestion. The hydrolysis degree of camel milk proteins exceeded by 13.18% that of bovine milk. The concentration and release rate of free amino groups in the camel milk digesta was higher than that of bovine milk powder, which was likely due to the higher β-/αs-casein ratio and larger casein micelle size in camel milk. Camel milk powder presented higher β-CN coverage and comparatively shorter bioactive peptides compared to bovine milk powder. The anti-inflammatory peptide KVLPVPQ displayed the highest abundance in camel milk powder. Outcomes of this study showed that camel milk proteins possessed superior digestibility and unique peptides, which outlined the potential nutritional implications of camel milk for infants.
Collapse
Affiliation(s)
- Hui Jiang
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yunxuan Xu
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Gangliang Chen
- Xinjiang Wangyuan Camel Milk Industrial Co., Ltd., Altay 836400, China
| | - Tianchong Liu
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Yitong Yang
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China
| | - Xueying Mao
- Key Laboratory of Functional Dairy, Ministry of Education, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100083, China.
| |
Collapse
|
9
|
Miltenburg J, Bastiaan-Net S, Hoppenbrouwers T, Wichers H, Hettinga K. Gastric clot formation and digestion of milk proteins in static in vitro infant gastric digestion models representing different ages. Food Chem 2024; 432:137209. [PMID: 37643515 DOI: 10.1016/j.foodchem.2023.137209] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2023] [Revised: 08/03/2023] [Accepted: 08/17/2023] [Indexed: 08/31/2023]
Abstract
Gastric digestion conditions change during infancy from newborn towards more adult digestion conditions, which can change gastric digestion kinetics. However, how these changes in gastric digestion conditions during infancy affect milk protein digestion has not been investigated. Therefore, we aimed to investigate milk protein digestion with static in vitro gastric digestion models representing one-, three- and six-month-old infants. With increasing age, gastric clots and soluble proteins were digested more extensively, which may partly be attributed to the looser gastric clot structure. Larger differences with increasing age were found for heated than unheated milk proteins, which might be caused by the presence of denatured whey proteins. Taken together, these findings show that gastric milk protein digestion increases during infancy. These in vitro gastric digestion models could be used to study how milk protein digestion changes with infant age, which may aid in developing infant formulas for different age stages.
Collapse
Affiliation(s)
- Julie Miltenburg
- Food Quality and Design, Wageningen University & Research, Wageningen, The Netherlands
| | - Shanna Bastiaan-Net
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Tamara Hoppenbrouwers
- Food Quality and Design, Wageningen University & Research, Wageningen, The Netherlands; Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Harry Wichers
- Wageningen Food & Biobased Research, Wageningen University & Research, Wageningen, The Netherlands
| | - Kasper Hettinga
- Food Quality and Design, Wageningen University & Research, Wageningen, The Netherlands.
| |
Collapse
|
10
|
Zhang W, Zhang Q, Cao Z, Zheng L, Hu W. Physiologically Based Pharmacokinetic Modeling in Neonates: Current Status and Future Perspectives. Pharmaceutics 2023; 15:2765. [PMID: 38140105 PMCID: PMC10747965 DOI: 10.3390/pharmaceutics15122765] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/07/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
Rational drug use in special populations is a clinical problem that doctors and pharma-cists must consider seriously. Neonates are the most physiologically immature and vulnerable to drug dosing. There is a pronounced difference in the anatomical and physiological profiles be-tween neonates and older people, affecting the absorption, distribution, metabolism, and excretion of drugs in vivo, ultimately leading to changes in drug concentration. Thus, dose adjustments in neonates are necessary to achieve adequate therapeutic concentrations and avoid drug toxicity. Over the past few decades, modeling and simulation techniques, especially physiologically based pharmacokinetic (PBPK) modeling, have been increasingly used in pediatric drug development and clinical therapy. This rigorously designed and verified model can effectively compensate for the deficiencies of clinical trials in neonates, provide a valuable reference for clinical research design, and even replace some clinical trials to predict drug plasma concentrations in newborns. This review introduces previous findings regarding age-dependent physiological changes and pathological factors affecting neonatal pharmacokinetics, along with their research means. The application of PBPK modeling in neonatal pharmacokinetic studies of various medications is also reviewed. Based on this, we propose future perspectives on neonatal PBPK modeling and hope for its broader application.
Collapse
Affiliation(s)
| | | | | | - Liang Zheng
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| | - Wei Hu
- Department of Clinical Pharmacology, The Second Affiliated Hospital of Anhui Medical University, Hefei 230601, China; (W.Z.); (Q.Z.); (Z.C.)
| |
Collapse
|
11
|
Barten LJC, Zuurveld M, Faber J, Garssen J, Klok T. Oral immunotherapy as a curative treatment for food-allergic preschool children: Current evidence and potential underlying mechanisms. Pediatr Allergy Immunol 2023; 34:e14043. [PMID: 38010006 DOI: 10.1111/pai.14043] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Revised: 10/10/2023] [Accepted: 10/19/2023] [Indexed: 11/29/2023]
Abstract
The worldwide rising prevalence of food allergy is a major public health concern. Standard care consists of allergen avoidance and rescue medication upon accidental exposure. Oral immunotherapy (OIT) is increasingly being studied as a treatment option. Although desensitization (an increased reaction threshold) is often achieved during OIT, sustained unresponsiveness (SU; clinical nonreactivity after finishing OIT) is not achieved in most patients. A few studies have investigated the effectiveness of OIT in children younger than 4 years of age (early = e-OIT) and have shown a much more favorable outcome in terms of SU development. Together with food allergy prevention studies, which have demonstrated high efficacy of early oral allergen exposure, the outcomes of e-OIT studies indicate an early-life window of opportunity to achieve SU, allowing unrestricted dietary intake. However, the underlying mechanism of the high effectiveness of e-OIT is not understood yet. Both cohort and OIT studies indicate early-life immune plasticity. An immature food-allergic response in the first years of life seems to be a major driver of this immune plasticity, along with a higher tolerogenic immunological state. Allergy maturation can likely be disrupted effectively by early intervention, preventing the development of persistent food allergy. Upcoming studies will provide important additional data on the safety, feasibility, and effectiveness of e-OIT. Combined with immune mechanistic studies, this should inform the implementation of e-OIT.
Collapse
Affiliation(s)
- Lieke J C Barten
- Pediatric Allergy Treatment Center, Deventer Hospital, Deventer, The Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Division Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Marit Zuurveld
- Utrecht Institute for Pharmaceutical Sciences, Division Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Joyce Faber
- Pediatric Allergy Treatment Center, Deventer Hospital, Deventer, The Netherlands
| | - Johan Garssen
- Utrecht Institute for Pharmaceutical Sciences, Division Pharmacology, Utrecht University, Utrecht, The Netherlands
| | - Ted Klok
- Pediatric Allergy Treatment Center, Deventer Hospital, Deventer, The Netherlands
| |
Collapse
|
12
|
Zhang Y, Zhou Q, Liu S, Quan X, Fang Z, Lin Y, Xu S, Feng B, Zhuo Y, Wu D, Che L. Partial Substitution of Whey Protein Concentrate with Spray-Dried Porcine Plasma or Soy Protein Isolate in Milk Replacer Differentially Modulates Ileal Morphology, Nutrient Digestion, Immunity and Intestinal Microbiota of Neonatal Piglets. Animals (Basel) 2023; 13:3308. [PMID: 37958063 PMCID: PMC10650022 DOI: 10.3390/ani13213308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 10/12/2023] [Accepted: 10/18/2023] [Indexed: 11/15/2023] Open
Abstract
Appropriate protein sources are vital for the growth, development and health of neonates. Twenty-four 2-day-old piglets were randomly divided into three groups and fed isoenergetic and isonitrogenous diets. The experimental diets included a milk replacer with 17.70% whey protein concentrate (WPC group), a milk replacer with 6% spray-dried porcine plasma isonitrogenously substituting WPC (SDPP group), and a milk replacer with 5.13% soy protein isolate isonitrogenously substituting WPC (SPI group). Neonatal piglets were fed milk replacer from postnatal day 2 (PND 2) to day 20 (PND 20). The growth performance, intestinal morphology, activities of digestive enzymes, plasma biochemical parameters, immunity-related genes, short-chain fatty acids (SCFA) and intestinal microbiota in the colonic chyme were determined. The results showed that SDPP-fed piglets had higher final BW (p = 0.05), ADG (p = 0.05) and F/G (p = 0.07) compared with WPC- and SPI-fed piglets, and SDPP-fed piglets had a lower diarrhea index (p < 0.01) from PND 2 to PND 8. SDPP-fed piglets had an increased ileal villus height (p = 0.04) and ratio of villus height to crypt depth (VCR) (p = 0.02), and increased activities of sucrase (p < 0.01), lactase (p = 0.02) and trypsin (p = 0.08) in the jejunum, compared with WPC- and SPI-fed piglets. Furthermore, SPI-fed piglets had an increased mRNA expression of IL-6 (p < 0.01) and concentration of plasma urea (p = 0.08). The results from LEfSe analysis showed that SDPP-fed piglets had a higher abundance of beneficial Butyricicoccus compared with WPC- and SPI-fed piglets, in which higher abundances of pathogenic bacteria such as Marinifilaceae, Fusobacterium and Enterococcus were observed. Moreover, SDPP-fed piglets had an increased concentration of butyric acid (p = 0.08) in the colonic chyme compared with WPC- and SPI-fed piglets. These results suggest that neonatal piglets fed milk replacer with SDPP partially substituting WPC had improved growth performance and intestinal morphology and function, associated with higher digestive enzyme activity and fewer pathogenic bacteria.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistant Nutrition of China Ministry of Education, Institute of Animal Nutrition, Sichuan Agricultural University, Chengdu 611130, China; (Y.Z.); (Q.Z.); (S.L.); (X.Q.); (Z.F.); (Y.L.); (S.X.); (B.F.); (Y.Z.); (D.W.)
