1
|
Nakajima A, Arzamasov AA, Sakanaka M, Murakami R, Kozakai T, Yoshida K, Katoh T, Ojima MN, Hirose J, Nagao S, Xiao JZ, Odamaki T, Rodionov DA, Katayama T. In vitro competition with Bifidobacterium strains impairs potentially pathogenic growth of Clostridium perfringens on 2'-fucosyllactose. Gut Microbes 2025; 17:2478306. [PMID: 40102238 PMCID: PMC11956901 DOI: 10.1080/19490976.2025.2478306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/06/2024] [Revised: 02/23/2025] [Accepted: 03/04/2025] [Indexed: 03/20/2025] Open
Abstract
Fortifying infant formula with human milk oligosaccharides, such as 2'-fucosyllactose (2'-FL), is a global trend. Previous studies have shown the inability of pathogenic gut microbes to utilize 2'-FL. However, the present study demonstrates that the type strain (JCM 1290T) of Clostridium perfringens, a pathobiont species often more prevalent and abundant in the feces of C-section-delivered infants, exhibits potentially pathogenic growth on 2'-FL. The expression of genes for α-toxin, an activator of NLRP3 inflammasome, and ethanolamine ammonia-lyase, a factor responsible for the progression of gas gangrene, was significantly upregulated during 2'-FL assimilation compared to growth on lactose. However, colony-forming unit of C. perfringens JCM 1290T markedly decreased when co-cultivated with selected strains of Bifidobacterium, a taxon frequently detected in the breastfed infant gut. Moreover, during co-cultivation, the expression of virulence-related genes, including the gene for perfringolysin O - another activator of NLRP3 inflammasome - were significantly downregulated, while the lactate oxidation genes were upregulated. This can occur through two different mechanisms: direct competition for 2'-FL between the two organisms, or cross-feeding of lactose, released from 2'-FL by C. perfringens JCM 1290T, to Bifidobacterium. Attenuation of α-toxin production by the selected Bifidobacterium strains was observed to varying extents in 2'-FL-utilizing C. perfringens strains clinically isolated from healthy infants. Our results warrant detailed in vivo studies using animal models with dysbiotic microbiota dominated by various types of C. perfringens strains to further validate the safety of 2'-FL for clinical interventions, particularly on vulnerable preterm infants.
Collapse
Affiliation(s)
- Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Aleksandr A. Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Ryuta Murakami
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Tomoya Kozakai
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Keisuke Yoshida
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Miriam N. Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Junko Hirose
- Department of Food and Nutrition, Kyoto Women’s University, Kyoto, Japan
| | | | - Jin-Zhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Innovative Research Institute, Morinaga Milk Industry Co, Ltd, Zama, Kanagawa, Japan
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| |
Collapse
|
2
|
Liu S, Zeng X, Li J, Li W, Gu Y, Li B, Wang J. Goat milk oligosaccharides: regulating infant immunity by intervention in the gut microbiota. Food Funct 2025; 16:2213-2229. [PMID: 40035489 DOI: 10.1039/d5fo00162e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/05/2025]
Abstract
The health status of the growing infant is closely related to the development of the gut microbiota during infancy, which is also a major stimulator of the immune system. Goat milk oligosaccharides (gMOs) are a class of bioactive compounds in goat milk, which have attracted extensive research interest in recent years. Recent studies have highlighted that gMOs as prebiotics can regulate the gut microbiota, exhibit multiple health effects, and act as immunomodulators. This article outlines the structure, classification, and functions of gMOs. In addition, we also deeply explored the mechanism of gMO interaction with infant gut microbiota and regulation of infant immunity. Finally, the possibility of gMOs as an effective substitute for natural prebiotics in breast milk is revisited. We concluded that gMOs improve infant immune function by regulating intestinal beneficial bacteria (Bifidobacteria, Lactobacilli, etc.) and their metabolism. Therefore, gMOs are significant to infant immune health and are expected to become a substitute for human milk oligosaccharides (HMOs).
Collapse
Affiliation(s)
- Sibo Liu
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Xiaoling Zeng
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Jing Li
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Wei Li
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| | - Yue Gu
- Food College, Northeast Agricultural University, Harbin 150030, China.
| | - Bailiang Li
- Food College, Northeast Agricultural University, Harbin 150030, China.
- Key Laboratory of Dairy Science, Ministry of Education, Northeast Agricultural University, Harbin 150030, China
| | - Jiaqi Wang
- Ausnutria Dairy (China) Co., Ltd, Changsha 410000, China.
| |
Collapse
|
3
|
Li C, Han T, Zhong P, Zhang Y, Zhao T, Wang S, Wang X, Tian Y, Gong G, Liu Y, Huang L, Lu Y, Wang Z. α2,6-linked sialylated oligosaccharides riched in goat milk alleviate food allergy by regulating the gut flora and mucin O-glycosylation. Carbohydr Polym 2025; 350:123049. [PMID: 39647952 DOI: 10.1016/j.carbpol.2024.123049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 11/17/2024] [Accepted: 11/19/2024] [Indexed: 12/10/2024]
Abstract
The nutritious goat milk has low allergenicity. Oligosaccharides represent one of the crucial functional constituents in goat milk, which are structurally similar to human milk oligosaccharides (HMOs). Currently, the anti-allergic activity of GMOs has not been reported. In this study, GMOs were efficiently separated into neutral (NGMOs) and sialylated (SGMOs) fractions, following by qualitative and quantitative analysis at the isomer level using online LC-MS/MS. Fifteen NGMOs and 28 SGMOs were detected in goat milk, with 10 SGMOs reported for the first time. Distinctly, α2,6-linked SGMOs were 3.9 times more abundant in goat milk than in bovine milk, with the total relative content of 6'SL, 3'SLN and 6'NGL in SGMOs approach to 60%, which is more similar to HMOs. Orally administering GMOs, especially α2,6-linked sialylated oligosaccharides, significantly alleviated food allergy in ovalbumin-induced BALB/c mice. SGMOs restored the balance of Lachnospiraceae, Erysipelotrichaceae, and Bacteroidaceae, reconstructed the intestinal mucosal barrier, especially restored the levels of fucosylation, sialylation, and sulfation of mucin O-glycans, increased the expression of four core type 2 O-glycans (F1H2N2, F2H2N2, S1F2H2N2, and A1F1H2N2) significantly. This is the first comprehensive study of the anti-allergic activity of GMOs, and the results lay the foundation for the development of GMOs-based natural anti-allergic components.
Collapse
Affiliation(s)
- Cheng Li
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tianjiao Han
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Peiyun Zhong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuyang Zhang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Tong Zhao
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Shukai Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yang Tian
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Guiping Gong
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yuxia Liu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Yu Lu
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| | - Zhongfu Wang
- Glycobiology and Glycotechnology Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
4
|
Zhang L, Ma Y, Hettinga K, Zhou P. Suckling Rat Pup Model: Do Caprine Milk Lactoferrin and Immunoglobulins Have Different Digestion and Absorption Properties from That of Human and Bovine Species? JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2025; 73:3069-3079. [PMID: 39873219 DOI: 10.1021/acs.jafc.4c10539] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2025]
Abstract
This study aimed to investigate the digestion and absorption properties of caprine milk serum proteins in comparison to human and bovine species by using rat pups to mimic preterm infants. The results indicate that caprine lactoferrin (LTF) had a shorter retention time in the intestine and released a greater number of fragments, resembling human milk LTF more closely. In contrast, caprine immunoglobulins (Igs) were similar to bovine Igs and both exhibited a longer retention time in the intestine. For absorption, caprine Igs could be absorbed intact, which was similar to human and bovine Igs, whereas caprine LTF fragments were found in jejunum but not in plasma of rat pups. This is similar to bovine LTF but differed from human LTF as human LTF could be absorbed intact in plasma of rat pups at 20 min. In addition, the absorption rate of peptides and amino acids from caprine milk serum was similar to that of human milk serum, which was higher than that from bovine milk serum. This study aimed to enhance our understanding of the differences in bioavailability of LTF and Igs derived from caprine, human milk, and bovine milk, thereby offering guidance for selecting protein sources for premature infants.
Collapse
Affiliation(s)
- Lina Zhang
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| | - Ying Ma
- National R&D Center for Se-rich Agricultural Products Processing, Hubei Engineering Research Center for Deep Processing of Green Se-rich Agricultural Products, School of Modern Industry for Selenium Science and Engineering, Wuhan Polytechnic University, Wuhan 430023, China
| | - Kasper Hettinga
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
- Food Quality & Design Group, Wageningen University, Wageningen 6708WG, The Netherlands
| | - Peng Zhou
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Laboratory for Dairy Science and Technology, Jiangnan University, Wuxi, Jiangsu 214122, China
| |
Collapse
|
5
|
Yang C, Pan J, Pang S, Hu S, Liu M, Zhang X, Song L, Ren X, Wang Z. Comparative analysis of the nutritional composition, digestibility, metabolomics profiles and growth influence of cow, goat and sheep milk powder diets in rat models. Front Nutr 2024; 11:1428938. [PMID: 39650706 PMCID: PMC11622695 DOI: 10.3389/fnut.2024.1428938] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2024] [Accepted: 11/01/2024] [Indexed: 12/11/2024] Open
Abstract
Introduction The diversity of dairy products and the increasing consumption levels have led to a growing interest in goat and sheep milk, which are rich in essential nutrients and functional components. The study aims to explore the nutritional composition, growth performance, digestibility, and serum metabolic differences of milk powders from cow, goat, and sheep using LC-MS/MS-based metabolomics in rat models. Methods Sixty male Sprague-Dawley rats were fed with whole cow, goat, and sheep milk powder samples , and their feces and urine were analyzed for fat and protein content. LC/MS analysis was conducted using a Dionex UltiMate 3000 UHPLC system coupled with a Thermo Q EXACTIVE mass spectrometer, with data processed using Wekemo Bioincloud for quality control, normalization, comparisons with the KEGG database, statistical analyses, and selection of differential metabolites. Results The sheep milk powder showed highest protein and fat content level, while cow and goat milk powders separately demonstrated higher lactose and carbohydrate levels. Each milk powder had a unique mineral profile, with sheep milk powder containing the highest calcium content. All groups exhibited consistent growth in body weight and high rates of protein and fat digestibility. Metabolomics analysis revealed distinct metabolic profiles, with goat milk powder linked to steroid hormone biosynthesis and sheep milk powder associated with hormone regulation and bile acid pathways. Conclusion This study offers valuable insights into the metabolic implications of different milk powder sources, informing dietary choices and facilitating the development of targeted public health strategies to optimize nutritional intake and promote overall well-being.
Collapse
Affiliation(s)
- Chun Yang
- School of Public Health, Capital Medical University, Beijing, China
| | - Jiancun Pan
- Feihe Research Institute,Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
| | - Shaojie Pang
- Feihe Research Institute,Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
| | - Shuang Hu
- Children’s Hospital of Nanjing Medical University, Nanjing, China
| | - Miao Liu
- School of Public Health, Capital Medical University, Beijing, China
| | - Xinyan Zhang
- Feihe Research Institute,Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
| | - Liping Song
- Feihe Research Institute,Heilongjiang Feihe Dairy Co., Ltd., Beijing, China
| | - Xiangnan Ren
- National Institute for Nutrition and Health, Chinese Center for Disease Control and Prevention, Beijing, China
| | - Zhongli Wang
- Department of Rehabilitation Medicine, The Second Affiliated Hospital of Jiaxing University, The Second Hospital of Jiaxing, Jiaxing, China
- School of Medicine and Nursing, Huzhou University, Huzhou, Zhejiang, China
| |
Collapse
|
6
|
Almasri RS, Bedir AS, Ranneh YK, El-Tarabily KA, Al Raish SM. Benefits of Camel Milk over Cow and Goat Milk for Infant and Adult Health in Fighting Chronic Diseases: A Review. Nutrients 2024; 16:3848. [PMID: 39599634 PMCID: PMC11597306 DOI: 10.3390/nu16223848] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 11/05/2024] [Accepted: 11/07/2024] [Indexed: 11/29/2024] Open
Abstract
The nutritional composition, antimicrobial properties, and health benefits of camel milk (CAM), cow milk (COM), and goat milk (GOM) have been extensively studied for their roles in managing diabetes and cardiovascular diseases (CVD). This review compares these milk types' nutritional and therapeutic properties, emphasizing their applications in chronic disease management. CAM is rich in insulin-like proteins, vitamins, minerals, and bioactive compounds that benefit glycemic control and cardiovascular health. It also exhibits potent antioxidants, anti-inflammatory, and lipid-lowering effects, which are crucial for managing diabetes and reducing CVD risk factors. While COM and GOM provide essential nutrients, their impact on metabolic health differs. GOM is known for its digestibility and antihypertensive properties, whereas COM's higher lactose content may be less suitable for diabetic patients. CAM's unique nutritional profile offers distinct therapeutic benefits, particularly for diabetes and CVD management. Further research is needed to clarify its mechanisms of action and optimize its clinical application for chronic disease prevention and management.
