51
|
Tang X, Liu C, Li T, Lin C, Hao Z, Zhang H, Zhao G, Chen Y, Guo A, Hu C. Gambogic acid alleviates inflammation and apoptosis and protects the blood-milk barrier in mastitis induced by LPS. Int Immunopharmacol 2020; 86:106697. [PMID: 32585608 DOI: 10.1016/j.intimp.2020.106697] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Revised: 06/09/2020] [Accepted: 06/10/2020] [Indexed: 11/28/2022]
Abstract
Mastitis is one of the most common diseases among dairy cows. There is still much debate worldwide as to whether antibiotic therapy should be given to dairy cows, or if natural products should be taken as a substitute for antibacterial therapy. As the antibiotic treatment leads to the bacterial resistance and drug residue in milk, introducing natural products for mastitis is becoming a trend. This study investigates the mechanisms of the protective effects of the natural product gambogic acid (GA) in lipopolysaccharide (LPS)-induced mastitis. For in vitro treatments, it was found that GA reduced IL-6, TNF-α, and IL-1β levels by inhibiting the phosphorylation of proteins in the nuclear factor κB (NF-κB) and the mitogen-activated protein kinase (MAPK) pathway. GA also maintained a stable membrane mitochondrial potential and inhibited the overproduction of reactive oxygen species, which protected the cells from apoptosis. On the other hand, in vivo treatments with GA were found to reduce pathological symptoms markedly, and protected the blood-milk barrier from damage induced by LPS. The results demonstrate that GA plays a vital role in suppressing inflammation, alleviating the apoptosis effect, and protecting the blood-milk barrier in mastitis induced by LPS. Thus, these results suggest that the natural product GA plays a potential role in mastitis treatment.
Collapse
Affiliation(s)
- Xin Tang
- Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Chang Liu
- Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Ting Li
- Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Changjie Lin
- Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Zhiyu Hao
- Department of Preventive Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Hui Zhang
- Department of Preventive Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Gang Zhao
- Department of Preventive Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Yingyu Chen
- State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Aizhen Guo
- Department of Preventive Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China; State Key Laboratory of Agricultural Microbiology, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China
| | - Changmin Hu
- Department of Clinical Veterinary Medicine, Faculty of Veterinary Medicine, Huazhong Agricultural University, Wuhan 430070, Hubei Province, China.
| |
Collapse
|
52
|
The Influence of DNA Extraction and Lipid Removal on Human Milk Bacterial Profiles. Methods Protoc 2020; 3:mps3020039. [PMID: 32429170 PMCID: PMC7359716 DOI: 10.3390/mps3020039] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2020] [Revised: 05/05/2020] [Accepted: 05/06/2020] [Indexed: 02/06/2023] Open
Abstract
Culture-independent molecular techniques have advanced the characterization of environmental and human samples including the human milk (HM) bacteriome. However, extraction of high-quality genomic DNA that is representative of the bacterial population in samples is crucial. Lipids removal from HM prior to DNA extraction is common practice, but this may influence the bacterial population detected. The objective of this study was to compare four commercial DNA extraction kits and lipid removal in relation to HM bacterial profiles. Four commercial DNA extraction kits, QIAamp® DNA Microbiome Kit, ZR Fungal/Bacterial DNA MiniPrep™, QIAsymphony DSP DNA Kit and ZymoBIOMICS™ DNA Miniprep Kit, were assessed using milk collected from ten healthy lactating women. The kits were evaluated based on their ability to extract high quantities of pure DNA from HM and how well they extracted DNA from bacterial communities present in a commercial mock microbial community standard spiked into HM. Finally, the kits were evaluated by assessing their extraction repeatability. Bacterial profiles were assessed using Illumina MiSeq sequencing targeting the V4 region of the 16S rRNA gene. The ZR Fungal/Bacterial DNA MiniPrep™ and ZymoBIOMICS™ DNA Miniprep (Zymo Research Corp., Irvine, CA, USA) kits extracted the highest DNA yields with the best purity. DNA extracted using ZR Fungal/Bacterial DNA MiniPrep™ best represented the bacteria in the mock community spiked into HM. In un-spiked HM samples, DNA extracted using the QIAsymphony DSP DNA kit showed statistically significant differences in taxa prevalence from DNA extracted using ZR Fungal/Bacterial DNA MiniPrep™ and ZymoBIOMICS™ DNA Miniprep kits. The only difference between skim and whole milk is observed in bacterial profiles with differing relative abundances of Enhydrobacter and Acinetobacter. DNA extraction, but not lipids removal, substantially influences bacterial profiles detected in HM samples, emphasizing the need for careful selection of a DNA extraction kit to improve DNA recovery from a range of bacterial taxa.
Collapse
|
53
|
Sipe LM, Chaib M, Pingili AK, Pierre JF, Makowski L. Microbiome, bile acids, and obesity: How microbially modified metabolites shape anti-tumor immunity. Immunol Rev 2020; 295:220-239. [PMID: 32320071 PMCID: PMC7841960 DOI: 10.1111/imr.12856] [Citation(s) in RCA: 42] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 03/02/2020] [Indexed: 02/06/2023]
Abstract
Bile acids (BAs) are known facilitators of nutrient absorption but recent paradigm shifts now recognize BAs as signaling molecules regulating both innate and adaptive immunity. Bile acids are synthesized from cholesterol in the liver with subsequent microbial modification and fermentation adding complexity to pool composition. Bile acids act on several receptors such as Farnesoid X Receptor and the G protein-coupled BA receptor 1 (TGR5). Interestingly, BA receptors (BARs) are expressed on immune cells and activation either by BAs or BAR agonists modulates innate and adaptive immune cell populations skewing their polarization toward a more tolerogenic anti-inflammatory phenotype. Intriguingly, recent evidence also suggests that BAs promote anti-tumor immune response through activation and recruitment of tumoricidal immune cells such as natural killer T cells. These exciting findings have redefined BA signaling in health and disease wherein they may suppress inflammation on the one hand, yet promote anti-tumor immunity on the other hand. In this review, we provide our readers with the most recent understanding of the interaction of BAs with the host microbiome, their effect on innate and adaptive immunity in health and disease with a special focus on obesity, bariatric surgery-induced weight loss, and immune checkpoint blockade in cancer.
Collapse
Affiliation(s)
- Laura M. Sipe
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Mehdi Chaib
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Ajeeth K. Pingili
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Joseph F. Pierre
- Department of Pediatrics, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
| | - Liza Makowski
- Division of Hematology and Oncology, Department of Medicine, College of Medicine, University of Tennessee Health Science Center, Memphis, TN, USA
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Tennessee Health Science Center, Memphis, TN, USA
- Center for Cancer Research, University of Tennessee Health Science Center, Memphis, TN, USA
| |
Collapse
|
54
|
Kapourchali FR, Cresci GAM. Early-Life Gut Microbiome-The Importance of Maternal and Infant Factors in Its Establishment. Nutr Clin Pract 2020; 35:386-405. [PMID: 32329544 DOI: 10.1002/ncp.10490] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2019] [Accepted: 03/10/2020] [Indexed: 12/17/2022] Open
Abstract
The early-life microbiome is gaining appreciation as a major influencer in human development and long-term health. Multiple factors are known to influence the initial colonization, development, and function of the neonatal gut microbiome. In addition, alterations in early-life gut microbial composition is associated with several chronic health conditions such as obesity, asthma, and allergies. In this review, we focus on both maternal and infant factors known to influence early-life gut colonization. Also reviewed is the important role of infant feeding, including evidence-based strategies for maternal and infant supplementation with the goal to protect and/or restore the infant gut microbiome.
Collapse
Affiliation(s)
| | - Gail A M Cresci
- Department of Inflammation and Immunity, Cleveland Clinic, Cleveland, Ohio, USA.,Department of Pediatric Gastroenterology, Cleveland Clinic, Cleveland, Ohio, USA.,Center for Human Nutrition, Cleveland Clinic, Cleveland, Ohio, USA
| |
Collapse
|
55
|
Lyons KE, Ryan CA, Dempsey EM, Ross RP, Stanton C. Breast Milk, a Source of Beneficial Microbes and Associated Benefits for Infant Health. Nutrients 2020; 12:E1039. [PMID: 32283875 PMCID: PMC7231147 DOI: 10.3390/nu12041039] [Citation(s) in RCA: 252] [Impact Index Per Article: 63.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2020] [Revised: 04/03/2020] [Accepted: 04/04/2020] [Indexed: 12/27/2022] Open
Abstract
Human breast milk is considered the optimum feeding regime for newborn infants due to its ability to provide complete nutrition and many bioactive health factors. Breast feeding is associated with improved infant health and immune development, less incidences of gastrointestinal disease and lower mortality rates than formula fed infants. As well as providing fundamental nutrients to the growing infant, breast milk is a source of commensal bacteria which further enhance infant health by preventing pathogen adhesion and promoting gut colonisation of beneficial microbes. While breast milk was initially considered a sterile fluid and microbes isolated were considered contaminants, it is now widely accepted that breast milk is home to its own unique microbiome. The origins of bacteria in breast milk have been subject to much debate, however, the possibility of an entero-mammary pathway allowing for transfer of microbes from maternal gut to the mammary gland is one potential pathway. Human milk derived strains can be regarded as potential probiotics; therefore, many studies have focused on isolating strains from milk for subsequent use in infant health and nutrition markets. This review aims to discuss mammary gland development in preparation for lactation as well as explore the microbial composition and origins of the human milk microbiota with a focus on probiotic development.
Collapse
Affiliation(s)
- Katríona E. Lyons
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- School of Microbiology, University College Cork, Cork T12 YN60, Ireland
| | - C. Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
| | - Eugene M. Dempsey
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
- Department of Neonatology, Cork University Maternity Hospital, Cork T12 YE02, Ireland
- INFANT Research Centre, University College Cork, Cork T12 DFK4, Ireland
| | - R. Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| | - Catherine Stanton
- Teagasc Food Research Centre, Moorepark, Fermoy, Co. Cork P61 C996, Ireland
- APC Microbiome Ireland, University College Cork, Cork T12 YT20, Ireland
| |
Collapse
|
56
|
Power ML, Snead C, Reed EG, Schulkin J. Integrating evolution into medical education for women's health care practitioners. Evol Med Public Health 2020; 2020:60-67. [PMID: 32382419 PMCID: PMC7196338 DOI: 10.1093/emph/eoaa009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/27/2020] [Accepted: 03/31/2020] [Indexed: 12/24/2022] Open
Abstract
Evolution is a fundamental principle in biology; however, it has been neglected in medical education. We argue that an evolutionary perspective is especially important for women's health care providers, as selection will act strongly on reproductive parameters, and the biological costs of female reproduction are generally more resource expensive than for men (e.g. due to gestation and lactation) with greater effects on health and wellbeing. An evolutionary perspective is needed to understand antibiotic resistance, disease and health risks associated with mismatches between our evolved adaptations and current conditions, the importance of the microbiome and the maternal role in how infants acquire and develop their early-life microbiome (vaginal birth, lactation), and the importance of breastmilk as a biochemical signal from mothers to their babies. We present data that obstetrician-gynecologists' views regarding the inclusion of evolution within their training is generally positive, but many barriers are perceived. Requiring coursework in evolutionary biology with an emphasis on evolutionary medicine prior to enrollment in medical school may be a solution.
