1
|
Liu B, Zhang Z, Zhao J, Li X, Wang Y, Liu L, Qiao W, Chen L. Lactiplantibacillus plantarum HM-P2 influences gestational gut microbiome and microbial metabolism. Front Nutr 2024; 11:1489359. [PMID: 39758313 PMCID: PMC11695228 DOI: 10.3389/fnut.2024.1489359] [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: 09/01/2024] [Accepted: 12/02/2024] [Indexed: 01/07/2025] Open
Abstract
Introduction Human milk-derived probiotics are beneficial bacteria that provide gestational health benefits, for both pregnant women and their offspring. The study aims to investigate whether the administration of human milk-derived probiotic L. plantarum HM-P2 could effectively influence gestational health. Methods The gestational humanized microbiome model was built by fecal microbiome transplant from gestational women into germ-free (GF) mice. Results HM-P2 was successfully planted and increased the top crypt depth of the colon, and microbes such as L. reuteri, Anaerofilum sp. An201, and Gemmiger were up-regulated in the HM-P2 group throughout gestation. HM-P2 significantly promoted the contents of intestinal caproic acid, bile acids, and tryptophan catabolites such as serotonin. Gut microbes were associated with these bile acids and tryptophans. Discussion HM-P2 could modulate the microbial community and microbial metabolites in gestational humanized GF mice. This probiotic strain could be a potential gestational dietary supplement with health benefits.
Collapse
Affiliation(s)
- Bin Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Zhenzhen Zhang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Junying Zhao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Xianping Li
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Yaru Wang
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lu Liu
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Weicang Qiao
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| | - Lijun Chen
- National Engineering Research Center of Dairy Health for Maternal and Child, Beijing Sanyuan Foods Co. Ltd., Beijing, China
- Beijing Engineering Research Center of Dairy, Beijing Technical Innovation Center of Human Milk Research, Beijing Sanyuan Foods Co. Ltd., Beijing, China
| |
Collapse
|
2
|
Tas GG, Sati L. Probiotic Lactobacillus rhamnosus species: considerations for female reproduction and offspring health. J Assist Reprod Genet 2024; 41:2585-2605. [PMID: 39172320 PMCID: PMC11535107 DOI: 10.1007/s10815-024-03230-6] [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: 06/26/2024] [Accepted: 08/08/2024] [Indexed: 08/23/2024] Open
Abstract
Lactobacillus rhamnosus is a type of bacteria known as a probiotic and is often used to support the health of the digestive system and vaginal flora. This type of bacteria has an important role, showing positive effects on female reproductive biology, particularly by maintaining the balance of microorganisms in the vagina, reducing the risk of infection, and strengthening the immune system to support maternal health during pregnancy. There are also studies showing that these probiotics prevent maternal obesity and gestational diabetes. Consuming probiotics containing Lactobacillus rhamnosus strains may support the intestinal health of breastfeeding mothers, but they may also contribute to the health of offspring. Therefore, this review focuses on the current available data for examining the effects of Lactobacillus rhamnosus strains on female reproductive biology and offspring health. A systematic search was conducted in the PubMed and Web of Science databases from inception to May 2024. The search strategy was performed using keywords and MeSH (Medical Subject Headings) terms. Inconsistent ratings were resolved through discussion. This review is strengthened by multiple aspects of the methodological approach. The systematic search strategy, conducted by two independent reviewers, enabled the identification and evaluation of all relevant literature. Although there is a limited number of studies with high heterogeneity, current literature highlights the important contribution of Lactobacillus rhamnosus probiotics in enhancing female reproductive health and fertility. Furthermore, the probiotic bacteria in breast milk may also support the intestinal health of newborn, strengthen the immune system, and protect them against diseases at later ages.
Collapse
Affiliation(s)
- Gizem Gamze Tas
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey
| | - Leyla Sati
- Department of Histology and Embryology, Akdeniz University School of Medicine, Campus, 07070, Antalya, Turkey.
| |
Collapse
|
3
|
Divakara N, Dempsey Z, Saraswati C, Garssen J, Silva D, Keelan JA, Christophersen CT, Cooper MN, Prescott SL, Palmer DJ, Verhasselt V, Macchiaverni P. Effect of maternal prebiotic supplementation on human milk immunological composition: Insights from the SYMBA study. Pediatr Allergy Immunol 2024; 35:e14226. [PMID: 39221598 DOI: 10.1111/pai.14226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 08/08/2024] [Accepted: 08/16/2024] [Indexed: 09/04/2024]
Abstract
BACKGROUND Immunomodulatory proteins in human milk (HM) can shape infant immune development. However, strategies to modulate their levels are currently unknown. This study investigated whether maternal prebiotic supplementation alters the levels of immunomodulatory proteins in HM. METHODS The study was nested within the SYMBA double-blind randomized controlled trial (ACTRN12615001075572), which investigated the effects of maternal prebiotic (short-chain galacto-oligosaccharides/long-chain fructo-oligosaccharides) supplementation from <21 weeks gestation during pregnancy until 6 months postnatal during lactation on child allergic disease risk. Mother-child dyads receiving prebiotics (n = 46) or placebo (n = 54) were included in this study. We measured the levels of 24 immunomodulatory proteins in HM collected at 2, 4, and 6 months. RESULTS Cluster analysis showed that the overall immunomodulatory protein composition of milk samples from both groups was similar. At 2 months, HM of prebiotic-supplemented women had decreased levels of TGF-β1 and TSLP (95% CI: -17.4 [-29.68, -2.28] and -57.32 [-94.22, -4.7] respectively) and increased levels of sCD14 (95% CI: 1.81 [0.17, 3.71]), when compared to the placebo group. At 4 months, IgG1 was lower in the prebiotic group (95% CI: -1.55 [-3.55, -0.12]) compared to placebo group. CONCLUSION This exploratory study shows that prebiotic consumption by lactating mothers selectively alters specific immunomodulatory proteins in HM. This finding is crucial for understanding how prebiotic dietary recommendations for pregnant and lactating women can modify the immune properties of HM and potentially influence infant health outcomes through immune support from breastfeeding.
Collapse
Affiliation(s)
- Nivedithaa Divakara
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, Medical School, University of Western Australia, Perth, Western Australia, Australia
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Zac Dempsey
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Chitra Saraswati
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Johan Garssen
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, Utrecht, The Netherlands
- Nutricia Research, Utrecht, The Netherlands
| | - Desiree Silva
- The Kids Research Institute Australia, Perth, Western Australia, Australia
- School of Medicine, UWA Health Campus, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Joondalup Health Campus, Joondalup, Western Australia, Australia
| | - Jeffrey A Keelan
- School of Medicine, UWA Health Campus, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- School of Biomedical Sciences, UWA Health Campus, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Claus T Christophersen
- Western Australian Human Microbiome Collaboration Centre, Curtin University, Bentley, Western Australia, Australia
- School of Medical and Health Sciences, Edith Cowan University, Joondalup, Western Australia, Australia
| | - Matthew N Cooper
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Susan L Prescott
- The Kids Research Institute Australia, Perth, Western Australia, Australia
- School of Medicine, UWA Health Campus, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
- Department of Immunology and Dermatology, Perth Children's Hospital, QEII Medical Centre, Nedlands, Western Australia, Australia
- Nova Institute for Health, Baltimore, Maryland, USA
| | - Debra J Palmer
- The Kids Research Institute Australia, Perth, Western Australia, Australia
- School of Medicine, UWA Health Campus, QEII Medical Centre, The University of Western Australia, Nedlands, Western Australia, Australia
| | - Valerie Verhasselt
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, Medical School, University of Western Australia, Perth, Western Australia, Australia
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| | - Patricia Macchiaverni
- Larsson-Rosenquist Foundation Centre for Immunology and Breastfeeding, Medical School, University of Western Australia, Perth, Western Australia, Australia
- The Kids Research Institute Australia, Perth, Western Australia, Australia
| |
Collapse
|
4
|
Gao T, Li R, Hu L, Hu Q, Wen H, Zhou R, Yuan P, Zhang X, Huang L, Zhuo Y, Xu S, Lin Y, Feng B, Che L, Wu D, Fang Z. Probiotic Lactobacillus rhamnosus GG improves insulin sensitivity and offspring survival via modulation of gut microbiota and serum metabolite in a sow model. J Anim Sci Biotechnol 2024; 15:89. [PMID: 38951898 PMCID: PMC11218078 DOI: 10.1186/s40104-024-01046-z] [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: 02/15/2024] [Accepted: 05/07/2024] [Indexed: 07/03/2024] Open
Abstract
BACKGROUND Sows commonly experience insulin resistance in late gestation and lactation, causing lower feed intake and milk production, which can lead to higher mortality rates in newborn piglets. The probiotic Lactobacillus rhamnosus GG (LGG) is known to improve insulin resistance. However, whether supplementing LGG can improve insulin sensitivity in sows and enhance lactation performance, particularly the early survival of offspring remains unclear. Hence, we explored the effects and mechanisms of supplementing LGG during late gestation and lactation on sow insulin sensitivity, lactation performance, and offspring survival. In total, 20 sows were randomly allocated to an LGG (n = 10) and control group (n = 10). RESULTS In sows, LGG supplementation significantly improved insulin sensitivity during late gestation and lactation, increased feed intake, milk production and colostrum lactose levels in early lactation, and enhanced newborn piglet survival. Moreover, LGG treatment significantly reshaped the gut microbiota in sows, notably increasing microbiota diversity and enriching the relative abundance of insulin sensitivity-associated probiotics such as Lactobacillus, Bifidobacterium, and Bacteroides. Serum metabolite and amino acid profiling in late-gestation sows also revealed decreased branched-chain amino acid and kynurenine serum levels following LGG supplementation. Further analyses highlighted a correlation between mitigated insulin resistance in late pregnancy and lactation by LGG and gut microbiota reshaping and changes in serum amino acid metabolism. Furthermore, maternal LGG enhanced immunity in newborn piglets, reduced inflammation, and facilitated the establishment of a gut microbiota. CONCLUSIONS We provide the first evidence that LGG mitigates insulin resistance in sows and enhances offspring survival by modulating the gut microbiota and amino acid metabolism.
Collapse
Affiliation(s)
- Tianle Gao
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Ran Li
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Food Science, Sichuan Agricultural University, Ya' an, 625014, China
| | - Liang Hu
- Key Laboratory of Agricultural Product Processing and Nutrition Health (Co-construction by Ministry and Province), Ministry of Agriculture and Rural Affairs, College of Food Science, Sichuan Agricultural University, Ya' an, 625014, China
| | - Quanfang Hu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Hongmei Wen
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Rui Zhou
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Peiqiang Yuan
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Xiaoling Zhang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Lingjie Huang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yong Zhuo
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Shengyu Xu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Yan Lin
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Bin Feng
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Lianqiang Che
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - De Wu
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China
| | - Zhengfeng Fang
- Key Laboratory for Animal Disease-Resistance Nutrition of China Ministry of Education, Animal Nutrition Institute, Sichuan Agricultural University, 211 Huimin Road, Wenjiang District, Chengdu, 611130, China.
| |
Collapse
|
5
|
Yu R, Ma Y, Luo Z, Qi C, Xie A, Jiang Y, Zhu B, Sun J. Maternal supplementation with Limosilactobacillus reuteri FN041 for preventing infants with atopic dermatitis: study protocol for a randomized controlled trial. Front Microbiol 2023; 14:1267448. [PMID: 37869669 PMCID: PMC10588667 DOI: 10.3389/fmicb.2023.1267448] [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: 07/26/2023] [Accepted: 08/28/2023] [Indexed: 10/24/2023] Open
Abstract
Background Atopic dermatitis (AD) has increased rapidly with rapid urbanization; however, the treatment options for AD are lacking because the commonly used therapies can only alleviate symptoms. Limosilactobacillus reuteri (L. reuteri), FN041 is a specific strain isolated from human breast milk, and its protective potential against AD has been confirmed. This study aims to assess the efficacy of maternal consumption of L. reuteri FN041 during late pregnancy and lactation in preventing infantile AD. Methods First, a randomized, double-blind, placebo-controlled intervention study will be conducted on 340 pregnant females with babies at high risk for AD. These subjects will be randomly divided into four groups of different doses of L. reuteri FN041 (1 × 109, 5 × 109, and 1 × 1010 CFU/d) along with a placebo. The safety and efficacy of maternal use of L. reuteri FN041 for preventing infantile AD will be analyzed, and the most efficient dosage of L. reuteri FN041 will be determined. Subsequently, a multicenter cohort study of 500 pregnant females with babies at high risk for AD will be conducted to promote the maternal application of L. reuteri FN041. These subjects will be administered L. reuteri FN041 at the optimal dose determined during the first stage of late pregnancy and lactation, and their babies will be analyzed for AD development. Recruitment was initiated in October 2022. Discussion The primary outcome is the cumulative incidence of AD at 24 months after maternal consumption of L. reuteri FN041 during late pregnancy and lactation, whereas the secondary outcome is the efficiency of L. reuteri FN041 transfer from the mother's gut to breast milk and then the infant's gut after oral supplementation. This study will demonstrate the efficacy of edible probiotics isolated from breast milk in preventing or treating AD in infants. Accordingly, we provide population-based advice for administering specific probiotics for the primary prevention of AD in pregnant females. Understanding the underlying mechanisms of probiotic strains derived from breast milk can promote their application in preventing infant diseases associated with intestinal microbiota imbalance and immune disorders. Clinical trial registration https://www.chictr.org.cn/, identifier [ChiCTR2300075611].