| |
Collapse
|
13
|
Shen R, Yang D, Zhang L, Yu Q, Ma X, Ma G, Guo Z, Chen C. Preparation of Complementary Food for Infants and Young Children with Beef Liver: Process Optimization and Storage Quality. Foods 2023; 12:2689. [PMID: 37509781 PMCID: PMC10379101 DOI: 10.3390/foods12142689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2023] [Revised: 06/30/2023] [Accepted: 07/04/2023] [Indexed: 07/30/2023] Open
Abstract
In this study, fuzzy mathematics and response surface modeling were applied to optimize the preparation process of beef liver paste and characterize the proximate composition, sensory and physicochemical qualities, and in vitro simulated digestive properties while refrigerated at 0-4 °C (0, 3, 7, 15, 30, 45, and 60 days). The results showed that the optimal preparation process was 4.8% potato starch, 99.4% water, 10.2% olive oil, and a 3:2 ratio of chicken breast and beef liver. The beef liver paste prepared contained essential amino acids for infants and children, with a protein content of 10.29 g/100 g. During storage, the pH of the beef liver paste decreased significantly (p < 0.05) on day 7, texture and rheological properties decreased significantly after 30 days, a* values increased, L* and b* values gradually decreased, and TVB-N and TBARS values increased significantly (p < 0.05) on day 7 but were below the limit values during the storage period (TVB-N value ≤ 15 mg/100 g, TBARS value ≤ 1 mg/Kg). In vitro simulated digestion tests showed better digestibility and digestive characteristics in the first 15 days. The results of this study provide a reference for the development of beef liver products for infant and child supplementation.
Collapse
Affiliation(s)
- Ruheng Shen
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Dawei Yang
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Li Zhang
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Qunli Yu
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Xiaotong Ma
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Guoyuan Ma
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Zhaobin Guo
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| | - Cheng Chen
- College of Food Science and Engineering, Gansu Agriculture University, Lanzhou 730070, China
| |
Collapse
|
14
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
15
|
Lee S, Jo K, Jeong HG, Choi YS, Jung S. Changes in beef protein digestibility in an in vitro infant digestion model with prefreezing temperatures and aging periods. Heliyon 2023; 9:e15611. [PMID: 37153398 PMCID: PMC10160746 DOI: 10.1016/j.heliyon.2023.e15611] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2022] [Revised: 04/10/2023] [Accepted: 04/17/2023] [Indexed: 05/09/2023] Open
Abstract
The protein digestibility of beef at three prefreezing temperatures (freezing at -20 °C, F20; freezing at -50 °C, F50; and freezing at -70 °C, F70) and aging periods (4, 14, and 28 days) was investigated using an in vitro infant digestion model. The increased cathepsin B activity in the frozen-then-aged treatments (P < 0.05) resulted in a higher content of 10% trichloroacetic acid-soluble α-amino groups than in the aged-only group on days 14 and 28 (P < 0.05). F50 had the most α-amino groups in the digesta and digested proteins under 3 kDa on day 28 (P < 0.05), with the disappearance of actin band in the digesta electrophoretogram. The secondary and tertiary structures of myofibrillar proteins revealed that F50 underwent irreversible denaturation (P < 0.05), especially in the myosin fraction, while F20 and F70 showed protein renaturation during aging (P < 0.05). In general, prefreezing at -50 °C then aging can improve the in vitro protein digestibility of beef through freezing-induced structural changes.
Collapse
Affiliation(s)
- Seonmin Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Kyung Jo
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Hyun Gyung Jeong
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, 34134, South Korea
| | - Yun-Sang Choi
- Research Group of Food Processing, Korea Food Research Institute, Wanju, 55365, South Korea
| | - Samooel Jung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, 34134, South Korea
- Corresponding author.
| |
Collapse
|
16
|
Byrne ME, Arranz E, Bot F, Gómez-Mascaraque LG, Tobin JT, O’Mahony JA, O’Callaghan TF. The Protein Composition and In Vitro Digestive Characteristics of Animal- versus Plant-Based Infant Nutritional Products. Foods 2023; 12:foods12071469. [PMID: 37048290 PMCID: PMC10094249 DOI: 10.3390/foods12071469] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2023] [Revised: 03/17/2023] [Accepted: 03/24/2023] [Indexed: 03/31/2023] Open
Abstract
The protein composition and digestive characteristics of four commercially available infant formulae (IF) manufactured using bovine (B-IF), caprine (C-IF), soy (S-IF), and rice (R-IF) as a protein source were examined in this study. Plant-based formulae had significantly higher crude protein and non-protein nitrogen (NPN) concentrations. Static in vitro gastrointestinal digestion of these formulae, and subsequent analysis of their digestates, revealed significantly higher proteolysis of B-IF at the end of gastrointestinal digestion compared to the other formulae, as indicated by the significantly higher concentration of free amine groups. Furthermore, differences in structure formation during the gastric phase of digestion were observed, with formation of a more continuous, firmer coagulum by C-IF, while R-IF demonstrated no curd formation likely due to the extensive hydrolysis of these proteins during manufacture. Differences in digestive characteristics between formulae manufactured from these different protein sources may influence the bio-accessibility and bioavailability of nutrients, warranting additional study.
Collapse
|
17
|
Developmental Pharmacokinetics of Antibiotics Used in Neonatal ICU: Focus on Preterm Infants. Biomedicines 2023; 11:biomedicines11030940. [PMID: 36979919 PMCID: PMC10046592 DOI: 10.3390/biomedicines11030940] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2023] [Revised: 03/06/2023] [Accepted: 03/07/2023] [Indexed: 03/22/2023] Open
Abstract
Neonatal Infections are among the most common reasons for admission to the intensive care unit. Neonatal sepsis (NS) significantly contributes to mortality rates. Empiric antibiotic therapy of NS recommended by current international guidelines includes benzylpenicillin, ampicillin/amoxicillin, and aminoglycosides (gentamicin). The rise of antibacterial resistance precipitates the growth of the use of antibiotics of the Watch (second, third, and fourth generations of cephalosporines, carbapenems, macrolides, glycopeptides, rifamycins, fluoroquinolones) and Reserve groups (fifth generation of cephalosporines, oxazolidinones, lipoglycopeptides, fosfomycin), which are associated with a less clinical experience and higher risks of toxic reactions. A proper dosing regimen is essential for effective and safe antibiotic therapy, but its choice in neonates is complicated with high variability in the maturation of organ systems affecting drug absorption, distribution, metabolism, and excretion. Changes in antibiotic pharmacokinetic parameters result in altered efficacy and safety. Population pharmacokinetics can help to prognosis outcomes of antibiotic therapy, but it should be considered that the neonatal population is heterogeneous, and this heterogeneity is mainly determined by gestational and postnatal age. Preterm neonates are common in clinical practice, and due to the different physiology compared to the full terms, constitute a specific neonatal subpopulation. The objective of this review is to summarize the evidence about the developmental changes (specific for preterm and full-term infants, separately) of pharmacokinetic parameters of antibiotics used in neonatal intensive care units.
