1
|
Bertin B, Foligne B, Ley D, Lesage J, Beghin L, Morcel J, Gottrand F, Hermann E. An Overview of the Influence of Breastfeeding on the Development of Inflammatory Bowel Disease. Nutrients 2023; 15:5103. [PMID: 38140362 PMCID: PMC10745409 DOI: 10.3390/nu15245103] [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: 11/22/2023] [Revised: 12/08/2023] [Accepted: 12/09/2023] [Indexed: 12/24/2023] Open
Abstract
The first 1000 days of life is a critical period that contributes significantly to the programming of an individual's future health. Among the many changes that occur during this period early in life, there is growing evidence that the establishment of healthy gut microbiota plays an important role in the prevention of both short- and long-term health problems. Numerous publications suggest that the quality of the gut microbiota colonisation depends on several dietary factors, including breastfeeding. In this respect, a relationship between breastfeeding and the risk of inflammatory bowel disease (IBD) has been suggested. IBDs are chronic intestinal diseases, and perinatal factors may be partly responsible for their onset. We review the existence of links between breastfeeding and IBD based on experimental and clinical studies. Overall, despite encouraging experimental data in rodents, the association between breastfeeding and the development of IBD remains controversial in humans, partly due to the considerable heterogeneity between clinical studies. The duration of exclusive breastfeeding is probably decisive for its lasting effect on IBD. Thus, specific improvements in our knowledge could support dietary interventions targeting the gut microbiome, such as the early use of prebiotics, probiotics or postbiotics, in order to prevent the disease.
Collapse
Affiliation(s)
- Benjamin Bertin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Benoit Foligne
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Delphine Ley
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Jean Lesage
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| | - Laurent Beghin
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Jules Morcel
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Frédéric Gottrand
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
- Univ. Lille, Inserm, CHU Lille, CIC-1403 Inserm-CHU, F-59000 Lille, France
| | - Emmanuel Hermann
- Univ. Lille, Inserm, CHU Lille, U1286-INFINITE-Institute for Translational Research in Inflammation, F-59000 Lille, France; (B.B.); (B.F.); (D.L.); (J.L.); (L.B.); (J.M.); (F.G.)
| |
Collapse
|
2
|
Correa F, Luise D, Negrini C, Ruggeri R, Bosi P, Trevisi P. Effect of two milk supplements and two ways of administration on growth performance, welfare and fecal microbial ecology of suckling piglets. Front Vet Sci 2023; 10:1050414. [PMID: 36923055 PMCID: PMC10008956 DOI: 10.3389/fvets.2023.1050414] [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: 09/21/2022] [Accepted: 02/09/2023] [Indexed: 03/02/2023] Open
Abstract
Introduction The aim of this study was to evaluate the effect of two MS formulas, DanMilk™ (AB Neo, Denmark) (MS1) and Neopigg® RescueMilk (Provimi, Netherlands) (MS2) administered manually and to compare two ways of administration (manual vs automatic) of MS1 on growth performance, health, fecal microbial profile, behavior, and skin lesions of piglets during suckling and post-weaning. Methods Forty litters (528 piglets) were divided into 4 groups: 1) Control group receiving no MS (CON); 2) MS1 administered automatically (A-MS1); 3) MS1 administered manually (Ma-MS1) 4) MS2 administered manually (Ma-MS2). All groups had access to sow milk and creep feed. On day 5 after birth (d0), litters were equalized (13.2 piglets/litter ± 0.8 SD), thereafter no cross-fostering was allowed. Piglets were weighed at day 5 after birth (d0), at the end of milk supplementation (d14), at weaning (d21 of the trial, 26 days of age) and ten days post-weaning (d31). Piglet welfare was assessed using behavioral and lesion measures at d4 and d10. Feces were collected at d14 and d21. Results and discussion During the suckling period, A-MS1 had lowest mortality (p < 0.05), while Ma-MS1 had lower mortality compared with CON and Ma-MS2 (p < 0.05). Negative social behavior at d4, was more frequent in MS groups (A-MS1, Ma-MS1, Ma-MS2) compared to CON group (p = 0.03). Growth performance and lesion prevalence were not affected by MS provision. During lactation, Ma-MS2 group had a higher percentage of piglets not eating during suckling at d18 compared with Ma-MS1 (p = 0.03). MS1 increased microbial diversity compared with CON at d14 (Chao1, p = 0.02; Shannon, p = 0.03) and compared with CON (Shannon, p < 0.05; InvSimpson, p = 0.01) and Ma-MS2 (Chao1, p < 0.05; Shannon, p = 0.05, InvSimpson p = 0.01) at d21. Groups that received MS1 were characterized by genera producing short-chain fatty acids (SCFAs), i.e., Lachnospiraceae (A-MS1) and Oscillospiraceae (Ma-MS1). MS composition and availability can contribute to reduce piglet's mortality during the suckling phase and can also affect intestinal microbiota by favoring the presence of SCFAs producing bacteria.
Collapse
Affiliation(s)
- Federico Correa
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
| | - Diana Luise
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
| | - Clara Negrini
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
| | - Roberta Ruggeri
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
- Agroscope, Pig Research Unit, Animal Production Systems and Animal Health, Posieux, Switzerland
| | - Paolo Bosi
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
| | - Paolo Trevisi
- Department of Agricultural and Food Sciences (DISTAL), University of Bologna, Bologna, Italy
| |
Collapse
|
3
|
Isoquinoline Alkaloids in Sows' Diet Reduce Body Weight Loss during Lactation and Increase IgG in Colostrum. Animals (Basel) 2021; 11:ani11082195. [PMID: 34438653 PMCID: PMC8388448 DOI: 10.3390/ani11082195] [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: 05/21/2021] [Revised: 07/20/2021] [Accepted: 07/21/2021] [Indexed: 11/17/2022] Open
Abstract
Simple Summary Plant extracts containing isoquinoline alkaloids (IQ) have been demonstrated to have anti-inflammatory properties. In pigs, IQ supplementation has been shown to downregulate the stress response and improve the digestibility of nutrients. The present experiment was conducted to test the hypothesis that supplementing sows’ diets with IQ during gestation would decrease stress at farrowing and improve colostrum quality, positively affecting the piglets’ health and performance. Abstract Isoquinoline alkaloids (IQ) exert beneficial antimicrobial and anti-inflammatory effects in livestock. Therefore, we hypothesized that supplementing sows’ diets with IQ during gestation would decrease farrowing stress, affecting the piglets’ development and performance. Sows were divided into: IQ1, supplemented with IQ from gestation day 80 (G80) to weaning; IQ2, supplemented from gestation day 110 (G110) to weaning, and a non-supplemented (NC) group. Sow body weight (BW), feed intake, back-fat thickness and back-muscle thickness were monitored. Cortisol, glucose and insulin were measured in sows’ blood collected 5 d before, during, and after 7 d farrowing. Protein, fat, IgA and IgG were analyzed in the colostrum and milk. Piglets were monitored for weight and diarrhea score, and for ileum histology and gene expression 5 d post-weaning. IQ-supplemented sows lost less BW during lactation. Glucose and insulin levels were lower in the IQ groups compared to NC-sows 5 d before farrowing and had higher levels of protein and IgG in their colostrum. No other differences were observed in sows, nor in the measured parameters in piglets. In conclusion, IQ supplementation affected sows’ metabolism, reducing body weight loss during lactation. Providing IQ to sows from their entrance into the maternity barn might be sufficient to induce these effects. IQ improved colostrum quality, increasing the protein and IgG content, improving passive immunity for piglets.
Collapse
|
4
|
Buffet-Bataillon S, Bellanger A, Boudry G, Gangneux JP, Yverneau M, Beuchée A, Blat S, Le Huërou-Luron I. New Insights Into Microbiota Modulation-Based Nutritional Interventions for Neurodevelopmental Outcomes in Preterm Infants. Front Microbiol 2021; 12:676622. [PMID: 34177860 PMCID: PMC8232935 DOI: 10.3389/fmicb.2021.676622] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/12/2021] [Indexed: 12/19/2022] Open
Abstract
Gut microbiota and the central nervous system have parallel developmental windows during pre and post-natal life. Increasing evidences suggest that intestinal dysbiosis in preterm infants predisposes the neonate to adverse neurological outcomes later in life. Understanding the link between gut microbiota colonization and brain development to tailor therapies aimed at optimizing initial colonization and microbiota development are promising strategies to warrant adequate brain development and enhance neurological outcomes in preterm infants. Breast-feeding has been associated with both adequate cognitive development and healthy microbiota in preterms. Infant formula are industrially produced substitutes for infant nutrition that do not completely recapitulate breast-feeding benefices and could be largely improved by the understanding of the role of breast milk components upon gut microbiota. In this review, we will first discuss the nutritional and bioactive component information on breast milk composition and its contribution to the assembly of the neonatal gut microbiota in preterms. We will then discuss the emerging pathways connecting the gut microbiota and brain development. Finally, we will discuss the promising microbiota modulation-based nutritional interventions (including probiotic and prebiotic supplementation of infant formula and maternal nutrition) for improving neurodevelopmental outcomes.
Collapse
Affiliation(s)
- Sylvie Buffet-Bataillon
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- Department of Clinical Microbiology, CHU Rennes, Rennes, France
| | - Amandine Bellanger
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- Department of Pediatrics-Neonatology, CHU Rennes, Rennes, France
| | - Gaelle Boudry
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | | | | - Alain Beuchée
- Department of Pediatrics-Neonatology, Univ Rennes, CHU Rennes, LTSI-UMR 1099, Rennes, France
| | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | | |
Collapse
|
5
|
Assay considerations for fluorescein isothiocyanate-dextran (FITC-d): an indicator of intestinal permeability in broiler chickens. Poult Sci 2021; 100:101202. [PMID: 34111612 PMCID: PMC8192867 DOI: 10.1016/j.psj.2021.101202] [Citation(s) in RCA: 25] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Revised: 03/12/2021] [Accepted: 03/26/2021] [Indexed: 12/16/2022] Open
Abstract
Fluorescein isothiocyanate-dextran (FITC-d) is being used as an indicator of intestinal paracellular permeability in poultry research. Especially with the industry moving toward antibiotic-free production, intestinal function and integrity issues have been a research focus. An increasing number of scientific conference abstracts and peer-reviewed journal publications have shown that 4-kDa FITC-d is an efficient marker candidate for measurement of intestinal permeability and can be applied in broiler research. However, experimental protocols vary by personnel, instruments used, and research institution, and potential concerns related to this assay have yet to receive the same amount of attention. Understanding protocol consistency within and across laboratories is vital for obtaining accurate, consistent, and comparable experimental results. This review is aimed to 1) summarize different FITC-d assays in broiler research from peer-reviewed publications during the past 6 yr and 2) discuss factors that can potentially affect intestinal permeability results when conducting the FITC-d assay. In summary, it is essential to pay attention to details, including gavage dose, fasting period, sample handling and lab analysis details when conducting the assay in broiler research. Differences in birds (breed/strain, age, and gender) and experimental design (diet, health status/challenge model, and sampling age) need to be considered when comparing serum FITC-d concentration results between different in vivo animal trials.