Collapse
Affiliation(s)
- Razan S. Almasri
- Department of Nutrition, College of Medicine and Health Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (R.S.A.); (A.S.B.)
| | - Alaa S. Bedir
- Department of Nutrition, College of Medicine and Health Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates; (R.S.A.); (A.S.B.)
| | - Yazan K. Ranneh
- Department of Pharmacy, College of Pharmacy, Al Ain University of Science and Technology, Al Ain 64141, United Arab Emirates;
| | - Khaled A. El-Tarabily
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| | - Seham M. Al Raish
- Department of Biology, College of Science, United Arab Emirates University, Al Ain 15551, United Arab Emirates;
| |
Collapse
|
7
|
Dargenio VN, Cristofori F, Brindicci VF, Schettini F, Dargenio C, Castellaneta SP, Iannone A, Francavilla R. Impact of Bifidobacterium longum Subspecies infantis on Pediatric Gut Health and Nutrition: Current Evidence and Future Directions. Nutrients 2024; 16:3510. [PMID: 39458503 PMCID: PMC11510697 DOI: 10.3390/nu16203510] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/08/2024] [Accepted: 10/09/2024] [Indexed: 10/28/2024] Open
Abstract
Background: the intestinal microbiota, a complex community vital to human health, is shaped by microbial competition and host-driven selective pressures. Among these microbes, Bifidobacterium plays a crucial role in early gut colonization during neonatal stages, where Bifidobacterium longum subspecies infantis (B. infantis) predominates and is particularly prevalent in healthy breastfed infants. Objectives: as we embark on a new era in nutrition of the pediatric population, this study seeks to examine the existing understanding regarding B. infantis, encompassing both preclinical insights and clinical evidence. Methods: through a narrative disceptation of the current literature, we focus on its genetic capacity to break down various substances that support its survival and dominance in the intestine. Results: using "omics" technologies, researchers have identified beneficial mechanisms of B. infantis, including the production of short-chain fatty acids, serine protease inhibitors, and polysaccharides. While B. infantis declines with age and in various diseases, it remains a widely used probiotic with documented benefits for infant and child health in numerous studies. Conclusions: the current scientific evidence underscores the importance for ongoing research and clinical trials for a deeper understanding of B. infantis's role in promoting long-term health.
Collapse
Affiliation(s)
- Vanessa Nadia Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Fernanda Cristofori
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Viviana Fara Brindicci
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Federico Schettini
- Neonatology and Neonatal Intensive Care, Santissima Annunziata Hospital, 74123 Taranto, Italy;
| | - Costantino Dargenio
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Stefania Paola Castellaneta
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| | - Andrea Iannone
- Gastroenterology Unit, Department of Emergency and Organ Transplantation, University of Bari “Aldo Moro”, 70126 Bari, Italy;
| | - Ruggiero Francavilla
- Interdisciplinary Department of Medicine, Pediatric Section, Children’s Hospital ‘Giovanni XXIII’, University of Bari “Aldo Moro”, 70126 Bari, Italy; (V.N.D.); (F.C.); (V.F.B.); (C.D.); (S.P.C.)
| |
Collapse
|
8
|
Mondragon Portocarrero ADC, Lopez-Santamarina A, Lopez PR, Ortega ISI, Duman H, Karav S, Miranda JM. Substitutive Effects of Milk vs. Vegetable Milk on the Human Gut Microbiota and Implications for Human Health. Nutrients 2024; 16:3108. [PMID: 39339708 PMCID: PMC11435326 DOI: 10.3390/nu16183108] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/20/2024] [Revised: 09/12/2024] [Accepted: 09/13/2024] [Indexed: 09/30/2024] Open
Abstract
Background: In the last two decades, the consumption of plant-based dairy substitutes in place of animal-based milk has increased in different geographic regions of the world. Dairy substitutes of vegetable origin have a quantitative composition of macronutrients such as animal milk, although the composition of carbohydrates, proteins and fats, as well as bioactive components, is completely different from that of animal milk. Many milk components have been shown to have relevant effects on the intestinal microbiota. Methods: Therefore, the aim of this review is to compare the effects obtained by previous works on the composition of the gut microbiota after the ingestion of animal milk and/or vegetable beverages. Results: In general, the results obtained in the included studies were very positive for animal milk intake. Thus, we found an increase in gut microbiota richness and diversity, increase in the production of short-chain fatty acids, and beneficial microbes such as Bifidobacterium, lactobacilli, Akkermansia, Lachnospiraceae or Blautia. In other cases, we found a significant decrease in potential harmful bacteria such as Proteobacteria, Erysipelotrichaceae, Desulfovibrionaceae or Clostridium perfingens after animal-origin milk intake. Vegetable beverages have also generally produced positive results in the gut microbiota such as the increase in the relative presence of lactobacilli, Bifidobacterium or Blautia. However, we also found some potential negative results, such as increases in the presence of potential pathogens such as Enterobacteriaceae, Salmonella and Fusobacterium. Conclusions: From the perspective of their effects on the intestinal microbiota, milks of animal origin appear to be more beneficial for human health than their vegetable substitutes. These different effects on the intestinal microbiota should be considered in those cases where the replacement of animal milks by vegetable substitutes is recommended.
Collapse
Affiliation(s)
- Alicia del Carmen Mondragon Portocarrero
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Campus Terra, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.d.C.M.P.); (A.L.-S.); (P.R.L.)
| | - Aroa Lopez-Santamarina
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Campus Terra, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.d.C.M.P.); (A.L.-S.); (P.R.L.)
| | - Patricia Regal Lopez
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Campus Terra, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.d.C.M.P.); (A.L.-S.); (P.R.L.)
| | - Israel Samuel Ibarra Ortega
- Área Académica de Química, Universidad Autónoma del Estado de Hidalgo, Carretera Pachuca-Tulancingo km. 4.5, Pachuca 42076, Hidalgo, Mexico;
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (H.D.); (S.K.)
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale 17000, Türkiye; (H.D.); (S.K.)
| | - Jose Manuel Miranda
- Laboratorio de Higiene Inspección y Control de Alimentos, Departamento de Química Analítica, Nutrición y Bromatología, Campus Terra, Universidade de Santiago de Compostela, 27002 Lugo, Spain; (A.d.C.M.P.); (A.L.-S.); (P.R.L.)
| |
Collapse
|
9
|
Li S, Liu Z, Zhang Q, Su D, Wang P, Li Y, Shi W, Zhang Q. The Antidiabetic Potential of Probiotics: A Review. Nutrients 2024; 16:2494. [PMID: 39125375 PMCID: PMC11313988 DOI: 10.3390/nu16152494] [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: 06/28/2024] [Revised: 07/26/2024] [Accepted: 07/26/2024] [Indexed: 08/12/2024] Open
Abstract
Diabetes has become one of the most prevalent global epidemics, significantly impacting both the economy and the health of individuals. Diabetes is associated with numerous complications, such as obesity; hyperglycemia; hypercholesterolemia; dyslipidemia; metabolic endotoxemia; intestinal barrier damage; insulin-secretion defects; increased oxidative stress; and low-grade, systemic, and chronic inflammation. Diabetes cannot be completely cured; therefore, current research has focused on developing various methods to control diabetes. A promising strategy is the use of probiotics for diabetes intervention. Probiotics are a class of live, non-toxic microorganisms that can colonize the human intestine and help improve the balance of intestinal microbiota. In this review, we summarize the current clinical studies on using probiotics to control diabetes in humans, along with mechanistic studies conducted in animal models. The primary mechanism by which probiotics regulate diabetes is improved intestinal barrier integrity, alleviated oxidative stress, enhanced immune response, increased short-chain fatty acid production, etc. Therefore, probiotic supplementation holds great potential for the prevention and management of diabetes.
Collapse
Affiliation(s)
- Shiming Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing 100193, China
| | - Zichao Liu
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qi Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Dan Su
- Department of Chemistry and Chemical Biology, Cornell University, Ithaca, NY 14850, USA;
| | - Pengjie Wang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Yixuan Li
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Wenbiao Shi
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| | - Qian Zhang
- Department of Nutrition and Health, China Agricultural University, Beijing 100193, China; (S.L.); (Z.L.); (Q.Z.); (P.W.); (Y.L.)
| |
Collapse
|
10
|
Arzamasov AA, Rodionov DA, Hibberd MC, Guruge JL, Kazanov MD, Leyn SA, Kent JE, Sejane K, Bode L, Barratt MJ, Gordon JI, Osterman AL. Integrative genomic reconstruction of carbohydrate utilization networks in bifidobacteria: global trends, local variability, and dietary adaptation. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.07.06.602360. [PMID: 39005317 PMCID: PMC11245093 DOI: 10.1101/2024.07.06.602360] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 07/16/2024]
Abstract
Bifidobacteria are among the earliest colonizers of the human gut, conferring numerous health benefits. While multiple Bifidobacterium strains are used as probiotics, accumulating evidence suggests that the individual responses to probiotic supplementation may vary, likely due to a variety of factors, including strain type(s), gut community composition, dietary habits of the consumer, and other health/lifestyle conditions. Given the saccharolytic nature of bifidobacteria, the carbohydrate composition of the diet is one of the primary factors dictating the colonization efficiency of Bifidobacterium strains. Therefore, a comprehensive understanding of bifidobacterial glycan metabolism at the strain level is necessary to rationally design probiotic or synbiotic formulations that combine bacterial strains with glycans that match their nutrient preferences. In this study, we systematically reconstructed 66 pathways involved in the utilization of mono-, di-, oligo-, and polysaccharides by analyzing the representation of 565 curated metabolic functional roles (catabolic enzymes, transporters, transcriptional regulators) in 2973 non-redundant cultured Bifidobacterium isolates and metagenome-assembled genomes (MAGs). Our analysis uncovered substantial heterogeneity in the predicted glycan utilization capabilities at the species and strain level and revealed the presence of a yet undescribed phenotypically distinct subspecies-level clade within the Bifidobacterium longum species. We also identified Bangladeshi isolates harboring unique gene clusters tentatively implicated in the breakdown of xyloglucan and human milk oligosaccharides. Predicted carbohydrate utilization phenotypes were experimentally characterized and validated. Our large-scale genomic analysis considerably expands the knowledge of carbohydrate metabolism in bifidobacteria and provides a foundation for rationally designing single- or multi-strain probiotic formulations of a given bifidobacterial species as well as synbiotic combinations of bifidobacterial strains matched with their preferred carbohydrate substrates.
Collapse
Affiliation(s)
- Aleksandr A Arzamasov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Dmitry A Rodionov
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Matthew C Hibberd
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Janaki L Guruge
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Marat D Kazanov
- Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, Turkey, 34956
| | - Semen A Leyn
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - James E Kent
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA
| | - Kristija Sejane
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, CA 92093, USA
| | - Lars Bode
- Department of Pediatrics, Larsson-Rosenquist Foundation Mother-Milk-Infant Center of Research Excellence (MOMI CORE), and the Human Milk Institute (HMI), University of California San Diego, La Jolla, CA 92093, USA
| | - Michael J Barratt
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Jeffrey I Gordon
- Edison Family Center for Genome Sciences and Systems Biology, Washington University School of Medicine, St. Louis, MO 63110, USA
- Center for Gut Microbiome and Nutrition Research, Washington University School of Medicine, St. Louis, MO 63110, USA
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Disease Center, Sanford Burnham Prebys Medical Discovery Institute, 10901 North Torrey Pines Rd, La Jolla, CA 92037, USA
| |
Collapse
|
11
|
Demmelmair H, Uhl O, Zhou SJ, Makrides M, Gibson RA, Prosser C, Gallier S, Koletzko B. Plasma Sphingomyelins and Carnitine Esters of Infants Consuming Whole Goat or Cow Milk-Based Infant Formulas or Human Milk. J Nutr 2024; 154:1781-1789. [PMID: 38615734 PMCID: PMC11217027 DOI: 10.1016/j.tjnut.2024.04.020] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2024] [Revised: 03/12/2024] [Accepted: 04/08/2024] [Indexed: 04/16/2024] Open
Abstract
BACKGROUND Infant formulas are typically manufactured using skimmed milk, whey proteins, and vegetable oils, which excludes milk fat globule membranes (MFGM). MFGM contains polar lipids, including sphingomyelin (SM). OBJECTIVE The objective of this study was comparison of infant plasma SM and acylcarnitine species between infants who are breastfed or receiving infant formulas with different fat sources. METHODS In this explorative study, we focused on SM and acylcarnitine species concentrations measured in plasma samples from the TIGGA study (ACTRN12608000047392), where infants were randomly assigned to receive either a cow milk-based infant formula (CIF) with vegetable oils only or a goat milk-based infant formula (GIF) with a goat milk fat (including MFGM) and vegetable oil mixture to the age ≥4 mo. Breastfed infants were followed as a reference group. Using tandem mass spectrometry, SM species in the study formulas and SM and acylcarnitine species in plasma samples collected at the age of 4 mo were analyzed. RESULTS Total SM concentrations (∼42 μmol/L) and patterns of SM species were similar in both formulas. The total plasma SM concentrations were not different between the formula groups but were 15 % (CIF) and 21% (GIF) lower in the formula groups than in the breastfed group. Between the formula groups, differences in SM species were statistically significant but small. Total carnitine and major (acyl) carnitine species were not different between the groups. CONCLUSIONS The higher total SM concentration in breastfed than in formula-fed infants might be related to a higher SM content in human milk, differences in cholesterol metabolism, dietary fatty acid intake, or other factors not yet identified. SM and acylcarnitine species composition in plasma is not closely related to the formula fatty acid composition. This trial was registered at Australian New Zealand Clinical Trials Registry as ACTRN12608000047392.