Collapse
|
18
|
Sabari S, Julmohammad N, Jahurul HAM, Matanjun P, Ab. Wahab N. In Vitro Infant Digestion of Whey Proteins Isolate-Lactose. Foods 2023; 12:667. [PMID: 36766193 PMCID: PMC9914322 DOI: 10.3390/foods12030667] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2022] [Revised: 12/31/2022] [Accepted: 01/06/2023] [Indexed: 02/05/2023] Open
Abstract
The model in vitro protein digestion technique has received greater attention due to providing significant advantages compared to in vivo experiments. This research employed an in vitro infant digestive static model to examine the protein digestibility of whey proteins isolate-lactose (WPI-Lac). The polyacrylamide gel electrophoresis (PAGE) pattern for alpha-lactalbumin of WPI at 60 min showed no detectable bands, while the alpha-lactalbumin of the WPI-Lac was completely digested after 5 min of gastric digestion. The beta-lactoglobulin of the WPI-Lac was found to be similar to the beta-lactoglobulin of the WPI, being insignificant at pH 3.0. The alpha-lactalbumin of the WPI decreased after 100 min of duodenal digestion at pH 6.5, and the WPI-Lac was completely digested after 60 min. The peptides were identified as ~2 kilodalton (kDa) in conjugated protein, which indicated that the level of degradation of the protein was high, due to the hydrolysis progress. The conjugated protein increased the responsiveness to digestive proteolysis, potentially leading to the release of immunogenic protein by lactose, and to the creation of hypoallergenic protein.
Collapse
Affiliation(s)
- Sarizan Sabari
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Norliza Julmohammad
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Haque Akanda Md Jahurul
- Department of Agriculture, School of Agriculture, University of Arkansas, 1200 North University Dr., M/S 4913, Pine Bluff, AR 71601, USA
| | - Patricia Matanjun
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| | - Noorakmar Ab. Wahab
- Faculty of Food Science and Nutrition, Universiti Malaysia Sabah, Kota Kinabalu 88400, Sabah, Malaysia
| |
Collapse
|
19
|
In vitro 1H MT and CEST MRI mapping of gastro-intestinal milk protein breakdown. FOOD STRUCTURE 2023. [DOI: 10.1016/j.foostr.2023.100314] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
|
20
|
Wang X, Roy D, Acevedo-Fani A, Ye A, Pundir S, Singh H. Challenges in simulating the biochemical environment of the infant stomach to assess the gastric digestion of infant formulae. Curr Opin Food Sci 2023. [DOI: 10.1016/j.cofs.2022.100984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
21
|
Choudhary D, Rideout TC, Cameron CE, Lehman HK, Oken E, Rifas-Shiman SL, Wen X. Egg Introduction during Infancy is Associated with Lower Fat Mass Index in Females at Early Adolescence. J Nutr 2023; 153:158-166. [PMID: 36913449 PMCID: PMC10196582 DOI: 10.1016/j.tjnut.2022.11.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2022] [Revised: 10/07/2022] [Accepted: 11/09/2022] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Egg consumption may play an important role in early-life growth given their high-quality protein, essential fatty acids, and micronutrients. OBJECTIVES Study objectives were to examine the longitudinal associations of infant age at egg introduction with obesity outcomes in early childhood, middle childhood (mid-childhood), and early adolescence. METHODS We used existing data from 1089 mother-child dyads from Project Viva to estimate age at egg introduction through a questionnaire completed by mothers at ∼1 y postpartum (mean ± SD, 13.3 ± 1.2 mo). Outcome measures included height and weight (early childhood, mid-childhood, and early adolescence), body composition including total fat mass, trunk fat mass, and lean mass (mid-childhood and early adolescence), and plasma adiponectin and leptin (early and mid-childhood and early adolescence). We defined childhood obesity as sex- and age-specific BMI ≥ 95th percentile. We estimated the associations of infant age at egg introduction with risk of obesity using multivariable logistic regression and multivariable linear regression models for BMI-z-score, body composition measures, and adiposity hormones; adjusted for maternal prepregnancy BMI and sociodemographics. RESULTS Among females, those introduced to egg by the 1-y survey had a lower total fat mass index (confounder-adjusted mean difference, -1.23 kg/m2; 95% CI: -2.14, -0.31), and trunk fat mass index (confounder-adjusted mean difference, -0.57 kg/m2; 95% CI: -1.01, -0.12) in early adolescence compared to those not introduced (reference group). However, no associations between infant age at egg introduction and risk of obesity were observed among males (confounder-adjusted odd ratio [aOR], 1.97; 95% CI: 0.90, 4.30) or females (aOR, 0.68; 95% CI: 0.38, 1.24) across all ages. Egg introduction in infancy was associated with lower plasma adiponectin among females (confounder-adjusted mean difference, -1.93 μg/mL; 95% CI: -3.70, -0.16) in early childhood only. CONCLUSIONS Egg introduction during infancy among females is associated with lower total fat mass index in early adolescence and plasma adiponectin in early childhood. This trial was registered at clinicaltrials.gov as NCT02820402.
Collapse
Affiliation(s)
- Divya Choudhary
- Division of Behavioral Medicine, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA; Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Todd C Rideout
- Department of Exercise and Nutrition Sciences, School of Public Health and Health Professions, State University of New York at Buffalo, Buffalo, NY, USA
| | - Claire E Cameron
- Department of Learning & Instruction, Graduate School of Education, State University of New York at Buffalo, Buffalo, NY, USA
| | - Heather K Lehman
- Division of Allergy/Immunology, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, John R. Oishei Children's Hospital, Buffalo, NY, USA
| | - Emily Oken
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Sheryl L Rifas-Shiman
- Division of Chronic Disease Research Across the Lifecourse, Department of Population Medicine, Harvard Medical School and Harvard Pilgrim Health Care Institute, Boston, MA, USA
| | - Xiaozhong Wen
- Division of Behavioral Medicine, Department of Pediatrics, Jacobs School of Medicine and Biomedical Sciences, State University of New York at Buffalo, Buffalo, NY, USA.
| |
Collapse
|
22
|
Ramiro-Cortijo D, Singh P, Herranz Carrillo G, Gila-Díaz A, Martín-Cabrejas MA, Martin CR, Arribas SM. Association of maternal body composition and diet on breast milk hormones and neonatal growth during the first month of lactation. Front Endocrinol (Lausanne) 2023; 14:1090499. [PMID: 36936154 PMCID: PMC10018215 DOI: 10.3389/fendo.2023.1090499] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/05/2022] [Accepted: 02/17/2023] [Indexed: 03/06/2023] Open
Abstract
INTRODUCTION Preterm birth is associated with altered growth patterns and an increased risk of cardiometabolic diseases, with breast milk (BM) being a counteracting factor. Preterm infants also show alterations in adipokines and gut hormones influencing appetite and metabolism. Since these hormones are present in BM, it is possible that their levels may equilibrate deficiencies improving infant growth. We aimed to assess 1) the BM levels of ghrelin, resistin, leptin, insulin, peptide YY, and the gastrointestinal peptide in women with preterm and term labor; 2) the relationship between BM hormones and neonatal growth; and 3) the influence of maternal body composition and diet on these BM hormones. METHODS BM from 48 women (30 term and 18 preterm labor) was collected at days 7, 14, and 28 of lactation. Maternal body composition was evaluated by bioimpedance, and neonate anthropometric parameters were collected from medical records. The maternal dietary pattern was assessed by a 72-h dietary recall at days 7 and 28 of lactation. BM hormones were analyzed by the U-Plex Ultra-sensitive method. Data were analyzed using linear regression models. BM from women with preterm labor had lower ghrelin levels, with the other hormones being significantly higher compared to women with term delivery. RESULTS In premature infants, growth was positively associated with BM ghrelin, while, in term infants, it was positively associated with insulin and negatively with peptide YY. In the first week of lactation, women with preterm labor had higher body fat compared to women with term labor. In this group, ghrelin levels were positively associated with maternal body fat and with fiber and protein intake. In women with term labor, no associations between anthropometric parameters and BM hormones were found, and fiber intake was negatively associated with peptide YY. DISCUSSION Preterm labor is a factor influencing the levels of BM adipokines and gut hormones, with BM ghrelin being a relevant hormone for premature infant growth. Since ghrelin is lower in BM from women with preterm labor and the levels are associated with maternal fat storage and some dietary components, our data support the importance to monitor diet and body composition in women who gave birth prematurely to improve the BM hormonal status.