Collapse
|
6
|
Käser T. Swine as biomedical animal model for T-cell research-Success and potential for transmittable and non-transmittable human diseases. Mol Immunol 2021; 135:95-115. [PMID: 33873098 DOI: 10.1016/j.molimm.2021.04.004] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 03/23/2021] [Accepted: 04/01/2021] [Indexed: 02/07/2023]
Abstract
Swine is biologically one of the most relevant large animal models for biomedical research. With its use as food animal that can be exploited as a free cell and tissue source for research and its high susceptibility to human diseases, swine additionally represent an excellent option for both the 3R principle and One Health research. One of the previously most limiting factors of the pig model was its arguably limited immunological toolbox. Yet, in the last decade, this toolbox has vastly improved including the ability to study porcine T-cells. This review summarizes the swine model for biomedical research with focus on T cells. It first contrasts the swine model to the more commonly used mouse and non-human primate model before describing the current capabilities to characterize and extend our knowledge on porcine T cells. Thereafter, it not only reflects on previous biomedical T-cell research but also extends into areas in which more in-depth T-cell analyses could strongly benefit biomedical research. While the former should inform on the successes of biomedical T-cell research in swine, the latter shall inspire swine T-cell researchers to find collaborations with researchers working in other areas - such as nutrition, allergy, cancer, transplantation, infectious diseases, or vaccine development.
Collapse
Affiliation(s)
- Tobias Käser
- Department of Population Health and Pathobiology, College of Veterinary Medicine, North Carolina State University, 1060 William Moore Drive, 27607 Raleigh, NC, USA.
| |
Collapse
|
7
|
Maternal Supplementation of Food Ingredient (Prebiotic) or Food Contaminant (Mycotoxin) Influences Mucosal Immune System in Piglets. Nutrients 2020; 12:nu12072115. [PMID: 32708852 PMCID: PMC7400953 DOI: 10.3390/nu12072115] [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: 06/10/2020] [Revised: 07/08/2020] [Accepted: 07/15/2020] [Indexed: 12/29/2022] Open
Abstract
The early life period is crucial for the maturation of the intestinal barrier, its immune system, and a life-long beneficial host-microbiota interaction. The study aims to assess the impact of a beneficial dietary (short-chain fructooligosaccharides, scFOS) supplementation vs. a detrimental dietary environment (such as mycotoxin deoxynivalenol, DON) on offspring intestinal immune system developmental profiles. Sows were given scFOS-supplemented or DON-contaminated diets during the last 4 weeks of gestation, whereas force-feeding piglets with DON was performed during the first week of offspring life. Intestinal antigen-presenting cell (APC) subset frequency was analyzed by flow cytometry in the Peyer's patches and in lamina propria and the responsiveness of intestinal explants to toll-like receptor (TLR) ligands was performed using ELISA and qRT-PCR from post-natal day (PND) 10 until PND90. Perinatal exposure with scFOS did not affect the ontogenesis of APC. While it early induced inflammatory responses in piglets, scFOS further promoted the T regulatory response after TLR activation. Sow and piglet DON contamination decreased CD16+ MHCII+ APC at PND10 in lamina propria associated with IFNγ inflammation and impairment of Treg response. Our study demonstrated that maternal prebiotic supplementation and mycotoxin contamination can modulate the mucosal immune system responsiveness of offspring through different pathways.
Collapse
|
8
|
Maternal dietary resistant starch does not improve piglet's gut and liver metabolism when challenged with a high fat diet. BMC Genomics 2020; 21:439. [PMID: 32590936 PMCID: PMC7318506 DOI: 10.1186/s12864-020-06854-x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Accepted: 06/19/2020] [Indexed: 12/14/2022] Open
Abstract
Background In the past several years, the use of resistant starch (RS) as prebiotic has extensively been studied in pigs, and this mostly in the critical period around weaning. RS is believed to exert beneficial effects on the gastrointestinal tract mainly due to higher levels of short chain fatty acids (SCFAs) and an improved microbiota profile. In this study, sows were fed digestible starch (DS) or RS during late gestation and lactation and the possible maternal effect of RS on the overall health of the progeny was assessed. Since RS is also described to have a positive effect on metabolism, and to investigate a metabolic programming of the progeny, half of the piglets per maternal diet were assigned to a high fat diet from weaning on to 10 weeks after. Results No bodyweight differences were found between the four experimental piglet groups. The high fat diet did however impact back fat thickness and meat percentage whereas maternal diet did not influence these parameters. The impact of the high fat diet was also reflected in higher levels of serum cholesterol. No major differences in microbiota could be distinguished, although higher levels of SCFA were seen in the colon of piglets born from RS fed sows, and some differences in SCFA production were observed in the caecum, mainly due to piglet diet. RNA-sequencing on liver and colon scrapings revealed minor differences between the maternal diet groups. Merely a handful of genes was differentially expressed between piglets from DS and RS sows, and network analysis showed only one significant cluster of genes in the liver due to the maternal diet that did not point to meaningful biological pathways. However, the high fat diet resulted in liver gene clusters that were significantly correlated with piglet diet, of which one is annotated for lipid metabolic processes. These clusters were not correlated with maternal diet. Conclusions There is only a minor impact of maternal dietary RS on the progeny, reflected in SCFA changes. A high fat diet given to the progeny directly evokes metabolic changes in the liver, without any maternal programming by a RS diet.
Collapse
|
9
|
Chen H, Liu Y, Li H, Fang Z, Lin Y, Xu S, Li J, Feng B, Wu D, Che L. Nutritional effects pre-weaning on growth performance, carcass traits and meat quality of pigs. ANIMAL PRODUCTION SCIENCE 2020. [DOI: 10.1071/an18368] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The objective of the present study was to investigate the effect of nutritional restriction during the suckling period on growth performance, carcass traits and meat quality of fattening pigs. A one-way experimental design was used. In total, 24 male pigs of normal birthweight (1.54 ± 0.05 kg) were randomly allocated at 7 days of age to three groups: control, fed ad libitum; 30% nutritional restriction (Re30%); and 60% nutritional restriction (Re60%). The Re30% and Re60% groups showed decreased average daily gain, average daily feed intake and feed conversion ratio from Day 7 to Day 28. After Day 28, differences in these parameters were observed only in the Re60% group relative to the control. With regard to hormone levels, the Re60% group showed decreased serum concentration of insulin-like growth factor-1 at Day 28 and increased serum concentration of growth hormone at Day 147. Furthermore, the Re60% group had decreased carcass weight, ham weight and dressing percentage, and increased carcass lean percentage relative to the control, as well as lower cross-sectional area and myofibre diameter of muscle. The Re60% group had lower levels of myosin heavy chain (MyHC) IIx and MyHC IIb mRNA and a higher percentage of MyHC I and MyHC IIa mRNA in longissimus dorsi muscle than the Re30% group. In conclusion, nutritional restriction during the suckling period decreased weaning weight, with post-weaning growth performance, carcass traits and myofibre type affected in the Re60% group rather than Re30% group.
Collapse
|
10
|
Arnaud AP, Rome V, Richard M, Formal M, David-Le Gall S, Boudry G. Post-natal co-development of the microbiota and gut barrier function follows different paths in the small and large intestine in piglets. FASEB J 2019; 34:1430-1446. [PMID: 31914707 DOI: 10.1096/fj.201902514r] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2019] [Revised: 11/04/2019] [Accepted: 11/13/2019] [Indexed: 12/17/2022]
Abstract
Gut microbiota and intestinal barrier co-develop after birth, establishing a homeostatic state whereby mucosal cells cohabit with commensal bacteria. We hypothesized that this post-natal co-development follows different timings depending on the intestinal site considered. Jejunal, ileal, and colonic luminal contents and mucosa were sampled in suckling piglets at post-natal day (PND) 0, 2, 7, 14, and 28. Jejunal, ileal, and colonic luminal microbiota (evaluated by 16S DNA sequencing followed by beta-diversity analysis) clustered at PND2 but colonic microbiota diverge afterwards (P < .05). Mucosal permeability, evaluated in Ussing chambers, increased with age in the jejunum and ileum (P < .05) but not the colon. Expression of pattern recognition receptor (PRR) exhibited different patterns (gradual or sharp increase, decrease, or no change with age, P < .05) depending on PRR and intestinal site considered. Principal component analysis of mucosa data revealed clear clustering of colonic samples, irrespective of the age and clustering of jejunal and ileal samples, with gradual changes with age. Correlation analysis highlighted three families correlating with mucosal parameters: Enterobacteriaceae in the jejunum, Peptostreptococcaceae in the ileum, and Micrococcaceae in the colon. In conclusion, small and large intestine display close microbiota composition early in life but distinct mucosal phenotype and follow very different post-natal development.