Collapse
Affiliation(s)
- Hans Demmelmair
- Department of Pediatrics, Division of Metabolic and Nutritional Medicine, Ludwig Maximilians University Munich, Dr. von Hauner Children's Hospital, Munich, Germany.
| | - Olaf Uhl
- Department of Pediatrics, Division of Metabolic and Nutritional Medicine, Ludwig Maximilians University Munich, Dr. von Hauner Children's Hospital, Munich, Germany
| | - Shao J Zhou
- Food and Wine, School of Agriculture, University of Adelaide, Adelaide, Australia
| | - Maria Makrides
- Woman's and Children's Health Research Institute, University of Adelaide, Adelaide, Australia
| | - Robert A Gibson
- Food and Wine, School of Agriculture, University of Adelaide, Adelaide, Australia
| | - Colin Prosser
- Science Department, Dairy Goat Co-operative (NZ) Ltd, Hamilton, New Zealand
| | - Sophie Gallier
- Science Department, Dairy Goat Co-operative (NZ) Ltd, Hamilton, New Zealand
| | - Berthold Koletzko
- Department of Pediatrics, Division of Metabolic and Nutritional Medicine, Ludwig Maximilians University Munich, Dr. von Hauner Children's Hospital, Munich, Germany
| |
Collapse
|
12
|
Wong CB, Huang H, Ning Y, Xiao J. Probiotics in the New Era of Human Milk Oligosaccharides (HMOs): HMO Utilization and Beneficial Effects of Bifidobacterium longum subsp. infantis M-63 on Infant Health. Microorganisms 2024; 12:1014. [PMID: 38792843 PMCID: PMC11124435 DOI: 10.3390/microorganisms12051014] [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: 04/24/2024] [Revised: 05/10/2024] [Accepted: 05/15/2024] [Indexed: 05/26/2024] Open
Abstract
A healthy gut microbiome is crucial for the immune system and overall development of infants. Bifidobacterium has been known to be a predominant species in the infant gut; however, an emerging concern is the apparent loss of this genus, in particular, Bifidobacterium longum subsp. infantis (B. infantis) in the gut microbiome of infants in industrialized nations, underscoring the importance of restoring this beneficial bacterium. With the growing understanding of the gut microbiome, probiotics, especially infant-type human-residential bifidobacteria (HRB) strains like B. infantis, are gaining prominence for their unique ability to utilize HMOs and positively influence infant health. This article delves into the physiology of a probiotic strain, B. infantis M-63, its symbiotic relationship with HMOs, and its potential in improving gastrointestinal and allergic conditions in infants and children. Moreover, this article critically assesses the role of HMOs and the emerging trend of supplementing infant formulas with the prebiotic HMOs, which serve as fuel for beneficial gut bacteria, thereby emulating the protective effects of breastfeeding. The review highlights the potential of combining B. infantis M-63 with HMOs as a feasible strategy to improve health outcomes in infants and children, acknowledging the complexities and requirements for further research in this area.
Collapse
Affiliation(s)
- Chyn Boon Wong
- International Division, Morinaga Milk Industry Co., Ltd., 5-2, Higashi Shimbashi 1-Chome, Minato-ku, Tokyo 105-7122, Japan
| | - Huidong Huang
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Yibing Ning
- Nutrition Research Institute, Junlebao Dairy Group Co., Ltd., 36 Shitong Road, Shijiazhuang 050221, China
| | - Jinzhong Xiao
- Morinaga Milk Industry (Shanghai) Co., Ltd., Room 509 Longemont Yes Tower, No. 369 Kaixuan Road, Changning District, Shanghai 200050, China
- Department of Microbiota Research, Graduate School of Medicine, Juntendo University, 2-1-1 Hongo, Bunkyo-ku, Tokyo 113-8421, Japan
- Research Center for Probiotics, Department of Nutrition and Health, China Agricultural University, Beijing 100093, China
| |
Collapse
|
13
|
Hilliard MA, Sela DA. Transmission and Persistence of Infant Gut-Associated Bifidobacteria. Microorganisms 2024; 12:879. [PMID: 38792709 PMCID: PMC11124121 DOI: 10.3390/microorganisms12050879] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2024] [Revised: 04/18/2024] [Accepted: 04/25/2024] [Indexed: 05/26/2024] Open
Abstract
Bifidobacterium infantis are the primary colonizers of the infant gut, yet scientific research addressing the transmission of the genus Bifidobacterium to infants remains incomplete. This review examines microbial reservoirs of infant-type Bifidobacterium that potentially contribute to infant gut colonization. Accordingly, strain inheritance from mother to infant via the fecal-oral route is likely contingent on the bifidobacterial strain and phenotype, whereas transmission via the vaginal microbiota may be restricted to Bifidobacterium breve. Additional reservoirs include breastmilk, horizontal transfer from the environment, and potentially in utero transfer. Given that diet is a strong predictor of Bifidobacterium colonization in early life and the absence of Bifidobacterium is observed regardless of breastfeeding, it is likely that additional factors are responsible for bifidobacterial colonization early in life.
Collapse
Affiliation(s)
- Margaret A. Hilliard
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
| | - David A. Sela
- Department of Food Science, University of Massachusetts, Amherst, MA 01003, USA;
- Organismic and Evolutionary Biology Graduate Program, University of Massachusetts, Amherst, MA 01003, USA
- Department of Nutrition, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology, University of Massachusetts, Amherst, MA 01003, USA
- Department of Microbiology & Physiological Systems and Center for Microbiome Research, University of Massachusetts Medical School, Worcester, MA 01605, USA
| |
Collapse
|
14
|
Tannock GW. Understanding the gut microbiota by considering human evolution: a story of fire, cereals, cooking, molecular ingenuity, and functional cooperation. Microbiol Mol Biol Rev 2024; 88:e0012722. [PMID: 38126754 PMCID: PMC10966955 DOI: 10.1128/mmbr.00127-22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2023] Open
Abstract
SUMMARYThe microbial community inhabiting the human colon, referred to as the gut microbiota, is mostly composed of bacterial species that, through extensive metabolic networking, degrade and ferment components of food and human secretions. The taxonomic composition of the microbiota has been extensively investigated in metagenomic studies that have also revealed details of molecular processes by which common components of the human diet are metabolized by specific members of the microbiota. Most studies of the gut microbiota aim to detect deviations in microbiota composition in patients relative to controls in the hope of showing that some diseases and conditions are due to or exacerbated by alterations to the gut microbiota. The aim of this review is to consider the gut microbiota in relation to the evolution of Homo sapiens which was heavily influenced by the consumption of a nutrient-dense non-arboreal diet, limited gut storage capacity, and acquisition of skills relating to mastering fire, cooking, and cultivation of cereal crops. The review delves into the past to gain an appreciation of what is important in the present. A holistic view of "healthy" microbiota function is proposed based on the evolutionary pathway shared by humans and gut microbes.
Collapse
Affiliation(s)
- Gerald W. Tannock
- Department of Microbiology and Immunology, University of Otago, Dunedin, New Zealand
| |
Collapse
|
15
|
Adamczak AM, Werblińska A, Jamka M, Walkowiak J. Maternal-Foetal/Infant Interactions-Gut Microbiota and Immune Health. Biomedicines 2024; 12:490. [PMID: 38540103 PMCID: PMC10967760 DOI: 10.3390/biomedicines12030490] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2024] [Revised: 02/14/2024] [Accepted: 02/19/2024] [Indexed: 01/03/2025] Open
Abstract
In recent years, the number of scientific publications on the role of intestinal microbiota in shaping human health, as well as the occurrence of intestinal dysbiosis in various disease entities, has increased dynamically. However, there is a gap in comprehensively understanding the factors influencing a child's gut microbiota. This review discusses the establishment of gut microbiota and the immunological mechanisms regulating children's microbiota, emphasising the importance of prioritising the development of appropriate gut microbiota in a child from the planning stages of pregnancy. The databases PubMed, Web of Sciences, Cochrane, Scopus and Google Scholar were searched to identify relevant articles. A child's gut microbiota composition is influenced by numerous factors, such as diet during pregnancy, antibiotic therapy, the mother's vaginal microbiota, delivery method, and, later, feeding method and environmental factors. During pregnancy, the foetus naturally acquires bacterial strains from the mother through the placenta, thereby shaping the newborn's immune system. Inappropriate maternal vaginal microbiota may increase the risk of preterm birth. Formula-fed infants typically exhibit a more diverse microbiota than their breastfed counterparts. These factors, among others, shape the maturation of the child's immune system, impacting the production of IgA antibodies that are central to cellular humoral immune defence. Further research should focus on identifying specific microbiota-immune system interactions influencing a child's immune health and developing personalised treatment strategies for immune-related disorders.
Collapse
Affiliation(s)
- Ada Maria Adamczak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| | - Alicja Werblińska
- Greater Poland Centre for Pulmonology and Thoracic Surgery Named after Eugenia and Janusz Zeyland, 62 Szamarzewskiego Street, 60-569 Poznań, Poland;
| | - Małgorzata Jamka
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, 27/33 Szpitalna Street, 60-572 Poznań, Poland; (A.M.A.); (M.J.)
| |
Collapse
|
16
|
Parkin K, Palmer DJ, Verhasselt V, Amenyogbe N, Cooper MN, Christophersen CT, Prescott SL, Silva D, Martino D. Metagenomic Characterisation of the Gut Microbiome and Effect of Complementary Feeding on Bifidobacterium spp. in Australian Infants. Microorganisms 2024; 12:228. [PMID: 38276213 PMCID: PMC10819277 DOI: 10.3390/microorganisms12010228] [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: 11/17/2023] [Revised: 01/12/2024] [Accepted: 01/15/2024] [Indexed: 01/27/2024] Open
Abstract
Complementary feeding induces dramatic ecological shifts in the infant gut microbiota toward more diverse compositions and functional metabolic capacities, with potential implications for immune and metabolic health. The aim of this study was to examine whether the age at which solid foods are introduced differentially affects the microbiota in predominantly breastfed infants compared with predominantly formula-fed infants. We performed whole-genome shotgun metagenomic sequencing of infant stool samples from a cohort of six-month-old Australian infants enrolled in a nested study within the ORIGINS Project longitudinal birth cohort. Infants born preterm or those who had been administered antibiotics since birth were excluded. The taxonomic composition was highly variable among individuals at this age. Predominantly formula-fed infants exhibited a higher microbiome diversity than predominantly breastfed infants. Among the predominantly breastfed infants, the introduction of solid foods prior to five months of age was associated with higher alpha diversity than solid food introduction after six months of age, primarily due to the loss of Bifidobacterium infantis. In contrast, the age at which solid food was introduced was not associated with the overall change in diversity among predominantly formula-fed infants but was associated with compositional changes in Escherichia abundance. Examining the functional capacity of the microbiota in relation to these changes, we found that the introduction of solid foods after six months of age was associated with elevated one-carbon compound metabolic pathways in both breastfed and formula-fed infants, although the specific metabolic sub-pathways differed, likely reflecting different taxonomic compositions. Our findings suggest that the age of commencement of solid foods influences the gut microbiota composition differently in predominantly breastfed infants than in predominantly formula-fed infants.
Collapse
Affiliation(s)
- Kimberley Parkin
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
- Medical School, University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Debra J. Palmer
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
- Medical School, University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Valerie Verhasselt
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, Medical School, University of Western Australia, Nedlands, Perth, WA 6009, Australia
| | - Nelly Amenyogbe
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
| | - Matthew N. Cooper
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
| | - Claus T. Christophersen
- School of Molecular Life Sciences, Curtin University, Bentley, Perth, WA 6102, Australia
- School of Medical and Health Sciences, Edith Cowen University, Joondalup, Perth, WA 6027, Australia
| | - Susan L. Prescott
- Medical School, University of Western Australia, Nedlands, Perth, WA 6009, Australia
- School of Molecular Life Sciences, Curtin University, Bentley, Perth, WA 6102, Australia
- Joondalup Health Campus, Joondalup, Perth, WA 6027, Australia
- Nova Institute for Health, Baltimore, MD 21231, USA
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- The ORIGINS Project, Telethon Kids Institute, Nedlands, Perth, WA 6009, Australia
| | - Desiree Silva
- Medical School, University of Western Australia, Nedlands, Perth, WA 6009, Australia
- School of Molecular Life Sciences, Curtin University, Bentley, Perth, WA 6102, Australia
- School of Medical and Health Sciences, Edith Cowen University, Joondalup, Perth, WA 6027, Australia
- Department of Family and Community Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
- The ORIGINS Project, Telethon Kids Institute, Nedlands, Perth, WA 6009, Australia
| | - David Martino
- Telethon Kids Institute, University of Western Australia, Nedlands, Perth, WA 6009, Australia; (K.P.)
- School of Molecular Science, University of Western Australia, Nedlands, Perth, WA 6009, Australia
| |
Collapse
|
17
|
Nogacka AM, Cuesta I, Gueimonde M, de los Reyes-Gavilán CG. 2-Fucosyllactose Metabolism by Bifidobacteria Promotes Lactobacilli Growth in Co-Culture. Microorganisms 2023; 11:2659. [PMID: 38004671 PMCID: PMC10673426 DOI: 10.3390/microorganisms11112659] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2023] [Revised: 10/25/2023] [Accepted: 10/27/2023] [Indexed: 11/26/2023] Open
Abstract
Breastfeeding is recognized as the gold standard in infant nutrition, not only because of breastmilk's intrinsic nutritional benefits but also due to the high content of different bioactive components such as 2-fucosyllactose (2'FL) in the mother's milk. It promotes the growth of its two major consumers, Bifidobacterium longum ssp. infantis and Bifidobacterium bifidum, but the effect on other intestinal microorganisms of infant microbiota remains incompletely understood. pH-uncontrolled fecal cultures from infants donors identified as "fast 2'FL -degrader" microbiota phenotype were used for the isolation of 2'FL-associated microorganisms. The use of specific selective agents allowed the successful isolation of B. bifidum IPLA20048 and of Lactobacillus gasseri IPLA20136. The characterization of 2'FL consumption and its moieties has revealed more pronounced growth, pH drop, and lactic acid production after 2'FL consumption when both microorganisms were grown together. The results point to an association between B. bifidum IPLA20048 and L. gasseri IPLA20136 in which L. gasseri is able to use the galactose from the lactose moiety after the hydrolysis of 2'FL by B. bifidum. The additional screening of two groups of bifidobacteria (n = 38), fast and slow degraders of 2'FL, in co-culture with lactobacilli confirmed a potential cross-feeding mechanism based on degradation products released from bifidobacterial 2'FL break-down. Our work suggests that this phenomenon may be widespread among lactobacilli and bifidobacteria in the infant gut. More investigation is needed to decipher how the ability to degrade 2'FL and other human milk oligosaccharides could influence the microbiota establishment in neonates and the evolution of the microbiota in adult life.