Collapse
Affiliation(s)
- Michael L Power
- Smithsonian National Zoological Park and Conservation Biology Institute, Washington, DC 20013-7012, USA
- American College of Obstetricians and Gynecologists, Washington, DC 20024-2188, USA
| | - Carrie Snead
- American College of Obstetricians and Gynecologists, Washington, DC 20024-2188, USA
| | - Eda G Reed
- Smithsonian National Zoological Park and Conservation Biology Institute, Washington, DC 20013-7012, USA
- Johns Hopkins University, Baltimore, MD 21205, USA
| | - Jay Schulkin
- Obstetrics & Gynecology, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
57
|
Demmelmair H, Jiménez E, Collado MC, Salminen S, McGuire MK. Maternal and Perinatal Factors Associated with the Human Milk Microbiome. Curr Dev Nutr 2020; 4:nzaa027. [PMID: 32270132 PMCID: PMC7127925 DOI: 10.1093/cdn/nzaa027] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2019] [Revised: 02/12/2020] [Accepted: 02/26/2020] [Indexed: 12/13/2022] Open
Abstract
Microbes are present in human milk regardless of the mother's health. The origins of the milk microbiota likely include the mother's skin, infant's mouth, and transfer from the maternal gastrointestinal (GI) tract. Prominent bacterial taxa in human milk are Staphylococcus and Streptococcus, but many other genera are also found including anaerobic Lactobacillus, Bifidobacterium, and Bacteroides. The milk microbiome is highly variable and potentially influenced by geographic location, delivery mode, time postpartum, feeding mode, social networks, environment, maternal diet, and milk composition. Mastitis alters the milk microbiome, and the intake of Lactobacilli has shown potential for mastitis treatment and prevention. Although milk and infant fecal microbiomes are different, their variations appear to be related - suggesting that milk is an important contributor of early GI colonization. Nonetheless, nothing is known regarding whether the milk microbiome influences infant health. Further research and clinical interventions are needed to determine if changes in the microbiomes of human milk and infant formula/food impact health.
Collapse
Affiliation(s)
- Hans Demmelmair
- Dr. von Hauner Children´s Hospital, University of Munich Medical Center, Munich, Germany
| | - Esther Jiménez
- ProbiSearch SLU, Madrid, Spain
- Department of Nutrition, Food Science, and Technology, University Complutense, Madrid, Spain
| | - Maria Carmen Collado
- Institute of Agrochemistry and Food Technology, Spanish National Research Council, Valencia, Spain
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Seppo Salminen
- Functional Foods Forum, University of Turku, Turku, Finland
| | - Michelle K McGuire
- Margaret Ritchie School of Family and Consumer Sciences, University of Idaho, Moscow, ID, USA
| |
Collapse
|
58
|
López-Moreno A, Aguilera M. Probiotics Dietary Supplementation for Modulating Endocrine and Fertility Microbiota Dysbiosis. Nutrients 2020; 12:E757. [PMID: 32182980 PMCID: PMC7146451 DOI: 10.3390/nu12030757] [Citation(s) in RCA: 34] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2020] [Revised: 03/06/2020] [Accepted: 03/09/2020] [Indexed: 12/15/2022] Open
Abstract
Human microbiota seems to play a key role in endocrine and reproductive systems. Fortunately, microbiota reproductive dysbiosis start to be treated by probiotics using typical species from genus Lactobacillus. This work presents the compiled and analysed results from the most up-to-date information from clinical trials regarding microbiota, fertility, probiotics and oral route administration, reviewing open access scientific documents. These studies analyse the clinical impact of probiotics administered on several endocrine disorders' manifestations in women: mastitis; vaginal dysbiosis; pregnancy complication disorders; and polycystic ovary syndrome. In all cases, the clinical modulation achieved by probiotics was evaluated positively through the improvement of specific disease outcomes with the exception of the pregnancy disorders studies, where the sample sizes results were statistically insufficient. High amounts of studies were discarded because no data were provided on specific probiotic strains, doses, impact on the individual autochthon microbiota, or data regarding specific hormonal values modifications and endocrine regulation effects. However, most of the selected studies with probiotics contained no protocolised administration. Therefore, we consider that intervention studies with probiotics might allocate the focus, not only in obtaining a final outcome, but in how to personalise the administration according to the disorder to be palliated.
Collapse
Affiliation(s)
- Ana López-Moreno
- Department of Microbiology, Faculty of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, 18016 Armilla, Granada, Spain
| | - Margarita Aguilera
- Department of Microbiology, Faculty of Pharmacy, University of Granada, Campus of Cartuja, 18071 Granada, Spain
- Institute of Nutrition and Food Technology “José Mataix”, Center of Biomedical Research, University of Granada, 18016 Armilla, Granada, Spain
- IBS: Instituto de Investigación Biosanitaria ibs., 18012 Granada, Spain
| |
Collapse
|
59
|
Barker M, Adelson P, Peters MDJ, Steen M. Probiotics and human lactational mastitis: A scoping review. Women Birth 2020; 33:e483-e491. [PMID: 32146088 DOI: 10.1016/j.wombi.2020.01.001] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2019] [Revised: 01/06/2020] [Accepted: 01/06/2020] [Indexed: 12/15/2022]
Abstract
PROBLEM Lactational mastitis is a common condition amongst breastfeeding women. It is associated with decreased breastfeeding rates and often treated with antibiotics. BACKGROUND The anti-inflammatory effects of probiotics have been identified as a potential treatment or prevention strategy for lactational mastitis leading to increased commercial and public interest. Despite the marketing of probiotics to women, evidence is still emerging as to its efficacy. AIM/METHODS This scoping review followed the Preferred Reporting Items for Systematic Reviews and Meta-Analyses extension for Scoping Reviews (PRISMA-ScR) to identify and examine the evidence around probiotic consumption and lactational mastitis. The review addressed the question; what is the evidence regarding probiotic consumption and human lactational mastitis? Studies were critically appraised using the Joanna Briggs Institute checklist for randomised control trials (RCTs). FINDINGS Five RCTs met the inclusion criteria; three concerned probiotic consumption for the treatment of mastitis, two for the prevention of mastitis. All reported a lower incidence of mastitis in the probiotic groups. DISCUSSION Although potentially promising results were reported across all studies there were significant methodological limitations concerning; appropriately described baseline characteristics, study hypotheses, lack of power calculations, definitional issues, and potential conflicts of interest. CONCLUSION Probiotics may have utility for the treatment or prevention of lactational mastitis. However only a few studies with significant limitations have been published to date. Well designed and conducted studies are needed before evidence-based recommendations can be made for use of probiotics in the treatment or prevention of lactational mastitis.
Collapse
Affiliation(s)
- Melissa Barker
- Rosemary Bryant AO Research Centre, Division of Health Sciences, University of South Australia, Adelaide, South Australia; The Women's and Children's Hospital, North Adelaide, Australia; Clinical and Health Sciences, University of South Australia, Adelaide, South Australia.
| | - Pamela Adelson
- Rosemary Bryant AO Research Centre, Division of Health Sciences, University of South Australia, Adelaide, South Australia
| | - Micah D J Peters
- Rosemary Bryant AO Research Centre, Division of Health Sciences, University of South Australia, Adelaide, South Australia; Adelaide Nursing School, Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia; The Centre for Evidence-based Practice South Australia (CEPSA): a Joanna Briggs Institute Centre of Excellence, Adelaide, Australia
| | - Mary Steen
- Rosemary Bryant AO Research Centre, Division of Health Sciences, University of South Australia, Adelaide, South Australia; The Women's and Children's Hospital, North Adelaide, Australia; Clinical and Health Sciences, University of South Australia, Adelaide, South Australia
| |
Collapse
|
60
|
Basım P, Özdenkaya Y. Can Traditional Fermented Food Products Protect Mothers Against Lactational Mastitis. Breastfeed Med 2020; 15:163-169. [PMID: 31977239 DOI: 10.1089/bfm.2019.0261] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Background/Objective: Various dietary factors, including the daily food intake during pregnancy and lactation can play a role in the development of lactational mastitis (LM). To investigate the effect of the most commonly consumed fermented foods (FF) during pregnancy and lactation in Turkey and other factors described in the literature regarding childbirth and breastfeeding on the development of LM. Materials and Methods: The study included 607 volunteers in the lactational period, of whom 303 had LM and 304 had no mastitis event. The data on sociodemographics, childbirth, and breastfeeding, and the consumption frequency of six types of FF were collected through a face-to-face questionnaire. Results: The variables significantly and independently associated with LM were birth week (odds ratio [OR] = 80.83, 95% confidence interval [CI]: 1.12-1.94), birth weight (OR = 0.63, CI: 1.24-1.79), time to breastfeeding after birth (OR = 0.79, CI: 1.62-2.31), breastfeeding length (OR = 0.12, CI: 2.25-2.78), breast preference (OR = 0.13, CI: 2.83-3.42), use of cream on nipples (OR = 0.81, CI: 1.31-1.97), use of oral probiotics (OR = 0.29, CI: 1.86-2.92), and receiving breastfeeding education from an expert (OR = 0.22, CI: 1.22-2.34). According to the multinominal logistic regression analysis, the daily consumption of kefir (OR = 0.69, CI: 1.18-2.22), homemade yogurt (OR = 0.78, CI: 1.14-1.87), conventional yogurt (OR = 0.81, CI: 1.24-2.46), boza (OR = 0.79, CI: 2.19-2.99), tarhana (OR = 0.52, CI: 2.47-2.81), and pickles (OR = 0.22, CI: 1.22-2.34) significantly reduced the risk of LM development. The diversity of consumed FF was also found to be protective against LM (OR = 0.34, CI: 1.34-2.35). Conclusion: Kefir, homemade and conventional yogurt, boza, tarhana, and pickles can protect breastfeeding mothers and also reduce the risk of LM development.
Collapse
Affiliation(s)
- Pelin Basım
- Department of General Surgery, Medipol University, Istanbul, Turkey
| | - Yaşar Özdenkaya
- Department of General Surgery, Medipol University, Istanbul, Turkey
| |
Collapse
|
61
|
Liu W, Chen M, Duo L, Wang J, Guo S, Sun H, Menghe B, Zhang H. Characterization of potentially probiotic lactic acid bacteria and bifidobacteria isolated from human colostrum. J Dairy Sci 2020; 103:4013-4025. [PMID: 32113772 DOI: 10.3168/jds.2019-17602] [Citation(s) in RCA: 69] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Accepted: 12/26/2019] [Indexed: 01/02/2023]
Abstract
Breast milk is the main source of nutrition for infants; it contains considerable microflora that can be transmitted to the infant endogenously or by breastfeeding, and it plays an important role in the maturation and development of the immune system. In this study, we isolated and identified lactic acid bacteria (LAB) from human colostrum, and screened 2 strains with probiotic potential. The LAB isolated from 40 human colostrum samples belonged to 5 genera: Lactobacillus, Bifidobacterium, Streptococcus, Enterococcus, and Staphylococcus. We also isolated Propionibacterium and Actinomyces. We identified a total of 197 strains of LAB derived from human colostrum based on their morphology and 16S rRNA sequence, among them 8 strains of Bifidobacterium and 10 strains of Lactobacillus, including 3 Bifidobacterium species and 4 Lactobacillus species. The physiological and biochemical characteristics of strains with good probiotic characteristics were evaluated. The tolerances of some of the Bifidobacterium and Lactobacillus strains to gastrointestinal fluid and bile salts were evaluated in vitro, using the probiotic strains Bifidobacterium lactis BB12 and Lactobacillus rhamnosus GG as controls. Among them, B. lactis Probio-M8 and L. rhamnosus Probio-M9 showed survival rates of 97.25 and 78.33% after digestion for 11 h in artificial gastrointestinal juice, and they exhibited growth delays of 0.95 and 1.87 h, respectively, in 0.3% bile salts. These two strains have the potential for application as probiotics and will facilitate functional studies of probiotics in breast milk and the development of human milk-derived probiotics.