Collapse
Affiliation(s)
- Renqiang Yu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yizhe Ma
- Department of Pediatric, Jiangyin People's Hospital of Nantong University, Wuxi, China
| | - Zichen Luo
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Ce Qi
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| | - Anni Xie
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Yifan Jiang
- School of Medicine, Nantong University, Nantong, China
| | - Baoli Zhu
- Department of Neonatology, Wuxi Maternity and Child Health Care Hospital, Women’s Hospital of Jiangnan University, Wuxi, China
| | - Jin Sun
- Institute of Nutrition and Health, Qingdao University, Qingdao, China
| |
Collapse
|
6
|
Chang C, Wang Q, Li X, Tan H, Huang G. The relationship between prebiotic intake and allergic rhinitis. Laryngoscope Investig Otolaryngol 2023; 8:1146-1153. [PMID: 37899852 PMCID: PMC10601581 DOI: 10.1002/lio2.1158] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2023] [Revised: 08/28/2023] [Accepted: 09/18/2023] [Indexed: 10/31/2023] Open
Abstract
Objectives Exploring the relationship between intake of probiotics and the prevalence of allergic rhinitis. Methods Based on data from the National Health and Nutrition Examination Survey, dietary supplement labels were examined to identify products containing probiotics and prebiotics. Statistical methods were used to analyze the factors influencing the prevalence of allergic rhinitis, and further stratified analysis was conducted to control for confounding factors. Results The proportion of individuals not consuming probiotics was significantly higher in the allergic rhinitis (AR) group than in those consuming them, suggesting a correlation between probiotics and AR. In the male subgroup with probiotic intake, the adjusted odds ratio (95% confidence interval) was 0.28 (0.10-0.75), p = .02, indicating that probiotic intake was a protective factor for AR in the male population. In the probiotic-intake group, the odds ratio for age < 65 was 0.26 (0.07-0.94), p = .04, and for age ≥ 80 was less than 1 with p < .0001, suggesting that probiotic intake was a protective factor for AR in age < 65 and age ≥ 80 populations, both with statistical significance. Conclusion Intake of probiotics is associated with a reduced prevalence of allergic rhinitis, particularly in the male population and individuals aged <65 years and ≥ 80 years. Level of Evidence Level 4.
Collapse
Affiliation(s)
- Chao Chang
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Qiuyang Wang
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Xiaodan Li
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Huazhang Tan
- Department of Otolaryngology, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangHubeiChina
| | - Guoxin Huang
- Department of Evidence‐Based Medicine Center, Xiangyang No.1 People's HospitalHubei University of MedicineXiangyangChina
- School of Public Health and ManagementHubei University of MedicineShiyanHubeiChina
| |
Collapse
|
7
|
Xiang Q, Yan X, Shi W, Li H, Zhou K. Early gut microbiota intervention in premature infants: Application perspectives. J Adv Res 2023; 51:59-72. [PMID: 36372205 PMCID: PMC10491976 DOI: 10.1016/j.jare.2022.11.004] [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: 08/08/2022] [Revised: 10/30/2022] [Accepted: 11/05/2022] [Indexed: 11/13/2022] Open
Abstract
BACKGROUND Preterm birth is the leading cause of death in children under the age of five. One of the major factors contributing to the high risk of diseases and deaths in premature infants is the incomplete development of the intestinal immune system. The gut microbiota has been widely recognized as a critical factor in promoting the development and function of the intestinal immune system after birth. However, the gut microbiota of premature infants is at high risk of dysbiosis, which is highly associated with adverse effects on the development and education of the early life immune system. Early intervention can modulate the colonization and development of gut microbiota and has a long-term influence on the development of the intestinal immune system. AIM OF REVIEW This review aims to summarize the characterization, interconnection, and underlying mechanism of gut microbiota and intestinal innate immunity in premature infants, and to discuss the status, applicability, safety, and prospects of different intervention strategies in premature infants, thus providing an overview and outlook of the current applications and remaining gaps of early intervention strategies in premature infants. KEY SCIENTIFIC CONCEPTS OF REVIEW This review is focused on three key concepts. Firstly, the gut microbiota of premature infants is at high risk of dysbiosis, resulting in dysfunctional intestinal immune system processes. Secondly, contributing roles of early intervention have been observed in improving the intestinal environment and promoting gut microbiota colonization, which is significant in the development and function of gut immunity in premature infants. Thirdly, different strategies of early intervention, such as probiotics, fecal microbiota transplantation, and nutrients, show different safety, applicability, and outcome in premature infants, and the underlying mechanism is complex and poorly understood.
Collapse
Affiliation(s)
- Quanhang Xiang
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Xudong Yan
- Department of Neonatal Intensive Care Unit, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Wei Shi
- Department of Obstetrics and Gynecology, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China
| | - Huiping Li
- Department of Respiratory and Critical Care Medicine, the first affiliated hospital of Southern University of Science and Technology of China, Shenzhen People's Hospital, Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China
| | - Kai Zhou
- Shenzhen Institute of Respiratory Diseases, the Second Clinical Medical College, Jinan University (Shenzhen People's Hospital), Shenzhen, China; The First Affiliated Hospital (Shenzhen People's Hospital), Southern University of Science and Technology, Shenzhen, China.
| |
Collapse
|
8
|
Mazziotta C, Tognon M, Martini F, Torreggiani E, Rotondo JC. Probiotics Mechanism of Action on Immune Cells and Beneficial Effects on Human Health. Cells 2023; 12:cells12010184. [PMID: 36611977 PMCID: PMC9818925 DOI: 10.3390/cells12010184] [Citation(s) in RCA: 192] [Impact Index Per Article: 96.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 12/12/2022] [Accepted: 12/29/2022] [Indexed: 01/03/2023] Open
Abstract
Immune cells and commensal microbes in the human intestine constantly communicate with and react to each other in a stable environment in order to maintain healthy immune activities. Immune system-microbiota cross-talk relies on a complex network of pathways that sustain the balance between immune tolerance and immunogenicity. Probiotic bacteria can interact and stimulate intestinal immune cells and commensal microflora to modulate specific immune functions and immune homeostasis. Growing evidence shows that probiotic bacteria present important health-promoting and immunomodulatory properties. Thus, the use of probiotics might represent a promising approach for improving immune system activities. So far, few studies have been reported on the beneficial immune modulatory effect of probiotics. However, many others, which are mainly focused on their metabolic/nutritional properties, have been published. Therefore, the mechanisms behind the interaction between host immune cells and probiotics have only been partially described. The present review aims to collect and summarize the most recent scientific results and the resulting implications of how probiotic bacteria and immune cells interact to improve immune functions. Hence, a description of the currently known immunomodulatory mechanisms of probiotic bacteria in improving the host immune system is provided.
Collapse
Affiliation(s)
- Chiara Mazziotta
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
| | - Mauro Tognon
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Fernanda Martini
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Laboratory for Technologies of Advanced Therapies (LTTA), University of Ferrara, 44121 Ferrara, Italy
| | - Elena Torreggiani
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| | - John Charles Rotondo
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
- Center for Studies on Gender Medicine, Department of Medical Sciences, University of Ferrara, 64/b, Fossato di Mortara Street, 44121 Ferrara, Italy
- Correspondence: (E.T.); (J.C.R.); Tel.: +39-053-2455-557 (E.T.); +39-053-245-5536 (J.C.R.)
| |
Collapse
|
9
|
Association between Maternal Characteristics and Immune Factors TGF-β1, TGF-β2, and IgA in Colostrum: An Exploratory Study in Japan. Nutrients 2022; 14:nu14163255. [PMID: 36014760 PMCID: PMC9416399 DOI: 10.3390/nu14163255] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/05/2022] [Accepted: 08/05/2022] [Indexed: 11/24/2022] Open
Abstract
Colostrum is the first food for newborns and it contains various crucial immune factors. The concentrations of immune factors in breast milk may change depending on maternal characteristics such as body mass index, collection day, and age at first pregnancy. In this exploratory study, we investigated the association between TGF-β1, TGF-β2, and IgA in colostrum and rarely studied factors that affect breast milk components, including the use of labor-inducing medication, colostrum secretion, sex of newborns, breast or nipple problems, and nipple care. Breast milk samples were collected from 42 mothers and analyzed for TGF-β1, TGF-β2, and IgA. The results suggest that parity and mode of delivery may be correlated with the concentrations of immune factors in colostrum. However, we found no association between the immune factors in colostrum and the use of labor-inducing medications, colostrum secretion, sex of newborns, breast or nipple problems, and nipple care. These findings have some implications for further analysis of the effects of immune factors in breast milk on the prognosis of allergies in children.
Collapse
|
10
|
Kwok KO, Fries LR, Silva-Zolezzi I, Thakkar SK, Iroz A, Blanchard C. Effects of Probiotic Intervention on Markers of Inflammation and Health Outcomes in Women of Reproductive Age and Their Children. Front Nutr 2022; 9:889040. [PMID: 35734372 PMCID: PMC9207510 DOI: 10.3389/fnut.2022.889040] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Accepted: 05/03/2022] [Indexed: 11/13/2022] Open
Abstract
The human intestinal microbiota has been shown to be modulated during inflammatory conditions. Probiotic administration has been shown to affect the immune system and cytokine expression which can affect inflammation and health outcomes. There seems to be an association between the mother's intestinal microbiota and inflammation biomarkers, both of which may contribute to newborn early life immune and metabolic programming and impact short and long-term health outcomes. Probiotic supplementation during pregnancy has been shown to influence metabolic health, immunity, and gastrointestinal health of the mother, and can also have carry-over benefits to infants such as infant allergy risk reduction. Therefore, this review focuses on the evidence of probiotic administration in women of reproductive age, including during pregnancy and its impact on inflammatory markers and on maternal and infant health. We performed a PubMed search for articles published in English in the last 20 years. Immune markers were narrowed to serum and breast milk levels of TNF-α, IL-6 and TGF-β, IgA, and IL-10. Studies that investigated the beneficial effects of interventions in women with gestational diabetes mellitus, polycystic ovarian syndrome, and infant allergy management are summarized. These results show a beneficial or neutral effect on selected health outcomes and that it is safe for woman and their infants. The effect of probiotics on modulation of inflammatory markers was probiotic specific. More research is needed to further our understanding of the mechanisms underlying the effects of probiotics on inflammation and how these effects improve health outcomes.
Collapse
Affiliation(s)
- Kah Onn Kwok
- Food Science and Technology Programme, National University of Singapore, Singapore, Singapore
- Nestlé Research, Singapore, Singapore
| | | | | | | | - Alison Iroz
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
| | - Carine Blanchard
- Nestlé Institute of Health Sciences, Nestlé Research, Société des Produits Nestlé S.A., Lausanne, Switzerland
- *Correspondence: Carine Blanchard
| |
Collapse
|
11
|
Jarosz ŁS, Ciszewski A, Marek A, Hejdysz M, Nowaczewski S, Grądzki Z, Michalak K, Kwiecień M, Rysiak A. The effect of the multi-strain probiotic preparation EM Bokashi® on selected parameters of the cellular immune response in pigs. FOOD AGR IMMUNOL 2022. [DOI: 10.1080/09540105.2021.2006611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Łukasz S. Jarosz
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Artur Ciszewski
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Agnieszka Marek
- Sub-Department of Preventive Veterinary and Avian Diseases, Faculty of Veterinary Medicine, Institute of Biological Bases of Animal Diseases, University of Life Sciences in Lublin, Lublin, Poland
| | - Marcin Hejdysz
- Department of Animal Breeding And Product Quality Assessment, Poznań University of Life Sciences, Poznań, Poland
| | - Sebastian Nowaczewski
- Department of Animal Breeding And Product Quality Assessment, Poznań University of Life Sciences, Poznań, Poland
| | - Zbigniew Grądzki
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Katarzyna Michalak
- Department of Epizootiology and Clinic of Infectious Diseases, Faculty of Veterinary Medicine, University of Life Sciences in Lublin, Lublin, Poland
| | - Małgorzata Kwiecień
- Faculty of Biology and Animal Breeding, Institute of Animal Nutrition and Bromatology, Department of Animal Nutrition, University of Life Sciences in Lublin, Lublin, Poland
| | - Anna Rysiak
- Department of Botany, Mycology, and Ecology, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
12
|
Kim E, Paik D, Ramirez RN, Biggs DG, Park Y, Kwon HK, Choi GB, Huh JR. Maternal gut bacteria drive intestinal inflammation in offspring with neurodevelopmental disorders by altering the chromatin landscape of CD4 + T cells. Immunity 2022; 55:145-158.e7. [PMID: 34879222 PMCID: PMC8755621 DOI: 10.1016/j.immuni.2021.11.005] [Citation(s) in RCA: 54] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/29/2021] [Accepted: 11/10/2021] [Indexed: 01/13/2023]
Abstract
Children with autism spectrum disorders often display dysregulated immune responses and related gastrointestinal symptoms. However, the underlying mechanisms leading to the development of both phenotypes have not been elucidated. Here, we show that mouse offspring exhibiting autism-like phenotypes due to prenatal exposure to maternal inflammation were more susceptible to developing intestinal inflammation following challenges later in life. In contrast to its prenatal role in neurodevelopmental phenotypes, interleukin-17A (IL-17A) generated immune-primed phenotypes in offspring through changes in the maternal gut microbiota that led to postnatal alterations in the chromatin landscape of naive CD4+ T cells. The transfer of stool samples from pregnant mice with enhanced IL-17A responses into germ-free dams produced immune-primed phenotypes in offspring. Our study provides mechanistic insights into why children exposed to heightened inflammation in the womb might have an increased risk of developing inflammatory diseases in addition to neurodevelopmental disorders.
Collapse
Affiliation(s)
- Eunha Kim
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Donggi Paik
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ricardo N Ramirez
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Delaney G Biggs
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Youngjun Park
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Ho-Keun Kwon
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Gloria B Choi
- The Picower Institute for Learning and Memory, Department of Brain and Cognitive Sciences, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Jun R Huh
- Department of Immunology, Blavatnik Institute, Harvard Medical School, Boston, MA 02115, USA; Evergrande Center for Immunologic Diseases, Harvard Medical School and Brigham and Women's Hospital, Boston, MA 02115, USA.
| |
Collapse
|
13
|
Cheng J, Laitila A, Ouwehand AC. Bifidobacterium animalis subsp. lactis HN019 Effects on Gut Health: A Review. Front Nutr 2022; 8:790561. [PMID: 34970580 PMCID: PMC8712437 DOI: 10.3389/fnut.2021.790561] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2021] [Accepted: 11/22/2021] [Indexed: 12/19/2022] Open
Abstract
Optimal gut motility is central to bowel function and gut health. The link between the gut dysmotility related disorders and dysfunctional-intestinal barriers has led to a hypothesis that certain probiotics could help in normalizing gut motility and maintain gut health. This review investigates the roles of Bifidobacterium animalis subsp. lactis HN019 (B. lactis HN019™) on gut health, and its mechanisms of action in various pre-clinical and clinical studies. Research supports the hypothesis that B. lactis HN019™ has a beneficial role in maintaining intestinal barrier function during gastrointestinal infections by competing and excluding potential pathogens via different mechanisms; maintaining normal tight junction function in vitro; and regulating host immune defense toward pathogens in both in vitro and human studies. This has been observed to lead to reduced incidence of diarrhea. Interestingly, B. lactis HN019™ also supports normal physiological function in immunosenescent elderly and competes and excludes potential pathogens. Furthermore, B. lactis HN019™ reduced intestinal transit time and increased bowel movement frequency in functional constipation, potentially by modulating gut–brain–microbiota axis, mainly via serotonin signaling pathway, through short chain fatty acids derived from microbial fermentation. B. lactis HN019™ is thus a probiotic that can contribute to relieving gut dysmotility related disorders.
