1
|
Manzoni P, Messina A, Germano C, Picone S, Masturzo B, Sainaghi PP, Sola D, Rizzi M. Lactoferrin Supplementation in Preventing and Protecting from SARS-CoV-2 Infection: Is There Any Role in General and Special Populations? An Updated Review of Literature. Int J Mol Sci 2024; 25:10248. [PMID: 39408576 PMCID: PMC11476995 DOI: 10.3390/ijms251910248] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2024] [Revised: 09/07/2024] [Accepted: 09/21/2024] [Indexed: 10/19/2024] Open
Abstract
At the beginning of the pandemic, SARS-CoV-2 infection represented a great medical burden worldwide, as targeted and effective therapeutic options were lacking. This resulted in the revival of existing molecules and the increasing popularity of over-the-counter nutritional supplements. Among the latter, lactoferrin has been investigated as an adjuvant in COVID-19 therapy with conflicting results, mainly depending on different study designs. Considering that lactoferrin is one of the main components of human breast milk with anti-microbial and anti-inflammatory activity, it is conceivable that such bioactive molecule could be effective in supporting anti-SARS-CoV-2 infection therapy, especially in infants and pregnant women, two subpopulations that have been poorly evaluated in the existing clinical trials. This narrative review is intended to offer insight into the existing literature on lactoferrin's biological functions and protective effects against COVID-19, with a special focus on pregnant women and their infants.
Collapse
Affiliation(s)
- Paolo Manzoni
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Alessandro Messina
- School of Medicine, University of Turin, 10124 Turin, Italy;
- Sant’Anna Hospital, Department of Surgical Sciences, University of Turin, 10126 Turin, Italy
| | - Chiara Germano
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Simonetta Picone
- Neonatology and Neonatal Intensive Care Unit, Policlinico Casilino, 00169 Rome, Italy
| | - Bianca Masturzo
- Department of Maternal, Neonatal and Infant Medicine, University Hospital “Degli Infermi”, 13875 Ponderano, Italy (B.M.)
- School of Medicine, University of Turin, 10124 Turin, Italy;
| | - Pier Paolo Sainaghi
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| | - Daniele Sola
- Department of Translational Medicine (DiMeT), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Laboratory of Metabolic Research, IRCCS Istituto Auxologico Italiano, S. Giuseppe Hospital, 28824 Piancavallo, Italy
| | - Manuela Rizzi
- IRCAD (Interdisciplinary Research Center of Autoimmune Diseases), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
- Department of Health Sciences (DiSS), Università del Piemonte Orientale (UPO), 28100 Novara, Italy
| |
Collapse
|
2
|
Sindi AS, Stinson LF, Lai CT, Gridneva Z, Leghi GE, Netting MJ, Wlodek ME, Muhlhausler BS, Zhou X, Payne MS, Geddes DT. Human milk lactoferrin and lysozyme concentrations vary in response to a dietary intervention. J Nutr Biochem 2024; 135:109760. [PMID: 39251146 DOI: 10.1016/j.jnutbio.2024.109760] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2024] [Revised: 08/08/2024] [Accepted: 09/04/2024] [Indexed: 09/11/2024]
Abstract
It is known that human milk (HM)1 antimicrobial protein composition varies during lactation. However, the impact of maternal diet on these antimicrobial proteins, particularly lactoferrin and lysozyme remains unknown. In addition, it is unclear whether daily, circadian, and between breast variations exist for lactoferrin and lysozyme concentrations. We investigated the impact of a low sugar, low fat, high fibre dietary intervention on HM lysozyme and lactoferrin concentrations. HM was sampled across a 3-week period; daily, at different times of day, and from both breasts to measure the level of intraindividual variation. The intervention significantly reduced maternal sugar, total fat, and saturated fat intake. HM lactoferrin concentration declined significantly over the course of the intervention however the effect size was relatively small. In addition, lactoferrin and lysozyme concentrations were variable over time, and differed significantly within and across the day but not between breasts.
Collapse
Affiliation(s)
- Azhar S Sindi
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, Western Australia, Australia; College of Applied Medical Sciences, Umm Al-Qura University, Makkah, Saudi Arabia; ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia
| | - Lisa F Stinson
- ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia; School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Ching Tat Lai
- ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia; School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Zoya Gridneva
- ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia; School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Gabriela E Leghi
- School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, South Australia, Australia
| | - Merryn J Netting
- Women and Kids Theme, South Australian Health and Medical Research Institute (SAHMRI), North Terrace, South Australia, Australia; Discipline of Paediatrics, The University of Adelaide, North Adelaide, South Australia, Australia; Women's and Children's Hospital, North Adelaide, South Australia, Australia
| | - Mary E Wlodek
- School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia; Department of Obstetrics and Gynaecology, University of Melbourne, Parkville, Victoria, Australia
| | - Beverly S Muhlhausler
- School of Agriculture, Food and Wine, The University of Adelaide, Urrbrae, South Australia, Australia; CSIRO, Adelaide, South Australia, Australia
| | - Xiaojie Zhou
- ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia; School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia
| | - Matthew S Payne
- Division of Obstetrics and Gynaecology, School of Medicine, The University of Western Australia, Subiaco, Western Australia, Australia
| | - Donna T Geddes
- ABREAST Network, Perth, Western Australia, Australia; UWA Centre for Human Lactation Research and Translation, Crawley, Western Australia, Australia; School of Molecular Sciences, The University of Western Australia, Crawley, Western Australia, Australia.
| |
Collapse
|
3
|
Marissen J, Reichert L, Härtel C, Fortmann MI, Faust K, Msanga D, Harder J, Zemlin M, Gomez de Agüero M, Masjosthusmann K, Humberg A. Antimicrobial Peptides (AMPs) and the Microbiome in Preterm Infants: Consequences and Opportunities for Future Therapeutics. Int J Mol Sci 2024; 25:6684. [PMID: 38928389 PMCID: PMC11203687 DOI: 10.3390/ijms25126684] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2024] [Revised: 06/07/2024] [Accepted: 06/14/2024] [Indexed: 06/28/2024] Open
Abstract
Antimicrobial peptides (AMPs) are crucial components of the innate immune system in various organisms, including humans. Beyond their direct antimicrobial effects, AMPs play essential roles in various physiological processes. They induce angiogenesis, promote wound healing, modulate immune responses, and serve as chemoattractants for immune cells. AMPs regulate the microbiome and combat microbial infections on the skin, lungs, and gastrointestinal tract. Produced in response to microbial signals, AMPs help maintain a balanced microbial community and provide a first line of defense against infection. In preterm infants, alterations in microbiome composition have been linked to various health outcomes, including sepsis, necrotizing enterocolitis, atopic dermatitis, and respiratory infections. Dysbiosis, or an imbalance in the microbiome, can alter AMP profiles and potentially lead to inflammation-mediated diseases such as chronic lung disease and obesity. In the following review, we summarize what is known about the vital role of AMPs as multifunctional peptides in protecting newborn infants against infections and modulating the microbiome and immune response. Understanding their roles in preterm infants and high-risk populations offers the potential for innovative approaches to disease prevention and treatment.
Collapse
Affiliation(s)
- Janina Marissen
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Lilith Reichert
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
| | - Christoph Härtel
- Department of Pediatrics, University Hospital Würzburg, 97080 Würzburg, Germany; (J.M.); (L.R.)
- German Center for Infection Research, Site Hamburg-Lübeck-Borstel-Riems, 23538 Lübeck, Germany
| | - Mats Ingmar Fortmann
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Kirstin Faust
- Department of Pediatrics, University Hospital Schleswig-Holstein, 23538 Lübeck, Germany; (M.I.F.); (K.F.)
| | - Delfina Msanga
- Department of Pediatrics, Bugando Hospital, Catholic University of Health and Allied Sciences, Mwanza 33109, Tanzania;
| | - Jürgen Harder
- Department of Dermatology, Venerology and Allergology, Quincke Research Center, Kiel University, 24105 Kiel, Germany;
| | - Michael Zemlin
- Department of General Pediatrics and Neonatology, Saarland University Medical Center, 66421 Homburg, Germany;
| | - Mercedes Gomez de Agüero
- Würzburg Institute of Systems Immunology, Max-Planck Research Group, University of Würzburg, 97078 Würzburg, Germany;
| | - Katja Masjosthusmann
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| | - Alexander Humberg
- Department of General Pediatrics, University Children’s Hospital Münster, 48149 Münster, Germany; (K.M.); (A.H.)
| |
Collapse
|
4
|
Wang W, An Q, Huang K, Dai Y, Meng Q, Zhang Y. Unlocking the power of Lactoferrin: Exploring its role in early life and its preventive potential for adult chronic diseases. Food Res Int 2024; 182:114143. [PMID: 38519174 DOI: 10.1016/j.foodres.2024.114143] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2023] [Revised: 02/05/2024] [Accepted: 02/17/2024] [Indexed: 03/24/2024]
Abstract
Nutrition during the early postnatal period exerts a profound impact on both infant development and later-life health. Breast milk, which contains lactoferrin, a dynamic protein, plays a crucial role in the growth of various biological systems and in preventing numerous chronic diseases. Based on the relationship between early infant development and chronic diseases later in life, this paper presents a review of the effects of lactoferrin in early life on neonates intestinal tract, immune system, nervous system, adipocyte development, and early intestinal microflora establishment, as well as the preventive and potential mechanisms of early postnatal lactoferrin against adult allergy, inflammatory bowel disease, depression, cancer, and obesity. Furthermore, we summarized the application status of lactoferrin in the early postnatal period and suggested directions for future research.