Collapse
Affiliation(s)
- Alicja M. Nogacka
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Isabel Cuesta
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
| | - Miguel Gueimonde
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| | - Clara G. de los Reyes-Gavilán
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), 33300 Villaviciosa, Asturias, Spain; (I.C.); (M.G.); (C.G.d.l.R.-G.)
- Institute of Health Research of the Principality of Asturias (ISPA), 33011 Oviedo, Asturias, Spain
| |
Collapse
|
18
|
Mazela J, Bartnicka A, Gallier S. Comment on Jankiewicz et al. The Effect of Goat-Milk-Based Infant Formulas on Growth and Safety Parameters: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 2110. Nutrients 2023; 15:4558. [PMID: 37960211 PMCID: PMC10649585 DOI: 10.3390/nu15214558] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2023] [Revised: 10/22/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
We have read the article entitled "The effect of goat-milk-based infant formulas on growth and safety parameters: a systematic review and meta-analysis" by Jankiewicz et al [...].
Collapse
Affiliation(s)
- Jan Mazela
- Department of Neonatology, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland (A.B.)
| | - Anna Bartnicka
- Department of Neonatology, Poznan University of Medical Sciences, 33 Polna Street, 60-535 Poznan, Poland (A.B.)
| | - Sophie Gallier
- Dairy Goat Co-operative (N.Z.) Ltd., Hamilton 3240, New Zealand
| |
Collapse
|
19
|
Jankiewicz M, van der Zee L, Szajewska H. Reply to Mazela et al. Comment on "Jankiewicz et al. The Effect of Goat-Milk-Based Infant Formulas on Growth and Safety Parameters: A Systematic Review and Meta-Analysis. Nutrients 2023, 15, 2110". Nutrients 2023; 15:4559. [PMID: 37960214 PMCID: PMC10650647 DOI: 10.3390/nu15214559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Accepted: 10/24/2023] [Indexed: 11/15/2023] Open
Abstract
Thank you for carefully reading and commenting [...].
Collapse
Affiliation(s)
- Mateusz Jankiewicz
- Department of Paediatrics, The Medical University of Warsaw, 02-091 Warsaw, Poland
| | | | - Hania Szajewska
- Department of Paediatrics, The Medical University of Warsaw, 02-091 Warsaw, Poland
| |
Collapse
|
20
|
van der Toorn M, Chatziioannou AC, Pellis L, Haandrikman A, van der Zee L, Dijkhuizen L. Biological Relevance of Goat Milk Oligosaccharides to Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2023; 71:13935-13949. [PMID: 37691562 PMCID: PMC10540210 DOI: 10.1021/acs.jafc.3c02194] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/05/2023] [Revised: 08/21/2023] [Accepted: 08/22/2023] [Indexed: 09/12/2023]
Abstract
Milk is often regarded as the gold standard for the nourishment of all mammalian offspring. The World Health Organization (WHO) recommends exclusive breastfeeding for the first 6 months of the life of the infant, followed by a slow introduction of complementary foods to the breastfeeding routine for a period of approximately 2 years, whenever this is possible ( Global Strategy for Infant and Young Child Feeding; WHO, 2003). One of the most abundant components in all mammals' milk, which is associated with important health benefits, is the oligosaccharides. The milk oligosaccharides (MOS) of humans and other mammals differ in terms of their concentration and diversity. Among those, goat milk contains more oligosaccharides (gMOS) than other domesticated dairy animals, as well as a greater range of structures. This review summarizes the biological functions of MOS found in both human and goat milk to identify the possible biological relevance of gMOS in human health and development. Based on the existing literature, seven biological functions of gMOS were identified, namely, MOS action as prebiotics, immune modulators, and pathogen traps; their modulation of intestinal cells; protective effect against necrotizing enterocolitis; improved brain development; and positive effects on stressor exposure. Overall, goat milk is a viable alternate supply of functional MOS that could be employed in a newborn formula.
Collapse
Affiliation(s)
| | - Anastasia Chrysovalantou Chatziioannou
- CarbExplore
Research BV, Groningen, 9747 AN The Netherlands
- Department
of Chemistry, Laboratory of Analytical Biochemistry, University of Crete, Heraklion, 70013, Greece
| | | | | | | | - Lubbert Dijkhuizen
- CarbExplore
Research BV, Groningen, 9747 AN The Netherlands
- Microbial
Physiology, Groningen Biomolecular Sciences and Biotechnology Institute
(GBB), University of Groningen, Groningen, 9747 AG, The Netherlands
| |
Collapse
|
21
|
Jung C, González Serrano A, Batard C, Seror E, Gelwane G, Poidvin A, Lavallée I, Elbez A, Brussieux M, Prosser C, Gallier S, Bellaïche M. Whole Goat Milk-Based Formula versus Whey-Based Cow Milk Formula: What Formula Do Infants Enjoy More?-A Feasibility, Double-Blind, Randomized Controlled Trial. Nutrients 2023; 15:4057. [PMID: 37764840 PMCID: PMC10537215 DOI: 10.3390/nu15184057] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 09/15/2023] [Accepted: 09/17/2023] [Indexed: 09/29/2023] Open
Abstract
(1) Background: While goat milk formula (GMF) is an alternative to cow milk formula (CMF), infants' preferences for one over the other have not been formally assessed. Specifically, our aim in this study was to determine whether infants experience fewer feeding behavior problems with whole milk-based GMF than with conventional whey-based CMF. (2) Methods: This was a multicenter, double-blind, randomized controlled trial with two-arm parallel assignment conducted in six pediatricians' offices in or near Paris, France, between June 2018 and 31 December 2021. Overall, 64 healthy infants (≤4 months old), predominantly formula-fed, were randomly assigned to either the whole milk-based GMF (n = 33) or whey-based CMF (n = 31) arm. Parents completed the Baby Eating Behavior Questionnaire (BEBQ) and the modified QUALIN questionnaire to evaluate infant feeding behavior and quality of life (psychomotor and socioemotional development), respectively, at inclusion (1 to 5 days before milk delivery) and the final visit (day 28 ± 3 after milk delivery). Informed consent was obtained for all recruited patients, and an ethical committee approved the study. (3) Results: Changes in BEBQ Enjoyment of Food and Slowness in Eating subscale scores from inclusion to final visit did not differ between arms. However, there were significant improvements in subscale scores for Food Responsiveness (GMF: 0.15 ± 1; CMF: -0.48 ± 0.81; p = 0.010) and General Appetite (GMF: 0.26 ± 1.2; CMF: -0.48 ± 0.88; p = 0.012), and modified QUALIN (GMF: 4.6 ± 9.4; CMF: -0.40 ± 7.6; p = 0.03) scores in favor of the GMF group. (4) Conclusions: In this double-blind, randomized controlled trial, GMF-fed infants exhibited a greater general appetite than CMF-fed infants, possibly due to differences in the composition of these formulas (i.e., protein and lipid profiles). In addition, GMF-fed infants enjoyed a better quality of life. There was no difference in food enjoyment between groups. These findings suggest that whole-milk-based GMF could be an attractive alternative to whey-based CMF. Clinical trial registration: NCT03488758 (clinicaltrials.gov).
Collapse
Affiliation(s)
- Camille Jung
- Clinical Research Center, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France
- Inserm, IMRB, Université Paris-Est-Créteil, 94000 Créteil, France;
| | | | | | - Elisa Seror
- Private Pediatric Practice, 75000 Paris, France
| | - Georges Gelwane
- Private Pediatric Practice, 92012 Boulogne-Billancourt, France
| | - Amélie Poidvin
- Private Pediatric Practice, 92035 La Garenne-Colombes, France
| | | | - Annie Elbez
- Private Pediatric Practice, 94700 Maisons-Alfort, France
| | - Maxime Brussieux
- Clinical Research Center, Centre Hospitalier Intercommunal de Créteil, 94000 Créteil, France
| | - Colin Prosser
- Dairy Goat Co-Operative (N.Z.) Ltd., Hamilton 3204, New Zealand (S.G.)
| | - Sophie Gallier
- Dairy Goat Co-Operative (N.Z.) Ltd., Hamilton 3204, New Zealand (S.G.)
| | - Marc Bellaïche
- Department of Pediatric Gastroenterology, Robert Debré Hospital, Assistance Publique-Hôpitaux de Paris, 75019 Paris, France;
| |
Collapse
|
22
|
Jacobs JP, Lee ML, Rechtman DJ, Sun AK, Autran C, Niklas V. Human milk oligosaccharides modulate the intestinal microbiome of healthy adults. Sci Rep 2023; 13:14308. [PMID: 37652940 PMCID: PMC10471580 DOI: 10.1038/s41598-023-41040-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2023] [Accepted: 08/18/2023] [Indexed: 09/02/2023] Open
Abstract
Human milk contains over 200 distinct oligosaccharides, which are critical to shaping the developing neonatal gut microbiome. To investigate whether a complex mixture of human milk oligosaccharides (HMOs) would similarly modulate the adult gut microbiome, HMO-Concentrate derived from pooled donor breast milk was administered orally to 32 healthy adults for 7 days followed by 21 days of monitoring. Fecal samples were collected for 16S rRNA gene sequencing, shotgun metagenomics, and metabolomics analyses. HMO-Concentrate induced dose-dependent Bifidobacterium expansion, reduced microbial diversity, and altered microbial gene content. Following HMO cessation, a microbial succession occurred with diverse taxonomic changes-including Bacteroides expansion-that persisted through day 28. This was associated with altered microbial gene content, shifts in serum metabolite levels, and increased circulating TGFβ and IL-10. Incubation of cultured adult microbiota with HMO-Concentrate induced dose-dependent compositional shifts that were not recapitulated by individual HMOs or defined mixtures of the 10 most abundant HMOs in HMO-Concentrate at their measured concentrations. These findings support that pooled donor HMOs can exert direct effects on adult gut microbiota and that complex mixtures including low abundance HMOs present in donor milk may be required for maximum effect.Registration: ClinicalTrials.gov NCT05516225.
Collapse
Affiliation(s)
- Jonathan P Jacobs
- The Vatche and Tamar Manoukian Division of Digestive Diseases, Department of Medicine, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA.
- Goodman-Luskin Microbiome Center, University of California Los Angeles, Los Angeles, CA, USA.
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA.
| | - Martin L Lee
- Fielding School of Public Health, University of California Los Angeles, Los Angeles, CA, USA
- Prolacta Bioscience, Duarte, CA, USA
| | | | | | | | - Victoria Niklas
- Department of Pediatrics, David Geffen School of Medicine at UCLA, Los Angeles, CA, USA
- Oak Hill Bio Ltd, Altrincham, Cheshire WA14 2DT, United Kingdom
| |
Collapse
|
23
|
Theophilus RJ, Taft DH. Antimicrobial Resistance Genes (ARGs), the Gut Microbiome, and Infant Nutrition. Nutrients 2023; 15:3177. [PMID: 37513595 PMCID: PMC10383493 DOI: 10.3390/nu15143177] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/14/2023] [Accepted: 07/14/2023] [Indexed: 07/30/2023] Open
Abstract
The spread of antimicrobial resistance genes (ARGs) is a major public health crisis, with the ongoing spread of ARGs leading to reduced efficacy of antibiotic treatments. The gut microbiome is a key reservoir for ARGs, and because diet shapes the gut microbiome, diet also has the potential to shape the resistome. This diet-gut microbiome-resistome relationship may also be important in infants and young children. This narrative review examines what is known about the interaction between the infant gut microbiome, the infant resistome, and infant nutrition, including exploring the potential of diet to mitigate infant ARG carriage. While more research is needed, diet has the potential to reduce infant and toddler carriage of ARGs, an important goal as part of maintaining the efficacy of available antibiotics and preserving infant and toddler health.
Collapse
Affiliation(s)
- Rufus J Theophilus
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| | - Diana Hazard Taft
- Food Science and Human Nutrition Department, University of Florida, Gainesville, FL 32611, USA
| |
Collapse
|
24
|
Eor JY, Lee CS, Moon SH, Cheon JY, Pathiraja D, Park B, Shin MJ, Kim JY, Kim S, Noh Y, Kim Y, Choi IG, Kim SH. Effect of Probiotic-Fortified Infant Formula on Infant Gut Health and Microbiota Modulation. Food Sci Anim Resour 2023; 43:659-673. [PMID: 37484007 PMCID: PMC10359846 DOI: 10.5851/kosfa.2023.e26] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 06/09/2023] [Accepted: 06/09/2023] [Indexed: 07/25/2023] Open
Abstract
Compared to infant formula, breast milk is the best source of nutrition for infants; it not only improves the neonatal intestinal function, but also regulates the immune system and gut microbiota composition. However, probiotic-fortified infant formula may further enhance the infant gut environment by overcoming the limitations of traditional infant formula. We investigated the probiotic formula administration for one month by comparing 118 Korean infants into the following three groups: infants in each group fed with breast milk (50), probiotic formula (35), or placebo formula-fed group (33). Probiotic formula improved stool consistency and defecation frequency compared to placebo formula-fed group. The probiotic formula helped maintaining the level of secretory immunoglobulin A (sIgA), which had remarkably decreased over time in placebo formula-fed infants (compared to weeks 0 and 4). Moreover, probiotic formula decreased the acidity of stool and considerably increased the butyrate concentration. Furthermore, the fecal microbiota of each group was evaluated at weeks 0 and 4. The microbial composition was distinct between each groups, and the abundance of health-promoting bacteria increased in the probiotic formula compared to the placebo formula-fed group. In summary, supplementation of probiotic infant formula can help optimize the infant gut environment, microbial composition, and metabolic activity of the microbiota, mimicking those of breast milk.