Collapse
Affiliation(s)
- David Ramiro-Cortijo
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Division of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Instituto Universitario de Estudios de la Mujer (IUEM), Universidad Autónoma de Madrid, Madrid, Spain
| | - Pratibha Singh
- Division of Gastroenterology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Gloria Herranz Carrillo
- Division of Neonatology, Hospital Clínico San Carlos, Instituto de Investigación Sanitaria del Hospital Clínico San Carlos (IdISSC), Madrid, Spain
| | - Andrea Gila-Díaz
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
| | - María A. Martín-Cabrejas
- Department of Agricultural Chemistry and Food Science, Institute of Food Science Research (CIAL), Universidad Autónoma de Madrid, Consejo Superior de Investigación Científica (CSIC), Madrid, Spain
| | - Camilia R. Martin
- Department of Neonatology, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
- Division of Translational Research, Beth Israel Deaconess Medical Center and Harvard Medical School, Boston, MA, United States
| | - Silvia M. Arribas
- Department of Physiology, Faculty of Medicine, Universidad Autónoma de Madrid, Madrid, Spain
- Instituto Universitario de Estudios de la Mujer (IUEM), Universidad Autónoma de Madrid, Madrid, Spain
- *Correspondence: Silvia M. Arribas,
| |
Collapse
|
23
|
Li W, Liu B, Lin Y, Xue P, Lu Y, Song S, Li Y, Szeto IMY, Ren F, Guo H. The application of lactoferrin in infant formula: The past, present and future. Crit Rev Food Sci Nutr 2022; 64:5748-5767. [PMID: 36533432 DOI: 10.1080/10408398.2022.2157792] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human milk is universally regarded as the gold standard to fulfill nutrition needs of infants. Lactoferrin (LF) is a major multiple bioactive glycoprotein in human milk but little is presented in infant formula. LF can resist digestion in the infant gastrointestinal tract and is absorbed into the bloodstream in an intact form to perform physiological functions. Evidence suggest that LF prevents pathogen infection, promotes immune system development, intestinal development, brain development and bone health, as well as ameliorates iron deficiency anemia. However, more clinical studies of LF need to be further elucidated to determine an appropriate dosage for application in infant formula. LF is sensitive to denaturation induced by processing of infant formula such as heat treatments and spay drying. Thus, further studies should be focus on maximizing the retention of LF activity in the infant formula process. This review summarizes the structural features of LF. Then the digestion, absorption and metabolism of LF in infants are discussed, followed by the function of LF for infants. Further, we summarize LF in infant formula and effects of processing of infant formula on bioactivities of LF, as well as future perspectives of LF research.
Collapse
Affiliation(s)
- Wusun Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Biao Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
| | - Yingying Lin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Peng Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Sijia Song
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
24
|
Rodríguez MD, León AE, Bustos MC. Starch Digestion in Infants: An Update of Available In Vitro Methods-A Mini Review. PLANT FOODS FOR HUMAN NUTRITION (DORDRECHT, NETHERLANDS) 2022; 77:345-352. [PMID: 35962846 DOI: 10.1007/s11130-022-01001-1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Accepted: 08/03/2022] [Indexed: 06/15/2023]
Abstract
Complementary feeding starts at around six months of age because neither breast milk nor formula assure the proper nutrition of infants. Therefore, along with breast milk, solid foods are gradually introduced, particularly cereal-based foods, which will provide starch as a new source of energy and nutrients. As a result, the need of an adequate in vitro digestion method to study the influence of different aspects of weaning period is unquestionable. This critical review summarizes the in vitro digestion methods available for the analysis of starch hydrolysis under infant conditions considering different features, namely, starch digestion, infant digestive conditions and in vitro models suitable for the study of starch digestion (static, semi-dynamic and dynamic). Key factors such as enzyme concentrations, transit time, oral, gastric and intestinal conditions and differences with current adult models, have been addressed. The need for standardized infant digestion models adapted to the complementary feeding period was discussed. Existing literature data demonstrate that more effort has to be done to improve the research on this issue, in order to obtain comparable results that would address a better understanding of the digestibility of different food nutrients under infant conditions facilitating the development of appropriate formulations that may assure proper infant nutrition.
Collapse
Affiliation(s)
| | - Alberto Edel León
- Facultad de Ciencias Agropecuarias, Universidad Nacional de Córdoba, Córdoba, Argentina
- Instituto de Ciencia Y Tecnología de los Alimentos-Córdoba (ICYTAC) CONICET-UNC, Córdoba, Argentina
| | - Mariela Cecilia Bustos
- Instituto de Ciencia Y Tecnología de los Alimentos-Córdoba (ICYTAC) CONICET-UNC, Córdoba, Argentina.
- Instituto de Ciencias Básicas Y Aplicadas, Universidad Nacional de Villa María, Villa María, Argentina.
| |
Collapse
|
25
|
Kijner S, Kolodny O, Yassour M. Human milk oligosaccharides and the infant gut microbiome from an eco-evolutionary perspective. Curr Opin Microbiol 2022; 68:102156. [DOI: 10.1016/j.mib.2022.102156] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 03/03/2022] [Accepted: 04/14/2022] [Indexed: 12/21/2022]
|
26
|
McKeen S, Roy NC, Mullaney JA, Eriksen H, Lovell A, Kussman M, Young W, Fraser K, Wall CR, McNabb WC. Adaptation of the infant gut microbiome during the complementary feeding transition. PLoS One 2022; 17:e0270213. [PMID: 35834499 PMCID: PMC9282554 DOI: 10.1371/journal.pone.0270213] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 06/03/2022] [Indexed: 11/18/2022] Open
Abstract
The infant gut microbiome progresses in composition and function during the introduction of solid foods throughout the first year of life. The purpose of this study was to characterize changes in healthy infant gut microbiome composition, metagenomic functional capacity, and associated metabolites over the course of the complementary feeding period. Fecal samples were obtained at three ‘snapshot’ timepoints from infants participating in the ‘Nourish to Flourish’ pilot study: before the introduction of solid foods at approximately 4 months of age, after introducing solid foods at 9 months of age, and after continued diet diversification at 12 months of age. KEGG and taxonomy assignments were correlated with LC-MS metabolomic profiles to identify patterns of co-abundance. The composition of the microbiome diversified during the first year of life, while the functional capacity present in the gut microbiome remained stable. The introduction of solid foods between 4 and 9 months of age corresponded to a larger magnitude of change in relative abundance of sequences assigned to KEGG pathways and taxonomic assignments, as well as to stronger correlations with metabolites, compared to the magnitude of changes and number of correlations seen during continued diet diversification between 9 and 12 months of age. Changes in aqueous fecal metabolites were more strongly correlated with KEGG pathway assignments, while changes in lipid metabolites associated with taxonomic assignments, particularly between 9 and 12 months of age. This study establishes trends in microbiome composition and functional capacity occurring during the complementary feeding period and identifies potential metabolite targets for future investigations.
Collapse
Affiliation(s)
- Starin McKeen
- Riddet Institute, Massey University, Palmerston North, New Zealand
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C. Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Jane Adair Mullaney
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Hannah Eriksen
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Amy Lovell
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Martin Kussman
- German Entrepreneurship, Cambridge, Massachusetts, United States of America
| | - Wayne Young
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Karl Fraser
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Clare R. Wall
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Warren C. McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
27
|
Sun N, Liu Y, Liu K, Wang S, Liu Q, Lin S. Gastrointestinal fate of food allergens and its relationship with allergenicity. Compr Rev Food Sci Food Saf 2022; 21:3376-3404. [PMID: 35751399 DOI: 10.1111/1541-4337.12989] [Citation(s) in RCA: 29] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2021] [Revised: 03/29/2022] [Accepted: 05/09/2022] [Indexed: 01/15/2023]
Abstract
Food allergens are closely related to their gastrointestinal digestion fate, but the changes in food allergens during digestion and related mechanisms are quite complicated. This review presents in detail digestion models for predicting allergenicity, the fates of food allergens in oral, gastric and duodenal digestion, and the applications of digestomics in mapping IgE-binding epitopes of digestion-resistant peptides. Moreover, this review highlights the structure-activity relationships of food allergens during gastrointestinal digestion. Digestion-labile allergens may share common structural characteristics, such as high flexibility, rendering them easier to be hydrolyzed into small fragments with decreased or eliminated allergenicity. In contrast, the presence of disulfide bonds, tightly wound α-helical structures, or hydrophobic domains in food allergens helps them resist gastrointestinal digestion, stabilizing IgE-binding epitopes, thus maintaining their sensitization. In rare cases, digestion leads to increased allergenicity due to exposure of new epitopes. Finally, the action of the food matrix and processing on the digestion and allergenicity of food allergens as well as the underlying mechanisms was overviewed. The food matrix can directly act on the allergen by forming complexes or new epitopes to affect its gastrointestinal digestibility and thereby alter its allergenicity or indirectly affect the allergenicity by competing for enzymatic cleavage or influencing gastrointestinal pH and microbial flora. Several processing techniques attenuate the allergenicity of food proteins by altering their conformation to improve susceptibility to degradation by digestive enzymes. Given the complexity of food components, the food itself rather than a single allergen should be used to obtain more accurate data for allergenicity assessment. PRACTICAL APPLICATION: The review article will help to understand the relationship between food protein digestion and allergenicity, and may provide fundamental information for evaluating and reducing the allergenicity of food proteins.