Collapse
Affiliation(s)
- Wenli Wang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Qin An
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Kunlun Huang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China
| | - Yunping Dai
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Qingyong Meng
- College of Biological Sciences, China Agricultural University, Beijing, China
| | - Yali Zhang
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, China.
| |
Collapse
|
5
|
Mitchell M, Suh M, Hooda N, Bylsma LC, Cohen SS. The effect of bovine dairy products and their components on the incidence and natural history of infection: a systematic literature review. Nutr J 2024; 23:26. [PMID: 38413931 PMCID: PMC10898086 DOI: 10.1186/s12937-024-00923-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2023] [Accepted: 01/30/2024] [Indexed: 02/29/2024] Open
Abstract
BACKGROUND Dairy products and their components may impact immune function, although the current evidence base has some research gaps. As part of a larger systematic literature review of dairy products/components (including probiotics, dairy proteins, and dairy fats) and immune function, we identified the available epidemiologic research on the impact of dairy products/components on incidence and natural history of infectious diseases. METHODS PubMed and Embase databases were systematically searched through May 2022 to identify eligible studies using pre-defined Population, Intervention, Comparator, Outcomes, and Study design criteria. Herein, we focused on describing the impacts of dairy product/component on infectious disease outcomes, including the effect on leukocyte and cytokine response in humans. Risk of bias assessment was performed using the Academy of Nutrition and Dietetics Quality Criteria Checklist. The Preferred Reporting Items for Systematic Reviews and Meta-analyses (PRISMA) guidelines were followed. RESULTS Among 9,832 studies identified from the larger literature search, 133 relevant publications from 128 studies reported on dairy product/component and infectious disease outcomes. Few studies are available on the impact of non-fermented milk and traditional yogurt on infectious disease. Evidence was identified to suggest milk and yogurt drinks fermented with Lactobacillus strains reduce the risk and burden of common infectious diseases (CIDs), although the findings are mixed and difficult to reconcile due to heterogenous study populations, bacterial strains, and study methods. Few studies are available on the impact of dairy products/components on the natural history of infection, with the available findings indicating probiotics may both improve gastrointestinal symptoms among HIV-infected persons and help eradicate and alleviate the symptoms of Heliobacter (H.) pylori. The available evidence also suggests lactoferrin may reduce the virological burden of COVID-19 and hepatitis C virus. No consistent changes in leukocytes or cytokine production were observed for any type of dairy product or their components, but probiotics appeared to enhance natural killer cell levels/activity and the phagocytic process. CONCLUSIONS Dairy products, particularly those with added probiotics, may represent an easily accessible nutritional intervention to prevent and improve the course of infectious diseases. This review highlights the need for additional research in this potentially impactful area. PROSPERO REGISTRATION CRD42022333780.
Collapse
Affiliation(s)
- Meghan Mitchell
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA.
| | - Mina Suh
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Naushin Hooda
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Lauren C Bylsma
- EpidStrategies, a division of ToxStrategies, LLC, 23501 Cinco Ranch Blvd, Suite B226, Katy, TX, 77494, USA
| | - Sarah S Cohen
- Independent contractor to ToxStrategies, LLC, Durham, NC, USA
| |
Collapse
|
6
|
González L, Paredes Sosa JL, Mosquito S, Filio Y, Romero PE, Ochoa TJ, Tsukayama P. Oral lactoferrin administration does not impact the diversity or composition of the infant gut microbiota in a Peruvian cohort. Microbiol Spectr 2023; 11:e0009623. [PMID: 37882571 PMCID: PMC10715004 DOI: 10.1128/spectrum.00096-23] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 09/05/2023] [Indexed: 10/27/2023] Open
Abstract
IMPORTANCE Previous studies have suggested that oral lactoferrin enhances diversity in the gut microbiota in infants while inhibiting the growth of opportunistic pathogens. However, the effect of lactoferrin on infant gut microbiota over time has yet to be thoroughly studied. Our study suggests that lactoferrin oral treatment in infants aged 12-18 months does not affect gut microbiome diversity and composition over time. To our knowledge, this is the first study to report the effect of lactoferrin on infant gut microbiome composition over time and helps elucidate its impact on infant health and its therapeutic potential.
Collapse
Affiliation(s)
- Luis González
- Laboratorio de Genómica Microbiana, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina Tropical Alexander von Humboldt, Lima, Peru
| | - Jose Luis Paredes Sosa
- Laboratorio de Genómica Microbiana, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Susan Mosquito
- Laboratorio de Genómica Microbiana, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Yesenia Filio
- Laboratorio de Genómica Microbiana, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Pedro E. Romero
- Facultad de Ciencias Biológicas, Universidad Nacional Mayor de San Marcos, Lima, Peru
| | - Theresa J. Ochoa
- Instituto de Medicina Tropical Alexander von Humboldt, Lima, Peru
| | - Pablo Tsukayama
- Laboratorio de Genómica Microbiana, Facultad de Ciencias e Ingeniería, Universidad Peruana Cayetano Heredia, Lima, Peru
- Instituto de Medicina Tropical Alexander von Humboldt, Lima, Peru
- Parasites and Microbes Programme, Wellcome Sanger Institute, Hinxton, United Kingdom
| |
Collapse
|
7
|
Zhang Y, Lin Y, He J, Song S, Luo Y, Lu Y, Chen S, Wang Q, Li Y, Ren F, Guo H. Milk-derived small extracellular vesicles: a new perspective on dairy nutrition. Crit Rev Food Sci Nutr 2023:1-22. [PMID: 37819268 DOI: 10.1080/10408398.2023.2263573] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/13/2023]
Abstract
Milk contains bioactive compounds that have multiple essential benefits. Milk-derived small extracellular vesicles (M-sEVs) have emerged as novel bioactive milk components with various beneficial biological functions and broad applications. The M-sEVs from different mammalian sources have similar composition and bioactive functions. The digestive stability and biocompatibility of the M-sEVs provide a good foundation for their physiological functions. Evidence suggests that M-sEVs promote intestinal, immune, bone, neural, liver, and heart health and show therapeutic effects against cancer, indicating their potential for use in functional foods. In addition, M-sEVs can be developed as natural delivery carriers owing to their superior structural characteristics. Further studies are needed to elucidate the relationship between the specific components and functions of M-sEVs, standardize their extraction processes, and refine relevant clinical trials to advance the future applications of M-sEVs. This review summarizes the structure and composition of M-sEVs isolated from different milk sources and discusses several common extraction methods. Since the introduction of M-sEVs for digestion and absorption, studies have been conducted on their biological functions. Furthermore, we outline the theoretical industrial production route, potential application scenarios of M-sEVs, and the future perspectives of M-sEV research.
Collapse
Affiliation(s)
- Yuning Zhang
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yingying Lin
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Jian He
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Sijia Song
- Food Laboratory of Zhongyuan, Luohe, PR China
| | - Yujia Luo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | | | - Qingyu Wang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Sciences, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Fazheng Ren
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- Key Laboratory of Functional Dairy, College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Precision Nutrition and Food Quality, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
8
|
Conesa C, Bellés A, Grasa L, Sánchez L. The Role of Lactoferrin in Intestinal Health. Pharmaceutics 2023; 15:1569. [PMID: 37376017 DOI: 10.3390/pharmaceutics15061569] [Citation(s) in RCA: 9] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/16/2023] [Accepted: 05/19/2023] [Indexed: 06/29/2023] Open
Abstract
The intestine represents one of the first barriers where microorganisms and environmental antigens come into tight contact with the host immune system. A healthy intestine is essential for the well-being of humans and animals. The period after birth is a very important phase of development, as the infant moves from a protected environment in the uterus to one with many of unknown antigens and pathogens. In that period, mother's milk plays an important role, as it contains an abundance of biologically active components. Among these components, the iron-binding glycoprotein, lactoferrin (LF), has demonstrated a variety of important benefits in infants and adults, including the promotion of intestinal health. This review article aims to provide a compilation of all the information related to LF and intestinal health, in infants and adults.
Collapse
Affiliation(s)
- Celia Conesa
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
| | - Andrea Bellés
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| | - Laura Grasa
- Departamento de Farmacología, Fisiología y Medicina Legal y Forense, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
- Instituto de Investigación Sanitaria de Aragón (IIS Aragón), 50009 Zaragoza, Spain
| | - Lourdes Sánchez
- Departamento de Producción Animal y Ciencia de los Alimentos, Facultad de Veterinaria, Universidad de Zaragoza, 50013 Zaragoza, Spain
- Instituto Agroalimentario de Aragón IA2 (UNIZAR-CITA), 50013 Zaragoza, Spain
| |
Collapse
|
9
|
Ohradanova-Repic A, Praženicová R, Gebetsberger L, Moskalets T, Skrabana R, Cehlar O, Tajti G, Stockinger H, Leksa V. Time to Kill and Time to Heal: The Multifaceted Role of Lactoferrin and Lactoferricin in Host Defense. Pharmaceutics 2023; 15:1056. [PMID: 37111542 PMCID: PMC10146187 DOI: 10.3390/pharmaceutics15041056] [Citation(s) in RCA: 10] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Revised: 03/16/2023] [Accepted: 03/21/2023] [Indexed: 04/29/2023] Open
Abstract
Lactoferrin is an iron-binding glycoprotein present in most human exocrine fluids, particularly breast milk. Lactoferrin is also released from neutrophil granules, and its concentration increases rapidly at the site of inflammation. Immune cells of both the innate and the adaptive immune system express receptors for lactoferrin to modulate their functions in response to it. On the basis of these interactions, lactoferrin plays many roles in host defense, ranging from augmenting or calming inflammatory pathways to direct killing of pathogens. Complex biological activities of lactoferrin are determined by its ability to sequester iron and by its highly basic N-terminus, via which lactoferrin binds to a plethora of negatively charged surfaces of microorganisms and viruses, as well as to mammalian cells, both normal and cancerous. Proteolytic cleavage of lactoferrin in the digestive tract generates smaller peptides, such as N-terminally derived lactoferricin. Lactoferricin shares some of the properties of lactoferrin, but also exhibits unique characteristics and functions. In this review, we discuss the structure, functions, and potential therapeutic uses of lactoferrin, lactoferricin, and other lactoferrin-derived bioactive peptides in treating various infections and inflammatory conditions. Furthermore, we summarize clinical trials examining the effect of lactoferrin supplementation in disease treatment, with a special focus on its potential use in treating COVID-19.
Collapse
Affiliation(s)
- Anna Ohradanova-Repic
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Romana Praženicová
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Laura Gebetsberger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Tetiana Moskalets
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| | - Rostislav Skrabana
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Ondrej Cehlar
- Laboratory of Structural Biology of Neurodegeneration, Institute of Neuroimmunology, Slovak Academy of Sciences, 845 10 Bratislava, Slovakia
| | - Gabor Tajti
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Hannes Stockinger
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
| | - Vladimir Leksa
- Institute for Hygiene and Applied Immunology, Center for Pathophysiology, Infectiology and Immunology, Medical University of Vienna, 1090 Vienna, Austria
- Laboratory of Molecular Immunology, Institute of Molecular Biology, Slovak Academy of Sciences, 845 51 Bratislava, Slovakia
| |
Collapse
|
10
|
Li W, Liu B, Lin Y, Xue P, Lu Y, Song S, Li Y, Szeto IMY, Ren F, Guo H. The application of lactoferrin in infant formula: The past, present and future. Crit Rev Food Sci Nutr 2022; 64:5748-5767. [PMID: 36533432 DOI: 10.1080/10408398.2022.2157792] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Human milk is universally regarded as the gold standard to fulfill nutrition needs of infants. Lactoferrin (LF) is a major multiple bioactive glycoprotein in human milk but little is presented in infant formula. LF can resist digestion in the infant gastrointestinal tract and is absorbed into the bloodstream in an intact form to perform physiological functions. Evidence suggest that LF prevents pathogen infection, promotes immune system development, intestinal development, brain development and bone health, as well as ameliorates iron deficiency anemia. However, more clinical studies of LF need to be further elucidated to determine an appropriate dosage for application in infant formula. LF is sensitive to denaturation induced by processing of infant formula such as heat treatments and spay drying. Thus, further studies should be focus on maximizing the retention of LF activity in the infant formula process. This review summarizes the structural features of LF. Then the digestion, absorption and metabolism of LF in infants are discussed, followed by the function of LF for infants. Further, we summarize LF in infant formula and effects of processing of infant formula on bioactivities of LF, as well as future perspectives of LF research.