Collapse
Affiliation(s)
- Ju Young Eor
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Chul Sang Lee
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | - Sung Ho Moon
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Ju Young Cheon
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Duleepa Pathiraja
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Byeonghyeok Park
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Min Jae Shin
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
| | - Jae-Young Kim
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | | | | | | | - In-Geol Choi
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| | - Sae Hun Kim
- College of Life Sciences and
Biotechnology, Korea University, Seoul 02841, Korea
- Institute of Life Sciences and Natural
Resources, Korea University, Seoul 02841, Korea
| |
Collapse
|
25
|
Elucidating gut microbiota and metabolite patterns shaped by goat milk-based infant formula feeding in mice colonized by healthy infant feces. Food Chem 2023; 410:135413. [PMID: 36623461 DOI: 10.1016/j.foodchem.2023.135413] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 12/15/2022] [Accepted: 01/04/2023] [Indexed: 01/07/2023]
Abstract
The gut microbiota plays an evolutionarily conserved role in host metabolism, which is influenced by diet. Here, we investigated differences in shaping the gut microbiota and regulating metabolism in cow milk-based infant formula, goat milk-based infant formula, and mix milk-based infant formula compared with pasteurized human milk. 16S rRNA results showed that goat milk-based infant formula selectively increased the relative abundance of Blautia, Roseburia, Alistites and Muribaculum in the gut compared to other infant formulas. Metabolomics identification indicated that goat milk-based infant formula mainly emphasized bile acid biosynthesis, arachidonic acid metabolism and steroid biosynthesis metabolic pathways. Metabolites associated with these metabolic pathways were positively associated with increased microorganisms in goat milk-based infant formula, particularly Alistipes. Furthermore, we found a deficiency of Akkermansia abundance in three infant formula-fed compared to pasteurizedhuman milk-fed. This study presents new insights into the improvement and application of goat milk-based infant formulas in terms of intestinal microecology.
Collapse
|
26
|
Urashima T, Horiuchi R, Sakanaka M, Katayama T, Fukuda K. Lactose or milk oligosaccharide: which is significant among mammals? Anim Front 2023; 13:14-23. [PMID: 37324204 PMCID: PMC10266760 DOI: 10.1093/af/vfad017] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/17/2023] Open
Affiliation(s)
- Tadasu Urashima
- Department of Food and Life Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Risa Horiuchi
- Department of Food and Life Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido 080-8555, Japan
| | - Mikiyasu Sakanaka
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto 606-8502, Japan
| | | |
Collapse
|
27
|
Al-Ishaq RK, Samuel SM, Büsselberg D. The Influence of Gut Microbial Species on Diabetes Mellitus. Int J Mol Sci 2023; 24:ijms24098118. [PMID: 37175825 PMCID: PMC10179351 DOI: 10.3390/ijms24098118] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2023] [Revised: 04/16/2023] [Accepted: 04/20/2023] [Indexed: 05/15/2023] Open
Abstract
Diabetes mellitus (DM) is a metabolic disorder with an alarming incidence rate and a considerable burden on the patient's life and health care providers. An increase in blood glucose level and insulin resistance characterizes it. Internal and external factors such as urbanization, obesity, and genetic mutations could increase the risk of DM. Microbes in the gut influence overall health through immunity and nutrition. Recently, more studies have been conducted to evaluate and estimate the role of the gut microbiome in diabetes development, progression, and management. This review summarizes the current knowledge addressing three main bacterial species: Bifidobacterium adolescentis, Bifidobacterium bifidum, and Lactobacillus rhamnosus and their influence on diabetes and its underlying molecular mechanisms. Most studies illustrate that using those bacterial species positively reduces blood glucose levels and activates inflammatory markers. Additionally, we reported the relationship between those bacterial species and metformin, one of the commonly used antidiabetic drugs. Overall, more research is needed to understand the influence of the gut microbiome on the development of diabetes. Furthermore, more efforts are required to standardize the model used, concentration ranges, and interpretation tools to advance the field further.
Collapse
Affiliation(s)
- Raghad Khalid Al-Ishaq
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Samson Mathews Samuel
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| | - Dietrich Büsselberg
- Department of Physiology and Biophysics, Weill Cornell Medicine-Qatar, Education City, Qatar Foundation, Doha 24144, Qatar
| |
Collapse
|
28
|
Liu Y, Hu X, Voglmeir J, Liu L. N-glycan profiles as a tool in qualitative and quantitative analysis of goat milk adulteration. Food Chem 2023; 423:136116. [PMID: 37182487 DOI: 10.1016/j.foodchem.2023.136116] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 03/24/2023] [Accepted: 04/04/2023] [Indexed: 05/16/2023]
Abstract
Goat milk is closer to human milk in some respects than cow milk, and therefore preferred by many consumers. Because of the short lactation period and consequently less milk production of goats, the price of goat milk is often higher than that of cow milk, so that adulteration of goat milk is common. N-glycans have stability and thus have a good potential for acting as a new biomarker for identifying dairy adulteration. In this study, the N-glycan structures of goat milk and cow milk were analyzed by Ultra-high performance liquid chromatography (UPLC) and MALDI-TOF-MS. Based on the high species specificity of N-glycans, a method for identifying goat milk mixed with cow milk was established. The adulteration content of 5% cow milk in goat milk could be qualitatively and quantitatively detected. A prediction model of adulteration in goat milk was established by using partial least squares (PLS).
Collapse
Affiliation(s)
- Yi Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Xiaojie Hu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China
| | - Josef Voglmeir
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Jiangsu Colleborative Innovation Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| | - Li Liu
- Glycomics and Glycan Bioengineering Research Center (GGBRC), College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China; Jiangsu Colleborative Innovation Center of Meat Production, Processing and Quality Control, College of Food Science and Technology, Nanjing Agricultural University, Nanjing, Jiangsu 210095, China.
| |
Collapse
|
29
|
Plaza-Diaz J, Ruiz-Ojeda FJ, Morales J, Martín-Masot R, Climent E, Silva Á, Martinez-Blanch JF, Enrique M, Tortajada M, Ramon D, Alvarez B, Chenoll E, Gil Á. Innova 2020: A Follow-Up Study of the Fecal Microbiota of Infants Using a Novel Infant Formula between 6 Months and 12 Months of Age. Int J Mol Sci 2023; 24:7392. [PMID: 37108555 PMCID: PMC10139017 DOI: 10.3390/ijms24087392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2023] [Revised: 04/10/2023] [Accepted: 04/16/2023] [Indexed: 04/29/2023] Open
Abstract
The World Health Organization recommends exclusive breastfeeding on demand until at least the sixth month of life. Breast milk or infant formula is the infant's primary food source until the age of one year, followed by the gradual introduction of other foods. During weaning, the intestinal microbiota evolves to a profile close to that of the adult, and its disruption can result in an increased incidence of acute infectious diseases. We aimed to determine whether a novel starting formula (INN) provides gut microbiota compositions more similar to those of breastfed (BF) infants from 6 to 12 months of age compared to a standard formula (STD). This study included 210 infants (70 per group) who completed the intervention until they reached the age of 12 months. In the intervention period, infants were divided into three groups. Group 1 received an INN formula with a lower protein content, a casein to whey protein ratio of approximately 70/30, twice as much docosahexaenoic acid as the STD formula, a thermally inactivated postbiotic (Bifidobacterium animalis subsp. lactis, BPL1TM HT), and twice as much arachidonic acid as the STD formula contained. The second group received the STD formula, while the third group was exclusively BF for exploratory purposes. In the course of the study, visits were conducted at 6 months and 12 months of age. Compared to the BF and STD groups, the Bacillota phylum levels in the INN group were significantly reduced after 6 months. At the end of 6 months, the alpha diversity indices of the BF and INN groups differed significantly from those of the STD group. At 12 months, the Verrucomicrobiota phylum levels in the STD group were significantly lower than those in the BF and INN groups. Based on the comparison between 6 and 12 months, the Bacteroidota phylum levels in the BF group were significantly higher than those in the INN and STD groups. When comparing the INN group with the BF and STD groups, Clostridium sensu stricto 1 was significantly higher in the INN group. The STD group had higher levels of calprotectin than the INN and BF groups at 6 months. The immunoglobulin A levels in the STD group were significantly lower than those in the INN and BF groups after 6 months. Both formulas had significantly higher levels of propionic acid than the BF group at 6 months. At 6 months, the STD group showed a higher quantification of all metabolic pathways than the BF group. The INN formula group exhibited similar behavior to the BF group, except for the superpathway of phospholipid biosynthesis (E. coli). We hypothesize that the novel INN formula may promote an intestinal microbiota that is more similar to the microbiota of an infant who consumes only human milk before the weaning period.
Collapse
Affiliation(s)
- Julio Plaza-Diaz
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- Children’s Hospital of Eastern Ontario Research Institute, Ottawa, ON K1H 8L1, Canada
| | - Francisco Javier Ruiz-Ojeda
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Neuherberg, 85764 Munich, Germany
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
| | - Javier Morales
- Product Development Department, Alter Farmacia SA, 28880 Madrid, Spain
| | - Rafael Martín-Masot
- Institute of Nutrition and Food Technology “José Mataix”, Centre of Biomedical Research, University of Granada, Avda. del Conocimiento s/n. Armilla, 18016 Granada, Spain
- Pediatric Gastroenterology and Nutrition Unit, Hospital Regional Universitario de Málaga, 29010 Málaga, Spain
| | - Eric Climent
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Ángela Silva
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | | | - María Enrique
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Marta Tortajada
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Daniel Ramon
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Beatriz Alvarez
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Empar Chenoll
- ADM-BIOPOLIS, Scientific Park Universitat de València, 46980 València, Spain
| | - Ángel Gil
- Department of Biochemistry and Molecular Biology II, School of Pharmacy, University of Granada, 18071 Granada, Spain
- Instituto de Investigación Biosanitaria IBS.GRANADA, Complejo Hospitalario Universitario de Granada, 18014 Granada, Spain
- RG Adipocytes and Metabolism, Institute for Diabetes and Obesity, Helmholtz Diabetes Center at Helmholtz Center Munich, Neuherberg, 85764 Munich, Germany
- CIBEROBN (CIBER Physiopathology of Obesity and Nutrition), Instituto de Salud Carlos III, 28029 Madrid, Spain
| |
Collapse
|
30
|
Ferry JM, Galera-Martínez R, Campoy C, Sáenz de Pipaón M, Jarocka-Cyrta E, Walkowiak J, Romańczuk B, Escribano J, Gispert M, Grattarola P, Gruszfeld D, Iglesia I, Grote V, Demmelmair H, Handel U, Gallier S, Koletzko B. Effects of infant feeding with goat milk formula or cow milk formula on atopic dermatitis: protocol of the randomised controlled Goat Infant Formula Feeding and Eczema (GIraFFE) trial. BMJ Open 2023; 13:e070533. [PMID: 37055203 PMCID: PMC10106058 DOI: 10.1136/bmjopen-2022-070533] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 04/15/2023] Open
Abstract
INTRODUCTION Atopic dermatitis (AD) is a chronic, inflammatory skin condition significantly affecting quality of life. A small randomised trial showed an approximately one-third lower incidence of AD in goat milk formula-fed compared with cow milk formula-fed infants. However, due to limited statistical power, AD incidence difference was not found to be significant. This study aims to explore a potential risk reduction of AD by feeding a formula based on whole goat milk (as a source of protein and fat) compared with a formula based on cow milk proteins and vegetable oils. METHODS AND ANALYSIS This two-arm (1:1 allocation), parallel, randomised, double-blind, controlled nutritional trial shall enrol up to 2296 healthy term-born infants until 3 months of age, if parents choose to start formula feeding. Ten study centres in Spain and Poland are participating. Randomised infants receive investigational infant and follow-on formulas either based on whole goat milk or on cow milk until the age of 12 months. The goat milk formula has a whey:casein ratio of 20:80 and about 50% of the lipids are milk fat from whole goat milk, whereas the cow milk formula, used as control, has a whey:casein ratio of 60:40 and 100% of the lipids are from vegetable oils. The energy and nutrient levels in both goat and cow milk formulas are the same. The primary endpoint is the cumulative incidence of AD until the age of 12 months diagnosed by study personnel based on the UK Working Party Diagnostic Criteria. The secondary endpoints include reported AD diagnosis, measures of AD, blood and stool markers, child growth, sleep, nutrition and quality of life. Participating children are followed until the age of 5 years. ETHICS AND DISSEMINATION Ethical approval was obtained from the ethical committees of all participating institutions. TRIAL REGISTRATION NUMBER NCT04599946.