Collapse
Affiliation(s)
- Alexis Pierre Arnaud
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France.,Service de chirurgie pédiatrique, CHU rennes, University of Rennes 1, Rennes, France
| | - Véronique Rome
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Marion Richard
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | - Michèle Formal
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| | | | - Gaëlle Boudry
- Institut NuMeCan, INRA, INSERM, Univ Rennes, St-Gilles, France
| |
Collapse
|
11
|
Lemaire M, Dou S, Cahu A, Formal M, Le Normand L, Romé V, Nogret I, Ferret-Bernard S, Rhimi M, Cuinet I, Canlet C, Tremblay-Franco M, Le Ruyet P, Baudry C, Gérard P, Le Huërou-Luron I, Blat S. Addition of dairy lipids and probiotic Lactobacillus fermentum in infant formula programs gut microbiota and entero-insular axis in adult minipigs. Sci Rep 2018; 8:11656. [PMID: 30076313 PMCID: PMC6076243 DOI: 10.1038/s41598-018-29971-w] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2018] [Accepted: 07/23/2018] [Indexed: 02/08/2023] Open
Abstract
Clinical and animal studies have demonstrated beneficial effects of early consumption of dairy lipids and a probiotic, Lactobacillus fermentum (Lf), on infant gut physiology. The objective of this study was to investigate their long-term effects on gut microbiota and host entero-insular axis and metabolism. Piglets were suckled with a milk formula containing only plant lipids (PL), a half-half mixture of plant lipids and dairy lipids (DL), or this mixture supplemented with Lf (DL + Lf). They were weaned on a standard diet and challenged with a high-energy diet until postnatal day 140. DL and DL + Lf modulated gut microbiota composition and metabolism, increasing abundance of several Clostridia genera. Moreover, DL + Lf specifically decreased the faecal content of 2-oxoglutarate and lysine compared to PL and 5-aminovalerate compared to PL and DL. It also increased short-chain fatty acid concentrations like propionate compared to DL. Furthermore, DL + Lf had a beneficial effect on the endocrine function, enhancing caecal GLP-1 and GLP-1 meal-stimulated secretion. Correlations highlighted the consistent relationship between microbiota and gut physiology. Together, our results evidence a beneficial programming effect of DL + Lf in infant formula composition on faecal microbiota and entero-insular axis function.
Collapse
Affiliation(s)
- Marion Lemaire
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
- Lactalis R&D, Retiers, France
| | - Samir Dou
- PEGASE, INRA, Agrocampus Ouest, Saint-Gilles, France
| | - Armelle Cahu
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | - Michèle Formal
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | - Laurence Le Normand
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | - Véronique Romé
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | - Isabelle Nogret
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France
| | | | - Moez Rhimi
- Micalis, INRA, AgroParisTech, Univ Paris-Saclay, Jouy-en-Josas, France
| | | | - Cécile Canlet
- Toxalim, INRA, Univ Toulouse, ENVT, INP-Purpan, UPS, PF MetaToul-AXIOM, Toulouse, France
| | - Marie Tremblay-Franco
- Toxalim, INRA, Univ Toulouse, ENVT, INP-Purpan, UPS, PF MetaToul-AXIOM, Toulouse, France
| | | | | | - Philippe Gérard
- Micalis, INRA, AgroParisTech, Univ Paris-Saclay, Jouy-en-Josas, France
| | | | - Sophie Blat
- INRA, INSERM, Univ Rennes, Nutrition Metabolisms and Cancer, NuMeCan, Rennes, France.
| |
Collapse
|
12
|
Le Bourgot C, Ferret‐Bernard S, Apper E, Taminiau B, Cahu A, Le Normand L, Respondek F, Le Huërou‐Luron I, Blat S. Perinatal short‐chain fructooligosaccharides program intestinal microbiota and improve enteroinsular axis function and inflammatory status in high‐fat diet‐fed adult pigs. FASEB J 2018; 33:301-313. [DOI: 10.1096/fj.201800108r] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Affiliation(s)
- Cindy Le Bourgot
- Tereos Marckolsheim France
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| | | | | | | | - Armelle Cahu
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| | | | | | | | - Sophie Blat
- INRAINSERMUniv RennesNutrition Metabolisms and CancerNuMeCan Rennes France
| |
Collapse
|
13
|
Leblois J, Massart S, Soyeurt H, Grelet C, Dehareng F, Schroyen M, Li B, Wavreille J, Bindelle J, Everaert N. Feeding sows resistant starch during gestation and lactation impacts their faecal microbiota and milk composition but shows limited effects on their progeny. PLoS One 2018; 13:e0199568. [PMID: 29969488 PMCID: PMC6029764 DOI: 10.1371/journal.pone.0199568] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/08/2018] [Indexed: 01/19/2023] Open
Abstract
Background Establishment of a beneficial microbiota profile for piglets as early in life as possible is important as it will impact their future health. In the current study, we hypothesized that resistant starch (RS) provided in the maternal diet during gestation and lactation will be fermented in their hindgut, which would favourably modify their milk and/or gut microbiota composition and that it would in turn affect piglets’ microbiota profile and their absorptive and immune abilities. Methods In this experiment, 33% of pea starch was used in the diet of gestating and lactating sows and compared to control sows. Their faecal microbiota and milk composition were determined and the colonic microbiota, short-chain fatty acids (SCFA) production and gut health related parameters of the piglets were measured two days before weaning. In addition, their overall performances and post-weaning faecal score were also assessed. Results The RS diet modulated the faecal microbiota of the sows during gestation, increasing the Firmicutes:Bacteroidetes ratio and the relative abundance of beneficial genera like Bifidobacterium but these differences disappeared during lactation and maternal diets did not impact the colonic microbiota of their progeny. Milk protein concentration decreased with RS diet and lactose concentration increased within the first weeks of lactation while decreased the week before weaning with the RS diet. No effect of the dietary treatment, on piglets’ bodyweight or diarrhoea frequency post-weaning was observed. Moreover, the intestinal morphology measured as villus height and crypt depths, and the inflammatory cytokines in the intestine of the piglets were not differentially expressed between maternal treatments. Only zonula occludens 1 (ZO-1) was more expressed in the ileum of piglets born from RS sows, suggesting a better closure of the mucosa tight junctions. Conclusion Changes in the microbiota transferred from mother to piglets due to the inclusion of RS in the maternal diet are rather limited even though milk composition was affected.
Collapse
Affiliation(s)
- Julie Leblois
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
- Research Foundation for Industry and Agriculture, National Scientific Research Foundation (FRIA-FNRS), Brussels, Belgium
- * E-mail: (JL); (NE)
| | - Sébastien Massart
- Laboratory of Urban and Integrated PhytoPathology, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Hélène Soyeurt
- Laboratory of Statistics, Informatics and Modelling Applied to Bioengineering, AGROBIOCHEM Department, Teaching and Research Centre (TERRA), Gembloux Agro-Bio Tech, University of Liège, Gembloux, Belgium
| | - Clément Grelet
- Valorisation of Agricultural Products Department, Walloon Agricultural Research Centre, Gembloux, Belgium
| | - Frédéric Dehareng
- Valorisation of Agricultural Products Department, Walloon Agricultural Research Centre, Gembloux, Belgium
| | - Martine Schroyen
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Bing Li
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - José Wavreille
- Production and Sectors Department, Walloon Agricultural Research Centre, Gembloux, Belgium
| | - Jérôme Bindelle
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
| | - Nadia Everaert
- Precision Livestock and Nutrition Unit, Gembloux Agro-Bio Tech, TERRA, Teaching and Research Centre, University of Liège, Gembloux, Belgium
- * E-mail: (JL); (NE)
| |
Collapse
|
14
|
Tari NR, Fan M, Archbold T, Kristo E, Guri A, Arranz E, Corredig M. Effect of milk protein composition of a model infant formula on the physicochemical properties of in vivo gastric digestates. J Dairy Sci 2018; 101:2851-2861. [DOI: 10.3168/jds.2017-13245] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Accepted: 11/27/2017] [Indexed: 11/19/2022]
|
15
|
Val‐Laillet D, Guérin S, Coquery N, Nogret I, Formal M, Romé V, Le Normand L, Meurice P, Randuineau G, Guilloteau P, Malbert C, Parnet P, Lallès J, Segain J. Oral sodium butyrate impacts brain metabolism and hippocampal neurogenesis, with limited effects on gut anatomy and function in pigs. FASEB J 2018; 32:2160-2171. [DOI: 10.1096/fj.201700547rr] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Affiliation(s)
- David Val‐Laillet
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
| | - Sylvie Guérin
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Nicolas Coquery
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Isabelle Nogret
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Michèle Formal
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Véronique Romé
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Laurence Le Normand
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Paul Meurice
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Gwénaëlle Randuineau
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | - Paul Guilloteau
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
| | | | - Patricia Parnet
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
- INRA, Unité Mixte de Recherche (UMR) 1280INRA‐Université de Nantes, Physiologie des Adaptations Nutritionnelles (PhAN)NantesFrance
- Institut des Maladies de l'Appareil DigestifCentre Hospitalier Universitaire (CHU) Ho tel‐DieuNantesFrance
| | - Jean‐Paul Lallès
- Nutrition Metabolisms and Cancer (NuMeCan)INRA, INSERM, Univ Rennes, Université Bretagne Loire (UBL)RennesFrance
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
| | - Jean‐Pierre Segain
- Centre de Recherche en Nutrition Humaine Ouest (CRNH‐Ouest)NantesFrance
- INRA, Unité Mixte de Recherche (UMR) 1280INRA‐Université de Nantes, Physiologie des Adaptations Nutritionnelles (PhAN)NantesFrance
- Institut des Maladies de l'Appareil DigestifCentre Hospitalier Universitaire (CHU) Ho tel‐DieuNantesFrance
| |
Collapse
|
16
|
A review on early gut maturation and colonization in pigs, including biological and dietary factors affecting gut homeostasis. Anim Feed Sci Technol 2017. [DOI: 10.1016/j.anifeedsci.2017.06.011] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
17
|
Ley D, Desseyn JL, Mischke M, Knol J, Turck D, Gottrand F. Early-life origin of intestinal inflammatory disorders. Nutr Rev 2017; 75:175-187. [PMID: 28340001 DOI: 10.1093/nutrit/nuw061] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
A growing body of evidence supports the concept of perinatal programming through which the perinatal environment affects the development of the fetus and infant, thereby modifying the risk profile for disease later in life. Increasing attention is focusing on the role of the early environment in the development of chronic intestinal disorders. Epidemiological studies have highlighted the link between perinatal factors, such as breastfeeding, cesarean delivery, and antibiotic use, and an increased risk for inflammatory bowel disease and/or celiac disease. These links are consistent with the concept of perinatal programming of intestinal inflammatory disorders. Animal models have shown that the early-life environment affects the development of the gastrointestinal tract, but further experimental studies are needed to confirm the long-term effects of the perinatal environment on susceptibility to chronic intestinal disorders later in life. Changes in the development and composition of the intestinal microbiota as well as epigenetic changes are emerging as key mechanisms through which the perinatal environment determines susceptibility to intestinal inflammatory disorders.