Collapse
Affiliation(s)
- Wenjun Liu
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Meixuan Chen
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Lana Duo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Jicheng Wang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Shuai Guo
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Haotian Sun
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China
| | - Bilige Menghe
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| | - Heping Zhang
- Key Laboratory of Dairy Biotechnology and Engineering, Ministry of Education of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Key Laboratory of Dairy Products Processing, Ministry of Agriculture and Rural Affairs of China, Inner Mongolia Agricultural University, Hohhot, 010018, China; Inner Mongolia Key Laboratory of Dairy Biotechnology and Engineering, Inner Mongolia Agricultural University, Hohhot, 010018, China.
| |
Collapse
|
62
|
Azagra-Boronat I, Massot-Cladera M, Knipping K, Garssen J, Ben Amor K, Knol J, Franch À, Castell M, Rodríguez-Lagunas MJ, Pérez-Cano FJ. Strain-Specific Probiotic Properties of Bifidobacteria and Lactobacilli for the Prevention of Diarrhea Caused by Rotavirus in a Preclinical Model. Nutrients 2020; 12:nu12020498. [PMID: 32075234 PMCID: PMC7071190 DOI: 10.3390/nu12020498] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2020] [Revised: 02/10/2020] [Accepted: 02/13/2020] [Indexed: 12/20/2022] Open
Abstract
Probiotic supplementation with different lactobacilli and bifidobacterial strains has demonstrated beneficial effects in infectious diarrhea caused by rotavirus (RV) in young children. Preclinical models of RV infection might be a good strategy to screen for the efficacy of new probiotic strains or to test their comparative efficacy. Neonatal Lewis rats were supplemented with Bifidobacterium breve M-16V, Lactobacillus acidophilus NCFM, Lactobacillus helveticus R0052, or Lactobacillus salivarius PS2 from days 2–14 of life. On day five, animals received RV SA-11 orally. Fecal samples were collected daily, weighed, and scored for the calculation of severity and incidence of diarrhea. In addition, fecal pH and fecal viral shedding were measured. Animals were sacrificed at the end of the study and their blood was obtained for the quantification of RV-specific immunoglobulins. RV infection was induced in ~90% of the animals. All probiotics caused a reduction of several clinical variables of severity and incidence of diarrhea, except L. salivarius PS2. L. acidophilus NCFM, B. breve M-16V, and L. helveticus R0052 seemed to be very effective probiotic strains. In addition, all Lactobacillus strains reduced the viral elimination one day post-inoculation. No differences were detected in the specific anti-RV humoral response. The present study highlights the strain-specific effects of probiotics and identifies promising probiotics for use in ameliorating and preventing RV-induced diarrhea in children, for example by including them in infant formulas.
Collapse
Affiliation(s)
- Ignasi Azagra-Boronat
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Malén Massot-Cladera
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Karen Knipping
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (K.K.); (J.G.); (K.B.A.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Johan Garssen
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (K.K.); (J.G.); (K.B.A.); (J.K.)
- Division of Pharmacology, Utrecht Institute for Pharmaceutical Sciences, Faculty of Science, Utrecht University, 3584 CA Utrecht, The Netherlands
| | - Kaouther Ben Amor
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (K.K.); (J.G.); (K.B.A.); (J.K.)
| | - Jan Knol
- Danone Nutricia Research, 3584 CT Utrecht, The Netherlands; (K.K.); (J.G.); (K.B.A.); (J.K.)
- Laboratory of Microbiology, Wageningen University, 6708 WE Wageningen, The Netherlands
| | - Àngels Franch
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - Margarida Castell
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| | - María J. Rodríguez-Lagunas
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
- Correspondence: ; Tel.: +34-934-024-505
| | - Francisco J. Pérez-Cano
- Physiology Section, Department of Biochemistry and Physiology, Faculty of Pharmacy and Food Science, University of Barcelona (UB), 08028 Barcelona, Spain; (I.A.-B.); (M.M.-C.); (À.F.); (M.C.); (F.J.P.-C.)
- Nutrition and Food Safety Research Institute (INSA-UB), 08921 Santa Coloma de Gramenet, Spain
| |
Collapse
|
63
|
Lagier JC, Raoult D. Faecal microbiota transplantations and urinary tract infections. Lancet 2020; 395:270-271. [PMID: 31982066 DOI: 10.1016/s0140-6736(19)32992-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/06/2019] [Accepted: 11/08/2019] [Indexed: 01/17/2023]
Affiliation(s)
- Jean Christophe Lagier
- Aix Marseille University, Institut de Recherche pour le Développement, Microbes Evolution Phylogeny and Infection, Assistance Publique Hôpitaux de Marseille, Institut Hospitalo Universitaire Méditerranée Infection, Marseille 13005, France
| | - Didier Raoult
- Aix Marseille University, Institut de Recherche pour le Développement, Microbes Evolution Phylogeny and Infection, Assistance Publique Hôpitaux de Marseille, Institut Hospitalo Universitaire Méditerranée Infection, Marseille 13005, France.
| |
Collapse
|
64
|
Walker M. Novel Innovations and Recent Findings in Lactation Support. CLINICAL LACTATION 2019. [DOI: 10.1891/2158-0782.10.4.175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
RationaleStaying current with the burgeoning amount of breastfeeding research can be a daunting task. It is incumbent upon the IBCLC and other lactation care providers to be cognizant of new research and innovations in lactation support.MethodRecent products and findings regarding neonatal jaundice, hypoglycemia, mastitis, and therapeutic taping are presented.FindingsNew research findings, techniques, and products have been gathered to augment the provider's knowledge and skill set regarding a broad range of items relevant to clinical lactation practice. Most are affordable options for practitioners and parents, and represent safe and effective options for screening and treatment.
Collapse
|
65
|
From the Table to the Tumor: The Role of Mediterranean and Western Dietary Patterns in Shifting Microbial-Mediated Signaling to Impact Breast Cancer Risk. Nutrients 2019; 11:nu11112565. [PMID: 31652909 PMCID: PMC6893457 DOI: 10.3390/nu11112565] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2019] [Revised: 10/14/2019] [Accepted: 10/15/2019] [Indexed: 12/16/2022] Open
Abstract
Diet is a modifiable component of lifestyle that could influence breast cancer development. The Mediterranean dietary pattern is considered one of the healthiest of all dietary patterns. Adherence to the Mediterranean diet protects against diabetes, cardiovascular disease, and cancer. Reported consumption of a Mediterranean diet pattern was associated with lower breast cancer risk for women with all subtypes of breast cancer, and a Western diet pattern was associated with greater risk. In this review, we contrast the available epidemiological breast cancer data, comparing the impact of consuming a Mediterranean diet to the Western diet. Furthermore, we will review the preclinical data highlighting the anticancer molecular mechanism of Mediterranean diet consumption in both cancer prevention and therapeutic outcomes. Diet composition is a major constituent shaping the gut microbiome. Distinct patterns of gut microbiota composition are associated with the habitual consumption of animal fats, high-fiber diets, and vegetable-based diets. We will review the impact of Mediterranean diet on the gut microbiome and inflammation. Outside of the gut, we recently demonstrated that Mediterranean diet consumption led to distinct microbiota shifts in the mammary gland tissue, suggesting possible anticancer effects by diet on breast-specific microbiome. Taken together, these data support the anti-breast-cancer impact of Mediterranean diet consumption.
Collapse
|
66
|
Maternal probiotic milk intake during pregnancy and breastfeeding complications in the Norwegian Mother and Child Cohort Study. Eur J Nutr 2019; 59:2219-2228. [PMID: 31506766 PMCID: PMC7351866 DOI: 10.1007/s00394-019-02072-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2019] [Accepted: 07/27/2019] [Indexed: 11/11/2022]
Abstract
Purpose During the time of breastfeeding, a third of all women contract (or: fall ill in) mastitis—the leading cause of precocious weaning. Recent studies indicate that probiotics intake may prevent mastitis by altering the breast’s bacterial flora. The aim of this study was to examine whether probiotic milk intake during pregnancy is associated with less breastfeeding complications and longer breastfeeding duration. Methods This study included 57,134 women, with live singleton term births, participating in the Norwegian Mother and Child Cohort Study. Probiotic milk intake during the first half of pregnancy was self-reported in a validated food frequency questionnaire at gestational week 22. At 6 month postpartum, women reported complications, including mastitis, and duration and exclusivity of breastfeeding. The association between probiotic milk intake and breastfeeding complications and duration was studied by adjusted logistic regression models. Results Probiotic milk intake was associated with increased risk for mastitis [adjusted odds ratio (aOR) 1.09, 95% confidence interval (CI) 1.02–1.16] and for any breastfeeding problems during the first month (aOR 1.19, 95% CI 1.10–1.21). However, cessation of predominant (aOR 0.95, 95% CI 0.91–0.96) or any (aOR 0.79, 95% CI 0.75–0.84) breastfeeding earlier than at 4 months was less frequent in probiotic milk consumers than in non-consumers. Conclusions Even though probiotic milk intake during the first half of pregnancy was statistically associated with increased risk for breastfeeding complications, including mastitis, the association is probably not causal. Probiotics intake was namely associated with longer breastfeeding duration and there was indication of socioeconomic confounding. Further studies, i.e., large randomized-controlled trials, are needed to understand the association between probiotic intake and breastfeeding complications. Electronic supplementary material The online version of this article (10.1007/s00394-019-02072-8) contains supplementary material, which is available to authorized users.
Collapse
|
67
|
Sakwinska O, Bosco N. Host Microbe Interactions in the Lactating Mammary Gland. Front Microbiol 2019; 10:1863. [PMID: 31456777 PMCID: PMC6701204 DOI: 10.3389/fmicb.2019.01863] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2019] [Accepted: 07/29/2019] [Indexed: 12/13/2022] Open
Abstract
The bacteria present in human milk constitute the human milk microbiome (hMM). Both the older culture-based work and the more recent studies using molecular detection of bacterial DNA have reached similar conclusions: the hMM mostly consists of commensal staphylococci such as Staphylococcus epidermidis, and streptococci. The prevalence of other bacterial groups such lactobacilli varies widely, while the abundance and prevalence of bifidobacteria is generally low. Recently, the hMM became accepted as a part of a physiologically normal state with suggested potential health benefits. Most research on the hMM has focused on its composition and potential effect on the breastfed infant. A major role as a microbiome inoculum for the infant gut has been proposed, but remains to be clearly demonstrated. Herein, we also discuss the emerging connection between the hMM and mammary gland physiology and lactation. Similarities between the mammary gland and mucosal interfaces are considerable, and in particular mucosal-like immune attributes of mammary gland. The potential role of hMM-host interactions in the mammary gland in maternal health is explored with a primary focus on lactational mastitis.