Collapse
Affiliation(s)
- Jing Cheng
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| | - Arja Laitila
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| | - Arthur C Ouwehand
- International Flavors & Fragrances Inc., Global Health and Nutrition Science, Danisco Sweeteners Oy, Kantvik, Finland
| |
Collapse
|
14
|
Taheri A, Raeisi T, Darand M, Jafari A, Janmohammadi P, Razi B, Mofidi Nejad M, Lafzi Ghazi M, Garousi N, Alizadeh S. Effects of Pre/Probiotic Supplementation on Breast Milk Levels of TGF-b1, TGF-b2, and IgA: A Systematic Review and Meta-Analysis of Randomized-Controlled Trial. Breastfeed Med 2022; 17:22-32. [PMID: 34714123 DOI: 10.1089/bfm.2021.0204] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Background: Previous studies have proposed that the maternal intake of pre/probiotics may affect the immune composition of breast milk. Nevertheless, the available findings are contradictory. This meta-analysis aimed to examine the impact of maternal supplementation with pre/probiotics on the levels of total immunoglobulin A (IgA), secretory IgA (SIgA), transforming growth factor beta 1 (TGF-β1), and TGF-2 in breast milk. Methods: PubMed and Scopus were systematically searched using a comprehensive search strategy for eligible randomized-controlled trials published up to February 2021. A random-effects model was applied to pool weighted mean difference and 95% confidence interval (CI) as effect size. Cochran's Q statistic and the I2 statistic were used to measure the between-study variance. Egger's regression test was used to assess publication bias. Results: A total of 12 different studies, with a total sample size of 1722 individuals (probiotic group: 858, placebo group: 864), were included in this meta-analysis. In the overall analysis, compared with placebo, maternal supplementation with pre/probiotics had no significant effect on concentrations of total IgA, SIgA, TGF-β1, and TGF-β2 in the breast milk. In the subgroup analysis, pre/probiotics did not affect total IgA, TGF-β1, and TGF-β2 in both colostrum/transitional and mature milk. However, a significant increase in SIgA was found in colostrum/transitional milk following pre/probiotic administration (WMD = 19.33, 95% CI: 0.83-37.83; p = 0.04), without evidence for remarkable heterogeneity (I2 = 0.0, p = 0.57). Conclusions: Maternal supplementation with pre/probiotics may increase SIgA in colostrum/transitional milk, without any effect on total IgA, TGF-β1, and TGF-β2.
Collapse
Affiliation(s)
- Akram Taheri
- Department of Nutrition, Faculty of Medicine, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Tahereh Raeisi
- Department of Medicine, Hormozgan University of Medical Sciences, Bandar Abbas, Iran
| | - Mina Darand
- Department of Clinical Nutrition and Dietetics, Faculty of Nutrition Sciences and Food Technology, National Nutrition and Food Technology Research Institute, ShahidBeheshti University of Medical Sciences, Tehran, Iran
| | - Alireza Jafari
- Department of Community Nutrition and School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Parisa Janmohammadi
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Bahman Razi
- Department of Hematology and Blood Banking, School of Medicine, Tarbiat Modares University (TMU), Tehran, Iran
| | - Maryam Mofidi Nejad
- Department of Community Nutrition and School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| | - Maryam Lafzi Ghazi
- Department of Exercise Physiology, Central Tehran Branch, Islamic Azad University, Tehran, Iran
| | - Nazila Garousi
- Department of Clinical Nutrition, School of Nutrition and Food Science, Isfahan University of Medical Sciences, Isfahan, Iran
| | - Shahab Alizadeh
- Department of Clinical Nutrition, School of Nutritional Sciences and Dietetics, Tehran University of Medical Sciences (TUMS), Tehran, Iran
| |
Collapse
|
15
|
Takahashi T, Fukudome H, Ueno HM, Watanabe-Matsuhashi S, Nakano T, Kobayashi T, Ishimaru K, Nakao A. Probiotic Supplementation and Human Milk Cytokine Profiles in Japanese Women: A Retrospective Study from an Open-Label Pilot Study. Nutrients 2021; 13:nu13072285. [PMID: 34209459 PMCID: PMC8308220 DOI: 10.3390/nu13072285] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2021] [Revised: 06/24/2021] [Accepted: 06/28/2021] [Indexed: 11/26/2022] Open
Abstract
The benefits of probiotic supplementation to lactating mothers on human milk cytokines are inconclusive. Thus, we performed a comprehensive open-label pilot trial analysis of 27 human milk cytokines in lactating women with allergies (one to three months postpartum) to determine the effect of supplementation with a mixture of new probiotic strains. Participants voluntarily joined the probiotic (n = 41) or no supplementation control (n = 19) groups. The probiotic group took three probiotic tablets (Lactobacillus casei LC5, Bifidobacterium longum BG7, and Bacillus coagulans SANK70258) daily for one to three months postpartum. Milk samples were collected at one, two, and three months postpartum, and cytokine levels were measured using multiplex assays. The effects were analyzed using multivariate regression models. Eleven cytokines showed a positive rate of over 50% in the milk samples throughout testing in both groups. The positive rates of IL-1 receptor antagonist and IL-7 changed significantly with lactation progression in logistic regression models after adjusting for time and supplementation, whereas rates of other cytokines showed no significant differences. The lactational change patterns of IL-10 concentrations differed significantly between the two groups. A short-term supplementation of probiotics affects human milk cytokine levels in lactating women with a possible placebo effect still existing. Future placebo-controlled studies are needed to support these results, based on the estimated sample sizes in this study.
Collapse
Affiliation(s)
- Tomoki Takahashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
- Correspondence: ; Tel.: +81-49-242-8138
| | - Hirofumi Fukudome
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Hiroshi M. Ueno
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Shiomi Watanabe-Matsuhashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Taku Nakano
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Toshiya Kobayashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama 350-1165, Japan; (H.F.); (H.M.U.); (S.W.-M.); (T.N.); (T.K.)
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (K.I.); (A.N.)
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi 409-3898, Japan; (K.I.); (A.N.)
| |
Collapse
|
16
|
Demers-Mathieu V, Mathijssen GB, DaPra C, Medo E. The effects of probiotic supplementation on the gene expressions of immune cell surface markers and levels of antibodies and pro-inflammatory cytokines in human milk. J Perinatol 2021; 41:1083-1091. [PMID: 33208844 DOI: 10.1038/s41372-020-00875-w] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/16/2020] [Revised: 10/07/2020] [Accepted: 11/04/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE This study investigated the impact of probiotic supplementation on the gene expressions of cluster of differentiation (CD) as cell markers and the concentrations of antibodies and cytokines in human milk. STUDY DESIGN Gene expressions of CD28, CD19, and CD38 were determined in milk from 15 women ingesting daily probiotics (from Greek yogurt) and 12 women who do not consume probiotics. Concentrations of antibodies and cytokines were measured using ELISA. RESULTS Gene expression of CD28 tended to be higher in milk from mothers ingesting daily probiotics than mothers who did not take probiotics. Interleukin-6 (IL-6) concentration in milk was higher in mothers ingesting probiotics than those who do not consume probiotics. The increase of IL-6 level in human milk was positively correlated with total IgA and IgG concentrations. CONCLUSIONS Probiotic supplementation could enhance the secretion of IL-6 in human milk. Human milk IL-6 may improve neonatal immunity due to its stimulation of total IgA and IgG.
Collapse
Affiliation(s)
- Veronique Demers-Mathieu
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA.
| | - Gabrielle B Mathijssen
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Ciera DaPra
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| | - Elena Medo
- Department of Neonatal Immunology and Microbiology, Medolac Laboratories A Public Benefit Corporation, Boulder City, NV, USA
| |
Collapse
|
17
|
Venter C, Palumbo MP, Sauder KA, Glueck DH, Starling AP, Ringham BM, O'Mahony L, Moore BF, Yang IV, Dabelea D. Examining Associations Between Dietary Inflammatory Index in Pregnancy, Pro-inflammatory Cytokine and Chemokine Levels at Birth, and Offspring Asthma and/or Wheeze by Age 4 Years. J Acad Nutr Diet 2021; 121:2003-2012.e3. [PMID: 33744233 DOI: 10.1016/j.jand.2021.02.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Revised: 02/14/2021] [Accepted: 02/16/2021] [Indexed: 12/28/2022]
Abstract
BACKGROUND Few studies have demonstrated associations between maternal dietary inflammatory index (DII) during pregnancy and offspring asthma and/or wheeze. OBJECTIVE The study aimed to assess associations between maternal DII during pregnancy and 1) offspring cord sera pro-inflammatory cytokines (interleukin [IL]-1β, IL-4, IL-6, IL-10, tumor necrosis factor-α) and chemokines (IL-8, monocyte chemoattractant protein-1) at birth and 2) offspring asthma and/or wheeze at age 4 years. DESIGN The Healthy Start study is a prospective prebirth longitudinal study that recruited pregnant women in Denver, Colorado and tracked their offspring. PARTICIPANTS AND SETTING This study used data from 1228 mother-child dyads enrolled in the Healthy Start study. Pregnant women were recruited in Denver, Colorado, between 2009 and 2014, and offspring tracked until age 4 years. MAIN OUTCOME MEASURES Cord sera cytokines and chemokines were analyzed with multiplex panel immunoassays. Offspring diagnosis of asthma and/or wheeze by age 4 years was extracted from electronic medical records. STATISTICAL ANALYSES PERFORMED Unadjusted and adjusted linear and logistic regression models were used to assess associations. Covariates included factors such as nulliparity, race/ethnicity, gestational smoking, and maternal history of asthma. RESULTS Unadjusted analysis showed that increasing maternal DII scores were associated with increased odds of child asthma and/or wheeze by 4 years (odds ratio = 1.17; 95% CI: 1.07-1.27), but the association was attenuated and no longer statistically significant in the adjusted model (odds ratio = 1.15; 95% CI: 0.99-1.33). There were no significant associations between DII scores and cord sera cytokine or chemokine levels. CONCLUSIONS The study showed that the inflammatory profile of the maternal diet was not associated with cytokines and chemokine levels at birth. The results suggested that a more inflammatory maternal diet was associated with increased odds of offspring asthma and/or wheeze by age 4 years, which could be considered of clinical relevance but the finding was not statistically significant at the .05 level.
Collapse
|
18
|
Wang K, Zhu Q, Kong X, Song M, Azad MAK, Xiong L, Zheng Y, He Q. Dietary Probiotics or Synbiotics Supplementation During Gestation, Lactation, and Nursery Periods Modifies Colonic Microbiota, Antioxidant Capacity, and Immune Function in Weaned Piglets. Front Vet Sci 2020; 7:597832. [PMID: 33381535 PMCID: PMC7767837 DOI: 10.3389/fvets.2020.597832] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2020] [Accepted: 11/23/2020] [Indexed: 12/11/2022] Open
Abstract
This study was conducted to investigate the effect of dietary probiotics or synbiotics supplementation on colonic microbiota, antioxidant capacity, and immune function in weaned piglets. A total of 64 pregnant Bama mini-sows and then 128 of their weaned piglets were randomly assigned into control group, antibiotics group, probiotics group, or synbiotics group. The results showed that colonic Firmicutes and Bifidobacterium abundances in the probiotics group and total bacteria, Bacteroidetes, and Lactobacillus abundances in the synbiotics group were increased (P < 0.05), while Escherichia coli abundance in the synbiotics group was decreased (P = 0.061) compared with the control group. Firmicutes, Bifidobacterium, and total bacteria abundances were increased (P < 0.05) in the probiotics and synbiotics groups compared with the antibiotics group. Probiotics supplementation up-regulated (P < 0.05) the mRNA expression of GPR109A compared with the control and antibiotics groups. Dietary probiotics or synbiotics supplementation improved the antioxidant capacity by increasing (P < 0.05) the colonic CAT, GSH-Px, SOD, and T-AOC levels and plasma CAT, GSH, GSH-Px, and SOD levels and by decreasing (P < 0.05) the colonic and plasma MDA and H2O2 levels. Compared to the control group, the colonic IL-10, IFN-α, and sIgA concentrations and plasma IgA and IgM concentrations were significantly increased (P < 0.05) in the probiotics and synbiotics groups. Spearman's correlation analysis showed that the changed colonic microbiota, such as Lactobacillus and Bifidobacterium were correlated with the alteration of antioxidant indexes, cytokines, and immunoglobulins. In conclusion, dietary probiotics or synbiotics supplementation during gestation, lactation, and nursery periods could be used as an alternative for antibiotics in terms of gut health of weaned piglets.