Collapse
Affiliation(s)
- Jill Marie Ferry
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Rafael Galera-Martínez
- Unit of Pediatric Gastroenterology and Nutrition, Torrecárdenas University Hospital, Almería, Spain
| | - Cristina Campoy
- Department of Paediatrics, EURISTIKOS Excellence Centre for Paediatric Research, School of Medicine, University of Granada, Granada, Spain
- Instituto Biosanitario de Granada (Ibs-Granada), Fundación Parque Tecnológico de Ciencias de la Salud de Granada, Granada, Spain
| | | | - Elzbieta Jarocka-Cyrta
- Department of Pediatrics, Gastroenterology, and Nutrition, Regional Specialized Children's Hospital in Olsztyn, Medical Faculty Collegium Medicum University of Warmia and Mazury, Olsztyn, Poland
| | - Jarosław Walkowiak
- Department of Pediatric Gastroenterology and Metabolic Diseases, Poznan University of Medical Sciences, Poznan, Poland
| | - Bartosz Romańczuk
- Department of Pediatrics, Medical College, University of Rzeszów, Rzeszów, Poland
| | - Joaquin Escribano
- IISPV, Pediatric Nutrition and Human Development Research Unit, Universitat Rovira i Virgili, Tarragona, Spain
- Department of Paediatric, Hospital Universitari Sant Joan de Reus, Reus, Spain
| | - Mariona Gispert
- IISPV, Pediatric Nutrition and Human Development Research Unit, Universitat Rovira i Virgili, Tarragona, Spain
| | | | - Dariusz Gruszfeld
- Department of Paediatric, Children's Memorial Health Institute in Warsaw, Warsaw, Poland
| | - Iris Iglesia
- Agrifood Institute of Aragon (IA2), Growth, Exercise, Nutrition and Development (GENUD) Research Group, University of Zaragoza, Zaragoza, Spain
- Instituto de Investigación Sanitaria Aragón (IIS-Aragón), Zaragoza, Spain
| | - Veit Grote
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Hans Demmelmair
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | - Uschi Handel
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| | | | - Berthold Koletzko
- Division of Metabolic and Nutritional Medicine, Department of Paediatrics, Dr von Hauner Children's Hospital, University Hospital, Ludwig Maximilian University Munich, Munich, Germany
| |
Collapse
|
31
|
Morozumi M, Wada Y, Tsuda M, Tabata F, Ehara T, Nakamura H, Miyaji K. Cross-feeding among bifidobacteria on glycomacropeptide. J Funct Foods 2023. [DOI: 10.1016/j.jff.2023.105463] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/11/2023] Open
|
32
|
Crost EH, Coletto E, Bell A, Juge N. Ruminococcus gnavus: friend or foe for human health. FEMS Microbiol Rev 2023; 47:fuad014. [PMID: 37015876 PMCID: PMC10112845 DOI: 10.1093/femsre/fuad014] [Citation(s) in RCA: 135] [Impact Index Per Article: 67.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 02/06/2023] [Accepted: 04/03/2023] [Indexed: 04/06/2023] Open
Abstract
Ruminococcus gnavus was first identified in 1974 as a strict anaerobe in the gut of healthy individuals, and for several decades, its study has been limited to specific enzymes or bacteriocins. With the advent of metagenomics, R. gnavus has been associated both positively and negatively with an increasing number of intestinal and extraintestinal diseases from inflammatory bowel diseases to neurological disorders. This prompted renewed interest in understanding the adaptation mechanisms of R. gnavus to the gut, and the molecular mediators affecting its association with health and disease. From ca. 250 publications citing R. gnavus since 1990, 94% were published in the last 10 years. In this review, we describe the biological characterization of R. gnavus, its occurrence in the infant and adult gut microbiota and the factors influencing its colonization of the gastrointestinal tract; we also discuss the current state of our knowledge on its role in host health and disease. We highlight gaps in knowledge and discuss the hypothesis that differential health outcomes associated with R. gnavus in the gut are strain and niche specific.
Collapse
Affiliation(s)
- Emmanuelle H Crost
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Erika Coletto
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Andrew Bell
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| | - Nathalie Juge
- Quadram Institute Bioscience, Rosalind Franklin Road, Colney, Norwich NR4 7UQ, United Kingdom
| |
Collapse
|
33
|
Therapeutic Potential of Gut Microbiota and Its Metabolite Short-Chain Fatty Acids in Neonatal Necrotizing Enterocolitis. Life (Basel) 2023; 13:life13020561. [PMID: 36836917 PMCID: PMC9959300 DOI: 10.3390/life13020561] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Revised: 01/31/2023] [Accepted: 02/15/2023] [Indexed: 02/19/2023] Open
Abstract
Short chain fatty acids (SCFAs), the principle end-products produced by the anaerobic gut microbial fermentation of complex carbohydrates (CHO) in the colon perform beneficial roles in metabolic health. Butyrate, acetate and propionate are the main SCFA metabolites, which maintain gut homeostasis and host immune responses, enhance gut barrier integrity and reduce gut inflammation via a range of epigenetic modifications in DNA/histone methylation underlying these effects. The infant gut microbiota composition is characterized by higher abundances of SCFA-producing bacteria. A large number of in vitro/vivo studies have demonstrated the therapeutic implications of SCFA-producing bacteria in infant inflammatory diseases, such as obesity and asthma, but the application of gut microbiota and its metabolite SCFAs to necrotizing enterocolitis (NEC), an acute inflammatory necrosis of the distal small intestine/colon affecting premature newborns, is scarce. Indeed, the beneficial health effects attributed to SCFAs and SCFA-producing bacteria in neonatal NEC are still to be understood. Thus, this literature review aims to summarize the available evidence on the therapeutic potential of gut microbiota and its metabolite SCFAs in neonatal NEC using the PubMed/MEDLINE database.
Collapse
|
34
|
Fan R, Xie S, Wang S, Yu Z, Sun X, Du Q, Yang Y, Han R. Identification markers of goat milk adulterated with bovine milk based on proteomics and metabolomics. Food Chem X 2023; 17:100601. [PMID: 36974185 PMCID: PMC10039227 DOI: 10.1016/j.fochx.2023.100601] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2022] [Revised: 02/09/2023] [Accepted: 02/14/2023] [Indexed: 02/18/2023] Open
Abstract
This study investigated the differences in proteins and metabolites from goat and bovine milk, and their mixtures, using data-independent-acquisition-based proteomics and metabolomics methods. In the skim milk, relative abundances of secretoglobin family 1D member (SCGB1D), polymeric immunoglobulin receptor, and glycosylation-dependent cell adhesion molecule 1 were increased, with an increase in the amount of 1-100 % bovine milk and served as markers at the 1 % adulteration level. In whey samples, β-lactoglobulin and α-2-HS-glycoprotein could be used to detect adulteration at the 0.1 % adulteration level, and SCGB1D and zinc-alpha-2-glycoprotein at the 1 % level. The metabolites of uric acid and N-formylkynurenine could be used to detect bovine milk at adulteration levels as low as 1 % based on variable importance at a projection value of > 1.0 and P-value of < 0.05. Our findings suggest novel markers of SCGB1D, uric acid, and N-formylkynurenine that can help to facilitate assessments of goat milk authenticity.
Collapse
Affiliation(s)
- Rongbo Fan
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Shubin Xie
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Shifeng Wang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Zhongna Yu
- Haidu College. Qingdao Agricultural University, Laiyang 265200, Shandong, China
| | - Xueheng Sun
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Qijing Du
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Yongxin Yang
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
| | - Rongwei Han
- College of Food Science and Engineering, Qingdao Agricultural University, Qingdao 266109, Shandong, China
- Corresponding author.
| |
Collapse
|
35
|
Cortés-Macías E, Selma-Royo M, Rio-Aige K, Bäuerl C, Rodríguez-Lagunas MJ, Martínez-Costa C, Pérez-Cano FJ, Collado MC. Distinct breast milk microbiota, cytokine, and adipokine profiles are associated with infant growth at 12 months: an in vitro host-microbe interaction mechanistic approach. Food Funct 2023; 14:148-159. [PMID: 36472137 DOI: 10.1039/d2fo02060b] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
Breast milk (BM) is important for adequate infant development, and it contains bioactive compounds, such as bacteria, cytokines and some adipokines which play a role in infant microbial, metabolic, and immunological maturation. However, little is known about its impact on growth and development in early life. The objective of this study was to evaluate the impact of milk microbiota, cytokine, and adipokine profiles on the risk of overweight at 12 months of life to find the possible mechanisms of host-microbe interactions. In this study, BM samples from 100 healthy women collected during 15 d after birth were included. BM microbiota was analysed by 16S rRNA gene sequencing, and cytokine and adipokine levels were measured by the Luminex approach. In addition, infant weight and length were recorded during the first 12 months and z-scores were obtained according to the WHO databases. Infants were classified as risk of overweight (ROW) and no-risk of overweight (NOROW) based on their body mass index z-score (BMIZ) and infant weight-for-length z-score (WLZ) at 12 months. In order to study host-microbe interactions, epithelial intestinal and mammary cell lines were exposed to milk microbes to assess the host response by interleukin (IL)-6 production as a potential inflammatory marker. BM was dominated by Staphylococcus and Streptococcus genera, and the most abundant cytokines were IL-6 and IL-18. Leptin levels were positively correlated with the pregestational body mass index (BMI). Higher relative abundance of the Streptococcus genus was associated with higher IL-10 and higher relative abundance of the Bifidobacterium genus was associated with lower IL-6 concentrations in milk. Infant WLZ at 12 months could be partially predicted by Streptococcus genus proportions and IL-10 and IL-18 levels in BM. BM microbiota significantly induced cytokine responses in mammary epithelial cells. Higher levels of IL-6 production were observed in mammary cells exposed to BM microbiota from mothers with ROW offspring compared to mothers with NOROW offspring. In conclusion, BM microbiota is related to the cytokine profile. IL-10 and IL-18 levels and the abundance of the Streptococcus genus could affect early infant development. Further research is needed to clarify the specific impact of BM microbiota and cytokines on infant growth and the risk of overweight.
Collapse
Affiliation(s)
- Erika Cortés-Macías
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - Marta Selma-Royo
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - Karla Rio-Aige
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Christine Bäuerl
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| | - María José Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Cecilia Martínez-Costa
- Department of Pediatrics, School of Medicine, University of Valencia, Valencia, Spain.,Pediatric Gastroenterology and Nutrition Section, Hospital Clínico Universitario Valencia, INCLIVA, Valencia, Spain
| | - Francisco J Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain.,Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Maria Carmen Collado
- Department of Biotechnology, Institute of Agrochemistry and Food Technology-, National Research Council (IATA-CSIC), Valencia, Spain.
| |
Collapse
|
36
|
Mao B, He Z, Chen Y, Stanton C, Ross RP, Zhao J, Chen W, Yang B. Effects of Bifidobacterium with the Ability of 2'-Fucosyllactose Utilization on Intestinal Microecology of Mice. Nutrients 2022; 14:nu14245392. [PMID: 36558551 PMCID: PMC9785880 DOI: 10.3390/nu14245392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2022] [Revised: 12/08/2022] [Accepted: 12/15/2022] [Indexed: 12/23/2022] Open
Abstract
In breast milk, 2'-Fucosyllactose (2'FL) is the most abundant breast milk oligosaccharide and can selectively promote the proliferation of bifidobacteria. This study aimed to explore the effect of ifidobacterial with different utilization capacities of 2'FL on the intestinal microecology of mice. Furthermore, the effects of ifidobacterial with different 2'FL utilization capabilities on mice gut microbiota under the competitive pressure of 2'FL as a carbon source were explored. Compared with the control group, 2'FL, Bifidobacterium (B.) bifidum M130R01M51 + 2'FL, B. longum subsp. Longum CCFM752, and CCFM752 + 2'FL treatments significantly decreased the food intake. Moreover, the water intake, body weight, and fecal water content in all groups showed no significant difference compared with the control group. The combination of B. longum subsp. longum CCFM752 and 2'FL can significantly increase the levels of pro-inflammatory and anti-inflammatory factors. B. bifidum M130R01M51 and mixed strains combined with 2'FL significantly increased the contents of acetic acid and isobutyric acid. The results showed that B. bifidum M130R01M51, B. breve FHuNCS6M1, B. longum subsp. longum CCFM752, and B. longum subsp. infantis SDZC2M4 combined with 2'FL significantly increased the species richness of the gut microbiota. Moreover, B. longum subsp. longum CCFM752 and B. longum subsp. infantis SDZC2M4 significantly increased the abundance of Faecalibaculum and Bifidobacterium, respectively. In conclusion, exploring the impact on intestinal microecology can provide theoretical guidance for the development of personalized prebiotics for different bifidobacteria, which has the potential to improve the ecological imbalance of infant gut microbiota.
Collapse
Affiliation(s)
- Bingyong Mao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Zhujun He
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Yang Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Catherine Stanton
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
- Teagasc Food Research Centre, Moorepark, Co., P61 C996 Cork, Ireland
| | - Reynolds Paul Ross
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- APC Microbiome Ireland, University College Cork, T12 R229 Cork, Ireland
| | - Jianxin Zhao
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
| | - Wei Chen
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- National Engineering Research Center for Functional Food, Jiangnan University, Wuxi 214122, China
| | - Bo Yang
- State Key Laboratory of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- School of Food Science and Technology, Jiangnan University, Wuxi 214122, China
- International Joint Research Center for Probiotics & Gut Health, Jiangnan University, Wuxi 214122, China
- Correspondence:
| |
Collapse
|
37
|
Arzamasov AA, Nakajima A, Sakanaka M, Ojima MN, Katayama T, Rodionov DA, Osterman AL. Human Milk Oligosaccharide Utilization in Intestinal Bifidobacteria Is Governed by Global Transcriptional Regulator NagR. mSystems 2022; 7:e0034322. [PMID: 36094076 PMCID: PMC9599254 DOI: 10.1128/msystems.00343-22] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Accepted: 08/23/2022] [Indexed: 12/24/2022] Open
Abstract
Bifidobacterium longum subsp. infantis is a prevalent beneficial bacterium that colonizes the human neonatal gut and is uniquely adapted to efficiently use human milk oligosaccharides (HMOs) as a carbon and energy source. Multiple studies have focused on characterizing the elements of HMO utilization machinery in B. longum subsp. infantis; however, the regulatory mechanisms governing the expression of these catabolic pathways remain poorly understood. A bioinformatic regulon reconstruction approach used in this study implicated NagR, a transcription factor from the ROK family, as a negative global regulator of gene clusters encoding lacto-N-biose/galacto-N-biose (LNB/GNB), lacto-N-tetraose (LNT), and lacto-N-neotetraose (LNnT) utilization pathways in B. longum subsp. infantis. This conjecture was corroborated by transcriptome profiling upon nagR genetic inactivation and experimental assessment of binding of recombinant NagR to predicted DNA operators. The latter approach also implicated N-acetylglucosamine (GlcNAc), a universal intermediate of LNT and LNnT catabolism, and its phosphorylated derivatives as plausible NagR transcriptional effectors. Reconstruction of NagR regulons in various Bifidobacterium lineages revealed multiple potential regulon expansion events, suggesting evolution from a local regulator of GlcNAc catabolism in ancestral bifidobacteria to a global regulator controlling the utilization of mixtures of GlcNAc-containing host glycans in B. longum subsp. infantis and Bifidobacterium bifidum. IMPORTANCE The predominance of bifidobacteria in the gut of breastfed infants is attributed to the ability of these bacteria to metabolize human milk oligosaccharides (HMOs). Thus, individual HMOs such as lacto-N-tetraose (LNT) and lacto-N-neotetraose (LNnT) are considered promising prebiotics that would stimulate the growth of bifidobacteria and confer multiple health benefits to preterm and malnourished children suffering from impaired (stunted) gut microbiota development. However, the rational selection of HMO-based prebiotics is hampered by the incomplete knowledge of regulatory mechanisms governing HMO utilization in target bifidobacteria. This study describes NagR-mediated transcriptional regulation of LNT and LNnT utilization in Bifidobacterium longum subsp. infantis. The elucidated regulatory network appears optimally adapted to simultaneous utilization of multiple HMOs, providing a rationale to add HMO mixtures (rather than individual components) to infant formulas. The study also provides insights into the evolutionary trajectories of complex regulatory networks controlling carbohydrate metabolism in bifidobacteria.