Collapse
Affiliation(s)
- Na Sun
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P. R. China
| | - Yao Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China
| | - Kexin Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China
| | - Shan Wang
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China
| | - Qiaozhen Liu
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P. R. China
| | - Songyi Lin
- School of Food Science and Technology, National Engineering Research Center of Seafood, Dalian Polytechnic University, Dalian, P. R. China.,Collaborative Innovation Center of Seafood Deep Processing, Dalian Polytechnic University, Dalian, P. R. China
| |
Collapse
|
28
|
Bozack AK, Colicino E, Rodosthenous RS, Bloomquist TR, Baccarelli AA, Wright RO, Wright RJ, Lee AG. Breast milk-derived extracellular vesicle miRNAs are associated with maternal asthma and atopy. Epigenomics 2022; 14:727-739. [PMID: 35638388 PMCID: PMC9280402 DOI: 10.2217/epi-2022-0090] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Background: Breast milk-derived extracellular vesicle (EV) miRNAs may program child health outcomes associated with maternal asthma and atopy. The authors investigated associations between maternal asthma/atopy and EV miRNAs in the Programming of Intergenerational Stress Mechanisms cohort. Methods: Breast milk-derived EV miRNAs collected 6.1 ± 5.9 weeks postnatally (n = 80 mothers) were profiled using the TaqMan OpenArray Human MicroRNA Panel. The authors assessed associations using adjusted robust regression. Results: Nine EV miRNAs were associated with asthma during pregnancy (a priori criteria: nominal p < 0.05; |Bregression| >0.2). miR-1290 was associated with asthma and atopy during pregnancy (p < 0.05; |Bregression| >0.2). Enriched Kyoto Encyclopedia of Genes and Genomes pathways included TGF-β signaling and extracellular matrix-receptor interaction (false discovery rate <0.05). Conclusion: In this study, maternal asthma and atopy were associated with breast milk-derived EV miRNAs. Additional studies are needed to understand whether EV miRNAs have direct effects on infant and child health.
Collapse
Affiliation(s)
- Anne K Bozack
- Department of Internal Medicine, Division of Pulmonary Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Division of Environmental Health Sciences, School of Public Health, University of California, Berkeley, Berkeley, CA, USA
| | - Elena Colicino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rodosthenis S Rodosthenous
- Institute for Molecular Medicine Finland, Helsinki Institute of Life Science, University of Helsinki, Helsinki, Finland
| | - Tessa R Bloomquist
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Andrea A Baccarelli
- Department of Environmental Health Sciences, Mailman School of Public Health, Columbia University, New York, NY, USA
| | - Robert O Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Rosalind J Wright
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| | - Alison G Lee
- Department of Internal Medicine, Division of Pulmonary Medicine, Icahn School of Medicine at Mount Sinai, New York, NY, USA.,Institute for Exposomic Research, Icahn School of Medicine at Mount Sinai, New York, NY, USA
| |
Collapse
|
29
|
Leite JA, Robinson RC, Salcedo J, Ract JN, Quintal VS, Tadini CC, Barile D. The effect of microwave-assisted heating on bioactive and immunological compounds in donor human milk. Lebensm Wiss Technol 2022. [DOI: 10.1016/j.lwt.2022.113306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
30
|
Luyckx T, Grootaert C, Monge‐Morera M, Delcour JA, Rousseau F, Schymkowitz J, Van Camp J. Bioavailability and Health Impact of Ingested Amyloid‐like Protein Fibrils and their Link with Inflammatory Status: a Need for More Research? Mol Nutr Food Res 2022; 66:e2101032. [DOI: 10.1002/mnfr.202101032] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 01/24/2022] [Indexed: 11/10/2022]
Affiliation(s)
- Trui Luyckx
- Laboratory of Food Chemistry and Human Nutrition Faculty of Bioscience Engineering Ghent University Ghent Belgium
| | - Charlotte Grootaert
- Laboratory of Food Chemistry and Human Nutrition Faculty of Bioscience Engineering Ghent University Ghent Belgium
| | - Margarita Monge‐Morera
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe) KU Leuven Leuven Belgium
| | - Jan A. Delcour
- Laboratory of Food Chemistry and Biochemistry and Leuven Food Science and Nutrition Research Centre (LFoRCe) KU Leuven Leuven Belgium
| | - Frederic Rousseau
- Switch Laboratory VIB‐KU Leuven Center for Brain & Disease Research Leuven Belgium
- Department of Cellular and Molecular Medicine KU Leuven Leuven Belgium
| | - Joost Schymkowitz
- Switch Laboratory VIB‐KU Leuven Center for Brain & Disease Research Leuven Belgium
- Department of Cellular and Molecular Medicine KU Leuven Leuven Belgium
| | - John Van Camp
- Laboratory of Food Chemistry and Human Nutrition Faculty of Bioscience Engineering Ghent University Ghent Belgium
| |
Collapse
|
31
|
Siddique A, Tayyaba T, Imran M, Rahman A. Biotechnology applications in precision food. BIOTECHNOLOGY IN HEALTHCARE 2022:197-222. [DOI: 10.1016/b978-0-323-90042-3.00013-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2025]
|
32
|
Jiang H, Gallier S, Feng L, Han J, Liu W. Development of the digestive system in early infancy and nutritional management of digestive problems in breastfed and formula-fed infants. Food Funct 2022; 13:1062-1077. [DOI: 10.1039/d1fo03223b] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Food digestion and absorption in infants are closely related to early growth and long-term health. Human milk and infant formula are the main food sources for 0-6 month-old infants. Due...
Collapse
|
33
|
Wang Y, Ye A, Hou Y, Jin Y, Xu X, Han J, Liu W. Microcapsule delivery systems of functional ingredients in infant formulae: Research progress, technology, and feasible application of liposomes. Trends Food Sci Technol 2022. [DOI: 10.1016/j.tifs.2021.11.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
34
|
Luo M, Shan K, Zhang M, Ke W, Zhao D, Nian Y, Wu J, Li C. Application of ultrasound treatment for improving the quality of infant meat puree. ULTRASONICS SONOCHEMISTRY 2021; 80:105831. [PMID: 34798524 PMCID: PMC8605278 DOI: 10.1016/j.ultsonch.2021.105831] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/02/2021] [Revised: 11/09/2021] [Accepted: 11/12/2021] [Indexed: 05/26/2023]
Abstract
Infant meat puree has an indispensable effect on the oral development and nutritional intake of infants. However, commercially available products have poor texture and relatively low digestibility. In this study, ultrasound (20 kHz and 200 W, 400 W, or 600 W) was applied to the pretreatment of raw meat for preparing infant meat puree for 15 min, 30 min, and 45 min. To assess the impact of ultrasound on infant meat puree, the viscosity, texture, water distribution, particle size and in vitro digestibility were determined. The results showed that, compared with control, viscosity and hardness of meat puree decreased and the texture was better in 400 W and 600 W groups. The content of immobilized water increased in comparison with the control. Ultrasound had no obvious effect on the digestibility of meat puree in gastric phase, but it increased the digestibility in intestinal phase with the highest digestibility (80.85%±3.33) in 600 W, 15 min group. Overall, the ultrasound parameters of 600 W for 15 min can be selected as the best condition to process infant meat puree. The findings provide a new perspective for the improvement of infant meat puree.