Collapse
Affiliation(s)
- Olga Sakwinska
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
| | - Nabil Bosco
- Nestlé Research, Nestlé Institute of Health Sciences, Lausanne, Switzerland
- Nestlé Research Singapore Hub, Singapore, Singapore
| |
Collapse
|
68
|
Mills S, Lane JA, Smith GJ, Grimaldi KA, Ross RP, Stanton C. Precision Nutrition and the Microbiome Part II: Potential Opportunities and Pathways to Commercialisation. Nutrients 2019; 11:E1468. [PMID: 31252674 PMCID: PMC6683087 DOI: 10.3390/nu11071468] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2019] [Revised: 06/05/2019] [Accepted: 06/05/2019] [Indexed: 12/11/2022] Open
Abstract
Modulation of the human gut microbiota through probiotics, prebiotics and dietary fibre are recognised strategies to improve health and prevent disease. Yet we are only beginning to understand the impact of these interventions on the gut microbiota and the physiological consequences for the human host, thus forging the way towards evidence-based scientific validation. However, in many studies a percentage of participants can be defined as 'non-responders' and scientists are beginning to unravel what differentiates these from 'responders;' and it is now clear that an individual's baseline microbiota can influence an individual's response. Thus, microbiome composition can potentially serve as a biomarker to predict responsiveness to interventions, diets and dietary components enabling greater opportunities for its use towards disease prevention and health promotion. In Part I of this two-part review, we reviewed the current state of the science in terms of the gut microbiota and the role of diet and dietary components in shaping it and subsequent consequences for human health. In Part II, we examine the efficacy of gut-microbiota modulating therapies at different life stages and their potential to aid in the management of undernutrition and overnutrition. Given the significance of an individual's gut microbiota, we investigate the feasibility of microbiome testing and we discuss guidelines for evaluating the scientific validity of evidence for providing personalised microbiome-based dietary advice. Overall, this review highlights the potential value of the microbiome to prevent disease and maintain or promote health and in doing so, paves the pathway towards commercialisation.
Collapse
Affiliation(s)
- Susan Mills
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Jonathan A Lane
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | - Graeme J Smith
- H&H Group, Technical Centre, Global Research and Technology Centre, Cork P61 C996, Ireland.
| | | | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork T12 K8AF, Ireland.
| | - Catherine Stanton
- APC Microbiome Ireland, Teagasc Food Research Centre, Fermoy P61 C996, Co Cork, Ireland.
| |
Collapse
|
69
|
Ruiz L, García-Carral C, Rodriguez JM. Unfolding the Human Milk Microbiome Landscape in the Omics Era. Front Microbiol 2019; 10:1378. [PMID: 31293535 PMCID: PMC6604669 DOI: 10.3389/fmicb.2019.01378] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Accepted: 06/03/2019] [Indexed: 12/31/2022] Open
Abstract
Studies conducted in the last years have demonstrated that human milk represents a continuous supply of beneficial bacteria to the infant gut, which contribute to the maturation of the digestive and immune functions in the developing infant. Nevertheless, the origin of bacterial populations in milk is not fully understood yet and they have been proposed to originate from maternal skin, infant’s mouth, and (or) endogenously, from the maternal digestive tract through a mechanism involving immune cells. Understanding the composition, functions and assembly of the human milk microbiota has important implications not only for the infant gut microbiota establishment, but also for the mammary health since dysbiosis in the milk bacteria may lead to mastitis. Besides, host, microbial, medical and environmental factors may affect the composition of the human milk microbiome, with implications for the mother-infant health. Application of both culture-dependent and -independent techniques to assess the milk microbiome faces some practical limitations but, together, have allowed providing novel and complementary views on its origin, composition and functioning as summarized in this minireview. In the next future, the application of the ultimate advances in next-generation sequencing and omics approaches, including culturomics, will allow a detailed and comprehensive understanding of the composition and functions of these microbial communities, including their interactions with other milk components, expanding the opportunities to design novel microbiome-based modulation strategies for this ecosystem.
Collapse
Affiliation(s)
- Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias (IPLA-CSIC), Villaviciosa, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| | - Juan Miguel Rodriguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
70
|
Ruiz L, Bacigalupe R, García-Carral C, Boix-Amoros A, Argüello H, Silva CB, de Los Angeles Checa M, Mira A, Rodríguez JM. Microbiota of human precolostrum and its potential role as a source of bacteria to the infant mouth. Sci Rep 2019; 9:8435. [PMID: 31182726 PMCID: PMC6557856 DOI: 10.1038/s41598-019-42514-1] [Citation(s) in RCA: 44] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2018] [Accepted: 04/02/2019] [Indexed: 12/21/2022] Open
Abstract
Human milk represents a source of bacteria for the initial establishment of the oral (and gut) microbiomes in the breastfed infant, however, the origin of bacteria in human milk remains largely unknown. While some evidence points towards a possible endogenous enteromammary route, other authors have suggested that bacteria in human milk are contaminants from the skin or the breastfed infant mouth. In this work 16S rRNA sequencing and bacterial culturing and isolation was performed to analyze the microbiota on maternal precolostrum samples, collected from pregnant women before delivery, and on oral samples collected from the corresponding infants. The structure of both ecosystems demonstrated a high proportion of taxa consistently shared among ecosystems, Streptococcus spp. and Staphylococcus spp. being the most abundant. Whole genome sequencing on those isolates that, belonging to the same species, were isolated from both the maternal and infant samples in the same mother-infant pair, evidenced that in 8 out of 10 pairs both isolates were >99.9% identical at nucleotide level. The presence of typical oral bacteria in precolostrum before contact with the newborn indicates that they are not a contamination from the infant, and suggests that at least some oral bacteria reach the infant's mouth through breastfeeding.
Collapse
Affiliation(s)
- Lorena Ruiz
- IPLA-CSIC, Department of Microbiology and Biochemistry of Dairy Products, Institute of Dairy Products of Asturias, Villaviciosa, Spain. .,Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.
| | - Rodrigo Bacigalupe
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain
| | - Cristina García-Carral
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.,Probisearch S.L., C/Santiago Grisolía, 2, 28760, Tres Cantos, Spain
| | - Alba Boix-Amoros
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain
| | - Héctor Argüello
- Grupo de Genómica y Mejora Animal, Departamento de Genética, Facultad de Veterinaria, Universidad de Córdoba, Córdoba, Spain
| | - Camilla Beatriz Silva
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.,Universidade de Uberaba, Uberaba, Brazil
| | | | - Alex Mira
- Centro Superior de Investigación en Salud Pública, Fundación FISABIO, Valencia, Spain.
| | - Juan M Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Avda. Puerta de Hierro, s/n, 28040, Madrid, Spain.
| |
Collapse
|
71
|
Targeting gut microbiota as a possible therapy for mastitis. Eur J Clin Microbiol Infect Dis 2019; 38:1409-1423. [PMID: 31079312 DOI: 10.1007/s10096-019-03549-4] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Accepted: 03/25/2019] [Indexed: 02/07/2023]
Abstract
Mastitis, a disease that affects both dairy herds and humans, is recognized as the most common source of losses in the dairy industry. Antibiotics have been used for years as the primary treatment for mastitis. However, abuse of antibiotics has led to the emergence of resistant strains and the presence of drug residues and has increased the difficulty of curing this disease. In addition, antibiotics kill most of the microbes that are present in the digestive tract, leading to imbalances in the gut microbiome and destruction of the ecosystem that is normally present in the gut. Gut microbiota play an important role in the host's health and could be considered the "second brain" of the body. In recent years, the gut microbiota and their metabolites, including lipopolysaccharide (LPS) and short-chain fatty acids (SCFAs), have been shown to participate in the development of mastitis. LPS is the main component of the cell walls of gram-negative bacteria. Overproduction of rumen-derived LPS injures the rumen epithelium, resulting in the entry of LPS into the blood and damaged liver function; once in the blood, it circulates into the mammary gland, increasing blood-barrier permeability and leading to mammary gland inflammation. SCFAs, which are produced by gut microbiota as fermentation products, have a protective effect on mammary gland inflammatory responses and help maintain the function of the blood-milk barrier. Recently, increasing attention has been focused on the use of probiotics as a promising alternative for the treatment of mastitis. This review summarizes the effects of the gut microbiome and its metabolites on mastitis as well as the current of probiotics in mastitis. This work may provide a valuable theoretical foundation for the development of fresh ideas for the prevention and treatment of mastitis.
Collapse
|
72
|
Martín V, Cárdenas N, Ocaña S, Marín M, Arroyo R, Beltrán D, Badiola C, Fernández L, Rodríguez JM. Rectal and Vaginal Eradication of Streptococcus agalactiae (GBS) in Pregnant Women by Using Lactobacillus salivarius CECT 9145, A Target-specific Probiotic Strain. Nutrients 2019; 11:E810. [PMID: 30974819 PMCID: PMC6521265 DOI: 10.3390/nu11040810] [Citation(s) in RCA: 31] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 04/02/2019] [Accepted: 04/05/2019] [Indexed: 12/14/2022] Open
Abstract
Streptococcus agalactiae (Group B Streptococci, GBS) can cause severe neonatal sepsis. The recto-vaginal GBS screening of pregnant women and intrapartum antibiotic prophylaxis (IAP) to positive ones is one of the main preventive options. However, such a strategy has some limitations and there is a need for alternative approaches. Initially, the vaginal microbiota of 30 non-pregnant and 24 pregnant women, including the assessment of GBS colonization, was studied. Among the Lactobacillus isolates, 10 Lactobacillus salivarius strains were selected for further characterization. In vitro characterization revealed that L. salivarius CECT 9145 was the best candidate for GBS eradication. Its efficacy to eradicate GBS from the intestinal and vaginal tracts of pregnant women was evaluated in a pilot trial involving 57 healthy pregnant women. All the volunteers in the probiotic group (n = 25) were GBS-positive and consumed ~9 log10 cfu of L. salivarius CECT 9145 daily from week 26 to week 38. At the end of the trial (week 38), 72% and 68% of the women in this group were GBS-negative in the rectal and vaginal samples, respectively. L. salivarius CECT 9145 seems to be an efficient method to reduce the number of GBS-positive women during pregnancy, decreasing the number of women receiving IAP during delivery.
Collapse
Affiliation(s)
- Virginia Martín
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Nivia Cárdenas
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Sara Ocaña
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain.