Collapse
Affiliation(s)
- Delphine Ley
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Jean-Luc Desseyn
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | | | - Jan Knol
- Nutricia Research, Utrecht, The Netherlands.,Laboratory of Microbiology, Wageningen University, The Netherlands
| | - Dominique Turck
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| | - Frédéric Gottrand
- Lille Inflammation Research International Center (LIRIC) - UMR 995 Inserm, University Lille, CHU Lille, Lille, France
| |
Collapse
|
18
|
Piccolo BD, Mercer KE, Bhattacharyya S, Bowlin AK, Saraf MK, Pack L, Chintapalli SV, Shankar K, Adams SH, Badger TM, Yeruva L. Early Postnatal Diets Affect the Bioregional Small Intestine Microbiome and Ileal Metabolome in Neonatal Pigs. J Nutr 2017; 147:1499-1509. [PMID: 28659406 DOI: 10.3945/jn.117.252767] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 05/10/2017] [Accepted: 06/05/2017] [Indexed: 11/14/2022] Open
Abstract
Background: Breastfeeding is known to be protective against gastrointestinal disorders and may modify gut development. Although the gut microbiome has been implicated, little is known about how early diet affects the small intestine microbiome.Objective: We hypothesized that disparate early diets would promote unique microbial profiles in the small intestines of neonatal pigs.Methods: Male and female 2-d-old White Dutch Landrace pigs were either sow fed or provided dairy (Similac Advance powder; Ross Products Abbott Laboratories) or soy (Enfamil Prosobee Lipil powder; Mead Johnson Nutritionals) infant formulas until day 21. Bacterial ecology was assessed in the contents of the small intestine through the use of 16S ribosomal RNA sequencing. α-Diversity, β-diversity, and differential abundances of operational taxonomic units were assessed by ANOVA, permutational ANOVA, and negative binomial regression, respectively. Ileum tissue metabolomics were measured by LC-mass spectrometry and assessed by weighted correlation network analysis.Results: Greater α-diversity was observed in the duodena of sow-fed compared with formula-fed neonatal pigs (P < 0.05). No differences were observed in the ilea. Firmicutes represented the most abundant phylum across all diets in duodena (78.8%, 80.1%, and 53.4% relative abundance in sow, dairy, and soy groups, respectively), followed by Proteobacteria in sow (12.2%) and dairy (12.4%) groups and Cyanobacteria in soy-fed (36.2%) pigs. In contrast to those in the duodenum, Proteobacteria was the dominant phylum in the ileum, with >60% relative abundance in all of the groups. In the duodenum, 77 genera were altered by diet, followed by 48 in the jejunum and 19 in the ileum. Metabolomics analyses revealed associations between ileum tissue metabolites (e.g., acylcarnitines, 3-aminoisobutyric acid) and diet-responsive microbial genera.Conclusions: These results indicate that the neonatal diet has regional effects on the small intestine microbiome in pigs, with the most pronounced effects occurring in the duodena. Regional effects may be important factors when considering gut tissue metabolism and development in the postnatal period.
Collapse
Affiliation(s)
- Brian D Piccolo
- Arkansas Children's Nutrition Center, Little Rock, AR; .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Kelly E Mercer
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Sudeepa Bhattacharyya
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Anne K Bowlin
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Manish K Saraf
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Lindsay Pack
- Arkansas Children's Nutrition Center, Little Rock, AR
| | - Sree V Chintapalli
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Kartik Shankar
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Sean H Adams
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Thomas M Badger
- Arkansas Children's Nutrition Center, Little Rock, AR.,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and
| | - Laxmi Yeruva
- Arkansas Children's Nutrition Center, Little Rock, AR; .,Department of Pediatrics, University of Arkansas for Medical Sciences, Little Rock, AR; and.,Arkansas Children's Research Institute, Little Rock, AR
| |
Collapse
|
19
|
The microbial community of the gut differs between piglets fed sow milk, milk replacer or bovine colostrum. Br J Nutr 2017; 117:964-978. [PMID: 28460652 DOI: 10.1017/s0007114517000216] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The aim of this study was to characterise the gut microbiota composition of piglets fed bovine colostrum (BC), milk replacer (MR) or sow milk (SM) in the post-weaning period. Piglets (n 36), 23-d old, were randomly allocated to the three diets. Faecal samples were collected at 23, 25, 27 and 30 d of age. Digesta from the stomach, ileum, caecum and mid-colon was collected at 30 d of age. Bacterial DNA from all samples was subjected to amplicon sequencing of the 16S rRNA gene. Bacterial enumerations by culture and SCFA analysis were conducted as well. BC-piglets had the highest abundance of Lactococcus in the stomach (P<0·0001) and ileal (P<0·0001) digesta, whereas SM-piglets had the highest abundance of Lactobacillus in the stomach digesta (P<0·0001). MR-piglets had a high abundance of Enterobacteriaceae in the ileal digesta (P<0·0001) and a higher number of haemolytic bacteria in ileal (P=0·0002) and mid-colon (P=0·001) digesta than SM-piglets. BC-piglets showed the highest colonic concentration of iso-butyric and iso-valeric acid (P=0·02). Sequencing and culture showed that MR-piglets were colonised by a higher number of Enterobacteriaceae, whereas the gut microbiota of BC-piglets was characterised by a change in lactic acid bacteria genera when compared with SM-piglets. We conclude that especially the ileal microbiota of BC-piglets had a closer resemblance to that of SM-piglets in regard to the abundance of potential enteric pathogens than did MR-piglets. The results indicate that BC may be a useful substitute for regular milk replacers, and as a feeding supplement in the immediate post-weaning period.
Collapse
|
20
|
Taranu I, Habeanu M, Gras MA, Pistol GC, Lefter N, Palade M, Ropota M, Sanda Chedea V, Marin DE. Assessment of the effect of grape seed cake inclusion in the diet of healthy fattening-finishing pigs. J Anim Physiol Anim Nutr (Berl) 2017; 102:e30-e42. [PMID: 28247575 DOI: 10.1111/jpn.12697] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2016] [Accepted: 01/14/2017] [Indexed: 01/06/2023]
Abstract
Modulatory capacity of bioactive compounds from different wastes has been scarcely investigated in pigs. This study aimed to evaluate the effects of dietary inclusion of grape seed cakes (GS diet) on performance and plasma biochemistry parameters as health indicators, as well as on several markers related to inflammation and antioxidant defence in the liver of fattening-finishing pigs. Twelve cross-bred pigs (TOPIG) were randomly assigned to one of two experimental diets: control and 5% grape seed cake diet during finishing period (24 days). No effect of GS diet on pig performance and blood biochemistry was observed. However, GS diet decreased significantly (-9.05%, p < .05) the cholesterol concentration (85.71 ± 0.94 mg/dl vs 94.24 ± 2.16 mg/dl) and increased IgA level (+49.90%, p < .05) in plasma (5.04 ± 0.5 mg/ml vs 3.36 ± 0.7 mg/ml). GS cakes decreased the inflammatory response in the liver of pigs fed with GS diet by lowering the Gene expression and protein concentration of pro-inflammatory cytokines (IL-1β, IL-8, TNF-α and IFN-γ) as well as the mRNA abundances of NF-κB signalling molecules. The antioxidant status was not increased by GS diet. The gene expression and activity of catalase decreased significantly. The gene expression of Nrf2, superoxide dismutase, glutathione peroxidase and heat-shock protein decreased, and no effect on their activity was observed with the exception of catalase activity which decreased. However, TBARS was reduced significantly. GS diet showed a modulatory effect on antioxidative status as well as anti-inflammatory and hypocholesterolic properties without effect on pig performance.
Collapse
Affiliation(s)
- I Taranu
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - M Habeanu
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - M A Gras
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - G C Pistol
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - N Lefter
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - M Palade
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - M Ropota
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - V Sanda Chedea
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| | - D E Marin
- INCDBNA-IBNA, National Institute of Research and Development for Biology and Animal Nutrition, Balotesti, Romania
| |
Collapse
|
21
|
Abstract
The present review examines the pig as a model for physiological studies in human subjects related to nutrient sensing, appetite regulation, gut barrier function, intestinal microbiota and nutritional neuroscience. The nutrient-sensing mechanisms regarding acids (sour), carbohydrates (sweet), glutamic acid (umami) and fatty acids are conserved between humans and pigs. In contrast, pigs show limited perception of high-intensity sweeteners and NaCl and sense a wider array of amino acids than humans. Differences on bitter taste may reflect the adaptation to ecosystems. In relation to appetite regulation, plasma concentrations of cholecystokinin and glucagon-like peptide-1 are similar in pigs and humans, while peptide YY in pigs is ten to twenty times higher and ghrelin two to five times lower than in humans. Pigs are an excellent model for human studies for vagal nerve function related to the hormonal regulation of food intake. Similarly, the study of gut barrier functions reveals conserved defence mechanisms between the two species particularly in functional permeability. However, human data are scant for some of the defence systems and nutritional programming. The pig model has been valuable for studying the changes in human microbiota following nutritional interventions. In particular, the use of human flora-associated pigs is a useful model for infants, but the long-term stability of the implanted human microbiota in pigs remains to be investigated. The similarity of the pig and human brain anatomy and development is paradigmatic. Brain explorations and therapies described in pig, when compared with available human data, highlight their value in nutritional neuroscience, particularly regarding functional neuroimaging techniques.