Collapse
Affiliation(s)
- Kai Wang
- Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China.,CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China.,Key Laboratory of Optoelectronic Devices and Systems of Ministry of Education and Guangdong Province, College of Optoelectronic Engineering, Shenzhen University, Shenzhen, China.,School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, China
| | - Qian Zhu
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Xiangfeng Kong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Mingtong Song
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Md Abul Kalam Azad
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Liang Xiong
- CAS Key Laboratory of Agro-ecological Processes in Subtropical Regions, Hunan Provincial Key Laboratory of Animal Nutritional Physiology and Metabolic Process, National Engineering Laboratory for Pollution Control and Waste Utilization in Livestock and Poultry Production, Institute of Subtropical Agriculture, Chinese Academy of Sciences, Changsha, China
| | - Yuzhong Zheng
- School of Food Engineering and Biotechnology, Hanshan Normal University, Chaozhou, China
| | - Qinghua He
- Department of Food Science and Engineering, College of Chemistry and Environmental Engineering, Shenzhen University, Shenzhen, China
| |
Collapse
|
19
|
Fernández L, Pannaraj PS, Rautava S, Rodríguez JM. The Microbiota of the Human Mammary Ecosystem. Front Cell Infect Microbiol 2020; 10:586667. [PMID: 33330129 PMCID: PMC7718026 DOI: 10.3389/fcimb.2020.586667] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 10/15/2020] [Indexed: 12/12/2022] Open
Abstract
Human milk contains a dynamic and complex site-specific microbiome, which is not assembled in an aleatory way, formed by organized microbial consortia and networks. Presence of some genera, such as Staphylococcus, Streptococcus, Corynebacterium, Cutibacterium (formerly known as Propionibacterium), Lactobacillus, Lactococcus and Bifidobacterium, has been detected by both culture-dependent and culture-independent approaches. DNA from some gut-associated strict anaerobes has also been repeatedly found and some studies have revealed the presence of cells and/or nucleic acids from viruses, archaea, fungi and protozoa in human milk. Colostrum and milk microbes are transmitted to the infant and, therefore, they are among the first colonizers of the human gut. Still, the significance of human milk microbes in infant gut colonization remains an open question. Clinical studies trying to elucidate the question are confounded by the profound impact of non-microbial human milk components to intestinal microecology. Modifications in the microbiota of human milk may have biological consequences for infant colonization, metabolism, immune and neuroendocrine development, and for mammary health. However, the factors driving differences in the composition of the human milk microbiome remain poorly known. In addition to colostrum and milk, breast tissue in lactating and non-lactating women may also contain a microbiota, with implications in the pathogenesis of breast cancer and in some of the adverse outcomes associated with breast implants. This and other open issues, such as the origin of the human milk microbiome, and the current limitations and future prospects are addressed in this review.
Collapse
Affiliation(s)
- Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Pia S. Pannaraj
- Department of Pediatrics and Molecular Microbiology and Immunology, Keck School of Medicine and Children’s Hospital, Los Angeles, CA, United States
| | - Samuli Rautava
- University of Helsinki and Helsinki University Hospital, New Children’s Hospital, Pediatric Research Center, Helsinki, Finland
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
20
|
Han SM, Binia A, Godfrey KM, El-Heis S, Cutfield WS. Do Human Milk Oligosaccharides Protect Against Infant Atopic Disorders and Food Allergy? Nutrients 2020; 12:nu12103212. [PMID: 33096669 PMCID: PMC7589050 DOI: 10.3390/nu12103212] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2020] [Revised: 10/14/2020] [Accepted: 10/20/2020] [Indexed: 12/28/2022] Open
Abstract
Atopic disorders (AD), often coexistent with food allergy (FA), start developing in early life and have lifelong health consequences. Breastfeeding is thought to be protective against AD and FA, but the data are controversial, and mechanisms are not well understood. Human milk oligosaccharides (HMOs) are complex carbohydrates that are abundant in human milk. These are thought to contribute to the development of the infant immune system by (i) promoting healthy microbiome, (ii) inhibiting pathogen binding to gut mucosa and (iii) modulating the immune system. Differences in microbiome composition between allergic and healthy infants have been observed, regardless of breastfeeding history. To date, limited studies have examined the preventive effects of HMOs on AD and FA in infants and current data relies on observation studies as trials of varying HMO intake through randomising individuals to breastfeeding are unethical. There is evidence for beneficial effects of breastfeeding on lowering the risks of FA, eczema and asthma but there are inconsistencies amongst studies in the duration of breastfeeding, diagnostic criteria for AD and the age at which the outcome was assessed. Furthermore, current analytical methods primarily used today only allow detection of 16-20 major HMOs while more than 100 types have been identified. More large-scale longitudinal studies are required to investigate the role of HMO composition and the impact of changes over the lactation period in preventing AD and FA later in life.
Collapse
Affiliation(s)
- Soo Min Han
- Liggins Institute, The University of Auckland, Auckland 1023, New Zealand;
| | - Aristea Binia
- Nestlé Research, Société des Produits Nestlé SA, 1000 Lausanne, Switzerland;
| | - Keith M. Godfrey
- NIHR Southampton Biomedical Research Centre, University of Southampton and University Hospital Southampton NHS Foundation Trust, Southampton SO17 1BJ, UK;
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO17 1BJ, UK;
| | - Sarah El-Heis
- MRC Lifecourse Epidemiology Unit, University of Southampton, Southampton SO17 1BJ, UK;
| | - Wayne S. Cutfield
- Liggins Institute, The University of Auckland, Auckland 1023, New Zealand;
- A Better Start—National Science Challenge, Liggins Institute, The University of Auckland, Auckland 1023, New Zealand
- Correspondence: ; Tel.: +64-9-923-4476
| |
Collapse
|
21
|
Guo TL, Chen Y, Xu HS, McDonough CM, Huang G. Gut microbiome in neuroendocrine and neuroimmune interactions: The case of genistein. Toxicol Appl Pharmacol 2020; 402:115130. [PMID: 32673657 DOI: 10.1016/j.taap.2020.115130] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2019] [Revised: 07/01/2020] [Accepted: 07/04/2020] [Indexed: 12/12/2022]
Abstract
The healthy and diverse microbes living in our gut provide numerous benefits to our health. It is increasingly recognized that the gut microbiome affects the host's neurobehavioral state through production of metabolites, modulation of intestinal immunity (e.g., cytokines) and other mechanisms (e.g., gut neuropeptides). By sending the sensed information (e.g., metabolic and immunologic mediators) about the state of the inner organs to the brain via afferent fibers, the vagus nerve maintains one of the connections between the brain and GI tract, and oversees many critical bodily functions (e.g., mood, immune response, digestion and heart rate). The microbiota-gut-brain axis is a bidirectional communication between the gut, its microbiome, and the nervous system. In the present review, the roles of microbiome in neuroendocrine and neuroimmune interactions have been discussed using naturally occurring isoflavones, particularly the phytoestrogen genistein, as there are sex differences in the interactions among the microbiome, hormones, immunity and disease susceptibility. A deep understanding of the mechanisms underlying the interactions among the endocrine modulators, brain, endocrine glands, gut immune cells, vagus nerve, enteric nervous system and gut microbiome will provide important knowledges that may ultimately lead to treatment and prevention of debilitating disorders characterized by deficits of microbiome-neuroendocrine-neuroimmune relationships.
Collapse
Affiliation(s)
- Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA.
| | - Yingjia Chen
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Hannah Shibo Xu
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | - Callie M McDonough
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine, University of Georgia, Athens, GA, USA
| | | |
Collapse
|
22
|
Amalia N, Orchard D, Francis KL, King E. Systematic review and meta‐analysis on the use of probiotic supplementation in pregnant mother, breastfeeding mother and infant for the prevention of atopic dermatitis in children. Australas J Dermatol 2019; 61:e158-e173. [DOI: 10.1111/ajd.13186] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2019] [Accepted: 08/31/2019] [Indexed: 12/11/2022]
Affiliation(s)
- Nasya Amalia
- Faculty of Medicine Universitas Indonesia Jakarta Indonesia
- Faculty of Medical Science University of Melbourne Melbourne Australia
| | - David Orchard
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| | - Kate Louise Francis
- Clinical Epidemiology and Biostatistics Unit Murdoch Children's Research Institute The Royal Children's Hospital Melbourne Australia
| | - Emma King
- Faculty of Medical Science University of Melbourne Melbourne Australia
- Dermatology DepartmentThe Royal Children's Hospital Melbourne Australia
| |
Collapse
|
23
|
Zielinska MA, Hamulka J. Protective Effect of Breastfeeding on the Adverse Health Effects Induced by Air Pollution: Current Evidence and Possible Mechanisms. INTERNATIONAL JOURNAL OF ENVIRONMENTAL RESEARCH AND PUBLIC HEALTH 2019; 16:E4181. [PMID: 31671856 PMCID: PMC6862650 DOI: 10.3390/ijerph16214181] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 10/22/2019] [Accepted: 10/25/2019] [Indexed: 02/08/2023]
Abstract
Air pollution is a major social, economic, and health problem around the world. Children are particularly susceptible to the negative effects of air pollution due to their immaturity and excessive growth and development. The aims of this narrative review were to: (1) summarize evidence about the protective effects of breastfeeding on the adverse health effects of air pollution exposure, (2) define and describe the potential mechanisms underlying the protective effects of breastfeeding, and (3) examine the potential effects of air pollution on breastmilk composition and lactation. A literature search was conducted using electronic databases. Existing evidence suggests that breastfeeding has a protective effect on adverse outcomes of indoor and outdoor air pollution exposure in respiratory (infections, lung function, asthma symptoms) and immune (allergic, nervous and cardiovascular) systems, as well as under-five mortality in both developing and developed countries. However, some studies reported no protective effect of breastfeeding or even negative effects of breastfeeding for under-five mortality. Several possible mechanisms of the breastfeeding protective effect were proposed, including the beneficial influence of breastfeeding on immune, respiratory, and nervous systems, which are related to the immunomodulatory, anti-inflammatory, anti-oxidant, and neuroprotective properties of breastmilk. Breastmilk components responsible for its protective effect against air pollutants exposure may be long chain polyunsaturated fatty acids (LC PUFA), antioxidant vitamins, carotenoids, flavonoids, immunoglobins, and cytokines, some of which have concentrations that are diet-dependent. However, maternal exposure to air pollution is related to increased breastmilk concentrations of pollutants (e.g., Polycyclic aromatic hydrocarbons (PAHs) or heavy metals in particulate matter (PM)). Nonetheless, environmental studies have confirmed that breastmilk's protective effects outweigh its potential health risk to the infant. Mothers should be encouraged and supported to breastfeed their infants due to its unique health benefits, as well as its limited ecological footprint, which is associated with decreased waste production and the emission of pollutants.
Collapse
Affiliation(s)
- Monika A Zielinska
- Department of Human Nutrition, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences-SGGW, 159C Nowoursynowska Street, 02-776 Warsaw, Poland.
| | - Jadwiga Hamulka
- Department of Human Nutrition, Institute of Human Nutrition Sciences, Warsaw University of Life Sciences-SGGW, 159C Nowoursynowska Street, 02-776 Warsaw, Poland.
| |
Collapse
|
24
|
Blyuss O, Cheung KY, Chen J, Parr C, Petrou L, Komarova A, Kokina M, Luzan P, Pasko E, Eremeeva A, Peshko D, Eliseev VI, Pedersen SA, Azad MB, Jarvinen KM, Peroni DG, Verhasselt V, Boyle RJ, Warner JO, Simpson MR, Munblit D. Statistical Approaches in the Studies Assessing Associations between Human Milk Immune Composition and Allergic Diseases: A Scoping Review. Nutrients 2019; 11:E2416. [PMID: 31658692 PMCID: PMC6836171 DOI: 10.3390/nu11102416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2019] [Revised: 09/27/2019] [Accepted: 10/01/2019] [Indexed: 01/06/2023] Open
Abstract
A growing number of studies are focusing on the associations between human milk (HM) immunological composition and allergic diseases. This scoping review aims to identify statistical methods applied in the field and highlight pitfalls and unmet needs. A comprehensive literature search in MEDLINE and Embase retrieved 13,607 unique records. Following title/abstract screening, 29 studies met the selection criteria and were included in this review. We found that definitions of colostrum and mature milk varied across the studies. A total of 17 out of 29 (59%) studies collected samples longitudinally, but only 12% of these used serial (longitudinal) analyses. Multivariable analysis was used in 45% of the studies, but statistical approaches to modelling varied largely across the studies. Types of variables included as potential confounding factors differed considerably between models. Discrimination analysis was absent from all studies and only a single study reported classification measures. Outcomes of this scoping review highlight lack of standardization, both in data collection and handling, which remains one of the main challenges in the field. Improved standardization could be obtained by a consensus group of researchers and clinicians that could recommend appropriate methods to be applied in future prospective studies, as well as already existing datasets.
Collapse
Affiliation(s)
- Oleg Blyuss
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London EC1M 6BQ, UK.
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Ka Yan Cheung
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
| | - Jessica Chen
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
| | - Callum Parr
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
| | - Loukia Petrou
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
| | - Alina Komarova
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Maria Kokina
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Polina Luzan
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Egor Pasko
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Alina Eremeeva
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Dmitrii Peshko
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
| | - Vladimir I Eliseev
- N. Polyakov Institute of Geotechnical Mechanics on the NAS of Ukraine, 49005 Dnipro, Ukraine.
| | - Sindre Andre Pedersen
- Library Section for Medicine and Health Sciences, NTNU-Norwegian University of Science and Technology, 7030 Trondheim, Norway.
| | - Meghan B Azad
- Department of Pediatrics and Child Health, Children's Hospital Research Institute of Manitoba, University of Manitoba, Winnipeg, MB R3E 3P4, Canada.
| | - Kirsi M Jarvinen
- Division of Pediatric Allergy and Immunology & Center for Food Allergy, University of Rochester School of Medicine and Dentistry, Rochester, New York, NY 14642, USA.
| | - Diego G Peroni
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, 56126 Pisa, Italy.
| | - Valerie Verhasselt
- School of Molecular Sciences, University of Western Australia, Perth, WA 6009, Australia.
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), West New York, NJ 10704, USA.
| | - Robert J Boyle
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
| | - John O Warner
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
- National Institute for Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, London SW10 9NH, UK.
| | - Melanie R Simpson
- Department of Public Health and General Practice, NTNU - Norwegian University of Science and Technology, 7030 Trondheim, Norway.
- Clinic of Laboratory Medicine, St Olavs Hospital, 7030 Trondheim, Norway.
| | - Daniel Munblit
- Department of Paediatrics and Paediatric Infectious Diseases, Institute of Child's Health, Sechenov First Moscow State Medical University (Sechenov University), 123337 Moscow, Russia.
- Department of Paediatrics, Imperial College London, London W2 1PG, UK.
- inVIVO Planetary Health, Group of the Worldwide Universities Network (WUN), West New York, NJ 10704, USA.