Collapse
Affiliation(s)
- Aleksandr A. Arzamasov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | - Miriam N. Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Dmitry A. Rodionov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| | - Andrei L. Osterman
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, California, USA
| |
Collapse
|
38
|
Kaplina A, Zaikova E, Ivanov A, Volkova Y, Alkhova T, Nikiforov V, Latypov A, Khavkina M, Fedoseeva T, Pervunina T, Skorobogatova Y, Volkova S, Ulyantsev V, Kalinina O, Sitkin S, Petrova N. Intestinal microbiome changes in an infant with right atrial isomerism and recurrent necrotizing enterocolitis: A case report and review of literature. World J Clin Cases 2022; 10:10583-10599. [PMID: 36312470 PMCID: PMC9602219 DOI: 10.12998/wjcc.v10.i29.10583] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2022] [Revised: 04/20/2022] [Accepted: 08/14/2022] [Indexed: 02/05/2023] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) is a multifactorial disease that predominantly affects premature neonates. Intestinal dysbiosis plays a critical role in NEC pathogenesis in premature neonates. The main risk factor for NEC in term infants is mesenteric hypoperfusion associated with ductal-dependent congenital heart disease (CHD) that eventually leads to intestinal ischemia. The incidence of NEC in neonates with critical CHD is 6.8%-13%. However, the role of the intestinal microbiome in NEC pathogenesis in infants with ductal-dependent CHD remains unclear. CASE SUMMARY A male term neonate with right atrial isomerism underwent modified Blalock-Taussig shunt placement on the 14th day of life and had persistent mesenteric hypoperfusion after surgery. The patient had episodes of NEC stage IIA on the 1st and 28th days after cardiac surgery. Fecal microbial composition was analyzed before and after cardiac surgery by sequencing region V4 of the 16S rRNA gene. Before surgery, species belonging to genera Veillonella and Clostridia and class Gammaproteobacteria were detected, Bifidobacteriaceae showed a low abundance. The first NEC episode was associated with postoperative hemodynamic instability, intestinal ischemia-reperfusion injury during cardiopulmonary bypass, and a high abundance of Clostridium paraputrificum (Clostridium sensu stricto I) (56.1%). Antibacterial therapy after the first NEC episode resulted in increased abundance of Gammaproteobacteria, decreased abundance of Firmicutes, and low alpha diversity. These changes in the microbial composition promoted the growth of Clostridium sensu stricto I (72.0%) before the second NEC episode. CONCLUSION A high abundance of Clostridium sensu stricto I and mesenteric hypoperfusion may have contributed to NEC in the present case.
Collapse
Affiliation(s)
- Aleksandra Kaplina
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Ekaterina Zaikova
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Artem Ivanov
- International Laboratory of Computer Technologies, ITMO University, St. Petersburg 197101, Russia
| | - Yulia Volkova
- Department of Cardiovascular Surgery for Children, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Alkhova
- Department of Neonatal Physiology with an ICU Ward, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Vladimir Nikiforov
- Pediatric Cardiac Intensive Care Unit, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Alexander Latypov
- Department of Cardiovascular Surgery for Children, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Marina Khavkina
- Neonatal and Preterm Special Care Unit (2nd Stage Care), Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Fedoseeva
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Tatiana Pervunina
- Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Yulia Skorobogatova
- Express Laboratory of Perinatal Centre, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Svetlana Volkova
- Clinical Diagnostic Laboratory, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Vladimir Ulyantsev
- International Laboratory of Computer Technologies, ITMO University, St. Petersburg 197101, Russia
| | - Olga Kalinina
- Research Laboratory of Autoimmune and Autoinflammatory Diseases, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| | - Stanislav Sitkin
- Epigenetics and Metagenomics Group, Institute of Perinatology and Pediatrics, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
- Department of Internal Diseases, Gastroenterology and Dietetics, North-Western State Medical University named after I.I. Mechnikov, St. Petersburg 191015, Russia
| | - Natalia Petrova
- Research Laboratory of Physiology and Diseases of Newborns, Almazov National Medical Research Centre, St. Petersburg 197341, Russia
| |
Collapse
|
39
|
Huang M, Cao X, He Q, Yang H, Chen Y, Zhao J, Ma H, Kang J, Liu J, Quang F. Alkaline semen diluent combined with R848 for separation and enrichment of dairy goat X-sperm. J Dairy Sci 2022; 105:10020-10032. [DOI: 10.3168/jds.2022-22115] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2022] [Accepted: 07/26/2022] [Indexed: 11/06/2022]
|
40
|
Arzamasov AA, Osterman AL. Milk glycan metabolism by intestinal bifidobacteria: insights from comparative genomics. Crit Rev Biochem Mol Biol 2022; 57:562-584. [PMID: 36866565 PMCID: PMC10192226 DOI: 10.1080/10409238.2023.2182272] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 01/11/2023] [Accepted: 02/15/2023] [Indexed: 03/04/2023]
Abstract
Bifidobacteria are early colonizers of the human neonatal gut and provide multiple health benefits to the infant, including inhibiting the growth of enteropathogens and modulating the immune system. Certain Bifidobacterium species prevail in the gut of breastfed infants due to the ability of these microorganisms to selectively forage glycans present in human milk, specifically human milk oligosaccharides (HMOs) and N-linked glycans. Therefore, these carbohydrates serve as promising prebiotic dietary supplements to stimulate the growth of bifidobacteria in the guts of children suffering from impaired gut microbiota development. However, the rational formulation of milk glycan-based prebiotics requires a detailed understanding of how bifidobacteria metabolize these carbohydrates. Accumulating biochemical and genomic data suggest that HMO and N-glycan assimilation abilities vary remarkably within the Bifidobacterium genus, both at the species and strain levels. This review focuses on the delineation and genome-based comparative analysis of differences in respective biochemical pathways, transport systems, and associated transcriptional regulatory networks, providing a foundation for genomics-based projection of milk glycan utilization capabilities across a rapidly growing number of sequenced bifidobacterial genomes and metagenomic datasets. This analysis also highlights remaining knowledge gaps and suggests directions for future studies to optimize the formulation of milk-glycan-based prebiotics that target bifidobacteria.
Collapse
Affiliation(s)
- Aleksandr A Arzamasov
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei L Osterman
- Infectious and Inflammatory Diseases Center, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| |
Collapse
|
41
|
Exploring Bacterial Attributes That Underpin Symbiont Life in the Monogastric Gut. Appl Environ Microbiol 2022; 88:e0112822. [PMID: 36036591 PMCID: PMC9499014 DOI: 10.1128/aem.01128-22] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The large bowel of monogastric animals, such as that of humans, is home to a microbial community (microbiota) composed of a diversity of mostly bacterial species. Interrelationships between the microbiota as an entity and the host are complex and lifelong and are characteristic of a symbiosis. The relationships may be disrupted in association with disease, resulting in dysbiosis. Modifications to the microbiota to correct dysbiosis require knowledge of the fundamental mechanisms by which symbionts inhabit the gut. This review aims to summarize aspects of niche fitness of bacterial species that inhabit the monogastric gut, especially of humans, and to indicate the research path by which progress can be made in exploring bacterial attributes that underpin symbiont life in the gut.
Collapse
|
42
|
Jackson PPJ, Wijeyesekera A, Rastall RA. Determining the metabolic fate of human milk oligosaccharides: it may just be more complex than you think? GUT MICROBIOME (CAMBRIDGE, ENGLAND) 2022; 3:e9. [PMID: 39295778 PMCID: PMC11406381 DOI: 10.1017/gmb.2022.8] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 05/10/2022] [Accepted: 08/26/2022] [Indexed: 09/21/2024]
Abstract
Human milk oligosaccharides (HMOs) are a class of structurally diverse and complex unconjugated glycans present in breast milk, which act as selective substrates for several genera of select microbes and inhibit the colonisation of pathogenic bacteria. Yet, not all infants are breastfed, instead being fed with formula milks which may or may not contain HMOs. Currently, formula milks only possess two HMOs: 2'-fucosyllactose (2'FL) and lacto-N-neotetraose (LNnT), which have been suggested to be similarly effective as human breast milk in supporting age-related growth. However, the in vivo evidence regarding their ability to beneficially reduce respiratory infections along with altering the composition of an infant's microbiota is limited at best. Thus, this review will explore the concept of HMOs and their metabolic fate, and summarise previous in vitro and in vivo clinical data regarding HMOs, with specific regard to 2'FL and LNnT.
Collapse
Affiliation(s)
| | - Anisha Wijeyesekera
- Department of Food and Nutritional Sciences, University of Reading, Reading, UK
| | | |
Collapse
|
43
|
Davis EC, Castagna VP, Sela DA, Hillard MA, Lindberg S, Mantis NJ, Seppo AE, Järvinen KM. Gut microbiome and breast-feeding: Implications for early immune development. J Allergy Clin Immunol 2022; 150:523-534. [PMID: 36075638 PMCID: PMC9463492 DOI: 10.1016/j.jaci.2022.07.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2022] [Revised: 07/21/2022] [Accepted: 07/21/2022] [Indexed: 11/16/2022]
Abstract
Establishment of the gut microbiome during early life is a complex process with lasting implications for an individual's health. Several factors influence microbial assembly; however, breast-feeding is recognized as one of the most influential drivers of gut microbiome composition during infancy, with potential implications for function. Differences in gut microbial communities between breast-fed and formula-fed infants have been consistently observed and are hypothesized to partially mediate the relationships between breast-feeding and decreased risk for numerous communicable and noncommunicable diseases in early life. Despite decades of research on the gut microbiome of breast-fed infants, there are large scientific gaps in understanding how human milk has evolved to support microbial and immune development. This review will summarize the evidence on how breast-feeding broadly affects the composition and function of the early-life gut microbiome and discuss mechanisms by which specific human milk components shape intestinal bacterial colonization, succession, and function.
Collapse
Affiliation(s)
- Erin C Davis
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | | | - David A Sela
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Department of Microbiology and Physiological Systems, University of Massachusetts Chan Medical School, Worcester, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Margaret A Hillard
- Department of Food Science, University of Massachusetts Amherst, Amherst, Mass; Organismic and Evolutionary Biology Graduate Program, University of Massachusetts Amherst, Amherst, Mass
| | - Samantha Lindberg
- Department of Biomedical Sciences, University of Albany, Rensselaer, NY
| | - Nicholas J Mantis
- Division of Infectious Diseases, New York State Department of Health, Albany, NY
| | - Antti E Seppo
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY
| | - Kirsi M Järvinen
- Division of Allergy and Immunology, Center for Food Allergy, Department of Pediatrics, University of Rochester School of Medicine and Dentistry, Golisano Children's Hospital, Rochester, NY; Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY; Department of Microbiology and Immunology, University of Rochester School of Medicine and Dentistry, Rochester, NY.
| |
Collapse
|
44
|
Ojima MN, Jiang L, Arzamasov AA, Yoshida K, Odamaki T, Xiao J, Nakajima A, Kitaoka M, Hirose J, Urashima T, Katoh T, Gotoh A, van Sinderen D, Rodionov DA, Osterman AL, Sakanaka M, Katayama T. Priority effects shape the structure of infant-type Bifidobacterium communities on human milk oligosaccharides. THE ISME JOURNAL 2022; 16:2265-2279. [PMID: 35768643 PMCID: PMC9381805 DOI: 10.1038/s41396-022-01270-3] [Citation(s) in RCA: 56] [Impact Index Per Article: 18.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 01/24/2022] [Revised: 06/05/2022] [Accepted: 06/09/2022] [Indexed: 11/10/2022]
Abstract
Bifidobacteria are among the first colonizers of the infant gut, and human milk oligosaccharides (HMOs) in breastmilk are instrumental for the formation of a bifidobacteria-rich microbiota. However, little is known about the assembly of bifidobacterial communities. Here, by applying assembly theory to a community of four representative infant-gut associated Bifidobacterium species that employ varied strategies for HMO consumption, we show that arrival order and sugar consumption phenotypes significantly affected community formation. Bifidobacterium bifidum and Bifidobacterium longum subsp. infantis, two avid HMO consumers, dominate through inhibitory priority effects. On the other hand, Bifidobacterium breve, a species with limited HMO-utilization ability, can benefit from facilitative priority effects and dominates by utilizing fucose, an HMO degradant not utilized by the other bifidobacterial species. Analysis of publicly available breastfed infant faecal metagenome data showed that the observed trends for B. breve were consistent with our in vitro data, suggesting that priority effects may have contributed to its dominance. Our study highlights the importance and history dependency of initial community assembly and its implications for the maturation trajectory of the infant gut microbiota.