Collapse
|
22
|
Bian G, Ma S, Zhu Z, Su Y, Zoetendal EG, Mackie R, Liu J, Mu C, Huang R, Smidt H, Zhu W. Age, introduction of solid feed and weaning are more important determinants of gut bacterial succession in piglets than breed and nursing mother as revealed by a reciprocal cross-fostering model. Environ Microbiol 2016; 18:1566-77. [DOI: 10.1111/1462-2920.13272] [Citation(s) in RCA: 122] [Impact Index Per Article: 15.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2015] [Accepted: 02/17/2016] [Indexed: 12/23/2022]
Affiliation(s)
- Gaorui Bian
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Shouqing Ma
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Zhigang Zhu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Yong Su
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Erwin G. Zoetendal
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Roderick Mackie
- Department of Animal Sciences; University of Illinois at Urbana-Champaign; IL USA
| | - Junhua Liu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Chunlong Mu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Ruihua Huang
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| | - Hauke Smidt
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
- Laboratory of Microbiology; Wageningen University; Wageningen The Netherlands
| | - Weiyun Zhu
- Laboratory of Gastrointestinal Microbiology, College of Animal Science and Technology; Nanjing Agricultural University; Nanjing P. R. China
| |
Collapse
|
23
|
Storme L, Luton D, Abdennebi-Najar L, Le Huërou-Luron I. [DOHaD: long-term impact of perinatal diseases (IUGR and prematurity)]. Med Sci (Paris) 2016; 32:74-80. [PMID: 26850610 DOI: 10.1051/medsci/20163201012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
The first epidemiological studies showing a link between low birth weight and chronic diseases in adults did not distinguish the origins of low birth weight. A low birth weight may be the result of a premature birth. It can also be caused by an intrauterine growth restriction (IUGR). A child can be both preterm and IUGR. It is clear now that prematurity is an independent risk factor for programming chronic adult diseases. However, unlike adults born IUGR, adults born prematurely do not have an increased risk to develop metabolic syndrome (dyslipidemia or obesity). An increased risk of neurodevelopmental and psychiatric morbidity and hypertension is found after a premature birth. Mechanisms of chronic diseases programming are multiple: they involve both the cause of prematurity and IUGR such as infection / inflammation or placental insufficiency, but also consequences for therapeutic or nutritional strategies needed to support these children. This chapter describes the possible prevention of perinatal programming of noncommunicable diseases.
Collapse
Affiliation(s)
- Laurent Storme
- EA4489, environnement périnatal et santé, faculté de médecine, université Lille 2, hôpital Jeanne de Flandre, CHRU de Lille, 1, rue Eugène Avinée, Lille, France
| | - Dominique Luton
- Maternité, hôpitaux universitaires Paris Nord Val-de-Seine, Assistance publique-hôpitaux de Paris, université Paris VII, Paris, France
| | - Latifa Abdennebi-Najar
- UP 2012.10.101, Expression des gènes et régulation épigénétique par l'aliment, institut polytechnique LaSalle, Beauvais, France
| | - Isabelle Le Huërou-Luron
- UR1341, alimentation et adaptations digestives, nerveuses et comportementales, Inra, Saint-Gilles, France
| |
Collapse
|
24
|
Desclée de Maredsous C, Oozeer R, Barbillon P, Mary-Huard T, Delteil C, Blachier F, Tomé D, van der Beek EM, Davila AM. High-Protein Exposure during Gestation or Lactation or after Weaning Has a Period-Specific Signature on Rat Pup Weight, Adiposity, Food Intake, and Glucose Homeostasis up to 6 Weeks of Age. J Nutr 2016; 146:21-9. [PMID: 26674762 DOI: 10.3945/jn.115.216465] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 10/26/2015] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Early-life nutrition has a programming effect on later metabolic health; however, the impact of exposure to a high-protein (HP) diet is still being investigated. OBJECTIVE This study evaluated the consequences on pup phenotype of an HP diet during gestation and lactation and after weaning. METHODS Wistar rat dams were separated into 2 groups fed an HP (55% protein) or normal protein (NP) (control; 20% protein) isocaloric diet during gestation, and each group subsequently was separated into 2 subgroups that were fed an HP or NP diet during lactation. After weaning, male and female pups from each mother subgroup were separated into 2 groups that were fed either an NP or HP diet until they were 6 wk old. Measurements included weight, food intake, body composition, blood glucose, insulin, glucagon, leptin, insulin-like growth factor I, and lipids. RESULTS Feeding mothers the HP diet during gestation or lactation induced lower postweaning pup weight (gestation diet × time, P < 0.0001; lactation diet × time, P < 0.0001). Regardless of dams' diets, pups receiving HP compared with NP diet after weaning had 7% lower weight (NP, 135.0 ± 2.6 g; HP, 124.4 ± 2.5 g; P < 0.0001), 16% lower total energy intake (NP, 777 ± 14 kcal; HP, 649 ± 13 kcal; P < 0.0001) and 31% lower adiposity (P < 0.0001). Pups receiving HP compared with NP diet after weaning had increased blood glucose, insulin, and glucagon when food deprived (P < 0.0001 for all). The HP compared with the NP diet during gestation induced higher blood glucose in food-deprived rats (NP, 83.2 ± 2.1 mg/dL; HP, 91.2 ± 2.1 mg/dL; P = 0.046) and increased plasma insulin in fed pups receiving the postweaning NP diet (gestation diet × postweaning diet, P = 0.02). CONCLUSION Increasing the protein concentration of the rat dams' diet during gestation, and to a lesser extent during lactation, and of the pups' diet after weaning influenced pup phenotype, including body weight, fat accumulation, food intake, and glucose tolerance at 6 wk of age.
Collapse
Affiliation(s)
- Caroline Desclée de Maredsous
- UMR 914 Nutrition Physiology and Ingestive Behavior, French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France; Danone Nutricia Research, Utrecht, Netherlands
| | | | - Pierre Barbillon
- UMR 518 Applied Mathematics and Informatics (MIA), French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France; and
| | - Tristan Mary-Huard
- UMR 518 Applied Mathematics and Informatics (MIA), French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France; and Quantitative Genetics Evolution Le Moulon, French National Institute for Agricultural Research (INRA), Paris-Sud University, Paris Saclay University, AgroParisTech, CNRS, Gif-sur-Yvette, France
| | - Corine Delteil
- UMR 914 Nutrition Physiology and Ingestive Behavior, French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France
| | - François Blachier
- UMR 914 Nutrition Physiology and Ingestive Behavior, French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France
| | - Daniel Tomé
- UMR 914 Nutrition Physiology and Ingestive Behavior, French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France
| | | | - Anne-Marie Davila
- UMR 914 Nutrition Physiology and Ingestive Behavior, French National Institute for Agricultural Research (INRA)/AgroParisTech, Paris Saclay University, Paris, France;
| |
Collapse
|
25
|
Chen L, Mao X, Han F, Yu B, He J, Zheng P, Yu J, Luo J, Chen D. The effect of high nutrient on the growth performance, adipose deposition and gene expression of lipid metabolism in the neonatal intrauterine growth-retarded piglets. JOURNAL OF APPLIED ANIMAL RESEARCH 2015. [DOI: 10.1080/09712119.2015.1091330] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/22/2022]
Affiliation(s)
- Lin Chen
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Xiangbing Mao
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Fei Han
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Bing Yu
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Jun He
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Ping Zheng
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Jie Yu
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Junqiu Luo
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| | - Daiwen Chen
- Key Laboratory of Animal Disease-Resistance Nutrition, Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, Ya'an, People's Republic of China
| |
Collapse
|
26
|
Impact of kefir derived Lactobacillus kefiri on the mucosal immune response and gut microbiota. J Immunol Res 2015; 2015:361604. [PMID: 25811034 PMCID: PMC4355334 DOI: 10.1155/2015/361604] [Citation(s) in RCA: 78] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Revised: 09/22/2014] [Accepted: 09/23/2014] [Indexed: 12/16/2022] Open
Abstract
The evaluation of the impact of probiotics on host health could help to understand how they can be used in the prevention of diseases. On the basis of our previous studies and in vitro assays on PBMC and Caco-2 ccl20:luc reporter system presented in this work, the strain Lactobacillus kefiri CIDCA 8348 was selected and administrated to healthy Swiss mice daily for 21 days. The probiotic treatment increased IgA in feces and reduced expression of proinflammatory mediators in Peyer Patches and mesenteric lymph nodes, where it also increased IL-10. In ileum IL-10, CXCL-1 and mucin 6 genes were upregulated; meanwhile in colon mucin 4 was induced whereas IFN-γ, GM-CSF, and IL-1β genes were downregulated. Moreover, ileum and colon explants showed the anti-inflammatory effect of L. kefiri since the LPS-induced increment of IL-6 and GM-CSF levels in control mice was significantly attenuated in L. kefiri treated mice. Regarding fecal microbiota, DGGE profiles allowed differentiation of experimental groups in two separated clusters. Quantitative PCR analysis of different bacterial groups revealed only significant changes in Lactobacillus population. In conclusion, L. kefiri is a good candidate to be used in gut inflammatory disorders.
Collapse
|
27
|
Arnal ME, Zhang J, Erridge C, Smidt H, Lallès JP. Maternal antibiotic-induced early changes in microbial colonization selectively modulate colonic permeability and inducible heat shock proteins, and digesta concentrations of alkaline phosphatase and TLR-stimulants in swine offspring. PLoS One 2015; 10:e0118092. [PMID: 25689154 PMCID: PMC4331088 DOI: 10.1371/journal.pone.0118092] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2014] [Accepted: 01/05/2015] [Indexed: 12/21/2022] Open
Abstract
Elevated intake of high energy diets is a risk factor for the development of metabolic diseases and obesity. High fat diets cause alterations in colonic microbiota composition and increase gut permeability to bacterial lipopolysaccharide, and subsequent low-grade chronic inflammation in mice. Chronic inflammatory bowel diseases are increasing worldwide and may involve alterations in microbiota-host dialog. Metabolic disorders appearing in later life are also suspected to reflect changes in early programming. However, how the latter affects the colon remains poorly studied. Here, we hypothesized that various components of colonic physiology, including permeability, ion exchange and protective inducible heat shock proteins (HSP) are influenced in the short- and long-terms by early disturbances in microbial colonization. The hypothesis was tested in a swine model. Offspring were born to control mothers (n = 12) or mothers treated with the antibiotic (ATB) amoxicillin around parturition (n = 11). Offspring were slaughtered between 14 and 42 days of age to study short-term effects. For long-term effects, young adult offspring from the same litters consumed a normal or a palm oil-enriched diet for 4 weeks between 140 and 169 days of age. ATB treatment transiently modified maternal fecal microbiota although the minor differences observed for offspring colonic microbiota were nonsignificant. In the short-term, consistently higher HSP27 and HSP70 levels and transiently increased horseradish peroxidase permeability in ATB offspring colon were observed. Importantly, long-term consequences included reduced colonic horseradish peroxidase permeability, and increased colonic digesta alkaline phosphatase (AP) and TLR2- and TLR4-stimulant concentrations in rectal digesta in adult ATB offspring. Inducible HSP27 and HSP70 did not change. Interactions between early ATB treatment and later diet were noted for paracellular permeability and concentrations of colonic digesta AP. In conclusion, our data suggest that early ATB-induced changes in bacterial colonization modulate important aspects of colonic physiology in the short- and long-terms.