- Solov'ev Research and Clinical Center for Neuropsychiatry, 115419 Moscow, Russia.
| |
Collapse
|
25
|
Lee HS. The interaction between gut microbiome and nutrients on development of human disease through epigenetic mechanisms. Genomics Inform 2019; 17:e24. [PMID: 31610620 PMCID: PMC6808642 DOI: 10.5808/gi.2019.17.3.e24] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 09/22/2019] [Indexed: 02/06/2023] Open
Abstract
Early environmental exposure is recognized as a key factor for long-term health based on the Developmental Origins of Health and Disease hypothesis. It considers that early-life nutrition is now being recognized as a major contributor that may permanently program change of organ structure and function toward the development of diseases, in which epigenetic mechanisms are involved. Recent researches indicate early-life environmental factors modulate the microbiome development and the microbiome might be mediate diet-epigenetic interaction. This review aims to define which nutrients involve microbiome development during the critical window of susceptibility to disease, and how microbiome modulation regulates epigenetic changes and influences human health and future prevention strategies.
Collapse
Affiliation(s)
- Ho-Sun Lee
- Forensic Toxicology Division, Daegu Institute, National Forensic Service, Chilgok 39872, Korea
| |
Collapse
|
26
|
Huang G, Xu J, Cai D, Chen SY, Nagy T, Guo TL. Exacerbation of Type 1 Diabetes in Perinatally Genistein Exposed Female Non-Obese Diabetic (NOD) Mouse Is Associated With Alterations of Gut Microbiota and Immune Homeostasis. Toxicol Sci 2019; 165:291-301. [PMID: 29982808 DOI: 10.1093/toxsci/kfy162] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Despite various hypothesized benefits of dietary isoflavone genistein (GEN) from soy-based products, many questions surrounding GEN's immunotoxic effects, especially during perinatal exposure, have yet to be answered. The objective of the study was to determine if there existed a sex-specific effect of GEN on type 1 diabetes (T1D) following perinatal exposure. We exposed offspring of non-obese diabetic (NOD) mice to GEN per oral at a physiological dose (20 mg/kg body weight) from embryonic day 7 to postnatal day (PND) 21. In female offspring, perinatal GEN dosing significantly increased the incidence of T1D at early time points, and the exacerbation was associated with decreased serum levels of interleukin (IL)-10, IgG2a, and IgM. In male offspring dosed with GEN, a decrease in serum IgG1 was also observed. Flow cytometric analysis in females suggested an increased pro-inflammatory splenic CD5+CD24- and CD4-CD8+ cell counts, while both %T cells and %CD4+ T cells were significantly decreased in males, suggesting an anti-inflammatory effect. Gut microbiota (GMB) analysis indicated that fecal microbiota from PND 90 female offspring exhibited an increased level of Enterobacteriales (suggesting a pro-inflammatory response), while the similar changes were not found in PND 30 females. Moreover, RNA sequencing showed that intestinal α-defensin expression was down-regulated in GEN-treated females, supporting a pro-inflammatory response. However, perinatal GEN administration perturbed GMB toward an anti-inflammatory response in PND 90 males. Taken together, a strong sex-specific effect was found in the perinatal GEN exposure window, and the T1D exacerbation in NOD females was associated with GMB-related immunomodulatory mechanisms.
Collapse
Affiliation(s)
- Guannan Huang
- Department of Environmental Health Sciences, College of Public Health
| | - Joella Xu
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine
| | - Dunpeng Cai
- Department of Physiology and Pharmacology, College of Veterinary Medicine
| | - Shi-You Chen
- Department of Physiology and Pharmacology, College of Veterinary Medicine
| | - Tamas Nagy
- Department of Pathology, College of Veterinary Medicine, University of Georgia, Athens, Georgia 30602
| | - Tai L Guo
- Department of Veterinary Biosciences and Diagnostic Imaging, College of Veterinary Medicine
| |
Collapse
|
27
|
Takahashi T, Fukudome H, Ueno HM, Watanabe-Matsuhashi S, Nakano T, Kobayashi T, Ishimaru K, Nakao A. Effects of Probiotic Supplementation on TGF-β1, TGF-β2, and IgA Levels in the Milk of Japanese Women: An Open-Label Pilot Study. Front Nutr 2019; 6:128. [PMID: 31552256 PMCID: PMC6735502 DOI: 10.3389/fnut.2019.00128] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2018] [Accepted: 07/29/2019] [Indexed: 12/11/2022] Open
Abstract
Background: Dietary probiotics supplementation in lactating mothers may help prevent allergic disease in infants. However, owing to a lack of consistency in nutritional and safety outcomes associated with probiotics, this topic remains controversial. Methods: In this open-label pilot trial conducted between April 2013 and December 2013, we evaluated the safety of probiotic supplementation with 5 × 109 CFU of Lactobacillus casei LC5, 5 × 109 CFU of Bifidobacterium longum BG7, and 2 × 108 CFU of Bacillus coagulans SANK70258 in lactating women who exhibited allergies for 2 months (1–3 months postpartum); we also evaluated the effects of probiotic supplementation on transforming growth factor-β (TGF-β) and immunoglobulin A (IgA) levels in human milk. Participants self-selected to join the probiotic (n = 41; age [median (interquartile range [IQR]), y] 33 [27–39], body mass index [BMI] [median (IQR), kg/m2] 21.8 [19.5–22.8]) or no supplementation control group (n = 19; age [median (IQR), y] 33 [23–43], BMI [median (IQR), kg/m2) 19.6 [18.4–22.1]). Probiotics (three tablets) received were taken as daily supplements. Milk samples were collected at 1, 2, and 3 months postpartum, and TGF-β1, TGF-β2, and IgA levels were measured. Results: No adverse effects were observed in the probiotic group, according to the self-recorded diary during the study period. Milk IgA decreased with increasing postpartum months in both groups. In contrast, TGF- β1 and β2 were not affected by lactation periods, and showed different patterns over time between the two groups. TGF-β1, TGF-β1, and IgA levels were significantly correlated at baseline (respectively p < 0.05). However, the correlation between TGF-β1 and IgA became non-significant by the end of the intervention (p = 0.063). Conclusion: Altogether, probiotic supplementation was tolerated with respect to no dropout and 91.5% adherence. Although probiotic supplementation might affect human milk TGF-β levels, a positive effect of probiotic supplementation was not entirely supported. Future placebo-controlled studies are needed to further support the efficacy and safety of probiotic supplementation. Clinical Trial Registration:www.umin.ac.jp/ctr/, identifier: UMIN000036059.
Collapse
Affiliation(s)
- Tomoki Takahashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama, Japan
| | - Hirofumi Fukudome
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama, Japan
| | - Hiroshi M Ueno
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama, Japan
| | | | - Taku Nakano
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama, Japan
| | - Toshiya Kobayashi
- Research and Development Department, Bean Stalk Snow Co., Ltd., Saitama, Japan
| | - Kayoko Ishimaru
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| | - Atsuhito Nakao
- Department of Immunology, Faculty of Medicine, University of Yamanashi, Yamanashi, Japan
| |
Collapse
|
28
|
Effect of Multi-Microbial Probiotic Formulation Bokashi on Pro- and Anti-Inflammatory Cytokines Profile in the Serum, Colostrum and Milk of Sows, and in a Culture of Polymorphonuclear Cells Isolated from Colostrum. Probiotics Antimicrob Proteins 2019; 11:220-232. [PMID: 29305686 PMCID: PMC6449489 DOI: 10.1007/s12602-017-9380-9] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
The use of probiotics in sows during pregnancy and lactation and their impact on the quality of colostrum and milk, as well as the health conditions of their offspring during the rearing period, are currently gaining the attention of researchers. The aim of the study was to determine the effect of Bokashi formulation on the concentrations of pro- and anti-inflammatory cytokines in the serum of sows during pregnancy, in their colostrum and milk, and in a culture of Con-A-stimulated polymorphonuclear cells (PMNs) isolated from the colostrum. The study was conducted on 60 sows aged 2–4 years. EM Bokashi were added to the sows’ feed. The material for the study consisted of peripheral blood, colostrum, and milk. Blood samples were collected from the sows on days 60 and 114 of gestation. Colostrum and milk samples were collected from all sows at 0, 24, 48, 72, 96, 120, 144, and 168 h after parturition. The results indicate that the use of Bokashi as feed additives resulted in increased concentrations of pro-inflammatory cytokines TNF-α and IL-6, which increase the protective capacity of the colostrum by stimulating cellular immune mechanisms protecting the sow and neonates against infection. At the same time, the increased concentrations of cytokines IL-4, IL-10, TGF-β, and of immunoglobulins in the colostrum and milk from sows in the experimental group demonstrate the immunoregulatory effect of Bokashi on Th2 cells and may lead to increased expression of regulatory T cells and polarization of the immune response from Th1 to Th2.
Collapse
|
29
|
Boix-Amorós A, Collado MC, Van't Land B, Calvert A, Le Doare K, Garssen J, Hanna H, Khaleva E, Peroni DG, Geddes DT, Kozyrskyj AL, Warner JO, Munblit D. Reviewing the evidence on breast milk composition and immunological outcomes. Nutr Rev 2019; 77:541-556. [PMID: 31111150 DOI: 10.1093/nutrit/nuz019] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
A large number of biologically active components have been found in human milk (HM), and in both human and animal models, studies have provided some evidence suggesting that HM composition can be altered by maternal exposures, subsequently influencing health outcomes for the breastfed child. Evidence varies from the research studies on whether breastfeeding protects the offspring from noncommunicable diseases, including those associated with immunological dysfunction. It has been hypothesized that the conflicting evidence results from HM composition variations, which contain many immune active molecules, oligosaccharides, lactoferrin, and lysozyme in differing concentrations, along with a diverse microbiome. Determining the components that influence infant health outcomes in terms of both short- and long-term sequelae is complicated by a lack of understanding of the environmental factors that modify HM constituents and thereby offspring outcomes. Variations in HM immune and microbial composition (and the differing infantile responses) may in part explain the controversies that are evidenced in studies that aim to evaluate the prevalence of allergy by prolonged and exclusive breastfeeding. HM is a "mixture" of immune active factors, oligosaccharides, and microbes, which all may influence early immunological outcomes. This comprehensive review provides an in-depth overview of existing evidence on the studied relationships between maternal exposures, HM composition, vaccine responses, and immunological outcomes.
Collapse
Affiliation(s)
- Alba Boix-Amorós
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, National Research Council (IATA-CSIC), Valencia, Spain
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
| | - Belinda Van't Land
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Wilhelmina Children's Hospital, University Medical Centre Utrecht, Utrecht, the Netherlands
| | - Anna Calvert
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
| | - Kirsty Le Doare
- Paediatric Infectious Diseases Research Group, St George's University of London, London, United Kingdom
- Imperial College London, London, United Kingdom
- Public Health England, Porton Down, United Kingdom, and the MRC Unit, Fajara, Gambia
| | - Johan Garssen
- Department of Immunology, Danone Nutricia Research, Utrecht, the Netherlands
- Utrecht Institute for Pharmaceutical Sciences, Utrecht University, Utrecht, the Netherlands
| | | | - Ekaterina Khaleva
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- University of Southampton, Southampton, UK
| | - Diego G Peroni
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Clinical and Experimental Medicine, Section of Paediatrics, University of Pisa, Pisa, Italy
| | - Donna T Geddes
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- School of Molecular Sciences, the University of Western Australia, Perth, Australia
| | - Anita L Kozyrskyj
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Department of Pediatrics, Department of Obstetrics & Gynecology, Faculty of Medicine & Dentistry, School of Public Health, University of Alberta, Alberta, Canada
| | - John O Warner
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- National Institute for Health Research, Collaboration for Leadership in Applied Health Research and Care for NW London, London, United Kingdom
| | - Daniel Munblit
- In-VIVO Global Network, an affiliate of the World Universities Network (WUN), New York, New York, United States
- Imperial College London, London, United Kingdom
- Department of Pediatrics, Sechenov University, Moscow, Russia, and the Solov'ev Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
30
|
Zhang Z, Chen X, Zhao J, Tian C, Wei X, Li H, Lin W, Jiang A, Feng R, Yuan J, Zhao X. Effects of a Lactulose-Rich Diet on Fecal Microbiome and Metabolome in Pregnant Mice. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2019; 67:7674-7683. [PMID: 31132256 DOI: 10.1021/acs.jafc.9b01479] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/09/2023]
Abstract
Lactulose, a safe and beneficial molecule, can be used in food as a prebiotic and as an osmotic laxative during pregnancy. This work evaluated the effects of dietary lactulose on the gut microenvironment of pregnant mice using the fecal microbiota and metabolomic profiling. After 2 weeks of feeding, the Bifidobacterium and Bacteroides abundances in the mouse feces were significantly increased in the LAC-high (the diet supplemented with 15% lactulose) group. A total of 15 metabolites, including 1-monoolein, glucose-6-phosphate, and short-chain fatty acids, were increased significantly in the LAC-high group. The serum glucose and total cholesterol concentrations were significantly decreased, while the progesterone level was significantly increased in the lactulose-fed mice. In the LAC-high group, the colonic pH and intestinal permeability were decreased, while the immunoglobulins in the colonic epithelial cells and the small intestinal absorption capacity were significantly increased. These findings indicated that lactulose supplementation benefitted pregnancy performance in mice.