Collapse
Affiliation(s)
- Wusun Li
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Biao Liu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
| | - Yingying Lin
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Peng Xue
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yao Lu
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Sijia Song
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
| | - Yixuan Li
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Ignatius Man-Yau Szeto
- Inner Mongolia Yili Industrial Group Co., Ltd, Hohhot, PR China
- National Center of Technology Innovation for Dairy, Hohhot, PR China
| | - Fazheng Ren
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| | - Huiyuan Guo
- College of Food Science and Nutritional Engineering, China Agricultural University, Beijing, PR China
- Key Laboratory of Functional Dairy, Department of Nutrition and Health, China Agricultural University, Beijing, PR China
| |
Collapse
|
11
|
Avalos-Gómez C, Ramírez-Rico G, Ruiz-Mazón L, Sicairos NL, Serrano-Luna J, de la Garza M. Lactoferrin: An Effective Weapon in the Battle Against Bacterial Infections. Curr Pharm Des 2022; 28:3243-3260. [PMID: 36284379 DOI: 10.2174/1381612829666221025153216] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 09/27/2022] [Indexed: 01/28/2023]
Abstract
The emergence of multidrug-resistant bacterial strains with respect to commercially available antimicrobial drugs has marked a watershed in treatment therapies to fight pathogens and has stimulated research on alternative remedies. Proteins of the innate immune system of mammals have been highlighted as potentially yielding possible treatment options for infections. Lactoferrin (Lf) is one of these proteins; interestingly, no resistance to it has been found. Lf is a conserved cationic nonheme glycoprotein that is abundant in milk and is also present in low quantities in mucosal secretions. Moreover, Lf is produced and secreted by the secondary granules of neutrophils at infection sites. Lf is a molecule of approximately 80 kDa that displays multiple functions, such as antimicrobial, anti-viral, anti-inflammatory, and anticancer actions. Lf can synergize with antibiotics, increasing its potency against bacteria. Lactoferricins (Lfcins) are peptides resulting from the N-terminal end of Lf by proteolytic cleavage with pepsin. They exhibit several anti-bacterial effects similar to those of the parental glycoprotein. Synthetic analog peptides exhibiting potent antimicrobial properties have been designed. The aim of this review is to update understanding of the structure and effects of Lf and Lfcins as anti-bacterial compounds, focusing on the mechanisms of action in bacteria and the use of Lf in treatment of infections in patients, including those studies where no significant differences were found. Lf could be an excellent option for prevention and treatment of bacterial diseases, mainly in combined therapies with antibiotics or other antimicrobials.
Collapse
Affiliation(s)
- Christian Avalos-Gómez
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de Mexico, Mexico
| | - Gerardo Ramírez-Rico
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de Mexico, Mexico.,Facultad de Estudios Superiores Cuautitlán, Universidad Nacional Autónoma de México (UNAM), Km 2.5 Carretera Cuautitlán- Teoloyucan, Cuautitlán Izcalli, 54714, Mexico
| | - Lucero Ruiz-Mazón
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de Mexico, Mexico
| | - Nidia León Sicairos
- Departamento de Investigación, Hospital Pediátrico de Sinaloa, Unidad de Investigación, Facultad de Medicina, Universidad Autónoma de Sinaloa, Mexico
| | - Jesús Serrano-Luna
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de Mexico, Mexico
| | - Mireya de la Garza
- Departamento de Biología Celular, Centro de Investigación y de Estudios Avanzados del IPN, Ciudad de Mexico, Mexico
| |
Collapse
|
12
|
Zuiderwijk MO, van der Burg M, Bekker V, Schoenaker MHD. Regulatory T Cells in Development and Prediction of Necrotizing Enterocolitis in Preterm Neonates: A Scoping Review. Int J Mol Sci 2022; 23:10903. [PMID: 36142816 PMCID: PMC9504949 DOI: 10.3390/ijms231810903] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2022] [Revised: 09/11/2022] [Accepted: 09/14/2022] [Indexed: 11/17/2022] Open
Abstract
Necrotizing enterocolitis (NEC) is a leading cause of mortality in premature infants. However, the pathophysiology and influence of regulatory T cells (Tregs) have not been sufficiently elucidated. We performed a scoping review to investigate current knowledge on the influence of Tregs in NEC, and to investigate the predictive value of Treg number in NEC development. Pubmed, Embase, Prospero and Cochrane Library were searched during December 2020. Primary research articles discussing Tregs and NEC development written in English were selected. Two reviewers screened title and abstract for relevance, after which full-text screening was performed. A total of 20 articles were selected-13 of the articles discussed studies performed in animal models, while 8 used human neonate data. One study discussed both animal and human data. It was shown that after NEC diagnosis or induction, Treg levels were decreased while Th17 levels were increased. No studies were found which investigated the predictive value of Treg number in NEC development. A reduced Treg level is found in animals and neonates with NEC. The question remains whether this effect is a factor on the causal pathway of NEC development or a bystander effect. Future research focusing on the pathophysiological timeline of NEC and the involvement of Tregs is required for better understanding of this disease.
Collapse
Affiliation(s)
- Mara O. Zuiderwijk
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Mirjam van der Burg
- Willem Alexander Children’s Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Vincent Bekker
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| | - Michiel H. D. Schoenaker
- Willem Alexander Children’s Hospital, Division of Neonatology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
- Willem Alexander Children’s Hospital, Laboratory for Pediatric Immunology, Department of Pediatrics, Leiden University Medical Center, 2333 ZA Leiden, The Netherlands
| |
Collapse
|
13
|
Martínez-García JJ, Canizalez-Roman A, Angulo-Zamudio UA, Velazquez-Roman J, Flores-Villaseñor H, Valdez-Flores MA, Rios-Burgueño E, Moran-Portela D, León-Sicairos N. Lactoferrin and Metoprolol Supplementation Increase Mouse Survival in an Experimental LPS-Induced Sepsis Model. Int J Pept Res Ther 2022; 28:141. [DOI: 10.1007/s10989-022-10447-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/13/2022] [Indexed: 11/27/2022]
|
14
|
Fitzgerald FC, Zingg W, Chimhini G, Chimhuya S, Wittmann S, Brotherton H, Olaru ID, Neal SR, Russell N, da Silva ARA, Sharland M, Seale AC, Cotton MF, Coffin S, Dramowski A. The Impact of Interventions to Prevent Neonatal Healthcare-associated Infections in Low- and Middle-income Countries: A Systematic Review. Pediatr Infect Dis J 2022; 41:S26-S35. [PMID: 35134037 PMCID: PMC8815829 DOI: 10.1097/inf.0000000000003320] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 08/12/2021] [Indexed: 12/14/2022]
Abstract
BACKGROUND Clinically suspected and laboratory-confirmed bloodstream infections are frequent causes of morbidity and mortality during neonatal care. The most effective infection prevention and control interventions for neonates in low- and middle-income countries (LMIC) are unknown. AIM To identify effective interventions in the prevention of hospital-acquired bloodstream infections in LMIC neonatal units. METHODS Medline, PUBMED, the Cochrane Database of Systematic Reviews, EMBASE and PsychInfo (January 2003 to October 2020) were searched to identify studies reporting single or bundled interventions for prevention of bloodstream infections in LMIC neonatal units. RESULTS Our initial search identified 5206 articles; following application of filters, 27 publications met the inclusion and Integrated Quality Criteria for the Review of Multiple Study Designs assessment criteria and were summarized in the final analysis. No studies were carried out in low-income countries, only 1 in Sub-Saharan Africa and just 2 in multiple countries. Of the 18 single-intervention studies, most targeted skin (n = 4) and gastrointestinal mucosal integrity (n = 5). Whereas emollient therapy and lactoferrin achieved significant reductions in proven neonatal infection, glutamine and mixed probiotics showed no benefit. Chlorhexidine gluconate for cord care and kangaroo mother care reduced infection in individual single-center studies. Of the 9 studies evaluating bundles, most focused on prevention of device-associated infections and achieved significant reductions in catheter- and ventilator-associated infections. CONCLUSIONS There is a limited evidence base for the effectiveness of infection prevention and control interventions in LMIC neonatal units; bundled interventions targeting device-associated infections were most effective. More multisite studies with robust study designs are needed to inform infection prevention and control intervention strategies in low-resource neonatal units.
Collapse
Affiliation(s)
- Felicity C. Fitzgerald
- From the Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
- Biomedical Research and Training Institute, Harare, Zimbabwe
| | - Walter Zingg
- Department of Infectious Diseases and Hospital Epidemiology, University Hospital Zurich, University of Zurich, Zurich, Switzerland
| | - Gwendoline Chimhini
- Department of Paediatrics and Child Health, University of Zimbabwe College of Health Sciences, Zimbabwe
| | - Simbarashe Chimhuya
- Department of Paediatrics and Child Health, University of Zimbabwe College of Health Sciences, Zimbabwe
| | - Stefanie Wittmann
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Helen Brotherton
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
- MRC Unit, The Gambia at the London School of Hygiene & Tropical Medicine, Fajara, The Gambia
| | - Ioana D. Olaru
- Biomedical Research and Training Institute, Harare, Zimbabwe
- Clinical Research Department, London School of Hygiene and Tropical Medicine, London, United Kingdom
| | - Samuel R. Neal
- From the Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Neal Russell
- Paediatric Infectious Diseases Research Group, St George’s University of London, United Kingdom
| | - André Ricardo Araujo da Silva
- Laboratory of Teaching of Prevention and Control of Healthcare-Associated Infections, Federal Fluminense University, Brazil
| | - Mike Sharland
- Paediatric Infectious Diseases Research Group, St George’s University of London, United Kingdom
| | - Anna C. Seale
- From the Department of Infection, Immunity and Inflammation, UCL Great Ormond Street Institute of Child Health, London, United Kingdom
| | - Mark F. Cotton
- Department of Paediatrics and Child Health, Division of Paediatric Infectious Diseases, Stellenbosch University, South Africa, and
| | - Susan Coffin
- Children’s Hospital of Philadelphia, Pennsylvania, Philadelphia
| | - Angela Dramowski
- Department of Paediatrics and Child Health, Division of Paediatric Infectious Diseases, Stellenbosch University, South Africa, and
| |
Collapse
|
15
|
Healy DB, Ryan CA, Ross RP, Stanton C, Dempsey EM. Clinical implications of preterm infant gut microbiome development. Nat Microbiol 2022; 7:22-33. [PMID: 34949830 DOI: 10.1038/s41564-021-01025-4] [Citation(s) in RCA: 58] [Impact Index Per Article: 29.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2021] [Accepted: 11/12/2021] [Indexed: 12/12/2022]
Abstract
Perturbations to the infant gut microbiome during the first weeks to months of life affect growth, development and health. In particular, assembly of an altered intestinal microbiota during infant development results in an increased risk of immune and metabolic diseases that can persist into childhood and potentially into adulthood. Most research into gut microbiome development has focused on full-term babies, but health-related outcomes are also important for preterm babies. The systemic physiological immaturity of very preterm gestation babies (born earlier than 32 weeks gestation) results in numerous other microbiome-organ interactions, the mechanisms of which have yet to be fully elucidated or in some cases even considered. In this Perspective, we compare assembly of the intestinal microbiome in preterm and term infants. We focus in particular on the clinical implications of preterm infant gut microbiome composition and discuss the prospects for microbiome diagnostics and interventions to improve the health of preterm babies.