Collapse
Affiliation(s)
- Marie-Edith Arnal
- Food and Digestive, Central and Behavioral Adaptation Department, French National Institute for Research in Agriculture, Saint-Gilles, France
| | - Jing Zhang
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Clett Erridge
- Department of Cardiovascular Sciences, Glenfield General Hospital, University of Leicester, Leicester, United Kingdom
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jean-Paul Lallès
- Food and Digestive, Central and Behavioral Adaptation Department, French National Institute for Research in Agriculture, Saint-Gilles, France
- * E-mail:
| |
Collapse
|
28
|
Mukhopadhya A, Noronha N, Bahar B, Ryan MT, Murray BA, Kelly PM, O'Loughlin IB, O'Doherty JV, Sweeney T. The anti-inflammatory potential of a moderately hydrolysed casein and its 5 kDa fraction in in vitro and ex vivo models of the gastrointestinal tract. Food Funct 2015; 6:612-21. [DOI: 10.1039/c4fo00689e] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
Bioactive peptides from milk can impart a wide range of physiological benefits without the allergies and intolerance associated with the consumption of whole milk.
Collapse
Affiliation(s)
- A. Mukhopadhya
- School of Veterinary Medicine
- UCD
- Belfield
- Dublin
- Food for Health Ireland (FHI)
| | - N. Noronha
- School of Agriculture & Food Science
- UCD
- Belfield
- Dublin
- Food for Health Ireland (FHI)
| | - B. Bahar
- School of Veterinary Medicine
- UCD
- Belfield
- Dublin
| | - M. T. Ryan
- School of Veterinary Medicine
- UCD
- Belfield
- Dublin
| | - B. A. Murray
- Teagasc Food Research Centre
- Moorepark
- Fermoy
- Co. Cork
- Ireland
| | - P. M. Kelly
- Teagasc Food Research Centre
- Moorepark
- Fermoy
- Co. Cork
- Ireland
| | | | | | - T. Sweeney
- School of Veterinary Medicine
- UCD
- Belfield
- Dublin
- Food for Health Ireland (FHI)
| |
Collapse
|
29
|
Fan W, Tang Y, Qu Y, Cao F, Huo G. Infant formula supplemented with low protein and high carbohydrate alters the intestinal microbiota in neonatal SD rats. BMC Microbiol 2014; 14:279. [PMID: 25403909 PMCID: PMC4243196 DOI: 10.1186/s12866-014-0279-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 10/30/2014] [Indexed: 12/26/2022] Open
Abstract
Background Infant microbiota is influenced by numerous factors, such as delivery mode, environment, prematurity and diet (breast milk or formula) and last but not least, the diet composition. In the diet composition, protein and carbohydrate are very important for the growth of microbiota, many infant fomulas (different ratio protein/carbohydrate) can regulate the development of gut microbiota by different metabolism. The effect of low-protein, high-carbohydrate infant formula on the establishment of microbiota remains unclear, and the effect of human breast milk on the gut microbiota of the rats has also not been reported. Results In a 7 d intervention, a total of 36 neonatal SD rats (14 d old) were randomly assigned to the following groups: (1) breast-fed group (A group); (2) low-protein, high-carbohydrate infant formula-fed group (B group); (3) human breast milk-fed group (C group). After 7 days, we selected 6 rats at random from each group to study. Microbial composition in the contents of the large intestines was analysed by Miseq Sequencing. Significantly different (p<0.05) microbial colonisation patterns were observed in the large intestines of breast-fed group from low-protein, high-carbohydrate infant formula-fed and human breast milk-fed rats, but the microbiota of low-protein, high-carbohydrate infant formula-fed group and human breast milk-fed group have high similarity. At the phylum level, the absolute quantity of Bacteroidetes, Firmicutes and Proteobacteria (p<0.001) significantly differentiated in breast-fed group from low- protein, high- carbohydrate infant formula-fed and human breast milk-fed groups. Lachnospiraceae, Bacteroidaceae, Porphyromonadaceae and Prevotellaceae were the 4 top families in breast-fed group, but the top 4 families in low-protein, high- carbohydrate infant formula-fed and human breast milk-fed groups were the same, which were Bacteroidaceae, Enterobacteriaceae, Porphyromonadaceae and Lachnospiraceae. At the genus level, Bacteroides was the most abundant division, their OTUS abundance in three groups was 14.91%, 35.94%, 43.24% respectively. Conclusions This study showed that infant formula closer resembling human milk was more different than rats’ breast milk and led to a microbiota profile similar to that for human breast milk-fed neonates. The finding could support a new thinking to develop infant formulas, and provide much more details than what is known previously. Electronic supplementary material The online version of this article (doi:10.1186/s12866-014-0279-2) contains supplementary material, which is available to authorized users.
Collapse
|
30
|
Taranu I, Gras M, Pistol GC, Motiu M, Marin DE, Lefter N, Ropota M, Habeanu M. ω-3 PUFA rich camelina oil by-products improve the systemic metabolism and spleen cell functions in fattening pigs. PLoS One 2014; 9:e110186. [PMID: 25303320 PMCID: PMC4193896 DOI: 10.1371/journal.pone.0110186] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2014] [Accepted: 09/08/2014] [Indexed: 12/31/2022] Open
Abstract
Camelina oil-cakes results after the extraction of oil from Camelina sativa plant. In this study, camelina oil-cakes were fed to fattening pigs for 33 days and its effect on performance, plasma biochemical analytes, pro-/anti-inflammatory mediators and antioxidant detoxifying defence in spleen was investigated in comparison with sunflower meal. 24 crossbred TOPIG pigs were randomly assigned to one of two experimental dietary treatments containing either 12% sunflower meal (treatment 1-T1), or 12.0% camelina oil-cakes, rich in polyunsaturated fatty acids ω-3 (ω-3 PUFA) (treatment 2-T2). The results showed no effect of T2 diet (camelina cakes) on feed intake, average weight gain or feed efficiency. Consumption of camelina diet resulted in a significant decrease in plasma glucose concentration (18.47%) with a trend towards also a decrease of plasma cholesterol. In spleen, T2 diet modulated cellular immune response by decreasing the protein and gene expression of pro-inflammatory markers, interleukin 1-beta (IL-1β), tumor necrosis factor alpha (TNF-α), interleukin 6 (IL-6) and interleukin (IL-8) and cyclooxigenase 2 (COX-2) in comparison with T1 diet. By contrast, T2 diet increased (P<0.05) in spleen the mRNA expression of antioxidant enzymes, catalase (CAT), superoxide dismutase (SOD), and glutathione peroxidase 1 (GPx1) by 3.43, 2.47 and 1.83 fold change respectively, inducible nitric oxide synthase (iNOS) (4.60 fold), endothelial nitric oxide synthase (eNOS) (3.23 fold) and the total antioxidant level (9.02%) in plasma. Camelina diet increased also peroxisome-proliferator activated receptor gamma (PPAR-γ) mRNA and decreased that of mitogen-activated protein kinase 14 (p38α MAPK) and nuclear factor of kappa light polypeptide gene enhancer in B-cells (NF-κB). At this level of inclusion (12%) camelina oil-cakes appears to be a potentially alternative feed source for pig which preserves a high content of ω-3 PUFA indicating antioxidant properties by the stimulation of detoxifying enzymes expression and the suppression of spleen pro-inflammatory markers.
Collapse
Affiliation(s)
- Ionelia Taranu
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Mihail Gras
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Gina Cecilia Pistol
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Monica Motiu
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Daniela E. Marin
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Nicoleta Lefter
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Mariana Ropota
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| | - Mihaela Habeanu
- INCDBNA-IBNA, National Institute of Research and development for Biology and Animal Nutrition, Balotesti, Romania
| |
Collapse
|
31
|
A high-protein formula increases colonic peptide transporter 1 activity during neonatal life in low-birth-weight piglets and disturbs barrier function later in life. Br J Nutr 2014; 112:1073-80. [DOI: 10.1017/s0007114514001901] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Dietary peptides are absorbed along the intestine through peptide transporter 1 (PepT-1) which is highly responsive to dietary protein level. PepT-1 is also involved in gut homeostasis, both initiating and resolving inflammation. Low-birth-weight (LBW) neonates are routinely fed a high-protein (HP) formula to enhance growth. However, the influence of this nutritional practice on PepT-1 activity is unknown. Intestinal PepT-1 activity was compared in normal-birth-weight (NBW) and LBW piglets. The effect of HP v. normal-protein (NP) formula feeding on PepT-1 activity and gut homeostasis in LBW piglets was evaluated, during the neonatal period and in adulthood. Flux of cephalexin (CFX) across the tissue mounted in Ussing chambers was used as an indicator of PepT-1 activity. CFX flux was greater in the ileum, but not jejunum or colon, of LBW than NBW piglets during the neonatal period. When LBW piglets were formula-fed, the HP formula increased colonic CFX during the 1st week of life. Later in life, intestinal CFX fluxes and barrier function were similar whether LBW pigs had been fed NP or HP formula. However, colonic permeability of HP- but not NP-fed pigs increased when luminal pH was brought to 6·0. The formyl peptide N-formyl methionyl-leucyl-phenylalanine conferred colonic barrier protection in HP-fed piglets. Heat shock protein 27 levels in the colonic mucosa of HP-fed LBW pigs correlated with the magnitude of response to the acidic challenge. In conclusion, feeding a HP formula enhanced colonic PepT-1 activity in LBW pig neonates and increased sensitivity of the colon to luminal stress in adulthood.