- Unidad de Reproducción, Fundación Hospital Alcorcón, 28922 Alcorcón, Spain.
| | - María Marín
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Rebeca Arroyo
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain.
| | - David Beltrán
- Centro de Diagnóstico Médico, Ayuntamiento de Madrid, 28006 Madrid, Spain.
| | - Carlos Badiola
- Laboratorios Casen Recordati S.L., Vía de las Dos Castillas, 33, 28224 Pozuelo de Alarcón, Madrid, Spain.
| | - Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, 28040 Madrid, Spain.
| | - Juan M Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, 28040 Madrid, Spain.
| |
Collapse
|
73
|
Parida S, Sharma D. The power of small changes: Comprehensive analyses of microbial dysbiosis in breast cancer. Biochim Biophys Acta Rev Cancer 2019; 1871:392-405. [PMID: 30981803 PMCID: PMC8769497 DOI: 10.1016/j.bbcan.2019.04.001] [Citation(s) in RCA: 67] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 12/22/2022]
Abstract
Disparate occurrence of breast cancer remains an intriguing question since only a subset of women with known risk factors develop cancer. Recent studies suggest an active role of local and distant microbiota in breast cancer initiation, progression, and overall prognosis. A dysbiotic microbiota predisposes the body to develop cancer by inducing genetic instability, initiating DNA damage and proliferation of the damaged progeny, eliciting favorable immune response, metabolic dysregulation and altered response to therapy. In this review, we present our analyses of the existing datasets and discuss the local dysbiosis observed in breast cancer patients and different aspects of breast carcinogenesis that can be potentially influenced by local breast microbiota. Striking differences between microbial community compositions in breast of cancer patients compared to healthy individuals were noted. Differences in microbiome were also apparent between benign and malignant disease and between nipple aspirate fluid of healthy individuals and breast survivors. We also discuss the identification of distinct bacterial, fungal, viral as well as parasite signatures for breast cancer. These microbes are capable of producing numerous secondary metabolites that can act as signaling mediators effecting breast cancer progression. We review how microbes potentially alter response to therapy affecting drug metabolism, pharmacokinetics, anti-tumor effects and toxicity. In conclusion, breast harbors a community of microbes that can communicate with the host cells inducing downstream signaling pathways and modulating various aspects of breast cancer growth and metastatic progression and an improved understanding of microbial dysbiosis can potentially reduce breast cancer risk and improve outcomes of breast cancer patients. The human microbiome, now referred to as, the "forgotten organ" contains a metagenome that is 100-fold more diverse compared to the human genome, thereby, is critically associated with human health [1,2]. With the revelations of the human microbiome project and advent of deep sequencing techniques, a plethora of information has been acquired in recent years. Body sites like stomach, bladder and lungs, once thought to be sterile, are now known to harbor millions of indigenous microbial species. Approximately 80% of the healthy microbiome consists of Firmicutes and Bacteroidetes accompanied by Verrucomicrobia, Actinobacteria, Proteobacteria, Tenericutes and Cyanobacteria [2-7]. The role of microbiome in diabetes, obesity and even neurodegenerative diseases was greatly appreciated in the last decade [1,7-14] and now it has been established that microbiome significantly contributes to many organ specific cancers [1,15,16].
Collapse
Affiliation(s)
- Sheetal Parida
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA
| | - Dipali Sharma
- Department of Oncology, Johns Hopkins University School of Medicine and the Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, Baltimore, MD, USA.
| |
Collapse
|
74
|
Guo W, Liu B, Yin Y, Kan X, Gong Q, Li Y, Cao Y, Wang J, Xu D, Ma H, Fu S, Liu J. Licochalcone A Protects the Blood-Milk Barrier Integrity and Relieves the Inflammatory Response in LPS-Induced Mastitis. Front Immunol 2019; 10:287. [PMID: 30858849 PMCID: PMC6398509 DOI: 10.3389/fimmu.2019.00287] [Citation(s) in RCA: 59] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2018] [Accepted: 02/04/2019] [Indexed: 12/18/2022] Open
Abstract
Background/Aims: Mastitis is an acute clinical inflammatory response. The occurrence and development of mastitis seriously disturb women's physical and mental health. Licochalcone A, a phenolic compound in Glycyrrhiza uralensis, has anti-inflammatory properties. Here, we examined the effect of licochalcone A on blood-milk barrier and inflammatory response in LPS-induced mice mastitis. Methods:In vivo, we firstly established mice models of mastitis by canal injection of LPS to mammary gland, and then detected the effect of licochalcone A on pathological indexes, inflammatory responses and blood-milk barrier in this model. In vivo, Mouse mammary epithelial cells (mMECs) were treated with licochalcone A prior to the incubation of LPS, and then the inflammatory responses, tight junction which is the basic structure of blood-milk barrier were analyzed. Last, we elucidated the anti-inflammatory mechanism by examining the activation of mitogen-activated protein kinase (MAPK) and AKT/NF-κB signaling pathways in vivo and in vitro. Result: The in vivo results showed that licochalcone A significantly decreased the histopathological impairment and the inflammatory responses, and improved integrity of blood-milk barrier. The in vitro results demonstrated that licochalcone A inhibited LPS-induced inflammatory responses and increase the protein levels of ZO-1, occludin, and claudin3 in mMECs. The in vivo and in vitro mechanistic study found that the anti-inflammatory effect of licochalcone A in LPS-induced mice mastitis was mediated by MAPK and AKT/NF-κB signaling pathways. Conclusions and Implications: Our experiments collectively indicate that licochalcone A protected against LPS-induced mice mastitis via improving the blood–milk barrier integrity and inhibits the inflammatory response by MAPK and AKT/NF-κB signaling pathways.
Collapse
Affiliation(s)
- Wenjin Guo
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Bingrun Liu
- Laboratory of Biomolecular Research, Division of Biology and Chemistry, Paul Scherrer Institute, Villigen, Switzerland
| | - Yunhou Yin
- School of Communication, Guizhou Minzu University, Guiyang, China
| | - Xingchi Kan
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Qian Gong
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Yanwei Li
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Yu Cao
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Jianfa Wang
- College of Animal Science and Veterinary Medicine, Heilongjiang Bayi Agricultural University, Daqing, China
| | - Dianwen Xu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - He Ma
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Shoupeng Fu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| | - Juxiong Liu
- College of Animal Science and Veterinary Medicine, Jilin University, Changchun, China
| |
Collapse
|
75
|
Grev J, Berg M, Soll R. Maternal probiotic supplementation for prevention of morbidity and mortality in preterm infants. Cochrane Database Syst Rev 2018; 12:CD012519. [PMID: 30548483 PMCID: PMC6516999 DOI: 10.1002/14651858.cd012519.pub2] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
BACKGROUND Inflammation may contribute to preterm birth and to morbidity of preterm infants. Preterm infants are at risk for alterations in the normal protective microbiome. Oral probiotics administered directly to preterm infants have been shown to decrease the risk for severe necrotizing enterocolitis (NEC) as well as the risk of death, but there are safety concerns about administration of probiotics directly to preterm infants. Through decreasing maternal inflammation, probiotics may play a role in preventing preterm birth and/or decreasing the inflammatory milieu surrounding delivery of preterm infants, and may alter the microbiome of the preterm infant when given to mothers during pregnancy. Probiotics given to mothers after birth of preterm infants may effect infant bacterial colonization, which could potentially reduce the incidence of NEC. OBJECTIVES 1. To compare the efficacy of maternal probiotic administration versus placebo or no intervention in mothers during pregnancy for the prevention of preterm birth and the prevention of morbidity and mortality of infants born preterm.2. To compare the efficacy of maternal probiotic administration versus placebo, no intervention, or neonatal probiotic administration in mothers of preterm infants after birth on the prevention of mortality and preterm infant morbidities such as NEC. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to search the Cochrane Central Register of Controlled Trials (CENTRAL 2017, Issue 2), MEDLINE via PubMed (1966 to 21 March 2017), Embase (1980 to 21 March 2017), and CINAHL (1982 to 21 March 2017). We also searched clinical trials databases, conference proceedings, and the reference lists of retrieved articles for randomized controlled trials and quasi-randomized trials. SELECTION CRITERIA We included randomized controlled trials in the review if they administered oral probiotics to pregnant mothers at risk for preterm birth, or to mothers of preterm infants after birth. Quasi-randomized trials were eligible for inclusion, but none were identified. Studies enrolling pregnant women needed to administer probiotics at < 36 weeks' gestation until the trimester of birth. Probiotics considered were of the genera Lactobacillus, Bifidobacterium or Saccharomyces. DATA COLLECTION AND ANALYSIS We used the standard methods of the Cochrane Collaboration and Cochrane Neonatal to determine the methodologic quality of studies, and for data collection and analysis. MAIN RESULTS We included 12 eligible trials with a total of 1450 mothers and 1204 known infants. Eleven trials administered probiotics to mothers during pregnancy and one trial administered probiotics to mothers after birth of their preterm infants. No studies compared maternal probiotic administration directly with neonatal administration. Included prenatal trials were highly variable in the indication for the trial, the gestational age and duration of administration of probiotics, as well as the dose and formulation of the probiotics. The pregnant women included in these trials were overall at low risk for preterm birth. In a meta-analysis of trial data, oral probiotic administration to pregnant women did not reduce the incidence of preterm birth < 37 weeks (typical risk ratio (RR) 0.92, 95% confidence interval (CI) 0.32 to 2.67; 4 studies, 518 mothers and 506 infants), < 34 weeks (typical risk difference (RD) 0.00, 95% CI -0.02 to 0.02; 2 studies, 287 mothers and infants), the incidence of infant mortality (typical RD 0.00, 95% CI -0.02 to 0.02; 2 studies, 309 mothers and 298 infants), or the gestational age at birth (mean difference (MD) 0.15, 95% CI -0.33 to 0.63; 2 studies, 209 mothers with 207 infants).One trial studied administration of probiotics to mothers after preterm birth and included 49 mothers and 58 infants. There were no significant differences in the risk of any NEC (RR 0.44, 95% CI 0.13 to 1.46; 1 study, 58 infants), surgery for NEC (RR 0.15, 95% CI 0.01 to 2.58; 1 study, 58 infants), death (RR 0.66, 95% CI 0.06 to 6.88; 1 study, 58 infants), and death or NEC (RR 0.53, 95% CI 0.19 to 1.49; 1 study, 58 infants). There was an improvement in time to reach 50% enteral feeds in infants whose mothers received probiotics, but the estimate is imprecise (MD -9.60 days, 95% CI -19.04 to -0.16 days; 58 infants). No other improvement in any neonatal outcomes were reported. The estimates were imprecise and do not exclude the possibility of meaningful harms or benefits from maternal probiotic administration. There were no cases of culture-proven sepsis with the probiotic organism. The GRADE quality of evidence was judged to be low to very low due to inconsistency and imprecision. AUTHORS' CONCLUSIONS There is insufficient evidence to conclude whether there is appreciable benefit or harm to neonates of either oral supplementation of probiotics administered to pregnant women at low risk for preterm birth or oral supplementation of probiotics to mothers of preterm infants after birth. Oral supplementation of probiotics to mothers of preterm infants after birth may decrease time to 50% enteral feeds, however, this estimate is extremely imprecise. More research is needed for post-natal administration of probiotics to mothers of preterm infants, as well as to pregnant mothers at high risk for preterm birth.
Collapse
Affiliation(s)
| | - Marie Berg
- Johns Hopkins All Children's HospitalPediatrics/Neonatology601 Fifth Street South, Suite 501St. PetersburgFloridaUSA33606
| | - Roger Soll
- Larner College of Medicine at the University of VermontDivision of Neonatal‐Perinatal Medicine, Department of Pediatrics111 Colchester AvenueBurlingtonVermontUSA05401
| | | |
Collapse
|
76
|
Fernández L, Ruiz L, Jara J, Orgaz B, Rodríguez JM. Strategies for the Preservation, Restoration and Modulation of the Human Milk Microbiota. Implications for Human Milk Banks and Neonatal Intensive Care Units. Front Microbiol 2018; 9:2676. [PMID: 30473683 PMCID: PMC6237971 DOI: 10.3389/fmicb.2018.02676] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Accepted: 10/19/2018] [Indexed: 12/11/2022] Open
Abstract
Studies carried in the last years have revealed that human milk contains a site-specific microbiota and constitutes a source of potentially beneficial bacteria to the infant gut. Once in the infant gut, these bacteria contribute to the assembly of a physiological gut microbiota and may play several functions, contributing to infant metabolism, protection against infections, immunomodulation or neuromodulation. Many preterm neonates are fed with pasteurized donor's human milk (DHM) or formula and, therefore, are devoid of contact with human milk microbes. As a consequence, new strategies are required to allow the exposition of a higher number of preterm infants to the human milk microbiota early in life. The first strategy would be to promote and to increase the use of own mother's milk (OMM) in Neonatal Intensive Care Units (NICUs). Even small quantities of OMM can be very valuable since they would be added to DHM in order to microbiologically "customize" it. When OMM is not available, a better screening of donor women, including routine cytomegalovirus (CMV) screening of milk, may help to avoid the pasteurization of the milk provided by, at least, a relevant proportion of donors. Finally, when pasteurized DHM or formula are the only feeding option, their supplementation with probiotic bacteria isolated from human milk, such as lactic acid bacteria or bifidobacteria, may be an alternative to try to restore a human milk-like microbiota before feeding the babies. In the future, the design of human milk bacterial consortia (minimal human milk microbiotas), including well characterized strains representative of a healthy human milk microbiota, may be an attractive strategy to provide a complex mix of strains specifically tailored to this target population.