Collapse
Affiliation(s)
- David B Healy
- APC Microbiome Ireland, University College Cork, Cork, Ireland. .,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.
| | - C Anthony Ryan
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland
| | - R Paul Ross
- APC Microbiome Ireland, University College Cork, Cork, Ireland
| | - Catherine Stanton
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Teagasc Food Research Centre, Moorepark, Fermoy, Ireland
| | - Eugene M Dempsey
- APC Microbiome Ireland, University College Cork, Cork, Ireland.,Department of Paediatrics and Child Health, University College Cork, Cork, Ireland.,INFANT Research Centre, Cork University Hospital, Cork, Ireland
| |
Collapse
|
16
|
Colonization of fecal microbiota from patients with neonatal necrotizing enterocolitis exacerbates intestinal injury in germfree mice subjected to necrotizing enterocolitis-induction protocol via alterations in butyrate and regulatory T cells. J Transl Med 2021; 19:510. [PMID: 34922582 PMCID: PMC8684079 DOI: 10.1186/s12967-021-03109-5] [Citation(s) in RCA: 26] [Impact Index Per Article: 8.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2021] [Accepted: 10/06/2021] [Indexed: 12/25/2022] Open
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) remains a life-threatening disease in neonates. Numerous studies have shown a correlation between the intestinal microbiota and NEC, but the causal link remains unclear. This study aimed to demonstrate the causal role of gut microbiota in NEC and explore potential mechanisms involved. METHODS Eighty-one fecal samples from patients with NEC and eighty-one matched controls (matched to the NEC infants by gestational age, birth weight, date of birth, mode of delivery and feeding patterns) were collected. To explore if altered gut microbiota contributes to the pathogenesis of NEC, fecal microbiota transplantation (FMT) was carried out in germ-free (GF) mice prior to a NEC-induction protocol that included exposure to hypoxia and cold stress. Butyric acid was also administered to demonstrate its role in NEC. The fecal microbiota from patients and mice were analyzed by 16S rRNA gene sequencing analysis. Short chain fatty acid (SCFA) levels were measured by gas chromatography-mass spectrometry (GC-MS). The ontogeny of T cells and regulatory T cells (Tregs) in lamina propria mononuclear cells (LPMC) from the ileum of patients and mice were isolated and analyzed by flow cytometry.The transcription of inflammatory cytokines was quantified by qRT-PCR. RESULTS NEC patients had increased Proteobacteria and decreased Firmicutes and Bacteroidetes compared to fecal control samples, and the level of butyric acid in the NEC group was lower than the control group. FMT in GF mice with samples from NEC patients achieved a higher histological injury scores when compared to mice that received FMT with control samples. Alterations in microbiota and butyrate levels were maintained in mice following FMT. The ratio of Treg/CD4+T (Thelper) cells was reduced in both NEC patients and mice modeling NEC following FMT. CONCLUSIONS The microbiota was found to have NEC and the microbial butyrate-Treg axis was identified as a potential mechanism for the observed effects.
Collapse
|
17
|
Zeinali LI, Giuliano S, Lakshminrusimha S, Underwood MA. Intestinal Dysbiosis in the Infant and the Future of Lacto-Engineering to Shape the Developing Intestinal Microbiome. Clin Ther 2021; 44:193-214.e1. [PMID: 34922744 DOI: 10.1016/j.clinthera.2021.11.005] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2021] [Revised: 11/06/2021] [Accepted: 11/12/2021] [Indexed: 12/16/2022]
Abstract
PURPOSE The goal of this study was to review the role of human milk in shaping the infant intestinal microbiota and the potential of human milk bioactive molecules to reverse trends of increasing intestinal dysbiosis and dysbiosis-associated diseases. METHODS This narrative review was based on recent and historic literature. FINDINGS Human milk immunoglobulins, oligosaccharides, lactoferrin, lysozyme, milk fat globule membranes, and bile salt-stimulating lipase are complex multifunctional bioactive molecules that, among other important functions, shape the composition of the infant intestinal microbiota. IMPLICATIONS The co-evolution of human milk components and human milk-consuming commensal anaerobes many thousands of years ago resulted in a stable low-diversity infant microbiota. Over the past century, the introduction of antibiotics and modern hygiene practices plus changes in the care of newborns have led to significant alterations in the intestinal microbiota, with associated increases in risk of dysbiosis-associated disease. A better understanding of mechanisms by which human milk shapes the intestinal microbiota of the infant during a vulnerable period of development of the immune system is needed to alter the current trajectory and decrease intestinal dysbiosis and associated diseases.
Collapse
Affiliation(s)
- Lida I Zeinali
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA
| | | | | | - Mark A Underwood
- Department of Pediatrics, UC Davis School of Medicine, Sacramento, CA, USA.
| |
Collapse
|
18
|
Wang Y, Ze X, Rui B, Li X, Zeng N, Yuan J, Li W, Yan J, Li M. Studies and Application of Sialylated Milk Components on Regulating Neonatal Gut Microbiota and Health. Front Nutr 2021; 8:766606. [PMID: 34859034 PMCID: PMC8631720 DOI: 10.3389/fnut.2021.766606] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2021] [Accepted: 10/18/2021] [Indexed: 12/13/2022] Open
Abstract
Breast milk is rich in sialic acids (SA), which are commonly combined with milk oligosaccharides and glycoconjugates. As a functional nutrient component, SA-containing milk components have received increasing attention in recent years. Sialylated human milk oligosaccharides (HMOs) have been demonstrated to promote the growth and metabolism of beneficial gut microbiota in infants, bringing positive outcomes to intestinal health and immune function. They also exhibit antiviral and bacteriostatic activities in the intestinal mucosa of new-borns, thereby inhibiting the adhesion of pathogens to host cells. These properties play a pivotal role in regulating the intestinal microbial ecosystem and preventing the occurrence of neonatal inflammatory diseases. In addition, some recent studies also support the promoting effects of sialylated HMOs on neonatal bone and brain development. In addition to HMOs, sialylated glycoproteins and glycolipids are abundant in milk, and are also critical to neonatal health. This article reviews the current research progress in the regulation of sialylated milk oligosaccharides and glycoconjugates on neonatal gut microbiota and health.
Collapse
Affiliation(s)
- Yushuang Wang
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xiaolei Ze
- Science and Technology Centre, By-Health Co., Ltd., Guangzhou, China
| | - Binqi Rui
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Xinke Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Nina Zeng
- Science and Technology Centre, By-Health Co., Ltd., Guangzhou, China
| | - Jieli Yuan
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Wenzhe Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| | - Jingyu Yan
- Key Laboratory of Separation Science for Analytical Chemistry, Dalian Institute of Chemical Physics, Chinese Academy of Sciences (CAS), Dalian, China
| | - Ming Li
- College of Basic Medical Science, Dalian Medical University, Dalian, China
| |
Collapse
|
19
|
Jang YS, Song HE, Seo GY, Jo HJ, Park S, Park HW, Kim TG, Kang SG, Yoon SI, Ko HJ, Lee GS, Park SR, Kim PH. Lactoferrin Potentiates Inducible Regulatory T Cell Differentiation through TGF-β Receptor III Binding and Activation of Membrane-Bound TGF-β. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2021; 207:2456-2464. [PMID: 34615735 DOI: 10.4049/jimmunol.2100326] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Accepted: 09/03/2021] [Indexed: 11/19/2022]
Abstract
Lactoferrin (LF) is known to possess anti-inflammatory activity, although its mechanisms of action are not well-understood. The present study asked whether LF affects the commitment of inducible regulatory T cells (Tregs). LF substantially promoted Foxp3 expression by mouse activated CD4+T cells, and this activity was further enhanced by TGF-β1. Interestingly, blocking TGF-β with anti-TGF-β Ab completely abolished LF-induced Foxp3 expression. However, no significant amount of soluble TGF-β was released by LF-stimulated T cells, suggesting that membrane TGF-β (mTGF-β) is associated. Subsequently, it was found that LF binds to TGF-β receptor III, which induces reactive oxygen species production and diminishes the expression of mTGF-β-bound latency-associated peptide, leading to the activation of mTGF-β. It was followed by phosphorylation of Smad3 and enhanced Foxp3 expression. These results suggest that LF induces Foxp3+ Tregs through TGF-β receptor III/reactive oxygen species-mediated mTGF-β activation, triggering canonical Smad3-dependent signaling. Finally, we found that the suppressive activity of LF-induced Tregs is facilitated mainly by CD39/CD73-induced adenosine generation and that this suppressor activity alleviates inflammatory bowel disease.