Collapse
|
32
|
Joly Condette C, Khorsi-Cauet H, Morlière P, Zabijak L, Reygner J, Bach V, Gay-Quéheillard J. Increased gut permeability and bacterial translocation after chronic chlorpyrifos exposure in rats. PLoS One 2014; 9:e102217. [PMID: 25019507 PMCID: PMC4096588 DOI: 10.1371/journal.pone.0102217] [Citation(s) in RCA: 73] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 06/17/2014] [Indexed: 12/15/2022] Open
Abstract
The epithelium's barrier function is crucial for maintaining homeostasis and preventing the passage of food antigens and luminal bacteria. This function is essentially subserved by tight junctions (TJs), multiprotein complexes located in the most apical part of the lateral membrane. Some gastrointestinal disease states are associated with elevated intestinal permeability to macromolecules. In a study on rats, we determined the influence of chronic, daily ingestion of chlorpyrifos (CPF, a pesticide that crosses the placental barrier) during pre- and postnatal periods on intestinal permeability and TJ characteristics in the pups. Fluorescein isothiocyanate (FITC)-dextran was used as a marker of paracellular transport and mucosal barrier dysfunction. Pups were gavaged with FITC-dextran solution and blood samples were collected every 30 min for 400 min and analyzed spectrofluorimetrically. At sacrifice, different intestinal segments were resected and prepared for analysis of the transcripts (qPCR) and localization (using immunofluorescence) of ZO-1, occludin and claudins (scaffolding proteins that have a role in the constitution of TJs). In rats that had been exposed to CPF in utero and after birth, we observed a progressive increase in FITC-dextran passage across the epithelial barrier from 210 to 325 min at day 21 after birth (weaning) but not at day 60 (adulthood). At both ages, there were significant changes in intestinal TJ gene expression, with downregulation of ZO-1 and occludin and upregulation of claudins 1 and 4. In some intestinal segments, there were changes in the cellular localization of ZO-1 and claudin 4 immunostaining. Lastly, bacterial translocation to the spleen was also observed. The presence of CPF residues in food may disturb epithelial homeostasis in rats. Changes in TJ protein expression and localization may be involved in gut barrier dysfunction in this model. Uncontrolled passage of macromolecules and bacteria across the intestinal epithelium may be a risk factor for digestive inflammatory diseases.
Collapse
Affiliation(s)
- Claire Joly Condette
- Peritox Laboratory, EA4285 UMI01 Ineris, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
| | - Hafida Khorsi-Cauet
- Peritox Laboratory, EA4285 UMI01 Ineris, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
| | - Patrice Morlière
- INSERMU1088, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
- Biochemistry Laboratory, Human Biology Centre, Amiens University Hospital, Amiens, France
| | | | - Julie Reygner
- Peritox Laboratory, EA4285 UMI01 Ineris, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
| | - Véronique Bach
- Peritox Laboratory, EA4285 UMI01 Ineris, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
| | - Jérôme Gay-Quéheillard
- Peritox Laboratory, EA4285 UMI01 Ineris, Faculty of Medicine, Jules Verne University of Picardy, Amiens, France
- * E-mail:
| |
Collapse
|
33
|
Desaldeleer C, Ferret-Bernard S, de Quelen F, Le Normand L, Perrier C, Savary G, Romé V, Michel C, Mourot J, Le Huërou-Luron I, Boudry G. Maternal 18:3n-3 favors piglet intestinal passage of LPS and promotes intestinal anti-inflammatory response to this bacterial ligand. J Nutr Biochem 2014; 25:1090-8. [PMID: 25087993 DOI: 10.1016/j.jnutbio.2014.05.014] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Revised: 05/22/2014] [Accepted: 05/29/2014] [Indexed: 11/15/2022]
Abstract
We recently observed that maternal 18:3n-3 increases piglet jejunal permeability. We hypothesized that this would favor intestinal lipopolysaccharide (LPS) passage and alter gut immune system education toward this bacterial ligand. Sows were fed 18:3n-3 or 18:2n-6 diets throughout gestation and lactation. In each litter, two piglets were given oral Gram-negative spectrum antibiotic from post-natal day (PND) 14 to 28. All piglets were weaned on a regular diet at PND28. 18:3n-3 piglets exhibited greater jejunal permeability to FITC-LPS at PND28. Levels of 18:3n-3 but neither 20:5n-3 nor 20:4n-6 were greater in mesenteric lymph nodes (MLN) of 18:3n-3 piglets. Jejunal explant or MLN cell cytokine responses to LPS were not influenced by the maternal diet. Antibiotic increased jejunal permeability to FITC-LPS and lowered the level of 20:5n-3 in MLN, irrespective of the maternal diet. At PND52, no long-lasting effect of the maternal diet or antibiotic treatment on jejunal permeability was noticed. 18:3n-3 and 20:4n-6 levels were greater and lower, respectively, in MLN of 18:3n-3 compared to 18:2n-6 piglets. IL-10 production by MLN cells in response to LPS was greater in the 18:3n-3 group, irrespective of the neonatal antibiotic treatment. IL-8 secretion by jejunal explants in response to LPS was lower in antibiotic-treated 18:3n-3 compared to 18:2n-6 piglets. Finally, proportion of MHC class II(+) antigen-presenting cells was greater in 18:3n-3 than 18:2n-6 MLN cells. In conclusion, maternal 18:3n-3 directs the intestinal immune response to LPS toward an anti-inflammatory profile beyond the breastfeeding period; microbiota involvement seems dependent of the immune cells considered.
Collapse
Affiliation(s)
- Cécile Desaldeleer
- Département de Médecine de l'Enfant et de l'Adolescent, CHU Rennes, Rennes, France; INRA UR1341 ADNC, Saint Gilles, France
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Boudry G, Hamilton M. 35. Milk formula and intestinal barrier function. HUMAN HEALTH HANDBOOKS 2014. [DOI: 10.3920/978-90-8686-223-8_35] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
|
35
|
Carasi P, Díaz M, Racedo SM, De Antoni G, Urdaci MC, Serradell MDLA. Safety characterization and antimicrobial properties of kefir-isolated Lactobacillus kefiri. BIOMED RESEARCH INTERNATIONAL 2014; 2014:208974. [PMID: 24955346 PMCID: PMC4052788 DOI: 10.1155/2014/208974] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 02/20/2014] [Revised: 04/17/2014] [Accepted: 04/21/2014] [Indexed: 01/18/2023]
Abstract
Lactobacilli are generally regarded as safe; however, certain strains have been associated with cases of infection. Our workgroup has already assessed many functional properties of Lactobacillus kefiri, but parameters regarding safety must be studied before calling them probiotics. In this work, safety aspects and antimicrobial activity of L. kefiri strains were studied. None of the L. kefiri strains tested caused α- or β-hemolysis. All the strains were susceptible to tetracycline, clindamycin, streptomycin, ampicillin, erythromycin, kanamycin, and gentamicin; meanwhile, two strains were resistant to chloramphenicol. On the other hand, all L. kefiri strains were able to inhibit both Gram(+) and Gram(-) pathogens. Regarding the in vitro results, L. kefiri CIDCA 8348 was selected to perform in vivo studies. Mice treated daily with an oral dose of 10(8) CFU during 21 days showed no signs of pain, lethargy, dehydration, or diarrhea, and the histological studies were consistent with those findings. Moreover, no differences in proinflammatory cytokines secretion were observed between treated and control mice. No translocation of microorganisms to blood, spleen, or liver was observed. Regarding these findings, L. kefiri CIDCA 8348 is a microorganism isolated from a dairy product with a great potential as probiotic for human or animal use.
Collapse
Affiliation(s)
- Paula Carasi
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, de La Plata, 47 y 115 s/n, CP, 1900 La Plata, Argentina
| | - Mariángeles Díaz
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, de La Plata, 47 y 115 s/n, CP, 1900 La Plata, Argentina
| | - Silvia M. Racedo
- Laboratoire de Microbiologie et Biochimie Appliquée (LBMA), Université de Bordeaux, UMR 5248, Bordeaux Sciences Agro, 1 Cours du Général de Gaulle, 33175 Gradignan, France
| | - Graciela De Antoni
- Cátedra de Microbiología, Departamento de Ciencias Biológicas, de La Plata, 47 y 115 s/n, CP, 1900 La Plata, Argentina
| | - María C. Urdaci
- Laboratoire de Microbiologie et Biochimie Appliquée (LBMA), Université de Bordeaux, UMR 5248, Bordeaux Sciences Agro, 1 Cours du Général de Gaulle, 33175 Gradignan, France
| | | |
Collapse
|
36
|
Chow J, Panasevich MR, Alexander D, Vester Boler BM, Rossoni Serao MC, Faber TA, Bauer LL, Fahey GC. Fecal Metabolomics of Healthy Breast-Fed versus Formula-Fed Infants before and during In Vitro Batch Culture Fermentation. J Proteome Res 2014; 13:2534-42. [DOI: 10.1021/pr500011w] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Affiliation(s)
- JoMay Chow
- Abbott Nutrition, Columbus, Ohio 43219, United States
| | - Matthew R. Panasevich
- Department
of Animal Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Danny Alexander
- Metabolon,
Inc., Durham, North Carolina 27713, United States
| | | | | | - Trevor A. Faber
- Department
of Animal Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - Laura L. Bauer
- Department
of Animal Sciences, University of Illinois, Urbana, Illinois 61801, United States
| | - George C. Fahey
- Department
of Animal Sciences, University of Illinois, Urbana, Illinois 61801, United States
| |
Collapse
|
37
|
Arnal ME, Zhang J, Messori S, Bosi P, Smidt H, Lallès JP. Early changes in microbial colonization selectively modulate intestinal enzymes, but not inducible heat shock proteins in young adult Swine. PLoS One 2014; 9:e87967. [PMID: 24505340 PMCID: PMC3913709 DOI: 10.1371/journal.pone.0087967] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Accepted: 01/02/2014] [Indexed: 12/30/2022] Open
Abstract
Metabolic diseases and obesity are developing worldwide in a context of plethoric intake of high energy diets. The intestine may play a pivotal role due to diet-induced alterations in microbiota composition and increased permeability to bacterial lipopolysaccharide inducing metabolic inflammation. Early programming of metabolic disorders appearing in later life is also suspected, but data on the intestine are lacking. Therefore, we hypothesized that early disturbances in microbial colonization have short- and long-lasting consequences on selected intestinal components including key digestive enzymes and protective inducible heat shock proteins (HSP). The hypothesis was tested in swine offspring born to control mothers (n = 12) or mothers treated with the antibiotic amoxicillin around parturition (n = 11), and slaughtered serially at 14, 28 and 42 days of age to assess short-term effects. To evaluate long-term consequences, young adult offspring from the same litters were offered a normal or a fat-enriched diet for 4 weeks between 140 and 169 days of age and were then slaughtered. Amoxicillin treatment transiently modified both mother and offspring microbiota. This was associated with early but transient reduction in ileal alkaline phosphatase, HSP70 (but not HSP27) and crypt depth, suggesting a milder or delayed intestinal response to bacteria in offspring born to antibiotic-treated mothers. More importantly, we disclosed long-term consequences of this treatment on jejunal alkaline phosphatase (reduced) and jejunal and ileal dipeptidylpeptidase IV (increased and decreased, respectively) of offspring born to antibiotic-treated dams. Significant interactions between early antibiotic treatment and later diet were observed for jejunal alkaline phosphatase and sucrase. By contrast, inducible HSPs were not affected. In conclusion, our data suggest that early changes in bacterial colonization not only modulate intestinal architecture and function transiently, but also exert site- and sometimes diet-specific long-term effects on key components of intestinal homeostasis.