Collapse
Affiliation(s)
- Leónides Fernández
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Lorena Ruiz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias – Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| | - Josué Jara
- Department of Galenic Pharmacy and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Belén Orgaz
- Department of Microbiology and Biochemistry of Dairy Products, Instituto de Productos Lácteos de Asturias – Consejo Superior de Investigaciones Científicas, Villaviciosa, Spain
| | - Juan M. Rodríguez
- Department of Nutrition and Food Science, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
77
|
Rainard P, Foucras G. A Critical Appraisal of Probiotics for Mastitis Control. Front Vet Sci 2018; 5:251. [PMID: 30364110 PMCID: PMC6191464 DOI: 10.3389/fvets.2018.00251] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2018] [Accepted: 09/19/2018] [Indexed: 01/13/2023] Open
Abstract
The urge to reduce antimicrobials use in dairy farming has prompted a search for alternative solutions. As infections of the mammary gland is a major reason for antibiotic administration to dairy ruminants, mammary probiotics have recently been presented as a possible alternative for the treatment of mastitis. To assess the validity of this proposal, we performed a general appraisal of the knowledge related to probiotics for mammary health by examining their potential modes of action and assessing the compatibility of these mechanisms with the immunobiology of mammary gland infections. Then we analyzed the literature published on the subject, taking into account the preliminary in vitro experiments and the in vivo trials. Preliminary experiments aimed essentially at exploring in vitro the capacity of putative probiotics, mainly lactic acid bacteria (LABs), to interfere with mastitis-associated bacteria or to interact with mammary epithelial cells. A few studies used LABs selected on the basis of bacteriocin production or the capacity to adhere to epithelial cells to perform in vivo experiments. Intramammary infusion of LABs showed that LABs are pro-inflammatory for the mammary gland, inducing an intense influx of neutrophils into milk during lactation and at drying-off. Yet, their capacity to cure mastitis remains to be established. A few preliminary studies tackle the possibility of using probiotics to interfere with the teat apex microbiota or to prevent the colonization of the teat canal by pathogenic bacteria. From the analysis of the published literature, it appears that currently there is no sound scientific foundation for the use of probiotics to prevent or treat mastitis. We conclude that the prospects for oral probiotics are not promising for ruminants, those for intramammary probiotics should be considered with caution, but that teat apex probiotics deserve further research.
Collapse
Affiliation(s)
- Pascal Rainard
- ISP, INRA, Université de Tours, UMR 1282, Nouzilly, France
| | - Gilles Foucras
- IHAP, Université de Toulouse, ENVT, INRA, UMR1225, Toulouse, France
| |
Collapse
|
78
|
Shively CA, Register TC, Appt SE, Clarkson TB, Uberseder B, Clear KYJ, Wilson AS, Chiba A, Tooze JA, Cook KL. Consumption of Mediterranean versus Western Diet Leads to Distinct Mammary Gland Microbiome Populations. Cell Rep 2018; 25:47-56.e3. [PMID: 30282037 PMCID: PMC6338220 DOI: 10.1016/j.celrep.2018.08.078] [Citation(s) in RCA: 99] [Impact Index Per Article: 16.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2018] [Revised: 07/23/2018] [Accepted: 08/24/2018] [Indexed: 12/16/2022] Open
Abstract
Recent identification of a mammary gland-specific microbiome led to studies investigating bacteria populations in breast cancer. Malignant breast tumors have lower Lactobacillus abundance compared with benign lesions, implicating Lactobacillus as a negative regulator of breast cancer. Diet is a main determinant of gut microbial diversity. Whether diet affects breast microbiome populations is unknown. In a non-human primate model, we found that consumption of a Western or Mediterranean diet modulated mammary gland microbiota and metabolite profiles. Mediterranean diet consumption led to increased mammary gland Lactobacillus abundance compared with Western diet-fed monkeys. Moreover, mammary glands from Mediterranean diet-fed monkeys had higher levels of bile acid metabolites and increased bacterial-processed bioactive compounds. These data suggest that diet directly influences microbiome populations outside the intestinal tract in distal sites such as the mammary gland. Our study demonstrates that diet affects the mammary gland microbiome, establishing an alternative mechanistic pathway for breast cancer prevention.
Collapse
Affiliation(s)
- Carol A Shively
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas C Register
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Susan E Appt
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Thomas B Clarkson
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Beth Uberseder
- Department of Pathology, Section of Comparative Medicine, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Kenysha Y J Clear
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Adam S Wilson
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Akiko Chiba
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Janet A Tooze
- Department of Biostatistical Sciences, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA
| | - Katherine L Cook
- Department of Surgery, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA; Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, NC 27157, USA.
| |
Collapse
|
79
|
Dronkers T, Krist L, Van Overveld F, Rijkers G. The ascent of the blessed: regulatory issues on health effects and health claims for probiotics in Europe and the rest of the world. Benef Microbes 2018; 9:717-723. [DOI: 10.3920/bm2017.0196] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The outcome of the first series of health claim applications for probiotics in Europe as evaluated by the European Food Safety Authority (EFSA) has, up to 2013 almost completely yielded negative results. All recent applications also have been rejected, including the latest on prevention of mastitis in breastfeeding mothers. In other developed countries, such as Switzerland, Japan and Canada, the health effects of probiotics, for which scientific evidence has been provided, can be communicated to potential consumers. The number of clinical trials with probiotics over recent years shows a trend to level off or even decline. At the same time, clinical research into the role of (gut) microbiota in a wide variety of diseases and conditions is booming. Ultimately, this may offer new indications for gut microbiota management by probiotics, prebiotics or other food supplements.
Collapse
Affiliation(s)
- T.M.G. Dronkers
- Science Department, University College Roosevelt, Lange Noordstraat 1, 4331 CB Middelburg, the Netherlands
| | - L. Krist
- Science Department, University College Roosevelt, Lange Noordstraat 1, 4331 CB Middelburg, the Netherlands
| | - F.J. Van Overveld
- Science Department, University College Roosevelt, Lange Noordstraat 1, 4331 CB Middelburg, the Netherlands
| | - G.T. Rijkers
- Science Department, University College Roosevelt, Lange Noordstraat 1, 4331 CB Middelburg, the Netherlands
| |
Collapse
|
80
|
Abstract
Mastitis can be an unwelcome and debilitating visitor to breastfeeding mothers. The mammary gland has its own microbiome that can be affected by reduced polymorphonuclear neutrophil recruitment during the first 3 months postpartum, as well as the receipt of antibiotics during the last trimester of pregnancy. This can leave the breast vulnerable to pathologic bacterial overgrowth. Mammary dysbiosis is a process whereby the population of potential pathogens increases at the expense of the normal mammary microbiota. Multiresistance to antibiotics plus tricky evasion techniques engaged in by bacterial agents can result in microbes that are elusive to antibiotic therapy. Therefore, new strategies are needed for the treatment of this threat to continued breastfeeding. Bacteriotherapy, targeting harmless bacteria to displace pathologic organisms, is an emerging therapeutic intervention that uses probiotics instead of antibiotics. Once more high-quality clinical trials of strain-specific probiotics have been conducted, bacteriotherapy may move into mainstream mastitis treatment.
Collapse
|
81
|
Zhang Z, Lv J, Pan L, Zhang Y. Roles and applications of probiotic Lactobacillus strains. Appl Microbiol Biotechnol 2018; 102:8135-8143. [PMID: 30032432 DOI: 10.1007/s00253-018-9217-9] [Citation(s) in RCA: 137] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/30/2018] [Accepted: 07/02/2018] [Indexed: 12/19/2022]
Abstract
Lactobacilli are recognized as probiotics on account of their health-promoting effects in the host. The aim of this review is to summarize current knowledge of the mechanisms of the adaption factors and main functions of lactobacilli that exert health-promoting effects in the host and to discuss important applications in animal and human health. The adaption mechanisms of lactobacilli facilitate interactions with the host and directly contribute to the beneficial nutritional, physiological, microbiological, and immunological effects in the host. Besides, the application of probiotic lactobacilli will increase our understanding of practical uses based on the roles of these organisms in immunoregulation, antipathogenic activities, and enhancement of the epithelial barrier.
Collapse
Affiliation(s)
- Zhongwang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China
| | - Jianliang Lv
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China
| | - Li Pan
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China. .,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China.
| | - Yongguang Zhang
- State Key Laboratory of Veterinary Etiological Biology, OIE/National Foot-and-Mouth Disease Reference Laboratory, Lanzhou Veterinary Research Institute, Chinese Academy of Agricultural Sciences, 1 Xujiaping, Yanchangbu, Lanzhou, 730046, Gansu, China.,Jiangsu Co-innovation Center for the Prevention and Control of Important Animal Infectious Diseases and Zoonoses, Yangzhou University, Yangzhou, 225009, Jiangsu, People's Republic of China
| |
Collapse
|
82
|
Chung HJ, Sim JH, Min TS, Choi HK. Metabolomics and Lipidomics Approaches in the Science of Probiotics: A Review. J Med Food 2018; 21:1086-1095. [PMID: 30004273 DOI: 10.1089/jmf.2017.4175] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The intestinal microflora plays important roles in the health of the host, such as nutrient processing and the modulation of intestinal immune responses. The constituents of the diet greatly affect the composition of the microbiota and its metabolites. The human intestinal microbiota is made up of around 100 trillion microbial cells encompassing at least 300 species. Consuming probiotics may lead to changes in the intestinal microflora that influence host health. Metabolomics is a powerful tool for revealing metabolic changes in biofluids, tissues, and organs of hosts induced by the consumption of probiotics, and lipidomics in particular is a technical approach that focuses on the analysis of lipids in various cells and biofluids. Metabolomics and lipidomics have been used to investigate intracellular and extracellular metabolites as well as for the nontargeted profiling and fingerprinting of metabolites. Based on metabolomics and lipidomics investigations, we reviewed the effects of consuming probiotics on metabolic profiles in controlled intestinal environments. We also discuss the associations between metabolic changes and human diseases after consuming probiotics in uncontrolled intestinal environments. In addition, we review the metabolic changes that take place within the food matrix during probiotic fermentation.