Collapse
Affiliation(s)
- Young-Saeng Jang
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea;
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| | - Ha-Eon Song
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Goo-Young Seo
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyeon-Ju Jo
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Sunhee Park
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Hui-Won Park
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Tae-Gyu Kim
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Seung-Goo Kang
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Sung-Il Yoon
- Division of Biomedical Convergence, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea
| | - Hyun-Jeong Ko
- College of Pharmacy, Kangwon National University, Chuncheon, Republic of Korea
| | - Geun-Shik Lee
- College of Veterinary Medicine, Kangwon National University, Chuncheon, Republic of Korea; and
| | - Seok-Rae Park
- Department of Microbiology, College of Medicine, Konyang University, Daejeon, Republic of Korea
| | - Pyeung-Hyeun Kim
- Department of Molecular Bioscience, College of Biomedical Science, Kangwon National University, Chuncheon, Republic of Korea;
- Institute of Bioscience and Biotechnology, Kangwon National University, Chuncheon, Republic of Korea
| |
Collapse
|
20
|
Ismail RIH, Awad HA, Imam SS, Gad GI, Aboushady NM, Abdou RM, Eissa DS, Azzam NT, Barakat MM, Yassin MM, Barakat NM. Gut priming with bovine colostrum and T regulatory cells in preterm neonates: a randomized controlled trial. Pediatr Res 2021; 90:650-656. [PMID: 33446924 DOI: 10.1038/s41390-020-01344-y] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/10/2020] [Revised: 11/25/2020] [Accepted: 12/09/2020] [Indexed: 01/29/2023]
Abstract
BACKGROUND Necrotizing enterocolitis (NEC) and neonatal sepsis are still considered major problems, especially in formula-fed preterm neonates. This study aimed to investigate the effect of bovine colostrum on T regulatory cells, NEC, and late-onset sepsis in preterm neonates ≤34 weeks. METHODS This prospective double-blind randomized controlled trial was conducted on 80 preterm infants who were randomly assigned to either the bovine colostrum group (n = 32) or control group (n = 48). T lymphocytes and their subsets, necrotizing enterocolitis, late-onset sepsis (LOS) and its severity, feeding tolerance, growth, length of hospital stay, and mortality were documented. RESULTS The bovine colostrum group showed higher follow-up levels of CD4+CD25+ FOXP3+ T lymphocyte % (FOXP3 Tregs). FOXP3 Tregs and its difference in change levels between baseline and follow-up were considered as the most related factors to the bovine colostrum. Bovine colostrum group showed positive trends for reduction of sepsis severity and mortality with no significant difference in the incidence of NEC, LOS, and length of hospital stay. CONCLUSIONS Preterm neonates who received bovine colostrum showed a higher FOXP3 Treg level. IMPACT Bovine colostrum has no significant effect on the incidence of necrotizing enterocolitis. FOXP3 T regulatory cells and their increased level between baseline and follow-up is considered as the most influencing factors related to the bovine colostrum. Positive trends were noted for reduction of sepsis severity and concomitant mortality, but the study lacked the power to assess these outcomes.
Collapse
Affiliation(s)
- Rania I H Ismail
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Hisham A Awad
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Safaa S Imam
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Ghada I Gad
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nancy M Aboushady
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Rania M Abdou
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Deena S Eissa
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nesmahar T Azzam
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | | | | | - Noha M Barakat
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt.
| |
Collapse
|
21
|
Zimecki M, Actor JK, Kruzel ML. The potential for Lactoferrin to reduce SARS-CoV-2 induced cytokine storm. Int Immunopharmacol 2021; 95:107571. [PMID: 33765614 PMCID: PMC7953442 DOI: 10.1016/j.intimp.2021.107571] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 03/05/2021] [Accepted: 03/05/2021] [Indexed: 02/07/2023]
Abstract
The COVID-19 pandemic is a serious global health threat caused by severe acute respiratory syndrome of coronavirus 2 (SARS-CoV-2). Symptoms of COVID-19 are highly variable with common hyperactivity of immune responses known as a "cytokine storm". In fact, this massive release of inflammatory cytokines into in the pulmonary alveolar structure is a main cause of mortality during COVID-19 infection. Current management of COVID-19 is supportive and there is no common clinical protocol applied to suppress this pathological state. Lactoferrin (LF), an iron binding protein, is a first line defense protein that is present in neutrophils and excretory fluids of all mammals, and is well recognized for its role in maturation and regulation of immune system function. Also, due to its ability to sequester free iron, LF is known to protect against insult-induced oxidative stress and subsequent "cytokine storm" that results in dramatic necrosis within the affected tissue. Review of the literature strongly suggests utility of LF to silence the "cytokine storm", giving credence to both prophylactic and therapeutic approaches towards combating COVID-19 infection.
Collapse
Affiliation(s)
- Michał Zimecki
- The Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, Wrocław, Poland
| | - Jeffrey K Actor
- University of Texas, Health Science Center Houston, Texas, USA.
| | - Marian L Kruzel
- University of Texas, Health Science Center Houston, Texas, USA
| |
Collapse
|
22
|
Nayak A, Tiozzo C, Lin X, Mejia C, Gurzenda E, Kim M, Hanna N. Is Lactoferrin Supplementation Beneficial for All Preterm Infants? Am J Perinatol 2021; 40:680-687. [PMID: 34058763 DOI: 10.1055/s-0041-1730433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
OBJECTIVE Human milk (HM) has antibacterial properties due to the presence of immune-modulators, including lactoferrin (LF). This study will determine effect(s) of HM maturation, fortification, and storage conditions on LF levels and its antibacterial properties. STUDY DESIGN HM samples (n = 30) were obtained from preterm and term mothers. The LF levels were analyzed by ELISA, and the antibacterial activity was measured after inoculation with Escherichia coli. RESULTS The highest level of LF in preterm HM was observed in the first week of lactation. However, storage of preterm HM at 4°C decreased LF levels significantly. Both LF levels and antibacterial activity in preterm HM was lower compared with term HM, but significantly higher than donor HM even after HM-based fortification. LF supplementation of donor HM improved its antibacterial activity. CONCLUSION Preterm infants fed donor HM, formula, or stored HM at 4°C may benefits from LF supplementation to improve HM antibacterial properties. KEY POINTS · Milk LF levels vary with storage and maturity.. · Donor milk is deficient in LF even after adding HM-based fortification.. · Donor HM and formula fed infants may benefit from LF..
Collapse
Affiliation(s)
- Amrita Nayak
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Caterina Tiozzo
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Xinhua Lin
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Claudia Mejia
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Ellen Gurzenda
- Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| | - Maureen Kim
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York
| | - Nazeeh Hanna
- Department of Pediatrics, NYU Langone Hospital-Long Island, Mineola, New York.,Women and Children Research Laboratory, NYU Long Island School of Medicine, Mineola, New York
| |
Collapse
|
23
|
de Lange IH, van Gorp C, Eeftinck Schattenkerk LD, van Gemert WG, Derikx JPM, Wolfs TGAM. Enteral Feeding Interventions in the Prevention of Necrotizing Enterocolitis: A Systematic Review of Experimental and Clinical Studies. Nutrients 2021; 13:1726. [PMID: 34069699 PMCID: PMC8161173 DOI: 10.3390/nu13051726] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 05/14/2021] [Accepted: 05/15/2021] [Indexed: 12/11/2022] Open
Abstract
Necrotizing enterocolitis (NEC), which is characterized by severe intestinal inflammation and in advanced stages necrosis, is a gastrointestinal emergency in the neonate with high mortality and morbidity. Despite advancing medical care, effective prevention strategies remain sparse. Factors contributing to the complex pathogenesis of NEC include immaturity of the intestinal immune defense, barrier function, motility and local circulatory regulation and abnormal microbial colonization. Interestingly, enteral feeding is regarded as an important modifiable factor influencing NEC pathogenesis. Moreover, breast milk, which forms the currently most effective prevention strategy, contains many bioactive components that are known to support neonatal immune development and promote healthy gut colonization. This systematic review describes the effect of different enteral feeding interventions on the prevention of NEC incidence and severity and the effect on pathophysiological mechanisms of NEC, in both experimental NEC models and clinical NEC. Besides, pathophysiological mechanisms involved in human NEC development are briefly described to give context for the findings of altered pathophysiological mechanisms of NEC by enteral feeding interventions.
Collapse
Affiliation(s)
- Ilse H. de Lange
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Charlotte van Gorp
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
| | - Laurens D. Eeftinck Schattenkerk
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Wim G. van Gemert
- European Surgical Center Aachen/Maastricht, Department of Pediatric Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), 6202 AZ Maastricht, The Netherlands; (I.H.d.L.); (W.G.v.G.)
- Department of Surgery, School for Nutrition, Toxicology and Metabolism (NUTRIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| | - Joep P. M. Derikx
- Department of Pediatric Surgery, Emma Children’s Hospital, Amsterdam UMC, University of Amsterdam and Vrije Universiteit Amsterdam, 1105 AZ Amsterdam, The Netherlands; (L.D.E.S.); (J.P.M.D.)
| | - Tim G. A. M. Wolfs
- Department of Pediatrics, School of Oncology and Developmental Biology (GROW), Maastricht University, 6202 AZ Maastricht, The Netherlands;
- Department of Biomedical Engineering (BMT), School for Cardiovascular Diseases (CARIM), Maastricht University, 6202 AZ Maastricht, The Netherlands
| |
Collapse
|
24
|
Razak A, Hussain A. Lactoferrin Supplementation to Prevent Late-Onset Sepsis in Preterm Infants: A Meta-Analysis. Am J Perinatol 2021; 38:283-290. [PMID: 31529448 DOI: 10.1055/s-0039-1696676] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
OBJECTIVE This study aimed to systematically review and meta-analyze the role of lactoferrin supplementation to prevent late-onset sepsis (LOS) in preterm infants. STUDY DESIGN Database search include PubMed, Web of Science, and Cochrane central for randomized clinical trial (RCTs). The Cochrane Grading of Recommendations Assessment, Development, and Evaluation methodology was used for summarizing the results. RESULTS Ten RCTs involving 3,679 infants were included. Lactoferrin supplementation with or without probiotics decreased all LOS (relative risk [RR]: 0.56; 95% confidence interval [CI]: 0.36-0.86; I 2 = 58%; 10 studies; 3,470 subjects; level of evidence [LOE]: low) significantly. Similarly, lactoferrin supplementation without probiotics decreased all LOS (RR: 0.43; 95% CI: 0.29-0.62; I 2 = 0%; 8 studies; 1,209 subjects; LOE: moderate) significantly. Lactoferrin supplementation did not significantly reduce necrotizing enterocolitis (RR: 0.62; 95% CI: 0.29-1.33; I 2 = 43%; 6 studies; 3,079 subjects; LOE: low), all-cause mortality (RR: 0.74; 95% CI: 0.36-1.53; I 2 = 53%; 8 studies; 3,395 subjects; LOE: very low), bronchopulmonary dysplasia (RR: 1; 95% CI: 0.90-1.13; I 2 = 0%; 4 studies; 2,570 subjects; LOE: moderate), and threshold retinopathy of prematurity eligible for surgical treatment (RR: 0.61; 95% CI: 0.25-1.51; I 2 = 74%; 2 studies; 2,481 subjects; LOE: very low). CONCLUSION Low to moderate quality evidence suggests that lactoferrin supplementation reduces LOS in preterm infants. Further research is needed to improve the certainty in the evidence.