Collapse
Affiliation(s)
- Mingyang Luo
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Kai Shan
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Miao Zhang
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Weixin Ke
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Di Zhao
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Yingqun Nian
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Juqing Wu
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China
| | - Chunbao Li
- Key Laboratory of Meat Processing and Quality Control, MOE|Key Laboratory of Meat Processing and Quality Control, MOE Key Laboratory of Meat Processing and Quality Control, MOE; Key Laboratory of Meat Processing, MARA, Jiangsu Collaborative Innovation Center of Meat Production and Processing, Quality and Safety Control, Nanjing Agricultural University, Nanjing 210095, PR China.
| |
Collapse
|
35
|
Adams K, Borkar D, Kaila R. 8-Day-old male infant with bilious emesis and hematochezia. Am J Emerg Med 2021; 54:325.e1-325.e2. [PMID: 34654600 DOI: 10.1016/j.ajem.2021.10.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2021] [Revised: 09/22/2021] [Accepted: 10/04/2021] [Indexed: 11/30/2022] Open
Abstract
The differential diagnosis for bilious emesis and hematochezia in newborns is broad and includes structural abnormalities (malrotation with volvulus, atresia, pyloric stenosis, intussusception), infectious colitis, necrotizing enterocolitis, milk protein intolerance, vascular malformations, and other bleeding disorders. Here we report a case of bilious emesis and hematochezia in an 8-day-old male infant who was ultimately found to have goat's milk protein intolerance after an extensive workup ruling out other pathology. There have been limited studies that examine goat's milk protein intolerance in pediatric patients, and to our knowledge, our paper presents the youngest patient presenting with bilious emesis related to goat's milk intake.
Collapse
Affiliation(s)
- Katelyn Adams
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States of America.
| | - Deeksha Borkar
- Department of Pediatrics, University of Minnesota Medical School, Minneapolis, MN, United States of America
| | - Rahul Kaila
- Division of Pediatric Emergency Medicine, University of Minnesota Medical School, Minneapolis, MN, United States of America
| |
Collapse
|
36
|
Jeong EW, Park GR, Kim J, Yun SY, Imm JY, Lee HG. Effect of Modified Casein to Whey Protein Ratio on Dispersion Stability, Protein Quality and Body Composition in Rats. Food Sci Anim Resour 2021; 41:855-868. [PMID: 34632404 PMCID: PMC8460334 DOI: 10.5851/kosfa.2021.e42] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2021] [Revised: 07/29/2021] [Accepted: 08/05/2021] [Indexed: 11/29/2022] Open
Abstract
The present study was designed to investigate the effects of protein formula with different casein (C) to whey protein (W) ratios on dispersion stability, protein quality and body composition in rats. Modification of the casein to whey protein (CW) ratio affected the extent of protein aggregation, and heated CW-2:8 showed a significantly increased larger particle (>100 μm) size distribution. The largest protein aggregates were formed by whey protein self-aggregation. There were no significant differences in protein aggregation when the CW ratios changed from 10:0 to 5:5. Based on the protein quality assessment (CW-10:0, CW-8:2, CW-5:5, and CW-2:8) for four weeks, CW-10:0 showed a significantly higher feed intake (p<0.05), but the high proportion of whey protein in the diet (CW-5:5 and CW-2:8) increased the feed efficiency ratio, protein efficiency ratio, and net protein ratio compared to other groups. Similarly, CW-2:8 showed greater true digestibility compared to other groups. No significant differences in fat mass and lean mass analyzed by dual-energy x-ray absorptiometry were observed. A significant difference was found in the bone mineral density between the CW-10:0 and CW-2:8 groups (p<0.05), but no difference was observed among the other groups. Based on the results, CW-5:5 improved protein quality without causing protein instability problems in the dispersion.
Collapse
Affiliation(s)
- Eun Woo Jeong
- Department of Food and Nutrition, Hanyang
University, Seoul 04763, Korea
| | - Gyu Ri Park
- Department of Food and Nutrition, Hanyang
University, Seoul 04763, Korea
| | - Jiyun Kim
- Department of Food and Nutrition, Hanyang
University, Seoul 04763, Korea
| | - So-Yul Yun
- Department of Foods and Nutrition, Kookmin
University, Seoul 02707, Korea
| | - Jee-Young Imm
- Department of Foods and Nutrition, Kookmin
University, Seoul 02707, Korea
| | - Hyeon Gyu Lee
- Department of Food and Nutrition, Hanyang
University, Seoul 04763, Korea,Corresponding author : Hyeon
Gyu Lee, Department of Food and Nutrition, Hanyang University, Seoul 04763,
Korea. Tel: +82-2-2220-1201, Fax: +82-2-2292-1226, E-mail:
| |
Collapse
|
37
|
Lee S, Jo K, Jeong HG, Choi YS, Yong HI, Jung S. Understanding protein digestion in infants and the elderly: Current in vitro digestion models. Crit Rev Food Sci Nutr 2021; 63:975-992. [PMID: 34346822 DOI: 10.1080/10408398.2021.1957765] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022]
Abstract
The last decades have witnessed a surge of interest in the fate of dietary proteins during gastrointestinal (GI) digestion. Although several in vitro digestion models are available as alternatives to clinical experiments, most of them focus on the digestive conditions of healthy young adults. This study investigates the static/dynamic models used to simulate digestion in infants and the elderly and considers the related in vivo conditions. The in vitro digestive protocols targeting these two groups are summarized, and the challenges associated with the further development of in vitro digestion models are discussed. Static models rely on several factors (e.g., enzyme concentration, pH, reaction time, and rotation speed) to differentiate digestive conditions depending on age. Dynamic models can more accurately simulate the complex digestion process and allow the inclusion of further parameters (sequential secretion of digestive fluids, gradual changes in pH, peristaltic mixing, GI emptying, and the inoculation of luminal microbiota). In the case of infants, age or growth stage clarification and the differentiation of digestive protocols between full-term and preterm infants are required, whereas protocols dealing with various health statuses are required in the case of the elderly, as this group is prone to oral cavity and GI function deterioration.
Collapse
Affiliation(s)
- Seonmin Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Kyung Jo
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Hyun Gyung Jeong
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| | - Yun-Sang Choi
- Research Group of Food Processing, Korea Food Research Institute, Wanju, Republic of Korea
| | - Hae In Yong
- Research Group of Food Processing, Korea Food Research Institute, Wanju, Republic of Korea
| | - Samooel Jung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon, Republic of Korea
| |
Collapse
|
38
|
Fournier E, Roussel C, Dominicis A, Ley D, Peyron MA, Collado V, Mercier-Bonin M, Lacroix C, Alric M, Van de Wiele T, Chassard C, Etienne-Mesmin L, Blanquet-Diot S. In vitro models of gut digestion across childhood: current developments, challenges and future trends. Biotechnol Adv 2021; 54:107796. [PMID: 34252564 DOI: 10.1016/j.biotechadv.2021.107796] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2021] [Revised: 07/05/2021] [Accepted: 07/06/2021] [Indexed: 02/08/2023]
Abstract
The human digestion is a multi-step and multi-compartment process essential for human health, at the heart of many issues raised by academics, the medical world and industrials from the food, nutrition and pharma fields. In the first years of life, major dietary changes occur and are concomitant with an evolution of the whole child digestive tract anatomy and physiology, including colonization of gut microbiota. All these phenomena are influenced by child exposure to environmental compounds, such as drugs (especially antibiotics) and food pollutants, but also childhood infections. Due to obvious ethical, regulatory and technical limitations, in vivo approaches in animal and human are more and more restricted to favor complementary in vitro approaches. This review summarizes current knowledge on the evolution of child gut physiology from birth to 3 years old regarding physicochemical, mechanical and microbial parameters. Then, all the available in vitro models of the child digestive tract are described, ranging from the simplest static mono-compartmental systems to the most sophisticated dynamic and multi-compartmental models, and mimicking from the oral phase to the colon compartment. Lastly, we detail the main applications of child gut models in nutritional, pharmaceutical and microbiological studies and discuss the limitations and challenges facing this field of research.