Collapse
Affiliation(s)
- Hyuk-Jin Chung
- 1 College of Pharmacy, Chung-Ang University , Seoul, Korea.,2 Korea Yakult Co., Ltd. , Yongin, Korea
| | | | - Tae-Sun Min
- 3 Faculty of Biotechnology, SARI, Jeju National University , Jeju, Korea
| | | |
Collapse
|
83
|
Wu WK, Chen CC, Panyod S, Chen RA, Wu MS, Sheen LY, Chang SC. Optimization of fecal sample processing for microbiome study - The journey from bathroom to bench. J Formos Med Assoc 2018; 118:545-555. [PMID: 29490879 DOI: 10.1016/j.jfma.2018.02.005] [Citation(s) in RCA: 93] [Impact Index Per Article: 15.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2017] [Revised: 01/23/2018] [Accepted: 02/05/2018] [Indexed: 12/30/2022] Open
Abstract
Although great interest has been displayed by researchers in the contribution of gut microbiota to human health, there is still no standard protocol with consensus to guarantee the sample quality of metagenomic analysis. Here we reviewed existing methodology studies and present suggestions for optimizing research pipeline from fecal sample collection to DNA extraction. First, we discuss strategies of clinical metadata collection as common confounders for microbiome research. Second, we propose general principles for freshly collected fecal sample and its storage and share a DIY stool collection kit protocol based on the manual procedure of Human Microbiome Project (HMP). Third, we provide a useful information of collection kit with DNA stabilization buffers and compare their pros and cons for multi-omic study. Fourth, we offer technical strategies as well as information of novel tools for sample aliquoting before long-term storage. Fifth, we discuss the substantial impact of different DNA extraction protocols on technical variations of metagenomic analysis. And lastly, we point out the limitation of current methods and the unmet needs for better quality control of metagenomic analysis. We hope the information provided here will help investigators in this exciting field to advance their studies while avoiding experimental artifacts.
Collapse
Affiliation(s)
- Wei-Kai Wu
- Department of Internal Medicine, National Taiwan University Hospital Bei-Hu Branch, Taipei, Taiwan; Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Chieh-Chang Chen
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan
| | - Suraphan Panyod
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Rou-An Chen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Ming-Shiang Wu
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan
| | - Lee-Yan Sheen
- Institute of Food Science and Technology, National Taiwan University, Taipei, Taiwan
| | - Shan-Chwen Chang
- Department of Internal Medicine, National Taiwan University Hospital, Taipei, Taiwan; College of Medicine, National Taiwan University, Taipei, Taiwan.
| |
Collapse
|
84
|
The microbiology and treatment of human mastitis. Med Microbiol Immunol 2018; 207:83-94. [PMID: 29350290 DOI: 10.1007/s00430-017-0532-z] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2017] [Accepted: 12/15/2017] [Indexed: 12/29/2022]
Abstract
Mastitis, which is generally described as an inflammation of breast tissue, is a common and debilitating disease which frequently results in the cessation of exclusive breastfeeding and affects up to 33% of lactating women. The condition is a primary cause of decreased milk production and results in organoleptic and nutritional alterations in milk quality. Recent studies employing culture-independent techniques, including metagenomic sequencing, have revealed a loss of bacterial diversity in the microbiome of mastitic milk samples compared to healthy milk samples. In those infected, the pathogens Staphylococcus aureus, Staphylococcus epidermidis and members of corynebacteria have been identified as the predominant etiological agents in acute, subacute and granulomatous mastitis, respectively. The increased incidence of antibiotic resistance in the causative species is also a key cause of concern for treatment of the disease, thus leading to the need to develop novel therapies. In this respect, probiotics and bacteriocins have revealed potential as alternative treatments.
Collapse
|
85
|
Physiological Translocation of Lactic Acid Bacteria during Pregnancy Contributes to the Composition of the Milk Microbiota in Mice. Nutrients 2017; 10:nu10010014. [PMID: 29295502 PMCID: PMC5793242 DOI: 10.3390/nu10010014] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2017] [Revised: 11/24/2017] [Accepted: 12/19/2017] [Indexed: 02/06/2023] Open
Abstract
The human milk microbiota is a complex and diverse ecosystem that seems to play a relevant role in the mother-to-infant transmission of microorganisms during early life. Bacteria present in human milk may arise from different sources, and recent studies suggest that at least some of them may be originally present in the maternal digestive tract and may reach the mammary gland through an endogenous route during pregnancy and lactation. The objective of this work was to elucidate whether some lactic acid bacteria are able to translocate and colonize the mammary gland and milk. For this purpose, two lactic acid bacteria strains (Lactococcus lactis MG1614 and Lactobacillus salivarius PS2) were transformed with a plasmid containing the lux genes; subsequently, the transformed strains were orally administered to pregnant mice. The murine model allowed the visualization, isolation, and Polymerase Chain Reaction (PCR)-detection of the transformed bacteria in different body locations, including mammary tissue and milk, reinforcing the hypothesis that physiological translocation of maternal bacteria during pregnancy and lactation may contribute to the composition of the mammary and milk microbiota.
Collapse
|
86
|
Wu H, Jiang K, Zhang T, Zhao G, Deng G. Hydroxytyrosol exerts an anti-inflammatory effect by suppressing Toll-like receptor 2 and TLR 2 downstream pathways in Staphylococcus aureus-induced mastitis in mice. J Funct Foods 2017. [DOI: 10.1016/j.jff.2017.06.035] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
|
87
|
Jiménez E, Arroyo R, Cárdenas N, Marín M, Serrano P, Fernández L, Rodríguez JM. Mammary candidiasis: A medical condition without scientific evidence? PLoS One 2017; 12:e0181071. [PMID: 28704470 PMCID: PMC5509296 DOI: 10.1371/journal.pone.0181071] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2016] [Accepted: 06/26/2017] [Indexed: 01/10/2023] Open
Abstract
Many physicians, midwives and lactation consultants still believe that yeasts (particularly Candida spp.) play an important role as an agent of nipple and breast pain despite the absolute absence of scientific proofs to establish such association. In this context, the objective of this study was to investigate the microorganisms involved in sore nipples and/or painful "shooting" breastfeeding by using a variety of microscopy techniques, as well as culture-dependent and-independent identification methods. Initially, 60 women (30 diagnosed as suffering "mammary candidiasis" and 30 with no painful breastfeeding) were recruited to elucidate the role of their pumps on the milk microbial profiles. After realizing the bias introduced by using such devices, manual expression was selected as the collection method for the microbiological analysis of milk samples provided by 529 women with symptoms compatible with "mammary candidiasis". Nipple swabs and nipple biopsy samples were also collected from the participating women. Results showed that the role played by yeasts in breast and nipple pain is, if any, marginal. In contrast, our results strongly support that coagulase-negative staphylococci and streptococci (mainly from the mitis and salivarius groups) are the agents responsible for such cases. As a consequence, and following the recommendations of the US Library of Medicine for the nomenclature of infectious diseases, the term "mammary candidiasis" or "nipple thrush" should be avoided when referring to such condition and replaced by "subacute mastitis".
Collapse
Affiliation(s)
- Esther Jiménez
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Rebeca Arroyo
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Nivia Cárdenas
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - María Marín
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Pilar Serrano
- Unidadde Endocrinología y Nutrición, Hospital Virgen del Rocío, Seville, Spain
| | - Leonides Fernández
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| | - Juan M. Rodríguez
- Dpt. Nutrition, Food Science and Food Technology, Complutense University of Madrid, Madrid, Spain
| |
Collapse
|
88
|
Bond DM, Morris JM, Nassar N. Study protocol: evaluation of the probiotic Lactobacillus Fermentum CECT5716 for the prevention of mastitis in breastfeeding women: a randomised controlled trial. BMC Pregnancy Childbirth 2017; 17:148. [PMID: 28525984 PMCID: PMC5437715 DOI: 10.1186/s12884-017-1330-8] [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] [Subscribe] [Scholar Register] [Received: 05/26/2016] [Accepted: 05/11/2017] [Indexed: 11/10/2022] Open
Abstract
Background Mastitis and accompanying pain have been associated with the cessation of breastfeeding. Mastitis is an inflammatory condition of the breast and may be a result of decreased immunity and a lowered resistance to infection. Mastitis affects up to one in five breastfeeding women with most episodes occurring in the first 6–8 weeks postpartum. Antibiotics are often used in the treatment of mastitis, but have not been popular or proven effective as a preventative agent. The WHO has highlighted significant concerns relating to adverse harms of antibiotic use with the production of antibiotic-resistant strains of disease organisms. Increasing research suggests that specific probiotic bacteria possess significant anti-inflammatory properties and supports their potential use as immunomodulatory agents. While animal studies have shown promising results in the use of probiotics for preventing mastitis, their use in human trials has had limited investigation. The aim of this study is to evaluate the effectiveness of oral probiotics for the prevention of mastitis in breastfeeding women. Methods APProve (CAn Probiotics ImProve Breastfeeding Outcomes?) is a double-blind randomised controlled trial designed to assess outcomes between breastfeeding women ingesting a probiotic versus a placebo daily for 8 weeks following birth. A total of 600 women (300 to each arm) who intend to breastfeed will be randomised after the birth of a term, healthy infant. Daily and weekly surveys for 8 weeks and follow-up surveys at 2, 6 and 12 months after birth will assess the primary outcome of mastitis in the first 8 weeks following birth as well as secondary maternal outcomes of breastfeeding duration (total/partial), antibiotic use, maternal health and well-being, and treatment compliance; and infant outcomes including gastroenteritis, infant health and well-being and growth and development. The acceptability and compliance using a novel mobile phone application system will also be evaluated. Discussion There is an urgent need to explore safe and effective alternatives for preventing mastitis in breastfeeding women. This trial seeks to provide evidence for such an alternative in the form of probiotics, which may also increase breastfeeding duration, providing long-term health, cognitive and developmental benefits for children. Decreased antibiotic usage also benefits the community and health system. Trial registration Australian New Zealand Clinical Trials Registry: ACTRN12615000923561. Date of registration: 4th September, 2015, retrospectively registered. Electronic supplementary material The online version of this article (doi:10.1186/s12884-017-1330-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Diana M Bond
- Clinical and Population Perinatal Health Research, Kolling Institute of Medical Research, University of Sydney, Level 2, Bldg 52, Royal North Shore Hospital Campus, St. Leonards, NSW, 2065, Australia. .,Menzies Centre for Health Policy, Charles Perkins Centre D17, The University of Sydney, Camperdown, NSW, 2006, Australia.
| | - Jonathan M Morris
- Clinical and Population Perinatal Health Research, Kolling Institute of Medical Research, University of Sydney, Level 2, Bldg 52, Royal North Shore Hospital Campus, St. Leonards, NSW, 2065, Australia.,Sydney Medical School - Northern, The University of Sydney, Kolling Building, Level 7, Royal North Shore Hospital, St Leonards, NSW, 2065, Australia
| | - Natasha Nassar
- Menzies Centre for Health Policy, Charles Perkins Centre D17, The University of Sydney, Camperdown, NSW, 2006, Australia
| |
Collapse
|
89
|
Chaves BD, Brashears MM, Nightingale KK. Applications and safety considerations of Lactobacillus salivarius as a probiotic in animal and human health. J Appl Microbiol 2017; 123:18-28. [PMID: 28256040 DOI: 10.1111/jam.13438] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2016] [Revised: 12/28/2016] [Accepted: 02/27/2017] [Indexed: 02/06/2023]
Abstract
The goals of this review are to summarize the current knowledge on the application of Lactobacillus salivarius as a probiotic in animals and humans, and to address safety concerns with its use on live hosts. Overall, several strains of L. salivarius are well established probiotics with multiple applications in animal health, particularly to reduce colonization by gastrointestinal pathogens, and to a lesser extent, as a production and quality aid. In humans, L. salivarius has been used to prevent and treat a variety of chronic diseases, including asthma, cancer, atopic dermatitis and halitosis, and to a much limited extent, to prevent or treat infections. Based on the results from primary research evidence, it seems that L. salivarius does not pose a health risk to animals or humans in the doses currently used for a variety of applications; however, there is a systematic lack of studies assuring the safety of many of the strains intended for clinical use. This review provides researchers in the field with up-to-date information regarding applications and safety of L. salivarius. Furthermore, it helps researchers identify knowledge gaps and potential opportunities for microbiological and clinical research.