Collapse
Affiliation(s)
- Abdul Razak
- Division of Neonatology, Department of Pediatrics, King Abdullah bin Abdulaziz University Hospital, Princess Nourah Bint Abdulrahman University, Riyadh, Saudi Arabia.,Department of Pediatrics, McMaster University, Hamilton, Ontario, Canada
| | - Asif Hussain
- Department of Orthopedics, Bhimrao Ambedkar University, Agra, India
| |
Collapse
|
25
|
Young L, McGuire W, Fowlie PW. Commentary on "Enteral Lactoferrin Supplementation for Prevention of Sepsis and Necrotizing Enterocolitis in Preterm Infants". Neonatology 2021; 118:139-142. [PMID: 33561861 DOI: 10.1159/000512988] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Accepted: 11/11/2020] [Indexed: 11/19/2022]
Affiliation(s)
- Lauren Young
- Department of Neonatal Medicine, Evelina London Children's Hospital, St Thomas' Hospital, London, United Kingdom
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, United Kingdom,
| | - Peter W Fowlie
- Department of Paediatrics, Ninewells Hospital and Medical School, Dundee, United Kingdom
| |
Collapse
|
26
|
Abstract
Mucosal surfaces are distinctive sites exposed to environmental, dietary, and microbial antigens. Particularly in the gut, the host continuously actively adapts via complex interactions between the microbiota and dietary compounds and immune and other tissue cells. Regulatory T cells (Tregs) are critical for tuning the intestinal immune response to self- and non-self-antigens in the intestine. Its importance in intestinal homeostasis is illustrated by the onset of overt inflammation caused by deficiency in Treg generation, function, or stability in the gut. A substantial imbalance in Tregs has been observed in intestinal tissue during pathogenic conditions, when a tightly regulated and equilibrated system becomes dysregulated and leads to unimpeded and chronic immune responses. In this chapter, we compile and critically discuss the current knowledge on the key factors that promote Treg-mediated tolerance in the gut, such as those involved in intestinal Treg differentiation, specificity and suppressive function, and their immunophenotype during health and disease. We also discuss the current state of knowledge on Treg dysregulation in human intestine during pathological states such as inflammatory bowel disease (IBD), necrotizing enterocolitis (NEC), graft-versus-host disease (GVHD), and colorectal cancer (CRC), and how that knowledge is guiding development of Treg-targeted therapies to treat or prevent intestinal disorders.
Collapse
|
27
|
Cutone A, Ianiro G, Lepanto MS, Rosa L, Valenti P, Bonaccorsi di Patti MC, Musci G. Lactoferrin in the Prevention and Treatment of Intestinal Inflammatory Pathologies Associated with Colorectal Cancer Development. Cancers (Basel) 2020; 12:E3806. [PMID: 33348646 PMCID: PMC7766217 DOI: 10.3390/cancers12123806] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Revised: 12/15/2020] [Accepted: 12/15/2020] [Indexed: 12/20/2022] Open
Abstract
The connection between inflammation and cancer is well-established and supported by genetic, pharmacological and epidemiological data. The inflammatory bowel diseases (IBDs), including Crohn's disease and ulcerative colitis, have been described as important promoters for colorectal cancer development. Risk factors include environmental and food-borne mutagens, dysbalance of intestinal microbiome composition and chronic intestinal inflammation, with loss of intestinal epithelial barrier and enhanced cell proliferation rate. Therapies aimed at shutting down mucosal inflammatory response represent the foundation for IBDs treatment. However, when applied for long periods, they can alter the immune system and promote microbiome dysbiosis and carcinogenesis. Therefore, it is imperative to find new safe substances acting as both potent anti-inflammatory and anti-pathogen agents. Lactoferrin (Lf), an iron-binding glycoprotein essential in innate immunity, is generally recognized as safe and used as food supplement due to its multifunctionality. Lf possesses a wide range of immunomodulatory and anti-inflammatory properties against different aseptic and septic inflammatory pathologies, including IBDs. Moreover, Lf exerts anti-adhesive, anti-invasive and anti-survival activities against several microbial pathogens that colonize intestinal mucosa of IBDs patients. This review focuses on those activities of Lf potentially useful for the prevention/treatment of intestinal inflammatory pathologies associated with colorectal cancer development.
Collapse
Affiliation(s)
- Antimo Cutone
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Giusi Ianiro
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| | - Maria Stefania Lepanto
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Luigi Rosa
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | - Piera Valenti
- Department of Public Health and Infectious Diseases, University of Rome La Sapienza, 00185 Rome, Italy; (M.S.L.); (L.R.); (P.V.)
| | | | - Giovanni Musci
- Department of Biosciences and Territory, University of Molise, 86090 Pesche, Italy; (A.C.); (G.I.)
| |
Collapse
|
28
|
Ochoa T, Loli S, Mendoza K, Carcamo C, Bellomo S, Cam L, Castaneda A, Campos M, Jacobs J, Cossey V, Zegarra J. Effect of bovine lactoferrin on prevention of late-onset sepsis in infants <1500 g: a pooled analysis of individual patient data from two randomized controlled trials. Biochem Cell Biol 2020; 99:14-19. [PMID: 32931708 DOI: 10.1139/bcb-2020-0046] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
We previously conducted two randomized controlled trials with bovine lactoferrin (bLF) for the prevention of late-onset sepsis (LOS) in infants with a birth weight <2500 g (Study 1) and <2000 g (Study 2). The aim of this study was to determine the preventative effects of bLF on culture-proven or probable LOS in infants with a birth weight <1500 g from both studies, and to determine the effect of bLF in relation to intake of human milk. Both trial designs had similar inclusion and exclusion criteria, the same dose of bLF [200 mg·(kg body mass)-1·day-1], and used the same control (maltodextrin). We fitted multivariate Cox regression models to estimate the effect of bLF on the risk of development of the composite outcome, adjusting for covariates. We included 335 neonates with a mean birth weight of 1162 ± 244 g; 27.5% were <1000 g. There were 33 first episodes of LOS in the bLF treatment group and 48 in the control group (19.5% vs. 28.9%). bLF had a protective effect on the risk of development of LOS [hazard ratio (HR) = 0.64; %95 CI = 0.41-0.99; p = 0.048]; particularly among infants weighing <1000 g [HR = 0.46; %95 CI = 0.22-0.96; p = 0.039] and infants with a low intake of human milk [HR = 0.40; %95 CI = 0.19-0.84; p = 0.015]. Therefore, bLF supplementation protects infants <1500 g from LOS, particularly those infants not receiving human milk.
Collapse
Affiliation(s)
- Theresa Ochoa
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA.,Doctoral School of Biomedial Sciences, KU Leuven, Belgium
| | - Sebastian Loli
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Karina Mendoza
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Carcamo
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,Hospital Cayetano Heredia, Lima, Peru
| | - Luis Cam
- Hospital Nacional Alberto Sabogal, Lima, Peru
| | | | - Miguel Campos
- Facultad de Ciencias y Filosofia, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jan Jacobs
- Department of Microbiology and Immunology, KU Leuven, Belgium.,Department of Clinical Sciences, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Cossey
- Department of Development and Regeneration, KU Leuven, Belgium
| | - Jaime Zegarra
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru.,Hospital Cayetano Heredia, Lima, Peru
| |
Collapse
|
29
|
Alford MA, Baquir B, Santana FL, Haney EF, Hancock REW. Cathelicidin Host Defense Peptides and Inflammatory Signaling: Striking a Balance. Front Microbiol 2020; 11:1902. [PMID: 32982998 PMCID: PMC7481365 DOI: 10.3389/fmicb.2020.01902] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 07/20/2020] [Indexed: 12/28/2022] Open
Abstract
Host-defense peptides (HDPs) are vital components of innate immunity in all vertebrates. While their antibacterial activity toward bacterial cells was the original focus for research, their ability to modulate immune and inflammatory processes has emerged as one of their major functions in the host and as a promising approach from which to develop novel therapeutics targeting inflammation and innate immunity. In this review, with particular emphasis on the cathelicidin family of peptides, the roles of natural HDPs are examined in managing immune activation, cellular recruitment, cytokine responses, and inflammation in response to infection, as well as their contribution(s) to various inflammatory disorders and autoimmune diseases. Furthermore, we discuss current efforts to develop synthetic HDPs as therapeutics aimed at restoring balance to immune responses that are dysregulated and contribute to disease pathologies.
Collapse
Affiliation(s)
- Morgan A. Alford
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Beverlie Baquir
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Felix L. Santana
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
- Departamento de Medicina Molecular y Bioprocesos, Instituto de Biotecnología, Universidad Nacional Autónoma de México, Cuernavaca, Mexico
| | - Evan F. Haney
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| | - Robert E. W. Hancock
- Centre for Microbial Diseases and Immunity Research, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
30
|
Itell HL, Berenz A, Mangan RJ, Permar SR, Kaufman DA. Systemic and mucosal levels of lactoferrin in very low birth weight infants supplemented with bovine lactoferrin. Biochem Cell Biol 2020; 99:25-34. [PMID: 32841570 DOI: 10.1139/bcb-2020-0238] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Lactoferrin supplementation may help prevent infections in preterm infants, but the efficacy has varied with different doses and products. We assessed the absorption and excretion of bovine lactoferrin (bLF) in 31 infants receiving 100, 200, or 300 mg·kg-1·day-1 of enteral bLF for 30 days. bLF and human lactoferrin (hLF) in infant saliva, blood, urine, and stool, as well as expressed (EBM) or donor breast milk (DBM) that were collected (i) before the treatment was initiated, (ii) at study day 22, and (iii) one week after treatment cessation, were measured using ELISA. During treatment, bLF was absorbed from the gastrointestinal tract and detected in plasma, saliva, and urine, as well as excreted in stool. Levels of bLF in the saliva and stool began to decline within 12 h after dosing, and bLF was undetectable in all samples one week after treatment. The concentrations of hLF exceeded those of bLF across sample types and time-points. Infants receiving EBM demonstrated higher levels of hLF in the saliva and stool than those receiving DBM. Neither bLF nor hLF levels varied by patient characteristics, bLF dosage, or infection status. This is the first study demonstrating bLF absorption into the bloodstream and distribution to saliva and urine in preterm infants. Future studies should further explore LF pharmacokinetics because higher and more frequent dosing may improve the clinical benefit of LF supplementation.
Collapse
Affiliation(s)
- Hannah L Itell
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Andrew Berenz
- Department of Pediatrics, Rush University Medical Center, Chicago, IL, USA
| | - Riley J Mangan
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, NC, USA
| | - David A Kaufman
- Division of Neonatology, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
31
|
Superti F. Lactoferrin from Bovine Milk: A Protective Companion for Life. Nutrients 2020; 12:nu12092562. [PMID: 32847014 PMCID: PMC7551115 DOI: 10.3390/nu12092562] [Citation(s) in RCA: 72] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Revised: 08/19/2020] [Accepted: 08/20/2020] [Indexed: 02/06/2023] Open
Abstract
Lactoferrin (Lf), an iron-binding multifunctional glycoprotein belonging to the transferrin family, is present in most biological secretions and reaches particularly high concentrations in colostrum and breast milk. A key function of lactoferrin is non-immune defence and it is considered to be a mediator linking innate and adaptive immune responses. Lf from bovine milk (bLf), the main Lf used in human medicine because of its easy availability, has been designated by the United States Food and Drug Administration as a food additive that is generally recognized as safe (GRAS). Among the numerous protective activities exercised by this nutraceutical protein, the most important ones demonstrated after its oral administration are: Antianemic, anti-inflammatory, antimicrobial, immunomodulatory, antioxidant and anticancer activities. All these activities underline the significance in host defence of bLf, which represents an ideal nutraceutical product both for its economic production and for its tolerance after ingestion. The purpose of this review is to summarize the most important beneficial activities demonstrated following the oral administration of bLf, trying to identify potential perspectives on its prophylactic and therapeutic applications in the future.