Collapse
Affiliation(s)
- Elora Fournier
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France; Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Charlène Roussel
- Laval University, INAF Institute of Nutrition and Functional Foods, G1V 0A6 Quebec, Canada
| | - Alessandra Dominicis
- European Reference Laboratory for E. coli, Istituto Superiore di Sanità, Rome, Italy
| | - Delphine Ley
- Université Lille 2, Faculté de Médecine, Inserm U995 Nutritional Modulation of Infection and Inflammation, 59045 Lille, France
| | - Marie-Agnès Peyron
- Université Clermont Auvergne, INRAE, UNH, Unité de Nutrition Humaine, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Valérie Collado
- Université Clermont Auvergne, EA 4847, CROC, Centre de Recherche en Odontologie Clinique, 63000 Clermont-Ferrand, France
| | - Muriel Mercier-Bonin
- Toxalim, Research Centre in Food Toxicology, INRAE, ENVT, INP-Purpan, UPS, Université de Toulouse, 31000 Toulouse, France
| | - Christophe Lacroix
- Laboratory of Food Biotechnology, Institute of Food, Nutrition and Health, ETH Zurich, 8092 Zürich, Switzerland
| | - Monique Alric
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Tom Van de Wiele
- Ghent University, Center for Microbial Ecology and Technology (CMET), Coupure Links 653, 9000 Ghent, Belgium
| | - Christophe Chassard
- Université Clermont Auvergne, INRAE, VetAgro Sup, UMRF, 15000 Aurillac, France
| | - Lucie Etienne-Mesmin
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France
| | - Stéphanie Blanquet-Diot
- Université Clermont Auvergne, INRAE, UMR 454 MEDIS, Microbiologie Environnement Digestif et Santé, CRNH Auvergne, 63000 Clermont-Ferrand, France.
| |
Collapse
|
39
|
Ye A. Gastric colloidal behaviour of milk protein as a tool for manipulating nutrient digestion in dairy products and protein emulsions. Food Hydrocoll 2021. [DOI: 10.1016/j.foodhyd.2021.106599] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
40
|
Phosanam A, Chandrapala J, Huppertz T, Adhikari B, Zisu B. In vitro digestion of infant formula model systems: Influence of casein to whey protein ratio. Int Dairy J 2021. [DOI: 10.1016/j.idairyj.2021.105008] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
|
41
|
Leroux C, Chervet ML, German JB. Perspective: Milk microRNAs as Important Players in Infant Physiology and Development. Adv Nutr 2021; 12:1625-1635. [PMID: 34022770 PMCID: PMC8483967 DOI: 10.1093/advances/nmab059] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/27/2020] [Revised: 01/08/2021] [Accepted: 04/14/2021] [Indexed: 12/16/2022] Open
Abstract
Evolutionary selective pressure on lactation has resulted in milk that provides far more than simply essential nutrients, delivering a complex repertoire of agents from hormones to intact cells. Human infants are born with low barrier integrity of their gut, which means that many of the complex biopolymer components of milk enter and circulate in lymph and blood, reaching organs throughout the body. Due to this state of gut maturation, all components of milk are potentially part of the crosstalk between mother and infants. This article highlights the functions of milk's complex biopolymers, more specifically the potential role of microRNAs (miRNAs) contained in extracellular vesicles in human milk. miRNAs are key effectors in the regulation of many biological processes during early-age development, and consequently milk-sourced miRNAs must be considered to provide unique biological assets to the infant during breastfeeding. This article interprets the evidence of the potential action of human milk miRNAs on infant development, taking into account their abundance in milk based on the literature and current knowledge. Human milk miRNAs appear to influence lipid and glucose metabolism, gut maturation, neurogenesis, and immunity. We also show growing evidence that human milk miRNAs are epigenetic modulators that play a pivotal role in the regulation of tissue-specific gene expression throughout life. Furthermore, this article addresses the ongoing debate regarding the potential influence of human milk miRNAs on viral infection as a new research area. This article highlights that these bioactive molecules are now being incorporated into our overall understanding of nutrient needs for healthy infant development, preparing each individual infant to succeed as a healthy and protected adult throughout its life. In essence, miRNAs are a new language in the Rosetta stone of health that is mammalian lactation.
Collapse
Affiliation(s)
| | - Mathilde Lea Chervet
- Foods for Health Institute, Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
| | - J Bruce German
- Foods for Health Institute, Department of Food Science and Technology, University of California, Davis, Davis, CA, USA
| |
Collapse
|
42
|
Bhat ZF, Morton JD, Bekhit AEDA, Kumar S, Bhat HF. Non-thermal processing has an impact on the digestibility of the muscle proteins. Crit Rev Food Sci Nutr 2021; 62:7773-7800. [PMID: 33939555 DOI: 10.1080/10408398.2021.1918629] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Muscle proteins undergo several processes before being ready in a final consumable form. All these processes affect the digestibility of muscle proteins and subsequent release of amino acids and peptides during digestion in the human gut. The present review examines the effects of different processing techniques, such as curing, drying, ripening, comminution, aging, and marination on the digestibility of muscle proteins. The review also examines how the source of muscle proteins alters the gastrointestinal protein digestion. Processing techniques affect the structural and functional properties of muscle proteins and can affect their digestibility negatively or positively depending on the processing conditions. Some of these techniques, such as aging and mincing, can induce favorable changes in muscle proteins, such as partial unfolding or exposure of cleavage sites, and increase susceptibility to hydrolysis by digestive enzymes whereas others, such as drying and marination, can induce unfavorable changes, such as severe cross-linking, protein aggregation, oxidation induced changes or increased disulfide (S-S) bond content, thereby decreasing proteolysis. The underlying mechanisms have been discussed in detail and the conclusions drawn in the light of existing knowledge provide information with potential industrial importance.
Collapse
Affiliation(s)
- Zuhaib F Bhat
- Division of Livestock Products Technology, SKUAST of Jammu, Jammu, India
| | - James D Morton
- Department of Wine Food and Molecular Biosciences, Faculty of Agriculture and Life Sciences, Lincoln University, Christchurch, New Zealand
| | | | - Sunil Kumar
- Division of Livestock Products Technology, SKUAST of Jammu, Jammu, India
| | - Hina F Bhat
- Division of Biotechnology, SKUAST of Kashmir, Srinagar, India
| |
Collapse
|
43
|
Lee S, Choi YS, Jo K, Yong HI, Jeong HG, Jung S. Improvement of meat protein digestibility in infants and the elderly. Food Chem 2021; 356:129707. [PMID: 33873143 DOI: 10.1016/j.foodchem.2021.129707] [Citation(s) in RCA: 30] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2021] [Revised: 02/24/2021] [Accepted: 03/23/2021] [Indexed: 01/11/2023]
Abstract
Meat is a valuable protein source with a balanced composition of essential amino acids and various nutrients. This review aims to identify methods to improve digestion of meat proteins, as well as evaluate the digestive characteristics of infants and the elderly. Immature digestive conditions in infants, including a high gastric pH and low protease concentration, can hinder protein digestion, thus resulting in inhibited growth and development. Likewise, gastrointestinal (GI) tract aging and chronic health problems, including tooth loss and atrophic gastritis, can lead to reduction in protein digestion and absorption in the elderly compared with those in young adults. Moderate heating and several non-thermal technologies, such as aging, enzymatic hydrolysis, ultrasound, high-pressure processing, and pulsed electric field can alter protein structure and improve protein digestion in individuals with low digestive capacity.
Collapse
Affiliation(s)
- Seonmin Lee
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, South Korea
| | - Yun-Sang Choi
- Research Group of Food Processing, Korea Food Research Institute, Wanju 55365, South Korea
| | - Kyung Jo
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, South Korea
| | - Hae In Yong
- Research Group of Food Processing, Korea Food Research Institute, Wanju 55365, South Korea
| | - Hyun Gyung Jeong
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, South Korea
| | - Samooel Jung
- Division of Animal and Dairy Science, Chungnam National University, Daejeon 34134, South Korea.
| |
Collapse
|
44
|
Gastric Enzyme Supplementation Inhibits Food Allergy in a BALB/c Mouse Model. Nutrients 2021; 13:nu13030738. [PMID: 33652629 PMCID: PMC7996948 DOI: 10.3390/nu13030738] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Revised: 02/08/2021] [Accepted: 02/22/2021] [Indexed: 02/07/2023] Open
Abstract
Impaired gastric digestion due to suppressed gastric acidity enhances the risk for food allergy development. In the current study, we aimed to evaluate the impact of a supported gastric digestion via application of a pharmaceutical gastric enzyme solution (GES) on food allergy development and allergic reactions in a BALB/c mouse model. The ability of the GES to restore hypoacidic conditions was tested in mice treated with gastric acid suppression medication. To evaluate the impact on allergic symptoms, mice were orally sensitized with ovalbumin (OVA) under gastric acid suppression and subjected to oral challenges with or without GES. The immune response was evaluated by measurement of antibody titers, cytokine levels, mucosal allergy effector cell influx and regulatory T-cell counts. Clinical response was objectified by core body temperature measurements after oral OVA challenge. Supplementation of GES transiently restored physiological pH levels in the stomach after pharmaceutical gastric acid suppression. During oral sensitization, supplementation of gastric enzymes significantly reduced systemic IgE, IgG1 and IgG2a levels and allergic symptoms. In food allergic mice, clinical symptoms were reduced by co-administration of the gastric enzyme solution. Support of gastric digestion efficiently prevents food allergy induction and alleviates clinical symptoms in our food allergy model.