Collapse
Affiliation(s)
- B D Chaves
- Department of Animal and Food Sciences, International Center for Food Industry Excellence, Texas Tech University, Lubbock, TX, USA
| | - M M Brashears
- Department of Animal and Food Sciences, International Center for Food Industry Excellence, Texas Tech University, Lubbock, TX, USA
| | - K K Nightingale
- Department of Animal and Food Sciences, International Center for Food Industry Excellence, Texas Tech University, Lubbock, TX, USA
| |
Collapse
|
90
|
Hurtado JA, Maldonado-Lobón JA, Díaz-Ropero MP, Flores-Rojas K, Uberos J, Leante JL, Affumicato L, Couce ML, Garrido JM, Olivares M, Fonollá J, the PROLAC Group. Oral Administration to Nursing Women of Lactobacillus fermentum CECT5716 Prevents Lactational Mastitis Development: A Randomized Controlled Trial. Breastfeed Med 2017. [PMCID: PMC5444431 DOI: 10.1089/bfm.2016.0173] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Objective: The objective of this study is to evaluate the preventive effect of oral administration of Lactobacillus fermentum CECT5716 on mastitis incidence in lactating women. Methods: A randomized double-blinded controlled trial that included 625 women was conducted. Women who received preventive dose of antibiotic in the context of delivery were recruited 1–6 days after childbirth and randomly assigned to a group. Probiotic group received 1 capsule/day containing L. fermentum 3 × 109 CFU, control group received 1 placebo capsule/day containing maltodextrin. The intervention period was 16 weeks. The primary outcome of the study was the incidence of clinical mastitis defined as at least two out of the three breast symptoms (pain, redness, and lump) and at least one of fever or flu-like symptoms (shivering, hot sweats, or aches). Results: Two hundred ninety-one women completed 16 weeks of treatment. Sixteen women in the probiotic group developed mastitis versus 30 women in the control group (odds ratio = 0.531; p = 0.058). Incidence rate of mastitis in the probiotic group was significantly lower than that in the control group (IR = 0.130 in the probiotic group versus IR = 0.263 in the control group; p = 0.021). Therefore, the oral administration of L. fermentum CECT5716 during lactation decreased by 51% the incidence rate of clinical mastitis. Staphylococcus spp. load at the end of intervention was significantly lower in breast milk of women in the probiotic group than in breast milk of women in the control group (p = 0.025). Conclusion: Consumption of the probiotic strain L. fermentum CECT5716 might be used during breastfeeding as an efficient strategy to prevent development of lactational mastitis in women. Trial registration: NCT02203877.
Collapse
Affiliation(s)
- José A. Hurtado
- Department of Neonatology, Hospital Materno-Infantil del CHU, Granada, Spain
| | | | | | | | - José Uberos
- Department of Pediatrics, School of Medicine, University of Granada, Spain
| | - José L. Leante
- Department of Neonatology, Hospital G.U. Santa Lucía, Cartagena, Murcia, Spain
| | | | - María Luz Couce
- Department of Neonatology, Hospital C.H.U., Santiago de Compostela, La Coruña, Spain
| | | | | | | | | |
Collapse
|
91
|
Power ML, Quaglieri C, Schulkin J. Reproductive Microbiomes: A New Thread in the Microbial Network. Reprod Sci 2017; 24:1482-1492. [DOI: 10.1177/1933719117698577] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Affiliation(s)
- Michael L. Power
- Research Department, American College of Obstetricians and Gynecologists, Washington, DC, USA
- Nutrition Laboratory, Conservation Ecology Center, Smithsonian Conservation Biology Institute, Washington, DC, USA
| | - Caroline Quaglieri
- Research Department, American College of Obstetricians and Gynecologists, Washington, DC, USA
| | - Jay Schulkin
- Research Department, American College of Obstetricians and Gynecologists, Washington, DC, USA
- Department of Obstetrics and Gynecology, University of Washington School of Medicine, Seattle, WA, USA
| |
Collapse
|
92
|
Kang MS, Lim HS, Oh JS, Lim YJ, Wuertz-Kozak K, Harro JM, Shirtliff ME, Achermann Y. Antimicrobial activity of Lactobacillus salivarius and Lactobacillus fermentum against Staphylococcus aureus. Pathog Dis 2017; 75:2966468. [DOI: 10.1093/femspd/ftx009] [Citation(s) in RCA: 56] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2016] [Accepted: 01/25/2017] [Indexed: 12/20/2022] Open
Affiliation(s)
- Mi-Sun Kang
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, 21201 MD, USA
- Oradentics Research Institute, Seoul 06157, South Korea
| | - Hae-Soon Lim
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, 21201 MD, USA
- Dental Science Research Institute, Chonnam National University, Gwangju 61186, South Korea
- Department of Dental Education, School of Dentistry, Chonnam National University, Gwangju 61186, South Korea
| | - Jong-Suk Oh
- Department of Microbiology, School of Medicine, Chonnam National University, Gwangju 61469, South Korea
| | - You-jin Lim
- Department of Nursing, Gwangju Health University, Gwangju 62287, South Korea
| | - Karin Wuertz-Kozak
- Institute for Biomechanics, ETH Zurich, 8093 Zurich, Switzerland
- Schön Clinic Munich Harlaching, Spine Center, Academic Teaching Hospital and Spine Research, Institute of the Paracelsus Medical University 5020 Salzburg (Austria), 81547 Munich, Germany
- Department of Health Sciences, University of Potsdam, 14469 Potsdam, Deutschland
| | - Janette M. Harro
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, 21201 MD, USA
| | - Mark E. Shirtliff
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, 21201 MD, USA
- Department of Microbiology and Immunology, School of Medicine, University of Maryland—Baltimore, Baltimore, 21201 MD, USA
| | - Yvonne Achermann
- Department of Microbial Pathogenesis, School of Dentistry, University of Maryland-Baltimore, Baltimore, 21201 MD, USA
- Department of Infectious Diseases, University and University Hospital Zurich, 8091 Zurich, Switzerland
| |
Collapse
|
93
|
Hieken TJ, Chen J, Hoskin TL, Walther-Antonio M, Johnson S, Ramaker S, Xiao J, Radisky DC, Knutson KL, Kalari KR, Yao JZ, Baddour LM, Chia N, Degnim AC. The Microbiome of Aseptically Collected Human Breast Tissue in Benign and Malignant Disease. Sci Rep 2016; 6:30751. [PMID: 27485780 PMCID: PMC4971513 DOI: 10.1038/srep30751] [Citation(s) in RCA: 269] [Impact Index Per Article: 33.6] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2016] [Accepted: 07/07/2016] [Indexed: 12/31/2022] Open
Abstract
Globally breast cancer is the leading cause of cancer death among women. The breast consists of epithelium, stroma and a mucosal immune system that make up a complex microenvironment. Growing awareness of the role of microbes in the microenvironment recently has led to a series of findings important for human health. The microbiome has been implicated in cancer development and progression at a variety of body sites including stomach, colon, liver, lung, and skin. In this study, we assessed breast tissue microbial signatures in intraoperatively obtained samples using 16S rDNA hypervariable tag sequencing. Our results indicate a distinct breast tissue microbiome that is different from the microbiota of breast skin tissue, breast skin swabs, and buccal swabs. Furthermore, we identify distinct microbial communities in breast tissues from women with cancer as compared to women with benign breast disease. Malignancy correlated with enrichment in taxa of lower abundance including the genera Fusobacterium, Atopobium, Gluconacetobacter, Hydrogenophaga and Lactobacillus. This work confirms the existence of a distinct breast microbiome and differences between the breast tissue microbiome in benign and malignant disease. These data provide a foundation for future investigation on the role of the breast microbiome in breast carcinogenesis and breast cancer prevention.
Collapse
Affiliation(s)
- Tina J Hieken
- Division of Subspecialty General Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jun Chen
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA.,Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Tanya L Hoskin
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Marina Walther-Antonio
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Stephen Johnson
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Sheri Ramaker
- Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA
| | - Jian Xiao
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Derek C Radisky
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Keith L Knutson
- Department of Immunology, Mayo Clinic, Jacksonville, FL, USA
| | - Krishna R Kalari
- Department of Health Sciences Research, Mayo Clinic, Rochester, MN, USA
| | - Janet Z Yao
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA
| | - Larry M Baddour
- Division of Infectious Diseases, Mayo Clinic, Rochester, MN, ville, USA
| | - Nicholas Chia
- Center for Individualized Medicine, Mayo Clinic, Rochester, MN, USA.,Division of Surgical Research, Department of Surgery, Mayo Clinic, Rochester, MN, USA.,Department of Physiology and Biomedical Engineering, Mayo Clinic, Rochester, MN, USA
| | - Amy C Degnim
- Division of Subspecialty General Surgery, Mayo Clinic, Rochester, MN, USA
| |
Collapse
|
94
|
Amir LH, Griffin L, Cullinane M, Garland SM. Probiotics and mastitis: evidence-based marketing? Int Breastfeed J 2016; 11:19. [PMID: 27446229 PMCID: PMC4955247 DOI: 10.1186/s13006-016-0078-5] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/27/2015] [Accepted: 07/04/2016] [Indexed: 11/25/2022] Open
Abstract
Probiotics are defined as live micro-organisms, which when administered in adequate amounts, confer health benefits on the host. Scientists have isolated various strains of Lactobacilli from human milk (such as Lactobacillus fermentum and Lactobacillus salivarius), and the presence of these organisms is thought to be protective against breast infections, or mastitis. Trials of probiotics for treating mastitis in dairy cows have had mixed results: some successful and others unsuccessful. To date, only one trial of probiotics to treat mastitis in women and one trial to prevent mastitis have been published. Although trials of probiotics to prevent mastitis in breastfeeding women are still in progress, health professionals in Australia are receiving marketing of these products. High quality randomised controlled trials are needed to assess the effectiveness of probiotics for the prevention and/or treatment of mastitis.
Collapse
Affiliation(s)
- Lisa H Amir
- Judith Lumley Centre, La Trobe University, Melbourne, VIC Australia
| | - Laura Griffin
- La Trobe Law School, La Trobe University, Bundoora, VIC Australia
| | - Meabh Cullinane
- Judith Lumley Centre, La Trobe University, Melbourne, VIC Australia
| | - Suzanne M Garland
- Department of Microbiology and Infectious Diseases, Western Pacific Regional HPV Reference Laboratory, Royal Women's Hospital, Parkville, VIC Australia ; Department of Infectious Diseases and Microbiology, Murdoch Childrens Research Institute, Melbourne, Australia ; Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, VIC Australia
| |
Collapse
|
95
|
The return of microbes. Clin Microbiol Infect 2016; 22:822-823. [PMID: 27102140 DOI: 10.1016/j.cmi.2016.04.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2016] [Revised: 04/05/2016] [Accepted: 04/07/2016] [Indexed: 11/22/2022]
|