Collapse
Affiliation(s)
- Zheng Zhang
- College of Food Science , South China Agricultural University , Guangzhou , Guangdong 510642 , People's Republic of China
| | - Xiao Chen
- College of Food Science , South China Agricultural University , Guangzhou , Guangdong 510642 , People's Republic of China
| | - Jiangtao Zhao
- Department of Histology and Embryology, School of Basic Medical Sciences , Zhengzhou University , Zhengzhou , Henan 450001 , People's Republic of China
| | - Changyu Tian
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| | - Xiao Wei
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| | - Huan Li
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| | - Weishi Lin
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| | - Aimin Jiang
- College of Food Science , South China Agricultural University , Guangzhou , Guangdong 510642 , People's Republic of China
| | - Ruo Feng
- Department of Histology and Embryology, School of Basic Medical Sciences , Zhengzhou University , Zhengzhou , Henan 450001 , People's Republic of China
| | - Jing Yuan
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| | - Xiangna Zhao
- Institute of Disease Control and Prevention , Chinese People's Liberation Army (PLA) , Beijing 100071 , People's Republic of China
| |
Collapse
|
31
|
Khaleva E, Gridneva Z, Geddes DT, Oddy WH, Colicino S, Blyuss O, Boyle RJ, Warner JO, Munblit D. Transforming growth factor beta in human milk and allergic outcomes in children: A systematic review. Clin Exp Allergy 2019; 49:1201-1213. [PMID: 31058363 DOI: 10.1111/cea.13409] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2019] [Revised: 03/25/2019] [Accepted: 04/20/2019] [Indexed: 12/21/2022]
Abstract
BACKGROUND Human milk (HM) transforming growth factor beta (TGF-β) is critical for inflammation regulation and oral tolerance promotion. Previous reports suggested that variations in HM TGF-β levels are associated with allergic outcomes. OBJECTIVE We undertook a systematic review (PROSPERO 2017 CRD42017069920) to reassess the evidence on the relationships between HM TGF-β and allergic outcomes in children. METHODS Electronic bibliographic databases (MEDLINE, EMBASE and Cochrane Library) were systematically searched. Two independent reviewers screened reference lists, extracted the data and assessed risk of bias using the National Institute for Clinical Excellence methodological checklist. RESULTS A total of 21 studies were identified. Sixteen studies assessed relationships between HM TGF-β and risk of eczema; 14, allergic sensitization; nine, wheezing/asthma; six, food allergy; three, allergic rhinitis/conjunctivitis. Five cohorts (5/18, 28%) reported a protective effect of TGF-β1, while 3 (3/10, 30%) suggested increased risk of allergic outcomes development and 1 (1/10, 10%), a protective effect of TGF-β2 on eczema. Meta-analysis was not possible due to significant heterogeneity in methodology, age of outcome assessment and differing statistical approaches. 71% (15/21) of studies carried a high risk of bias. CONCLUSION AND CLINICAL RELEVANCE In contrast with previous findings, we did not find strong evidence of associations between HM TGF-β and allergic outcomes. Differences in studies' methodology and outcomes do not allow unconditional rejection or acceptance of the hypothesis that HM TGF-β influences the risk of allergy development. Future studies on diverse populations employing standardized methods, accurate phenotyping of outcomes and evaluation of the effect of TGF-β in combination with other HM immune markers, microbiome and oligosaccharides are required.
Collapse
Affiliation(s)
- Ekaterina Khaleva
- University of Southampton, Southampton, UK.,inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey
| | - Zoya Gridneva
- inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey.,School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Donna T Geddes
- inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey.,School of Molecular Sciences, The University of Western Australia, Perth, Western Australia, Australia
| | - Wendy H Oddy
- Nutritional Epidemiology, Menzies Institute for Medical Research, University of Tasmania, Hobart, Tasmania, Australia
| | - Silvia Colicino
- The National Heart and Lung Institute, Imperial College London, London, UK
| | - Oleg Blyuss
- Wolfson Institute of Preventive Medicine, Queen Mary University of London, London, UK.,Department of Applied Mathematics, Lobachevsky State University of Nizhny Novgorod, Nizhny Novgorod, Russia.,Department of Pediatrics, Faculty of Pediatrics, Sechenov University, Moscow, Russia
| | - Robert J Boyle
- inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey.,Department of Paediatrics, Imperial College London, London, UK.,Centre of Evidence-based Dermatology, University of Nottingham, Nottingham, UK
| | - John O Warner
- inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey.,Department of Paediatrics, Imperial College London, London, UK.,Paediatrics and NIHR Collaboration for Leadership in Applied Health Research and Care for NW London, London, UK
| | - Daniel Munblit
- inVIVO Planetary Health, Research Group of the Worldwide Universities Network, West New York, New Jersey.,Department of Pediatrics, Faculty of Pediatrics, Sechenov University, Moscow, Russia.,Department of Paediatrics, Imperial College London, London, UK.,Healthcare Department of Moscow, Moscow Research and Clinical Center for Neuropsychiatry, Moscow, Russia
| |
Collapse
|
32
|
Li L, Han Z, Niu X, Zhang G, Jia Y, Zhang S, He C. Probiotic Supplementation for Prevention of Atopic Dermatitis in Infants and Children: A Systematic Review and Meta-analysis. Am J Clin Dermatol 2019; 20:367-377. [PMID: 30465329 DOI: 10.1007/s40257-018-0404-3] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
BACKGROUND Probiotic supplementation in early life may be effective in preventing atopic dermatitis (AD); however, results regarding efficacy have been controversial. OBJECTIVE The aim of our study was to investigate the effect of probiotic supplementation on the risk of AD. METHODS We systematically searched PubMed, EBSCO, Embase and Web of Science databases up to 8 March 2018 for potentially relevant studies regarding probiotic supplementation and AD. Included infants and children were those with probiotic exposure in utero and/or after birth who were not previously diagnosed with AD. We calculated the odds ratios (ORs) and 95% confidence intervals (CIs) and used the Jadad and Newcastle-Ottawa scales to assess methodologic quality. RESULTS A total of 28 studies met the inclusion criteria. Compared with controls, probiotic treatment was associated with a reduced risk of AD (OR 0.69; 95% CI 0.58-0.82, P < 0.0001). The use of probiotics during both the prenatal and the postnatal period significantly reduced the incidence of AD (OR 0.67; 95% CI 0.54-0.82); however, analysis of studies of probiotics given prenatally only or postnatally only did not reach statistical significance. CONCLUSIONS Our meta-analysis showed that probiotic supplementation during both the prenatal and the postnatal period reduced the incidence of AD in infants and children. Our findings suggest that starting probiotic treatment during gestation and continuing through the first 6 months of the infant's life may be of benefit in the prevention of AD.
Collapse
Affiliation(s)
- Lin Li
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Zhen Han
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Xiaoping Niu
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Guozheng Zhang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Yuliang Jia
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Shunguo Zhang
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China
| | - Chiyi He
- Department of Gastroenterology, Yijishan Hospital of Wannan Medical College, Wuhu, 241001, Anhui, People's Republic of China.
| |
Collapse
|
33
|
Cingi C, Bayar Muluk N, Scadding GK. Will every child have allergic rhinitis soon? Int J Pediatr Otorhinolaryngol 2019; 118:53-58. [PMID: 30580075 DOI: 10.1016/j.ijporl.2018.12.019] [Citation(s) in RCA: 15] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/23/2018] [Revised: 12/13/2018] [Accepted: 12/13/2018] [Indexed: 02/08/2023]
Abstract
OBJECTIVES Given the increasing prevalence of AR amongst children, we aimed to review the literature regarding the future of AR in this population. METHODS We searched the PubMed, Google and Proquest Central databases at Kırıkkale University Library. Search terms used were: "allergic rhinitis", "children", "paediatric", "allergy", "future", "risk factors", "treatment", "pharmacotherapy" and/or "allergen - specific immunotherapy". With regard to risk factors for allergic rhinitis, the terms "Environmental factors", "Improved hygiene", "Increased indoor allergen exposure", "Farms, villages, worms, and other parasites", "Environmental toxicants", "Diet", "Lifestyle changes", "Air pollution" and "Climate factors" were searched for. "Prevention of allergic diseases" and "Allergen-specific immunotherapy in the future" were also included in the search. RESULTS AR has a high prevalence and causes considerable morbidity, has associated comorbidity and features specific complications. The principal treatments rely on avoiding the allergens responsible, and administering drug treatment or immunotherapy, which targets specific antigens. Genetic drift does not explain the rising prevalence of allergic disorders, but multifactorial environmental factors are likely culprits. Amongst such environmental factors to consider are the rise in caesarean births, decreases in breast feeding, dietary changes resulting in less fresh produce being consumed, the eradication of intestinal worm infestations, alterations in the way homes are aired and heated, children taking less exercise and being outdoors for shorter periods, whilst also having more contact with pollution. CONCLUSION Barring substantial lifestyle alterations, more and more children are likely to develop AR. It may prove feasible to stop allergy developing in the first place through manipulation of the microbiome, but the exact format such a modification should involve remains to be discovered. Molecular allergological techniques do offer the prospect of more precisely targeted immunotherapy, the sole disease modifier at present. However, at present the complexity and cost of such interventions prevents their widespread use and research in this area is still needed. The majority of children with AR are going to be managed using nasal saline sprays, since they are the most straightforward and least risky alternative for first line treatment.
Collapse
Affiliation(s)
- Cemal Cingi
- Eskisehir Osmangazi University, Medical Faculty, Department of Otorhinolaryngology, Eskisehir, Turkey.
| | - Nuray Bayar Muluk
- Kirikkale University, Medical Faculty, Department of Otorhinolaryngology, Kirikkale, Turkey.
| | - Glenis K Scadding
- Honorary Consultant Allergist & Rhinologist, RNTNE Hospital, University College Hospitals, London, United Kingdom.
| |
Collapse
|
34
|
Alsharnoubi J, Ishaak M, Elsheikh S, Ezzat S. Transforming Growth Factor Beta-1 in Human Breast Milk and Its Correlation with Infants' Parameters. Breastfeed Med 2019; 14:404-407. [PMID: 30990330 DOI: 10.1089/bfm.2018.0214] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Background: Breastfeeding provides optimal nutrition and health protection for the infant; it contains many anti-inflammatory factors, including transforming growth factor beta-1 (TGF-β1). Our study aimed to measure the level of TGF-β1 in human milk and to find its correlation with some infant anthropometric characteristics. Subjects and Methods: A milk sample was collected from 84 mothers and the level of TGF-β1 was measured using enzyme-linked immunosorbent assay. Results: TGF-β1 was significantly higher in vegetarian mothers compared with nonvegetarian mothers (p = 0.044). Additionally, the mean value of breast milk TGF-β1 was significantly higher in mothers using contraceptive pills compared with those who do not (p = 0.021). Also, the mean value of TGF-β1 was significantly higher in infants 3-6 months than those <3 months (p = 0.010); also there was a significant difference regarding infants' weight and length with average weight and length (p = 0.042) and (p = 0.009), respectively. Conclusions: TGF-β1 in human milk may play a role in infants' growth and development; mothers' diet is known to influence TGF-β1 level and its relation to infants' age and weight. Contraceptive method could have an influence on TGF-β1 levels during breastfeeding.
Collapse
Affiliation(s)
- Jehan Alsharnoubi
- 1 Department of Pediatrics, National Institute of Laser Enhanced Sciences, Cairo University, Cairo, Egypt
| | - Marian Ishaak
- 2 Faculty of Medicine, Cairo University, Cairo, Egypt
| | | | | |
Collapse
|
35
|
Shah R, Newcomb DC. Sex Bias in Asthma Prevalence and Pathogenesis. Front Immunol 2018; 9:2997. [PMID: 30619350 PMCID: PMC6305471 DOI: 10.3389/fimmu.2018.02997] [Citation(s) in RCA: 125] [Impact Index Per Article: 17.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2018] [Accepted: 12/04/2018] [Indexed: 12/24/2022] Open
Abstract
Sex-related differences in asthma prevalence are well established and change through the reproductive phases of life. As children, boys have increased prevalence of asthma compared to girls. However, as adults, women have increased prevalence of asthma compared to men. Many factors, including genetics, environment, immunological responses, and sex hormones, affect the sex disparity associated with the development and control of asthma and other allergic diseases. Fluctuations of hormones during puberty, menstruation, pregnancy, and menopause, alter asthma symptoms and severity. In this article, we review clinical and epidemiological studies that examined the sex disparity in asthma and other allergic diseases as well as the role of sex hormones on asthma pathogenesis.
Collapse
Affiliation(s)
- Ruchi Shah
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States
| | - Dawn C Newcomb
- Department of Medicine, Vanderbilt University Medical Center, Nashville, TN, United States.,Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN, United States
| |
Collapse
|
36
|
Wickens K, Barthow C, Mitchell EA, Kang J, van Zyl N, Purdie G, Stanley T, Fitzharris P, Murphy R, Crane J. Effects of Lactobacillus rhamnosus HN001 in early life on the cumulative prevalence of allergic disease to 11 years. Pediatr Allergy Immunol 2018; 29:808-814. [PMID: 30430649 DOI: 10.1111/pai.12982] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 08/02/2018] [Accepted: 08/09/2018] [Indexed: 12/16/2022]
Abstract
BACKGROUND In a two-centre randomized placebo-controlled trial of Lactobacillus rhamnosus HN001 (HN001) (6 × 109 colony-forming units [cfu]) or Bifidobacterium lactis HN019 (HN019) (9 × 109 cfu) taken daily from 35-week gestation to 6 months' post-partum in mothers while breastfeeding and from birth to age 2 years in infants, we showed that HN001 significantly protected against eczema development at 2, 4 and 6 years and atopic sensitization at 6 years. There was no effect of HN019. We report here the findings for 11 year outcomes. METHODS At age 11 years, eczema was defined as previously using the UK Working Party's Diagnostic Criteria. Asthma, wheeze, hay fever and rhinitis were defined based on the International Study of Asthma and Allergies in Childhood (ISAAC) questions. Atopic sensitization was defined as one or more positive responses (mean wheal diameter ≥3 mm) to a panel of food and aeroallergens. Analysis was intention-to-treat using hazard ratios to assess probiotic effects on the 11-year lifetime prevalence and relative risks for point or 12-month prevalence at 11 years. RESULTS Early childhood HN001 supplementation was associated with significant reductions in the 12-month prevalence of eczema at age 11 years (relative risk [RR] = 0.46, 95% CI 0.25-0.86, P = 0.015) and hay fever (RR = 0.73, 95% CI 0.53-1.00, P = 0.047). For the lifetime prevalence, HN001 was associated with a significant reduction in atopic sensitization (hazard ratio [HR] = 0.71, 95% CI 0.51-1.00, P = 0.048), eczema (HR = 0.58, 95% CI 0.41-0.82, P = 0.002) and wheeze (HR = 0.76, 95% CI 0.57-0.99, P = 0.046). HN019 had no significant effect on these outcomes. CONCLUSION This is the first early probiotic intervention to show positive outcomes for at least the first decade of life across the spectrum of allergic disease.