Collapse
Affiliation(s)
- Marie-Edith Arnal
- Food and Digestive, Central and Behavioral Adaptation Department, French National Institute for Research in Agriculture, Saint-Gilles, France
| | - Jing Zhang
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Stefano Messori
- Department of Agricultural and Food Sciences, University of Bologna, Reggio Emilia, Italy
| | - Paolo Bosi
- Department of Agricultural and Food Sciences, University of Bologna, Reggio Emilia, Italy
| | - Hauke Smidt
- Laboratory of Microbiology, Wageningen University, Wageningen, The Netherlands
| | - Jean-Paul Lallès
- Food and Digestive, Central and Behavioral Adaptation Department, French National Institute for Research in Agriculture, Saint-Gilles, France
| |
Collapse
|
38
|
Boudry G, Jamin A, Chatelais L, Gras-Le Guen C, Michel C, Le Huërou-Luron I. Dietary protein excess during neonatal life alters colonic microbiota and mucosal response to inflammatory mediators later in life in female pigs. J Nutr 2013; 143:1225-32. [PMID: 23761650 DOI: 10.3945/jn.113.175828] [Citation(s) in RCA: 43] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The interplay between the colonic microbiota and gut epithelial and immune cells during the neonatal period, which establishes the structure of the microbiota and programs mucosal immunity, is affected by the diet. We hypothesized that protein-enriched milk formula would disturb this interplay through greater flux of protein entering the colon, with consequences later in life. Piglets were fed from postnatal day (PND) 2 to 28 either a normal-protein formula (NP; 51 g protein/L) or high-protein formula (HP; 77 g protein/L) and weaned at PND28, when they received standard diets until PND160. HP feeding transiently increased the quantity of protein entering the colon (PND7) but did not change the microbiota composition at PND28, except for a higher production of branched-chain fatty acids (BCFAs) in an in vitro fermentation test (P < 0.05). HP piglets had greater colonic mucosa densities of cluster of differentiation (CD) 3(+) and CD172(+) cells and lower Il-1β and Tnfα mRNA levels at PND28 (P < 0.05). Later in life (PND160), HP females, but not males, had a higher increase in colonic permeability after ex vivo oxidative stress and higher cytokine secretion in response to lipopolysaccharide in colonic explant cultures than NP females (P < 0.05). HP females also had lower colonic amounts of F. prausnitzii and BCFAs (P < 0.05). BCFAs displayed a dose-dependent protection against inflammation-induced alteration of barrier function in Caco-2 cells (P < 0.05). In conclusion, protein-enriched formula had little impact on colonic microbiota, but it modified colonic immune cell development and had a long-term effect on adult colonic mucosa sensitivity to inflammatory insults, probably through microbiotal and hormonal factors.
Collapse
|
39
|
Lallès JP. Long term effects of pre- and early postnatal nutrition and environment on the gut. J Anim Sci 2013; 90 Suppl 4:421-9. [PMID: 23365399 DOI: 10.2527/jas.53904] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
The Developmental Origins of Health and Disease hypothesis formulated in the early 1990 s has stimulated research on long-term effects of early nutrition and environment over the last decades. Long-term is understood in this review as physiologically relevant periods such as after weaning, around sexual maturity, and in adulthood, as opposed to early developmental periods. The small and large intestines as targets for the study of long-term effects have received little attention until recent years and the stomach has been considered very rarely. Data have accumulated for laboratory animal models but they are still scarce in the swine species. Following the epidemics of metabolic diseases and obesity in western countries, experimental evidence has been published showing that nutritional factors, including energy, fat and fatty acids, protein, and micronutrients impact various facets of gut function. These include alterations in intestinal digestive, absorptive, secretory, barrier, and defense systems, often in a way potentially detrimental to the host. Environmental factors with long-term influence include stress (e.g., maternal deprivation, neonatal gut irritation), chemical pollutants (e.g., bisphenol A), and gut microbiota disturbances (e.g., by antibiotics). Examples of such long-term effects on the gut are provided in both laboratory animals and pigs together with underlying physiological mechanisms whenever available. Experimental evidence for the involvement of underlying epigenetic modifications (e.g., genomic DNA methylation) in long-term studies has just started to emerge with regard to the gastrointestinal tract. Also, interactions between the microbiota and the host are being considered pivotal in the early programming of gut functions. Finally, suggestions for future research are provided in order to better understand and then control early programming as an attempt to optimize vital functions of the gastrointestinal tract throughout adult life.
Collapse
Affiliation(s)
- J P Lallès
- Institut National de la Recherche Agronomique, UR1341 ADNC, Department of Nutrition & Digestive, Nervous and Behavioral Adaptations, F-35590 Saint-Gilles, France.
| |
Collapse
|
40
|
Effects of high nutrient intake on the growth performance, intestinal morphology and immune function of neonatal intra-uterine growth-retarded pigs. Br J Nutr 2013; 110:1819-27. [PMID: 23596997 DOI: 10.1017/s0007114513001232] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Intra-uterine growth-retarded (IUGR) neonates have shown an impairment of postnatal intestinal development and function. We hypothesised that the immune function of IUGR neonates might be affected by increased nutrient intake (NI) during the suckling period. Therefore, we investigated the effects of high NI (HNI) on the growth performance, intestinal morphology and immunological response of IUGR and normal-birth weight (NBW) piglets. A total of twelve pairs of IUGR and NBW piglets (7 d old) were randomly assigned to two different nutrient-level formula milk groups. After 21 d of rearing, growth performance, the composition of peripheral leucocytes, serum cytokines and intestinal innate immune-related genes involved in the Toll-like receptor (TLR)-4–myeloid differentiation factor 88–NF-κB pathway were determined. The results indicated that IUGR decreased the average daily DM intake (ADMI) and the average daily growth (ADG). However, the ADMI and ADG were increased by HNI, irrespective of body weight. Likewise, serum cytokines (TNF-α and IL-1β) and ileal gene expressions (TLR-4, TLR-9, TRAF-6 and IL-1β) were lower in IUGR piglets, whereas HNI significantly increased blood lymphocyte percentage and serum IL-10 concentrations, but decreased neutrophil percentage, serum IL-1β concentrations and ileal gene expressions (NF-kB and IL-1β). Furthermore, IUGR piglets with HNI exhibited lower serum concentrations of TNF-α and IL-1β than NBW piglets, and these alterations in the immune traits of IUGR piglets receiving HNI were accompanied by decreasing ileal gene expressions of TLR-4, TLR-9, NF-κB and IL-1β that are related to innate immunity. In conclusion, the present findings suggest that increased NI during the suckling period impaired the immune function of neonatal piglets with IUGR.
Collapse
|
41
|
Cousin FJ, Foligné B, Deutsch SM, Massart S, Parayre S, Le Loir Y, Boudry G, Jan G. Assessment of the probiotic potential of a dairy product fermented by Propionibacterium freudenreichii in piglets. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:7917-7927. [PMID: 22823107 DOI: 10.1021/jf302245m] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Dairy propionibacteria, including Propionibacterium freudenreichii , display promising probiotic properties, including immunomodulation. These properties are highly strain-dependent and rarely studied in a fermented dairy product. We screened 10 strains, grown in a newly developed fermented milk ultrafiltrate, for immunomodulatory properties in vitro. The most anti-inflammatory strain, P. freudenreichii BIA129, was further tested on piglets. P. freudenreichii -fermented product improved food intake and growth of piglets. Colonic mucosa explants of treated pigs secreted less interleukin 8 (-25%, P < 0.05) and tumor necrosis factor α (-20%, P < 0.05), either in basal conditions or after a lipopolysaccharide challenge. By contrast, the gut structure, barrier function (measured ex vivo in Ussing chambers), microbial diversity (assessed by 16S rRNA pyrosequencing), and colonic short-chain fatty acid content were unchanged, assuming maintenance of normal intestinal physiology. In conclusion, this work confirms in vivo probiotic properties of dairy propionibacteria-fermented products, which are promising for the prevention or healing of inflammatory bowel diseases.
Collapse
Affiliation(s)
- Fabien J Cousin
- UMR 1253, Science et Technologie du Lait et de l'Œuf, INRA, F-35042 Rennes, France
| | | | | | | | | | | | | | | |
Collapse
|
42
|
Accelerated growth rate induced by neonatal high-protein milk formula is not supported by increased tissue protein synthesis in low-birth-weight piglets. J Nutr Metab 2012; 2012:545341. [PMID: 22315674 PMCID: PMC3270429 DOI: 10.1155/2012/545341] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2011] [Revised: 08/26/2011] [Accepted: 10/13/2011] [Indexed: 12/19/2022] Open
Abstract
Low-birth-weight neonates are routinely fed a high-protein formula to promote catch-up growth and antibiotics are usually associated to prevent infection. Yet the effects of such practices on tissue protein metabolism are unknown. Baby pigs were fed from age 2 to 7 or 28 d with high protein formula with or without amoxicillin supplementation, in parallel with normal protein formula, to determine tissue protein metabolism modifications. Feeding high protein formula increased growth rate between 2 and 28 days of age when antibiotic was administered early in the first week of life. This could be explained by the occurrence of diarrhea when piglets were fed the high protein formula alone. Higher growth rate was associated with higher feed conversion and reduced protein synthesis rate in the small intestine, muscle and carcass, whereas proteolytic enzyme activities measured in these tissues were unchanged. In conclusion, accelerated growth rate caused by high protein formula and antibiotics was not supported by increased protein synthesis in muscle and carcass.
Collapse
|
43
|
Alterations in ileal and colonic permeability by chronic intake of high-lipid diets enriched with omega 3, omega 6 or saturated fat. Proc Nutr Soc 2011. [DOI: 10.1017/s002966511100437x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|