Collapse
|
45
|
Increased protein digestibility of beef with aging in an infant in vitro digestion model. Meat Sci 2020; 169:108210. [DOI: 10.1016/j.meatsci.2020.108210] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Revised: 04/24/2020] [Accepted: 06/04/2020] [Indexed: 01/30/2023]
|
46
|
Corrigan B, Brodkorb A. The effect of pre-treatment of protein ingredients for infant formula on their in vitro gastro-intestinal behaviour. Int Dairy J 2020. [DOI: 10.1016/j.idairyj.2020.104810] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
|
47
|
Elwakiel M, Boeren S, Wang W, Schols HA, Hettinga KA. Degradation of Proteins From Colostrum and Mature Milk From Chinese Mothers Using an in vitro Infant Digestion Model. Front Nutr 2020; 7:162. [PMID: 33117838 PMCID: PMC7557360 DOI: 10.3389/fnut.2020.00162] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2020] [Accepted: 08/10/2020] [Indexed: 12/14/2022] Open
Abstract
This study provided insights into the degradation of human milk proteins in an in vitro infant digestion model by comparing colostrum (week 1) and mature milk (week 4) of 7 Chinese mothers individually. In this study, we adapted the exiting INFOGEST in vitro model, to conditions representative to infants (0 to 3 month-old). The level of undigested proteins was analyzed by LC-MS/MS after gel-electrophoretic separation and in-gel digestion. The BCA protein assay showed that the total undigested milk protein content decreased from the start to the end of digestion with variations between mothers, especially in the gastric phase (25–80%). Undigested proteins could also still be found after the intestinal phase, ranging from 0.5 to 4.2% of initial protein content. Based on LC-MS/MS analysis, milk protein digestion varied between the mothers individually, especially during the gastric phase. No differences could be observed between protein digestion from colostrum and mature milk after the intestinal phase. The highest levels of proteins remaining after intestinal digestion can be linked to the group immune-active proteins, for all mothers. The level of protease inhibitors and total protein content in the milk did not correlate with the overall proteolysis during digestion. The results also showed that milk serum proteins partly remained after the gastric phase, with 33% remaining from colostrum and 37% remaining from mature milk. More than 40 milk serum proteins were detected after the intestinal phase. Some of the highly abundant milk serum proteins (lactoferrin, serum albumin, bile salt-activated lipase, immunoglobulins, α1-antichymotrypsin) were still partially present intact after the intestinal phase, for all mothers. Caseins were also not completely digested in the gastric phase, with 35% remaining from colostrum and 13% remaining from mature milk. Caseins, on the other hand, were almost completely digested after the intestinal phase. The complete degradation of caseins into peptides might be related to their structural features. Overall, this study showed that digestion differed for the various human milk proteins by adapting an in vitro digestion model to infant physiological conditions, with the main differences between digestion of the milk from individual mothers being observed after gastric digestion.
Collapse
Affiliation(s)
- Mohèb Elwakiel
- Food Quality and Design Group, Wageningen University & Research, Wageningen, Netherlands.,Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Wendan Wang
- Inner Mongolia Yili Industrial Group Co., Ltd., Hohhot, China
| | - Henk A Schols
- Laboratory of Food Chemistry, Wageningen University & Research, Wageningen, Netherlands
| | - Kasper A Hettinga
- Food Quality and Design Group, Wageningen University & Research, Wageningen, Netherlands
| |
Collapse
|
48
|
He X, McClorry S, Hernell O, Lönnerdal B, Slupsky CM. Digestion of human milk fat in healthy infants. Nutr Res 2020; 83:15-29. [PMID: 32987285 DOI: 10.1016/j.nutres.2020.08.002] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2020] [Revised: 08/02/2020] [Accepted: 08/07/2020] [Indexed: 02/07/2023]
Abstract
Lipid digestion is critical for infant development, and yet, the interconnection between lipid digestion and the microbiota is largely understudied. This review focuses on digestion of the human milk fat globule and summarizes the current understanding of the mechanisms underlying this process in infants. We first discuss the partial hydrolysis of milk fat in the stomach, which leads to rearrangement of lipid droplets, creating a lipid-water interface necessary for duodenal lipolysis. In the first few months of life, secretion of pancreatic triglyceride lipase, phospholipase A2, and bile salts is immature. The dominant lipases aiding fat digestion in the newborn small intestine are therefore pancreatic lipase-related protein 2 and bile salt-stimulated lipase from both the exocrine pancreas and milk. We summarize the interaction between ionic fatty acids and cations to form insoluble fatty acid soaps and how it is influenced by various factors, including cation availability, pH, and bile salt concentration, as well as saturation and chain length of fatty acids. We further argue that the formation of the soap complex does not contribute to lipid bioavailability. Next, the possible roles that the gut microbiota plays in lipid digestion and absorption are discussed. Finally, we provide a perspective on how the manufacturing process of infant formula and dairy products may alter the physical properties and structure of lipid droplets, thereby altering the rate of lipolysis.
Collapse
Affiliation(s)
- Xuan He
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Shannon McClorry
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Olle Hernell
- Department of Clinical Sciences, Pediatrics, Umeå University, SE 901 85 Umeå, Sweden
| | - Bo Lönnerdal
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA
| | - Carolyn M Slupsky
- Department of Nutrition, Davis, One Shields Ave, Davis, CA 95616, USA; Department of Food Science and Technology, University of California, Davis, One Shields Ave, Davis, CA 95616, USA.
| |
Collapse
|
49
|
Cheng L, Wong H. Food Effects on Oral Drug Absorption: Application of Physiologically-Based Pharmacokinetic Modeling as a Predictive Tool. Pharmaceutics 2020; 12:pharmaceutics12070672. [PMID: 32708881 PMCID: PMC7408216 DOI: 10.3390/pharmaceutics12070672] [Citation(s) in RCA: 72] [Impact Index Per Article: 14.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2020] [Revised: 07/15/2020] [Accepted: 07/15/2020] [Indexed: 12/13/2022] Open
Abstract
The bioavailability of an orally administered small molecule is often dictated by drug-specific physicochemical characteristics and is influenced by many biological processes. For example, in fed or fasted conditions, the transit time within the gastrointestinal tract can vary, confounding the ability to predict the oral absorption. As such, the effects of food on the pharmacokinetics of compounds in the various biopharmaceutics classification system (BCS) classes need to be assessed. The consumption of food leads to physiological changes, including fluctuations in the gastric and intestinal pH, a delay in gastric emptying, an increased bile secretion, and an increased splanchnic and hepatic blood flow. Despite the significant impact of a drug's absorption and dissolution, food effects have not been fully studied and are often overlooked. Physiologically-based pharmacokinetic (PBPK) models can be used to mechanistically simulate a compound's pharmacokinetics under fed or fasted conditions, while integrating drug properties such as solubility and permeability. This review discusses the PBPK models published in the literature predicting the food effects, the models' strengths and shortcomings, as well as future steps to mitigate the current knowledge gap. We observed gaps in knowledge which limits the ability of PBPK models to predict the negative food effects and food effects in the pediatric population. Overall, the further development of PBPK models to predict food effects will provide a mechanistic basis to understand a drug's behavior in fed and fasted conditions, and will help enable the drug development process.
Collapse
|
50
|
Alifariki LO, Kusnan A, Asriati, Binekada IMC, Saida, Usman AN. The proxy determinant of complementary feeding of the breastfed child delivery in less than 6 months old infant in the fishing community of Buton tribe. ENFERMERIA CLINICA 2020. [DOI: 10.1016/j.enfcli.2019.10.135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
|