Collapse
Affiliation(s)
- Kristin Wickens
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Christine Barthow
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Edwin A Mitchell
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
| | - Janice Kang
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Noleen van Zyl
- Department of Paediatrics, Child and Youth Health, The University of Auckland, Auckland, New Zealand
| | - Gordon Purdie
- Dean's Department, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | - Thorsten Stanley
- Department of Paediatrics, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| | | | - Rinki Murphy
- Department of Medicine, University of Auckland, Auckland, New Zealand
| | - Julian Crane
- Wellington Asthma Research Group, Wellington School of Medicine and Health Sciences, University of Otago, Wellington, New Zealand
| |
Collapse
|
37
|
Probiotics SOD inhibited food allergy via downregulation of STAT6-TIM4 signaling on DCs. Mol Immunol 2018; 103:71-77. [DOI: 10.1016/j.molimm.2018.09.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2017] [Revised: 09/03/2018] [Accepted: 09/04/2018] [Indexed: 12/16/2022]
|
38
|
Zhang H, Yeh C, Jin Z, Ding L, Liu BY, Zhang L, Dannelly HK. Prospective study of probiotic supplementation results in immune stimulation and improvement of upper respiratory infection rate. Synth Syst Biotechnol 2018; 3:113-120. [PMID: 29900424 PMCID: PMC5995450 DOI: 10.1016/j.synbio.2018.03.001] [Citation(s) in RCA: 70] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2018] [Revised: 03/04/2018] [Accepted: 03/06/2018] [Indexed: 01/31/2023] Open
Abstract
The human gut microbiota is an important environmental factor for human health with evolutionarily conserved roles in immunity, metabolism, development, and behavior of the host. Probiotic organisms are claimed to offer several functional properties including stimulation of immune system. The purpose of this study is to investigate the effects of a probiotic supplementation on adult volunteers who have contracted the common cold four or more times in the past year. This study is a single center, double-blind, randomized, controlled, prospective trial. Subjects received a probiotic drink containing Lactobacillus paracasei (at least 3 × 107 colony forming units (CFU) ml-1), Lactobacillus casei 431® (at least 3 × 107 CFU ml-1) and Lactobacillus fermentium PCC® (at least 3 × 106 CFU ml-1) or an identical placebo without probiotics for a 12-week study period. The consumption of probiotics significantly reduced the incidence of upper respiratory infection (p < 0.023) and flu-like symptoms with an oral temperature higher than 38 °C (p < 0.034) as compared to the placebo group. Subjects that consumed probiotics demonstrated a significantly higher level of IFN-γ in the serum (p < 0.001) and sIgA in the gut (p < 0.010) as compared to the placebo group and a significant higher level of serum IFN-γ (p < 0.001) and gut sIgA (p < 0.001) as compared to their baseline test results. In contrast, there were no significant differences in the serum IL-4, IL-10, IgA, IgG or IgM between the probiotics and the placebo groups. Results of this study demonstrated that probiotics were safe and effective for fighting the common cold and influenza-like respiratory infections by boosting the immune system.
Collapse
Affiliation(s)
- Hong Zhang
- Beijing Chao-Yang Hospital affiliated to Capital Medical University, 8 Gongti South Road, Chaoyang District, Beijing, 10020, China
| | - Chiajung Yeh
- Hangzhou Weiquan Foods Co., LTD R&D Center, 1688 Wu Zhong Road, Minhang District, Shanghai, 201100, China
| | - Zonglian Jin
- College of Applied Arts and Science of Beijing Union University, 197 Bei Tu Cheng West Road, Haitian District, Beijing, 100108, China
| | - Liwei Ding
- Hangzhou Weiquan Foods Co., LTD R&D Center, 1688 Wu Zhong Road, Minhang District, Shanghai, 201100, China
| | - Bryan Y. Liu
- Indiana State University, Department of Biology, 600 Chestnut Street, Terre Haute, IN 47809, USA
| | - Li Zhang
- SPRIM China, 100 Zunyi Road, Changning District, Shanghai, 200336, China
| | - H. Kathleen Dannelly
- Indiana State University, Department of Biology, 600 Chestnut Street, Terre Haute, IN 47809, USA
| |
Collapse
|
39
|
Malik SS, Saeed A, Baig M, Asif N, Masood N, Yasmin A. Anticarcinogenecity of microbiota and probiotics in breast cancer. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2018. [DOI: 10.1080/10942912.2018.1448994] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Affiliation(s)
- Saima Shakil Malik
- Department of Environmental Sciences, Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
- Surgery Department, Armed Forces Institute of Pathology, Rawalpindi, Pakistan
| | - Ayesha Saeed
- Department of Environmental Sciences, Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| | - Mehreen Baig
- Chemical Pathology and Endocrinology, Fauji Foundation Hospital, Rawalpindi, Pakistan
| | - Naveed Asif
- Surgery Department, Armed Forces Institute of Pathology, Rawalpindi, Pakistan
| | - Nosheen Masood
- Department of Environmental Sciences, Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| | - Azra Yasmin
- Department of Environmental Sciences, Microbiology and Biotechnology Research Lab, Fatima Jinnah Women University, Rawalpindi, Pakistan
| |
Collapse
|
40
|
Wickens K, Barthow C, Mitchell EA, Stanley TV, Purdie G, Rowden J, Kang J, Hood F, van den Elsen L, Forbes-Blom E, Franklin I, Barnes P, Fitzharris P, Maude RM, Stone P, Abels P, Murphy R, Crane J. Maternal supplementation alone with Lactobacillus rhamnosus HN001 during pregnancy and breastfeeding does not reduce infant eczema. Pediatr Allergy Immunol 2018; 29:296-302. [PMID: 29415330 DOI: 10.1111/pai.12874] [Citation(s) in RCA: 47] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 01/30/2018] [Indexed: 12/25/2022]
Abstract
BACKGROUND In a randomized placebo-controlled trial, we previously found that the probiotic Lactobacillus rhamnosus HN001 (HN001) taken by mothers from 35 weeks of gestation until 6 months post-partum if breastfeeding and their child from birth to age 2 years halved the risk of eczema during the first 2 years of life. We aimed to test whether maternal supplementation alone is sufficient to reduce eczema and compare this to our previous study when both the mother and their child were supplemented. METHODS In this 2-centre, parallel double-blind, randomized placebo-controlled trial, the same probiotic as in our previous study (HN001, 6 × 109 colony-forming units) was taken daily by mothers from 14-16 weeks of gestation till 6 months post-partum if breastfeeding, but was not given directly to the child. Women were recruited from the same study population as the first study, where they or their partner had a history of treated asthma, eczema or hay fever. RESULTS Women were randomized to HN001 (N = 212) or placebo (N = 211). Maternal-only HN001 supplementation did not significantly reduce the prevalence of eczema, SCORAD ≥ 10, wheeze or atopic sensitization in the infant by 12 months. This contrasts with the mother and child intervention study, where HN001 was associated with reductions in eczema (hazard ratio (HR): 0.39, 95% CI 0.19-0.79, P = .009) and SCORAD (HR = 0.61, 95% 0.37-1.02). However, differences in the HN001 effect between studies were not significant. HN001 could not be detected in breastmilk from supplemented mothers, and breastmilk TGF-β/IgA profiles were unchanged. CONCLUSION Maternal probiotic supplementation without infant supplementation may not be effective for preventing infant eczema.
Collapse
Affiliation(s)
| | | | | | | | | | - Judy Rowden
- University of Auckland, Auckland, New Zealand
| | - Janice Kang
- University of Otago, Wellington, New Zealand
| | - Fiona Hood
- University of Otago, Wellington, New Zealand
| | | | | | | | | | | | - Robyn M Maude
- Victoria University of Wellington, Wellington, New Zealand
| | - Peter Stone
- University of Auckland, Auckland, New Zealand
| | - Peter Abels
- University of Otago, Wellington, New Zealand
| | | | | |
Collapse
|
41
|
Morita Y, Campos-Alberto E, Yamaide F, Nakano T, Ohnisi H, Kawamoto M, Kawamoto N, Matsui E, Kondo N, Kohno Y, Shimojo N. TGF-β Concentration in Breast Milk is Associated With the Development of Eczema in Infants. Front Pediatr 2018; 6:162. [PMID: 29911097 PMCID: PMC5992274 DOI: 10.3389/fped.2018.00162] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/25/2018] [Accepted: 05/15/2018] [Indexed: 01/28/2023] Open
Abstract
Background: Transforming growth factor (TGF)-β in breast milk is crucial for mucosal immune system in the neonatal period. We hypothesized that the level of exposure to TGF-β from breast milk in the first month of life is related to the development of eczema later in life. Thus, the present study investigated whether changes in TGF-β levels between colostrum and mature milk are associated with such occurrence in a birth cohort study. Methods: Colostrum and 1-month breast milk samples were collected from mothers who participated in our birth cohort study. TGF-β1 and TGF-β2 levels in breast milk were measured using a commercial ELISA kit. The development of eczema in the first 6 months after birth was assessed based on parent's response to a questionnaire. Levels of TGF-β1 and TGF-β2 were compared in breast milk from mothers of infants with and without eczema. Results: In children with eczema, TGF-β1 levels were higher in colostrum, but lower in 1-month milk. A lower TGF-β1 ratio (1-month milk/colostrum) was related to the development of eczema during the first 6 months of life. There was no difference in TGF-β2 ratio (1-month milk/colostrum) between eczema group and control group. Conclusions: Concentration of TGF-β1 but not TGF-β2 in breast milk during the first month after birth may be associated with eczema later in life. Factors that increase TGF-β1 levels in breast milk may play a role in preventing allergic disease.
Collapse
Affiliation(s)
- Yoshinori Morita
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | | | - Fumiya Yamaide
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Taiji Nakano
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Hidenori Ohnisi
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Minako Kawamoto
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Norio Kawamoto
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Eiko Matsui
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Naomi Kondo
- Department of Pediatrics, Graduate School of Medicine, Gifu University, Gifu, Japan
| | - Yoichi Kohno
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Naoki Shimojo
- Department of Pediatrics, Graduate School of Medicine, Chiba University, Chiba, Japan
| |
Collapse
|
42
|
Abstract
Cytokines are required for normal growth and development of the mammary gland and TGF-β prominently represents an established effector of apoptosis, e.g., during involution of the mammary gland. By the control of intracellular signaling pathways, including JAK/STAT, MAPK, PI-3K, and NF-κB, cytokines efficiently regulate cell proliferation and inflammation in the breast. Therefore, cytokines are discussed also in a context of malignant mammary growth. As a group of tissue hormones produced by somatic cells or by cells from the immune system, cytokines are defined by their immunomodulatory potential. Over the past 40 years, multiple cytokines were identified in colostrum and milk. Importantly, cytokines derived from mammary secretions after birth are required for maturation of the immune system in the developing gastrointestinal tract from the suckling. Moreover, recent studies have further assessed the particular interactions between probiotic bacterial strains and cytokines. In light of the increasing prevalence of inflammatory diseases of the gastrointestinal system, the effects of probiotic microorganisms during milk fermentation may have immunotherapeutic potential in the future.
Collapse
Affiliation(s)
- Julia Brenmoehl
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Daniela Ohde
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Elisa Wirthgen
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany
| | - Andreas Hoeflich
- Leibniz Institute for Farm Animal Biology (FBN), Dummerstorf, Germany.
| |
Collapse
|
43
|
Forsberg A, West CE, Prescott SL, Jenmalm MC. Pre- and probiotics for allergy prevention: time to revisit recommendations? Clin Exp Allergy 2017; 46:1506-1521. [PMID: 27770467 DOI: 10.1111/cea.12838] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Reduced intensity and diversity of microbial exposure is considered a major factor driving abnormal postnatal immune maturation and increasing allergy prevalence, particularly in more affluent regions. Quantitatively, the largest important source of early immune-microbial interaction, the gut microbiota, is of particular interest in this context, with variations in composition and diversity in the first months of life associated with subsequent allergy development. Attempting to restore the health consequences of the 'dysbiotic drift' in modern society, interventions modulating gut microbiota for allergy prevention have been evaluated in several randomized placebo-controlled trials. In this review, we provide an overview of these trials and discuss recommendations from international expert bodies regarding prebiotic, probiotic and synbiotic interventions. Recent guidelines from the World Allergy Organization recommend the use of probiotics for the primary prevention of eczema in pregnant and breastfeeding mothers of infants at high risk for developing allergy and in high-risk infants. It is however stressed that these recommendations are conditional, based on very low-quality evidence and great heterogeneity between studies, which also impedes specific and practical advice to consumers on the most effective regimens. We discuss how the choice of probiotic strains, timing and duration of administration can critically influence the outcome due to different effects on immune modulation and gut microbiota composition. Furthermore, we propose strategies to potentially improve allergy-preventive effects and enable future evidence-based implementation.
Collapse
Affiliation(s)
- A Forsberg
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| | - C E West
- International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden.,Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden
| | - S L Prescott
- International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden.,School of Paediatrics and Child Health, University of Western Australia and Princess Margaret Hospital for Children, Perth, WA, Australia
| | - M C Jenmalm
- Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden.,International Inflammation (in-FLAME) network of the World Universities Network, Umeå, Sweden
| |
Collapse
|
44
|
Koleva PT, Tun HM, Konya T, Guttman DS, Becker AB, Mandhane PJ, Turvey SE, Subbarao P, Sears MR, Scott JA, Kozyrskyj AL. Sex-specific impact of asthma during pregnancy on infant gut microbiota. Eur Respir J 2017; 50:50/5/1700280. [PMID: 29167295 PMCID: PMC5898939 DOI: 10.1183/13993003.00280-2017] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 08/16/2017] [Indexed: 12/27/2022]
Abstract
Asthma during pregnancy is associated with retardation of fetal growth in a sex-specific manner. Lactobacilli microbes influence infant growth. This study aimed to determine whether lactobacilli and other microbes are reduced in the gut of infants born to an asthmatic mother, and whether this differs by the sex of the infant. Mother-infant pairs (N=1021) from the Canadian Healthy Infant Longitudinal Development full-term cohort were studied. The abundance of infant faecal microbiota at 3–4 months, profiled by gene sequencing, was compared between both women with and without asthma treatment during pregnancy. Infant sex, maternal ethnicity, pre-pregnancy overweight and atopy status, birth mode, breastfeeding status and intrapartum antibiotic treatment were tested as covariates. Independent of birth mode and other covariates, male, Caucasian infants born to women with prenatal asthma harboured fewer lactobacilli in the gut at 3–4 months of age. If asthmatic mothers had pre-pregnancy overweight, the abundance of Lactobacillus in males was further reduced in the infant gut, whereas the microbiota of female infants was enriched with Bacteroidaceae. Similar differences in infant gut microbial composition according to maternal prenatal asthma status were also more evident among women with food or environmental allergies. Gut lactobacilli were less abundant in male infants, but Bacteroidaceae were more abundant in female infants at 3–4 months of age, following maternal asthma during pregnancy. Gut lactobacilli are less abundant at 3–4 months in male but not female infants following maternal prenatal asthmahttp://ow.ly/jXnl30fU9xH
Collapse
Affiliation(s)
- Petya T Koleva
- Dept of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Hein M Tun
- Dept of Pediatrics, University of Alberta, Edmonton, AB, Canada
| | - Theodore Konya
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | - David S Guttman
- Cell and Systems Biology, University of Toronto, Toronto, ON, Canada
| | - Allan B Becker
- Dept of Pediatrics and Child Health, University of Manitoba, Winnipeg, MB, Canada
| | | | - Stuart E Turvey
- Dept of Pediatrics, University of British Columbia, Vancouver, BC, Canada
| | - Padmaja Subbarao
- Dept of Pediatric Respiratory Medicine, University of Toronto, Toronto, ON, Canada
| | | | - James A Scott
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | |
Collapse
|
45
|
Jennewein MF, Abu-Raya B, Jiang Y, Alter G, Marchant A. Transfer of maternal immunity and programming of the newborn immune system. Semin Immunopathol 2017; 39:605-613. [PMID: 28971246 DOI: 10.1007/s00281-017-0653-x] [Citation(s) in RCA: 87] [Impact Index Per Article: 10.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022]
Abstract
As placental mammals, the pregnant women and the fetus have intense and prolonged interactions during gestation. There is increasing evidence that multiple molecular as well as cellular components originating in pregnant women are transferred to the fetus. The transfer of maternal antibodies has long been recognized as a central component of newborn immunity against pathogens. More recent studies indicate that inflammatory mediators, micronutrients, microbial products and maternal cells are transferred in utero and influence the fetal immune system. Together, these multiple signals are likely to form a complex network of interactions that program the neonatal immune system and tune its homeostatic regulation. Maternal disorders, in particular infectious diseases, modify these signals and may thereby alter immunity in early life. Understanding maternal programming of the newborn immune system could provide a basis for interventions promoting child health.