Collapse
Affiliation(s)
- Miriam N Ojima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| | - Lin Jiang
- School of Biological Sciences, Georgia Institute of Technology, Atlanta, GA, USA
| | | | - Keisuke Yoshida
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Toshitaka Odamaki
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Jinzhong Xiao
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
- Next Generation Science Institute, Morinaga Milk Industry Co., Ltd., Kanagawa, Japan
| | - Aruto Nakajima
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | | | - Junko Hirose
- School of Human Cultures, The University of Shiga Prefecture, Hikone, Shiga, Japan
- Department of Food and Nutrition, Kyoto Women's University, Kyoto, Japan
| | - Tadasu Urashima
- Department of Food and Life Science, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, Hokkaido, Japan
| | - Toshihiko Katoh
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Aina Gotoh
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan
| | - Douwe van Sinderen
- APC Microbiome Ireland and School of Microbiology, Food Science Building, University College Cork, Cork, Ireland
| | - Dmitry A Rodionov
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | - Andrei L Osterman
- Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA, USA
| | | | - Takane Katayama
- Graduate School of Biostudies, Kyoto University, Kyoto, Japan.
| |
Collapse
|
45
|
McKeen S, Roy NC, Mullaney JA, Eriksen H, Lovell A, Kussman M, Young W, Fraser K, Wall CR, McNabb WC. Adaptation of the infant gut microbiome during the complementary feeding transition. PLoS One 2022; 17:e0270213. [PMID: 35834499 PMCID: PMC9282554 DOI: 10.1371/journal.pone.0270213] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 06/03/2022] [Indexed: 11/18/2022] Open
Abstract
The infant gut microbiome progresses in composition and function during the introduction of solid foods throughout the first year of life. The purpose of this study was to characterize changes in healthy infant gut microbiome composition, metagenomic functional capacity, and associated metabolites over the course of the complementary feeding period. Fecal samples were obtained at three ‘snapshot’ timepoints from infants participating in the ‘Nourish to Flourish’ pilot study: before the introduction of solid foods at approximately 4 months of age, after introducing solid foods at 9 months of age, and after continued diet diversification at 12 months of age. KEGG and taxonomy assignments were correlated with LC-MS metabolomic profiles to identify patterns of co-abundance. The composition of the microbiome diversified during the first year of life, while the functional capacity present in the gut microbiome remained stable. The introduction of solid foods between 4 and 9 months of age corresponded to a larger magnitude of change in relative abundance of sequences assigned to KEGG pathways and taxonomic assignments, as well as to stronger correlations with metabolites, compared to the magnitude of changes and number of correlations seen during continued diet diversification between 9 and 12 months of age. Changes in aqueous fecal metabolites were more strongly correlated with KEGG pathway assignments, while changes in lipid metabolites associated with taxonomic assignments, particularly between 9 and 12 months of age. This study establishes trends in microbiome composition and functional capacity occurring during the complementary feeding period and identifies potential metabolite targets for future investigations.
Collapse
Affiliation(s)
- Starin McKeen
- Riddet Institute, Massey University, Palmerston North, New Zealand
- School of Food and Advanced Technology, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Nicole C. Roy
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - Jane Adair Mullaney
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Hannah Eriksen
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Amy Lovell
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Martin Kussman
- German Entrepreneurship, Cambridge, Massachusetts, United States of America
| | - Wayne Young
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Karl Fraser
- Riddet Institute, Massey University, Palmerston North, New Zealand
- AgResearch Ltd, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
| | - Clare R. Wall
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- Department of Nutrition and Dietetics, The University of Auckland, Auckland, New Zealand
| | - Warren C. McNabb
- Riddet Institute, Massey University, Palmerston North, New Zealand
- High-Value Nutrition National Science Challenge, Auckland, New Zealand
- * E-mail:
| |
Collapse
|
46
|
Liu Y, Zhang F. Comparison of whole goat milk and its major fractions regarding the modulation of gut microbiota. JOURNAL OF THE SCIENCE OF FOOD AND AGRICULTURE 2022; 102:3618-3627. [PMID: 34873691 DOI: 10.1002/jsfa.11708] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/24/2021] [Revised: 10/13/2021] [Accepted: 12/06/2021] [Indexed: 06/13/2023]
Abstract
BACKGROUND Goat milk can be important for human nutrition because of its nutritional value, which may be attributed to its richness in protein, lactose, fat, and other bioactive components. This study compared the diversity and composition of gut microbiota in response to whole goat milk and its major fractions (milk fat, casein, milk whey, whey protein, and whey supernatant). Goat milk, its major fractions, and sterile distilled water (for the control group) were administered to mice intragastrically, and gut microbiota were compared in these groups using metagenomic analysis. RESULTS We observed distinct patterns of gut microbiota from different diet groups. The sample distance heatmap showed that, compared with other goat milk fractions, gut microbiota in the casein group was more similar to that in the whole goat-milk group. The relative abundance of the genus Lactobacillus increased significantly after whole goat-milk treatment; the milk whey fraction increased the abundance of Blautia; milk fat and milk whey related fractions treatment promoted the population of Bacteroides. The network analysis showed that genera Lactobacillus and Lactococcus were negatively associated with Helicobacter and Acinetobacter, respectively. CONCLUSION Fractions of goat milk could contain different gut microbiota from whole goat milk. Consumption of certain goat milk fractions could increase the ingestion of beneficial bacteria and inhibit the growth of some pathogenic bacteria. Our results could provide the basis for the research into and development of goat-milk based functional foods. © 2021 Society of Chemical Industry.
Collapse
Affiliation(s)
- Yufang Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| | - Fuxin Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an, China
| |
Collapse
|
47
|
Liu Y, Zhang F. Changes of antibiotic resistance genes and gut microbiota after the ingestion of goat milk. J Dairy Sci 2022; 105:4804-4817. [PMID: 35346469 DOI: 10.3168/jds.2021-21325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2021] [Accepted: 01/28/2022] [Indexed: 11/19/2022]
Abstract
Antibiotic resistance genes, as newly emerging contaminants, have become a serious challenge to public health through the food chain. The gut of humans and animals is an important reservoir for the development and dissemination of antibiotic resistance genes because of the great abundance and diversity of intestinal microbiota. In the present study, we evaluated the influence of goat milk on the diversity and abundance of antibiotic resistance genes and gut microbial communities, especially pathogenic bacteria. Male mice were used, 12 for each of the 2 groups: a control group that received sterile distilled water and a treated group that received goat milk, and gut microbiota and antibiotic resistance genes were compared in these groups using metagenomic analysis. The results revealed that ingestion of goat milk decreased the diversity and abundance of antibiotic resistance genes in the mice gut. The relative abundance of fluoroquinolone, peptide, macrolide, and β-lactam resistance genes in the total microbial genes significantly decreased after the intervention. Goat milk intake also significantly reduced the abundance of pathogenic bacteria, such as Clostridium bolteae, Clostridium symbiosum, Helicobacter cinaedi, and Helicobacter bilis. Therefore, goat milk intake might decrease the transfer potential of antibiotic resistance gene to pathogenic bacteria in the gut. In addition, bacteria with multiple resistance mechanisms accounted for approximately 4.5% of total microbial communities in the control group, whereas it was not detectable in the goat milk group, indicating the total inhibition by goat milk intake. This study highlights the influence of goat milk on antibiotic resistome and microbial communities in the gut, and provides a new insight into the function of goat milk for further study.
Collapse
Affiliation(s)
- Yufang Liu
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China
| | - Fuxin Zhang
- College of Food Engineering and Nutritional Science, Shaanxi Normal University, Xi'an 710119, China.
| |
Collapse
|
48
|
Liu F, He S, Yan J, Yan S, Chen J, Lu Z, Zhang B, Lane J. Longitudinal changes of human milk oligosaccharides, breastmilk microbiome and infant gut microbiome are associated with maternal characteristics. Int J Food Sci Technol 2022. [DOI: 10.1111/ijfs.15324] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Affiliation(s)
- Feitong Liu
- H&H Group Global Research and Technology Center Guangzhou 510700 China
| | - Shiting He
- H&H Group Global Research and Technology Center Guangzhou 510700 China
- College of Life Science and Technology Beijing University of Chemical Technology Beijing 100029 China
| | - Jingyu Yan
- Dalian Institute of Chemical Physics Chinese Academy of Sciences Dalian 116023 China
| | - Shuyuan Yan
- Child Health Care Center Changsha Hospital for Maternal and Child Care Changsha 410007 China
| | - Juchun Chen
- H&H Group Global Research and Technology Center Guangzhou 510700 China
| | - Zerong Lu
- H&H Group Global Research and Technology Center Guangzhou 510700 China
| | - Bin Zhang
- School of Food Science and Engineering South China University of Technology Guangzhou 510640 China
| | - Jonathan Lane
- H&H Group Global Research and Technology Center Cork P61 C996 Ireland
| |
Collapse
|
49
|
Alshehri DB, Sindi HH, AlMusalami IM, Rozi IH, Shagrani M, Kamal NM, Alahmadi NS, Alfuraikh SS, Vandenplas Y. Saudi Experts Consensus on Diagnosis and Management of Pediatric Functional Constipation. Pediatr Gastroenterol Hepatol Nutr 2022; 25:163-179. [PMID: 35611377 PMCID: PMC9110844 DOI: 10.5223/pghn.2022.25.3.163] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2021] [Revised: 01/02/2022] [Accepted: 03/07/2022] [Indexed: 11/14/2022] Open
Abstract
Although functional gastrointestinal disorders (FGIDs) are very common in pediatric patients, there is a scarcity of published epidemiologic data, characteristics, and management patterns from Saudi Arabia, which is the 2nd largest Arabic country in terms of area and the 6th largest Arabic country in terms of population, with 10% of its population aged <5 years. Functional constipation (FC) is an FGID that has shown a rising prevalence among Saudi infants and children in the last few years, which urges us to update our clinical practices. Nine pediatric consultants attended two advisory board meetings to discuss and address current challenges, provide solutions, and reach a Saudi national consensus for the management of pediatric constipation. The pediatric consultants agreed that pediatricians should pay attention to any alarming signs (red flags) found during history taking or physical examinations. They also agreed that the Rome IV criteria are the gold standard for the diagnosis of pediatric FC. Different therapeutic options are available for pediatric patients with FC. Dietary treatment is recommended for infants with constipation for up to six months of age. When non-pharmacological interventions fail to improve FC symptoms, pharmacological treatment with laxatives is indicated. First, the treatment is aimed at disimpaction to remove fecal masses. This is achieved by administering a high dose of oral polyethylene glycol (PEG) or lactulose for a few days. Subsequently, maintenance therapy with PEG should be initiated to prevent the re-accumulation of feces. In addition to PEG, several other options may be used, such as Mg-rich formulas or stimulant laxatives. However, rectal enemas and suppositories are usually reserved for cases that require acute pain relief. In contrast, infant formulas that contain prebiotics or probiotics have not been shown to be effective in infant constipation, while the use of partially hydrolyzed formula is inconclusive. These clinical practice recommendations are intended to be adopted by pediatricians and primary care physicians across Saudi Arabia.
Collapse
Affiliation(s)
- Dhafer B. Alshehri
- Department of Pediatrics, Faculty of Medicine, Najran University, Najran, Saudi Arabia
| | | | | | | | - Mohamed Shagrani
- King Faisal Specialist Hospital and Research Centre, Pediatric Gastroenterology, Riyadh, Saudi Arabia
| | - Naglaa M. Kamal
- Pediatric Department, Kasr Alainy Faculty of Medicine, Cairo University, Cairo, Egypt
- Pediatric Hepatology, Gastroenterology and Nutrition, Alhada Armed Forces Hospital, Taif, Saudi Arabia
| | - Najat Saeid Alahmadi
- Pediatric Department, King Salman Medical City, Ministry of Health, Almadinah Almonawarah, Saudi Arabia
| | - Samia Saud Alfuraikh
- King Abdul Aziz Hospital, Ministry of National Guard Health Affairs, Eastern Region, Saudi Arabia
| | - Yvan Vandenplas
- Vrije Universiteit Brussel, UZ Brussel, KidZ Health Castle, Brussels, Belgium
| |
Collapse
|
50
|
Beta-diversity distance matrices for microbiome sample size and power calculations - How to obtain good estimates. Comput Struct Biotechnol J 2022; 20:2259-2267. [PMID: 35664226 PMCID: PMC9133771 DOI: 10.1016/j.csbj.2022.04.032] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2021] [Revised: 04/13/2022] [Accepted: 04/22/2022] [Indexed: 12/12/2022] Open
Abstract
In microbiome studies, researchers often wish to compare the taxa count distributions between groups of samples. Commonly-used corresponding methods of analysis are built on examining distance matrices, where distances describe the beta-diversity between samples. Analyses then compare the distribution of distances within groups to the distributions between groups. However, when performing a priori sample size or power calculations for such study designs, appropriate within and between group distance distributions can be challenging to obtain. When available, pilot study data, or data from prior studies of similar design should provide realistic distance estimates. However, when these are not available, distances can be extracted from available studies where one can assume similar beta-diversity. Alternatively, distances can be generated by simulation methods. Here, we describe and illustrate these three strategies for obtaining realistic distance matrices. For simulation methods, we illustrate the procedures required starting from existing benchmark data, as well as how to simulate directly from population assumptions. Using data from the American Gut project, we provide tables of observed distances for use by researchers planning their own studies, as well as R codes for generating similar matrices in other datasets. Furthermore, for simulated data, we compare methods, provide R codes, and demonstrate how challenging it is to obtain realistic distance distributions without any benchmark data. This code and illustrative distance tables are provided by the IMPACTT Consortium as a resource to the microbiome research community.
Collapse
|