Collapse
Affiliation(s)
- Fabiana Superti
- National Centre for Innovative Technologies in Public Health, National Institute of Health, Viale Regina Elena 299, 00161 Rome, Italy
| |
Collapse
|
32
|
Berrington JE, McGuire W, Embleton ND. ELFIN, the United Kingdom preterm lactoferrin trial: interpretation and future questions 1. Biochem Cell Biol 2020; 99:1-6. [PMID: 32830532 DOI: 10.1139/bcb-2020-0073] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Results from previous studies have suggested that supplemental bovine lactoferrin (BLF) given to preterm infants (<32 weeks gestation) reduces late-onset sepsis (LOS) and necrotising enterocolitis (NEC). The Enteral Lactoferrin in Neonates (ELFIN) study, performed in the UK, aimed to further address this issue with a well powered double-blind placebo controlled trial of >2200 preterm infants. The results from ELFIN did not demonstrate a reduction in LOS or NEC, or several other clinically important measures. Of the 1093 infants, 316 (29%) in the intervention group developed late-onset sepsis versus 334 (31%) of 1089 in the control group, with an adjusted risk ratio of 0.95 (95% CI = 0.86-1.04; p = 0.233). Reasons for the differences in ELFIN trial results and other studies may include population differences, the routine use of antifungal prophylaxis in the UK, timing of administration of the lactoferrin in relation to disease onset, or specific properties of the lactoferrin used in the different trials. The UK National Institutes for Health Research funded "Mechanisms Affecting the Guts of Preterm Infants in Enteral feeding trials" (MAGPIE) study is further exploring the use of lactoferrin, and the results should be available soon.
Collapse
Affiliation(s)
- Janet Elizabeth Berrington
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, and Translational and Clinical Medicine, Newcastle University, Newcastle, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York YO10 5DD, UK
| | - Nicholas David Embleton
- Newcastle Upon Tyne Hospitals NHS Foundation Trust, and Population Health Sciences, Newcastle University, Newcastle, UK
| |
Collapse
|
33
|
Gao Y, Hou L, Lu C, Wang Q, Pan B, Wang Q, Tian J, Ge L. Enteral Lactoferrin Supplementation for Preventing Sepsis and Necrotizing Enterocolitis in Preterm Infants: A Meta‑Analysis With Trial Sequential Analysis of Randomized Controlled Trials. Front Pharmacol 2020; 11:1186. [PMID: 32848789 PMCID: PMC7426497 DOI: 10.3389/fphar.2020.01186] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2019] [Accepted: 07/21/2020] [Indexed: 12/21/2022] Open
Abstract
Background Several clinical trials investigated the effects of enteral lactoferrin supplementation on the prevention of sepsis and necrotizing enterocolitis (NEC) in preterm infants, but the efficacy and safety remain disputed. Therefore, we systematically evaluated the effect of enteral lactoferrin supplementation in preterm infants through a meta‑analysis with trial sequential analysis (TSA). Methods We searched six databases to identify randomized controlled trials (RCTs) that evaluated the effects of lactoferrin supplementation compared with placebo or no intervention in preterm infants. RevMan version 5.3 software was used to estimate pooled relative risks (RRs) with the random-effects model. TSA, subgroup analyses, and meta-regression analyses were also performed. Results Nine RCTs with 3515 samples were included. With low to moderate quality of evidence, compared with placebo, enteral lactoferrin supplementation did not significantly decrease the incidences of late-onset sepsis (RR = 0.63, 95% CI: 0.38 to 1.02, P = 0.06), NEC stage II or III (RR = 0.68, 95% CI: 0.30 to 1.52, P = 0.35), all-cause mortality (RR = 0.89, 95% CI: 0.51 to 1.57, P = 0.69), bronchopulmonary dysplasia (RR = 1.01, 95% CI: 0.90 to 1.13, P = 0.92), retinopathy of prematurity (RR = 0.80, 95% CI: 0.49 to 1.32, P = 0.38), invasive fungal infection (RR = 0.27, 95% CI: 0.02 to 3.94, P = 0.34), intraventricular hemorrhage (RR = 1.40, 95% CI: 0.39 to 5.08, P = 0.61), and urinary tract infection (RR = 0.35, 95% CI: 0.11 to 1.06, P = 0.06). Subgroup analysis revealed that lactoferrin significantly reduced the incidence of sepsis in infants with a birth weight below 1500 g (RR = 0.43, 95% CI: 0.22 to 0.84, P = 0.01). TSAs of the primary outcomes showed that the evidence is insufficient and further data is required. Conclusions Limited evidence suggested that enteral lactoferrin supplementation was associated with a reduction of late-onset sepsis in infants with a birth weight below 1500g, however, did not decrease the incidence of NEC stage II or III, all-cause mortality, and other adverse events in preterm infants. The present evidence was insufficient to inform clinical practice.
Collapse
Affiliation(s)
- Ya Gao
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Liangying Hou
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, China
| | - Cuncun Lu
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Qi Wang
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, China
| | - Bei Pan
- Gansu Provincial Hospital, Lanzhou, China
| | - Quan Wang
- Department of Gastrointestinal Surgery & Laboratory of Surgical Oncology, Peking University People's Hospital, Peking University, Beijing, China
| | - Jinhui Tian
- Evidence-Based Medicine Center, School of Basic Medical Sciences, Lanzhou University, Lanzhou, China.,Key Laboratory of Evidence-Based Medicine and Knowledge Translation of Gansu Province, Lanzhou, China
| | - Long Ge
- Evidence-Based Social Science Research Center, School of Public Health, Lanzhou University, Lanzhou, China.,Department of Social Medicine and Health Management, School of Public Health, Lanzhou University, Lanzhou, China
| |
Collapse
|
34
|
Pammi M, Preidis GA, Tarnow-Mordi WO. Evidence from systematic reviews of randomized trials on enteral lactoferrin supplementation in preterm neonates. Biochem Cell Biol 2020; 99:20-24. [PMID: 32721215 DOI: 10.1139/bcb-2020-0136] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
In this commentary, we summarize the current evidence from randomized controlled trials on enteral lactoferrin supplementation in preterm neonates. Our recently completed systematic review includes 12 randomized controlled trials performed all over the world. Our meta-analysis suggests clinical benefit in decreasing late-onset sepsis, late-onset fungal sepsis, length of stay in the hospital and urinary tract infections. There were no adverse effects. There was no statistically significant decrease in necrotizing enterocolitis, mortality or neurodevelopmental impairment in lactoferrin supplemented preterm infants. There was significant statistical heterogeneity in the effects of lactoferrin on late-onset sepsis between larger and smaller studies, which may reflect either small study biases, differences in the effectiveness, dose or duration of supplemental lactoferrin products, or differences in underlying population risk, or any or all of these.
Collapse
Affiliation(s)
- Mohan Pammi
- Department of Pediatrics, Section of Neonatology, Baylor College of Medicine & Texas Children's Hospital, Houston, TX 77030, USA
| | - Geoffrey A Preidis
- Department of Pediatrics, Section of Gastroenterology, Hepatology & Nutrition, Baylor College of Medicine & Texas Children's Hospital, Houston, TX 77030, USA
| | - William O Tarnow-Mordi
- WINNER Centre for Newborn Research, NHMRC Clinical Trials Centre, University of Sydney, Camperdown, NSW 2050, Australia
| |
Collapse
|
35
|
Tarnow-Mordi WO, Abdel-Latif ME, Martin A, Pammi M, Robledo K, Manzoni P, Osborn D, Lui K, Keech A, Hague W, Ghadge A, Travadi J, Brown R, Darlow BA, Liley H, Pritchard M, Kochar A, Isaacs D, Gordon A, Askie L, Cruz M, Schindler T, Dixon K, Deshpande G, Tracy M, Schofield D, Austin N, Sinn J, Simes RJ. The effect of lactoferrin supplementation on death or major morbidity in very low birthweight infants (LIFT): a multicentre, double-blind, randomised controlled trial. THE LANCET CHILD & ADOLESCENT HEALTH 2020; 4:444-454. [PMID: 32407710 DOI: 10.1016/s2352-4642(20)30093-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2019] [Revised: 03/06/2020] [Accepted: 03/23/2020] [Indexed: 12/24/2022]
Abstract
BACKGROUND Very low birthweight or preterm infants are at increased risk of adverse outcomes including sepsis, necrotising enterocolitis, and death. We assessed whether supplementing the enteral diet of very low-birthweight infants with lactoferrin, an antimicrobial protein, reduces all-cause mortality or major morbidity. METHODS We did a multicentre, double-blind, pragmatic, randomised superiority trial in 14 Australian and two New Zealand neonatal intensive care units. Infants born weighing less than 1500 g and aged less than 8 days, were eligible and randomly assigned (1:1) using minimising web-based randomisation to receive once daily 200 mg/kg pasteurised bovine lactoferrin supplements or no lactoferrin supplement added to breast or formula milk until 34 weeks' post-menstrual age (or for 2 weeks, if longer), or until discharge from the study hospital if that occurred first. Designated nurses preparing the daily feeds were not masked to group assignment, but other nurses, doctors, parents, caregivers, and investigators were unaware. The primary outcome was survival to hospital discharge or major morbidity (defined as brain injury, necrotising enterocolitis, late-onset sepsis at 36 weeks' post-menstrual age, or retinopathy treated before discharge) assessed in the intention-to-treat population. Safety analyses were by treatment received. We also did a prespecified, PRISMA-compliant meta-analysis, which included this study and other relevant randomised controlled trials, to estimate more precisely the effects of lactoferrin supplementation on late-onset sepsis, necrotising enterocolitis, and survival. This trial is registered with the Australian and New Zealand Clinical Trials Registry, ACTRN12611000247976. FINDINGS Between June 27, 2014, and Sept 1, 2017, we recruited 1542 infants; 771 were assigned to the intervention group and 771 to the control group. One infant who had consent withdrawn before beginning lactoferrin treatment was excluded from analysis. In-hospital death or major morbidity occurred in 162 (21%) of 770 infants in the intervention group and in 170 (22%) of 771 infants in the control group (relative risk [RR] 0·95, 95% CI 0·79-1·14; p=0·60). Three suspected unexpected serious adverse reactions occurred; two in the lactoferrin group, namely unexplained late jaundice and inspissated milk syndrome, but were not attributed to the intervention and one in the control group had fatal inspissated milk syndrome. Our meta-analysis identified 13 trials completed before Feb 18, 2020, including this Article, in 5609 preterm infants. Lactoferrin supplements significantly reduced late-onset sepsis (RR 0·79, 95% CI 0·71-0·88; p<0·0001; I2=58%), but not necrotising enterocolitis or all-cause mortality. INTERPRETATION Lactoferrin supplementation did not improve death or major morbidity in this trial, but might reduce late-onset sepsis, as found in our meta-analysis of over 5000 infants. Future collaborative studies should use products with demonstrated biological activity, be large enough to detect moderate and clinically important effects reliably, and assess greater doses of lactoferrin in infants at increased risk, such as those not exclusively receiving breastmilk or infants of extremely low birthweight. FUNDING Australian National Health and Medical Research Council.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Kei Lui