Collapse
Affiliation(s)
| | - Bahaa Abu-Raya
- Vaccine Evaluation Center, BC Children's Hospital, Department of Pediatrics, Division of Infectious Diseases, University of British Columbia, Vancouver, BC, Canada
| | - Yiwei Jiang
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium
| | - Galit Alter
- Ragon Institute of MGH, MIT and Harvard, Cambridge, MA, 02139, USA
| | - Arnaud Marchant
- Institute for Medical Immunology, Université Libre de Bruxelles, Rue Adrienne Bolland 8, 6041 Gosselies, Charleroi, Belgium.
| |
Collapse
|
46
|
West CE, Dzidic M, Prescott SL, Jenmalm MC. Bugging allergy; role of pre-, pro- and synbiotics in allergy prevention. Allergol Int 2017; 66:529-538. [PMID: 28865967 DOI: 10.1016/j.alit.2017.08.001] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2017] [Revised: 08/02/2017] [Accepted: 08/02/2017] [Indexed: 02/07/2023] Open
Abstract
Large-scale biodiversity loss and complex changes in social behaviors are altering human microbial ecology. This is increasingly implicated in the global rise in inflammatory diseases, most notably the "allergy epidemic" in very early life. Colonization of human ecological niches, particularly the gastrointestinal tract, is critical for normal local and systemic immune development and regulation. Disturbances in composition, diversity and timing of microbial colonization have been associated with increased allergy risk, indicating the importance of strategies to restore a dysbiotic gut microbiota in the primary prevention of allergic diseases, including the administration of probiotics, prebiotics and synbiotics. Here, we summarize and discuss findings of randomized clinical trials that have examined the effects of these microbiome-related strategies on short and long-term allergy preventative effects - including new guidelines from the World Allergy Organization which now recommend probiotics and prebiotics for allergy prevention under certain conditions. The relatively low quality evidence, limited comparative studies and large heterogeneity between studies, have collectively hampered recommendations on specific probiotic strains, specific timing and specific conditions for the most effective preventive management. At the same time the risk of using available products is low. While further research is needed before specific practice guidelines on supplement probiotics and prebiotics, it is equally important that the underlying dietary and lifestyle factors of dysbiosis are addressed at both the individual and societal levels.
Collapse
Affiliation(s)
- Christina E West
- Department of Clinical Sciences, Pediatrics, Umeå University, Umeå, Sweden; inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA.
| | - Majda Dzidic
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden; Institute of Agrochemistry and Food Technology, Spanish National Research Council (IATA-CSIC), Department of Biotechnology, Unit of Lactic Acid Bacteria and Probiotics, Valencia, Spain
| | - Susan L Prescott
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; School of Paediatrics and Child Health, University of Western Australia and Princess Margaret Hospital for Children, Perth, Australia
| | - Maria C Jenmalm
- inFLAME Global Network (Worldwide Universities Network), West New York, NJ, USA; Division of Neuro and Inflammation Sciences, Department of Clinical and Experimental Medicine, Linköping University, Linköping, Sweden
| |
Collapse
|
47
|
Allergy prevention by breastfeeding: possible mechanisms and evidence from human cohorts. Curr Opin Allergy Clin Immunol 2017; 16:427-33. [PMID: 27518839 DOI: 10.1097/aci.0000000000000303] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
PURPOSE OF REVIEW Allergy is a modern disease which does not seem to benefit from breast milk preventive effects. We propose that maternal milk composition has not adapted to the needs of allergy prevention because of the recent and rapid increase of allergy. Modulation of breast milk composition may be the best strategy to counteract allergy development. We will review recent advances in understanding of allergy physiopathology and how breast milk factors may be specifically appropriate to interfere with allergy development in early life. RECENT FINDINGS There is strong evidence both from rodent and human studies that breast milk factors may impact on parameters which are now recognized to be essential for allergy physiopathology: infant gut barrier function, microbiota metabolites production, and oral tolerance induction. Data from human cohorts support the possibility to modify breast milk composition by selected interventions and to impact health outcomes in offspring. SUMMARY Nutritional intervention in lactating mothers should endow breast milk with the capacity to combat allergy epidemics in addition to infectious disease.
Collapse
|
48
|
Human Milk and Allergic Diseases: An Unsolved Puzzle. Nutrients 2017; 9:nu9080894. [PMID: 28817095 PMCID: PMC5579687 DOI: 10.3390/nu9080894] [Citation(s) in RCA: 89] [Impact Index Per Article: 11.1] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 08/01/2017] [Accepted: 08/01/2017] [Indexed: 02/07/2023] Open
Abstract
There is conflicting evidence on the protective role of breastfeeding in relation to the development of allergic sensitisation and allergic disease. Studies vary in methodology and definition of outcomes, which lead to considerable heterogeneity. Human milk composition varies both within and between individuals, which may partially explain conflicting data. It is known that human milk composition is very complex and contains variable levels of immune active molecules, oligosaccharides, metabolites, vitamins and other nutrients and microbial content. Existing evidence suggests that modulation of human breast milk composition has potential for preventing allergic diseases in early life. In this review, we discuss associations between breastfeeding/human milk composition and allergy development.
Collapse
|
49
|
Ruiz L, Espinosa-Martos I, García-Carral C, Manzano S, McGuire MK, Meehan CL, McGuire MA, Williams JE, Foster J, Sellen DW, Kamau-Mbuthia EW, Kamundia EW, Mbugua S, Moore SE, Kvist LJ, Otoo GE, Lackey KA, Flores K, Pareja RG, Bode L, Rodríguez JM. What's Normal? Immune Profiling of Human Milk from Healthy Women Living in Different Geographical and Socioeconomic Settings. Front Immunol 2017; 8:696. [PMID: 28713365 PMCID: PMC5492702 DOI: 10.3389/fimmu.2017.00696] [Citation(s) in RCA: 77] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Accepted: 05/29/2017] [Indexed: 12/31/2022] Open
Abstract
Human milk provides a very wide range of nutrients and bioactive components, including immune factors, human milk oligosaccharides, and a commensal microbiota. These factors are essential for interconnected processes including immunity programming and the development of a normal infant gastrointestinal microbiome. Newborn immune protection mostly relies on maternal immune factors provided through milk. However, studies dealing with an in-depth profiling of the different immune compounds present in human milk and with the assessment of their natural variation in healthy women from different populations are scarce. In this context, the objective of this work was the detection and quantification of a wide array of immune compounds, including innate immunity factors (IL1β, IL6, IL12, INFγ, TNFα), acquired immunity factors (IL2, IL4, IL10, IL13, IL17), chemokines (IL8, Groα, MCP1, MIP1β), growth factors [IL5, IL7, epidermal growth factor (EGF), granulocyte colony-stimulating factor, granulocyte-macrophage colony-stimulating factor, TGFβ2], and immunoglobulins (IgA, IgG, IgM), in milk produced by healthy women of different ethnicities living in different geographic, dietary, socioeconomic, and environmental settings. Among the analyzed factors, IgA, IgG, IgM, EGF, TGFβ2, IL7, IL8, Groα, and MIP1β were detected in all or most of the samples collected in each population and, therefore, this specific set of compounds might be considered as the "core" soluble immune factors in milk produced by healthy women worldwide. This approach may help define which immune factors are (or are not) common in milk produced by women living in various conditions, and to identify host, lifestyle, and environmental factors that affect the immunological composition of this complex biological fluid. Clinical Trial Registration: www.ClinicalTrials.gov, identifier NCT02670278.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Irene Espinosa-Martos
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
- Probisearch S.L., C/Santiago Grisolía, Tres Cantos, Spain
| | - Cristina García-Carral
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Susana Manzano
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Michelle K. McGuire
- School of Biological Sciences, Washington State University, Pullman, WA, United States
- Paul G. Allen School for Global Animal Health, Washington State University, Pullman, WA, United States
| | - Courtney L. Meehan
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | - Mark A. McGuire
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - Janet E. Williams
- Department of Animal and Veterinary Science, University of Idaho, Moscow, ID, United States
| | - James Foster
- Department of Biological Sciences, University of Idaho, Moscow, ID, United States
| | - Daniel W. Sellen
- Dalla Lana School of Public Health, University of Toronto, Toronto, ON, Canada
| | | | | | - Samwel Mbugua
- Department of Human Nutrition, Egerton University, Nakuru, Kenya
| | - Sophie E. Moore
- Division of Women’s Health, King’s College London, London, United Kingdom
- MRC Unit, Serekunda, Gambia
| | | | - Gloria E. Otoo
- Department of Nutrition and Food Science, University of Ghana, Accra, Ghana
| | - Kimberly A. Lackey
- School of Biological Sciences, Washington State University, Pullman, WA, United States
| | - Katherine Flores
- Department of Anthropology, Washington State University, Pullman, WA, United States
| | | | - Lars Bode
- Department of Pediatrics, and Mother Milk Infant Center of Research Excellence (MoMICoRE), University of California, San Diego, La Jolla, CA, United States
| | - Juan M. Rodríguez
- Department of Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
50
|
Ricoldi MST, Furlaneto FAC, Oliveira LFF, Teixeira GC, Pischiotini JP, Moreira ALG, Ervolino E, de Oliveira MN, Bogsan CSB, Salvador SL, Messora MR. Effects of the probiotic Bifidobacterium animalis subsp. lactis on the non-surgical treatment of periodontitis. A histomorphometric, microtomographic and immunohistochemical study in rats. PLoS One 2017; 12:e0179946. [PMID: 28662142 PMCID: PMC5491108 DOI: 10.1371/journal.pone.0179946] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Accepted: 06/07/2017] [Indexed: 12/05/2022] Open
Abstract
Lactobacillus probiotics have been investigated in periodontitis. However, the effects of the genus Bifidobacterium on periodontitis are hardly known. This study evaluated the effects of the probiotic (PROB) Bifidobacterium animalis subsp. lactis (B. lactis) HN019 as an adjunct to scaling and root planing (SRP) in rats with experimental periodontitis (EP). At baseline, 32 rats were assigned to 4 groups: C (control), PROB, EP-SRP and EP-SRP-PROB. In groups EP-SRP and EP-SRP-PROB, the mandibular first molars of the animals received a ligature. At day 14, the ligatures were removed and SRP was performed. Animals of groups PROB and EP-SRP-PROB were orally administered with 10 mL/day of 109 colony forming units of B. lactis HN019 for 15 days, starting at day 14. Animals were euthanized at day 29. Histomorphometric, microtomographic and immunohistochemical analyses were performed. Microbiological effects of B. lactis on biofilm were also evaluated. Data were statistically analyzed (ANOVA, Tukey; Kruskal-Wallis, Dunn’s; Two-tailed t-test; p<0.05). Group EP-SRP-PROB presented reduced alveolar bone resorption and attachment loss when compared with Group EP-SRP (p<0.05). Group EP-SRP-PROB showed significantly fewer osteoclasts, increased expression of anti-inflammatory cytokines and reduced expression of proinflammatory cytokines compared with Group EP-SRP (p<0.05). B. lactis promoted a higher ratio between aerobic and anaerobic bacteria in biofilm samples (p<0.05). B. lactis HN019 may have a role in the treatment of EP in rats, as an adjunct to SRP.
Collapse
Affiliation(s)
- Milla S. T. Ricoldi
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
| | - Flávia A. C. Furlaneto
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
| | - Luiz F. F. Oliveira
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
| | - Gustavo C. Teixeira
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
| | - Jéssica P. Pischiotini
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
| | | | - Edilson Ervolino
- Department of Basic Sciences, Division of Histology, Dental School of Aracatuba, UNESP-Univ Estadual Paulista, Aracatuba/ SP, Brazil
| | - Maricê N. de Oliveira
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo–USP, Sao Paulo/ SP, Brazil
| | - Cristina S. B. Bogsan
- Department of Biochemical and Pharmaceutical Technology, School of Pharmaceutical Sciences, University of Sao Paulo–USP, Sao Paulo/ SP, Brazil
| | - Sérgio L. Salvador
- Department of Clinical Analyses, School of Pharmaceutical Sciences of Ribeirao Preto, University of São Paulo—USP, Ribeirão Preto/ SP, Brazil
| | - Michel R. Messora
- Department of Oral and Maxillofacial Surgery and Periodontology, School of Dentistry of Ribeirao Preto, University of Sao Paulo–USP, Ribeirao Preto/ SP, Brazil
- * E-mail:
| |
Collapse
|