- University of New South Wales, Kensington, NSW, Australia
| | | | - Wendy Hague
- University of Sydney, Sydney, NSW, Australia
| | | | | | | | | | - Helen Liley
- University of Queensland, Brisbane, QLD, Australia
| | | | - Anu Kochar
- University of Adelaide, Adelaide, SA, Australia
| | | | | | - Lisa Askie
- University of Sydney, Sydney, NSW, Australia
| | - Melinda Cruz
- Miracle Babies Foundation, Chipping Norton, NSW, Australia
| | - Tim Schindler
- University of New South Wales, Kensington, NSW, Australia
| | - Kelly Dixon
- University of Queensland, Brisbane, QLD, Australia
| | | | - Mark Tracy
- University of Sydney, Sydney, NSW, Australia
| | | | | | - John Sinn
- University of Sydney, Sydney, NSW, Australia
| | | | | |
Collapse
|
36
|
Ochoa TJ, Zegarra J, Bellomo S, Carcamo CP, Cam L, Castañeda A, Villavicencio A, Gonzales J, Rueda MS, Turin CG, Zea-Vera A, Guillen D, Campos M, Ewing-Cobbs L. Randomized Controlled Trial of Bovine Lactoferrin for Prevention of Sepsis and Neurodevelopment Impairment in Infants Weighing Less Than 2000 Grams. J Pediatr 2020; 219:118-125.e5. [PMID: 32037149 PMCID: PMC7096260 DOI: 10.1016/j.jpeds.2019.12.038] [Citation(s) in RCA: 32] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/31/2019] [Revised: 11/25/2019] [Accepted: 12/17/2019] [Indexed: 12/15/2022]
Abstract
OBJECTIVES To determine the effect of bovine lactoferrin on prevention of late-onset sepsis (LOS) and neurodevelopment delay. STUDY DESIGN Randomized, double-blind, controlled trial in neonates with a birth weight of 500-2000 g in 3 neonatal units in Lima, Peru, comparing bovine lactoferrin 200 mg/kg/day with placebo administered for 8 weeks. The primary outcome was the first episode of culture-proven LOS or sepsis-associated death. Neurodevelopment delay was assessed by the Mullen Scales at 24 months corrected age. RESULTS Of the 414 infants enrolled, 209 received bovine lactoferrin and 205 received placebo. LOS or sepsis-associated death occurred in 22 infants (10.5%) in the bovine lactoferrin group vs 30 (14.6%) in the placebo group; there was no difference after adjusting for hospital and birth weight; hazard ratio 0.73 (95% CI, 0.42-1.26). For infants with birth weights of <1500 g the hazard ratio was 0.69 (95% CI, 0.39-1.25). The mean age-adjusted normalized Mullen composite score at 24 months was 83.3 ± 13.6 in the bovine lactoferrin group vs 82.6 ± 13.1 in the placebo group. Growth outcomes and rehospitalization rates during the 2-year follow-up were similar in both groups, except for significantly less bronchiolitis in the bovine lactoferrin group (rate ratio, 0.34; 95% CI, 0.14-0.86). CONCLUSIONS Supplementation with bovine lactoferrin did not decrease the incidence of sepsis in infants with birth weights of <2000 g. Growth and neurodevelopment outcomes at 24 months of age were similar. Neonatal bovine lactoferrin supplementation had no adverse effects. TRIAL REGISTRATION ClinicalTrials.gov: NCT01525316.
Collapse
Affiliation(s)
- Theresa J. Ochoa
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Instituto de Medicina Tropical “Alexander von Humboldt”, Universidad Peruana Cayetano Heredia, Lima, Peru,Center for Infectious Diseases, School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | - Jaime Zegarra
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Cesar P. Carcamo
- School of Public Health and Administration, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Luis Cam
- Hospital Nacional Alberto Sabogal, Lima, Peru
| | | | - Aasith Villavicencio
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jorge Gonzales
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Maria S. Rueda
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Christie G. Turin
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Alonso Zea-Vera
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Daniel Guillen
- Department of Pediatrics, School of Medicine, Universidad Peruana Cayetano Heredia, Lima, Peru,Hospital Cayetano Heredia, Lima, Peru
| | - Miguel Campos
- Department of Mathematics, School of Science and Philosophy, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Linda Ewing-Cobbs
- Department of Pediatrics and Children’s Learning Institute, School of Medicine, University of Texas Health Science Center at Houston, Houston, Texas, United States
| | | |
Collapse
|
37
|
Pammi M, Suresh G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2020; 3:CD007137. [PMID: 32232984 PMCID: PMC7106972 DOI: 10.1002/14651858.cd007137.pub6] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
BACKGROUND Lactoferrin, a normal component of human colostrum and milk, can enhance host defenses and may be effective for prevention of sepsis and necrotizing enterocolitis (NEC) in preterm neonates. OBJECTIVES To assess the safety and effectiveness of lactoferrin supplementation to enteral feeds for prevention of sepsis and NEC in preterm neonates. Secondarily, we assessed the effects of lactoferrin supplementation to enteral feeds on the duration of positive-pressure ventilation, development of chronic lung disease (CLD) or periventricular leukomalacia (PVL), length of hospital stay to discharge among survivors, and adverse neurological outcomes at two years of age or later. SEARCH METHODS We used the standard search strategy of Cochrane Neonatal to update our search. We searched the Cochrane Central Register of Controlled Trials (CENTRAL 2019, Issue 9), MEDLINE via PubMed (1966 to 20 January 2020), PREMEDLINE (1996 to 20 January 2020), Embase (1980 to 20 January 2020), and CINAHL (1982 to 20 January 2020). 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 In our search, we included randomized controlled trials (RCTs) evaluating enteral lactoferrin supplementation at any dose or duration to prevent sepsis or NEC in preterm neonates. DATA COLLECTION AND ANALYSIS We used the standard methods of Cochrane Neonatal and the GRADE approach to assess the certainty of evidence. MAIN RESULTS Meta-analysis of data from twelve randomized controlled trials showed that lactoferrin supplementation to enteral feeds decreased late-onset sepsis (typical RR 0.82, 95% CI 0.74 to 0.91; typical RD -0.04, 95% CI, -0.06, -0.02; NNTB 25, 95% CI 17 to 50; 12 studies, 5425 participants, low-certainty evidence) and decreased length of hospital stay (MD -2.38, 95% CI, -4.67, -0.09; 3 studies, 1079 participants, low-certainty evidence). Sensitivity analysis including only good methodological certainty studies suggested a decrease in late-onset sepsis with enteral lactoferrin supplementation (typical RR 0.87, 95% CI, 0.78, 0.97; typical RD -0.03, 95% CI, -0.05, -0.0; 9 studies, 4702 participants, low-certainty evidence). There were no differences in NEC stage II or III (typical RR 1.10, 95% CI, 0.86, 1.41; typical RD -0.00, 95% CI, -0.02, 0.01; 7 studies, 4874 participants; low-certainty evidence) or 'all-cause mortality' (typical RR 0.90, 95% CI 0.69, 1.17; typical RD -0.00, 95% CI, -0.01, 0.01; 11 studies, 5510 participants; moderate-certainty evidence). One study reported no differences in neurodevelopmental testing by Mullen's or Bayley III at 24 months of age after enteral lactoferrin supplementation (one study, 292 participants, low-certainty evidence). Lactoferrin supplementation to enteral feeds with probiotics decreased late-onset sepsis (RR 0.25, 95% CI 0.14 to 0.46; RD -0.13, 95% CI -0.18 to -0.08; NNTB 8, 95% CI 6 to 13; 3 studies, 564 participants; low-certainty evidence) and NEC stage II or III (RR 0.04, 95% CI 0.00 to 0.62; RD -0.05, 95% CI -0.08 to -0.03; NNTB 20, 95% CI 12.5 to 33.3; 1 study, 496 participants; very low-certainty evidence), but not 'all-cause mortality' (very low-certainty evidence). Lactoferrin supplementation to enteral feeds with or without probiotics had no effect on CLD, duration of mechanical ventilation or threshold retinopathy of prematurity (low-certainty evidence). Investigators reported no adverse effects in the included studies. AUTHORS' CONCLUSIONS We found low-certainty evidence from studies of good methodological quality that lactoferrin supplementation of enteral feeds decreases late-onset sepsis but not NEC ≥ stage II or 'all cause mortality' or neurodevelopmental outcomes at 24 months of age in preterm infants without adverse effects. Low- to very low-certainty evidence suggests that lactoferrin supplementation of enteral feeds in combination with probiotics decreases late-onset sepsis and NEC ≥ stage II in preterm infants without adverse effects, however, there were few included studies of poor methodological quality. The presence of publication bias and small studies of poor methodology that may inflate the effect size make recommendations for clinical practice difficult.
Collapse
Affiliation(s)
- Mohan Pammi
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| | - Gautham Suresh
- Baylor College of Medicine, Section of Neonatology, Department of Pediatrics, 6621, Fannin, MC.WT 6-104, Houston, Texas, USA, 77030
| |
Collapse
|
38
|
Ochoa TJ, Mendoza K, Carcamo C, Zegarra J, Bellomo S, Jacobs J, Cossey V. Is Mother's Own Milk Lactoferrin Intake Associated with Reduced Neonatal Sepsis, Necrotizing Enterocolitis, and Death? Neonatology 2020; 117:167-174. [PMID: 32053823 PMCID: PMC7381382 DOI: 10.1159/000505663] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/16/2019] [Accepted: 12/28/2019] [Indexed: 11/19/2022]
Abstract
INTRODUCTION Lactoferrin (LF) is a protective protein present in milk with anti-infective and immune-modulating properties. OBJECTIVES The aim of this study was to determine the association of maternal LF intake and mother's own milk intake in the first 10 days of life on the prevention of late-onset sepsis (LOS), necrotizing enterocolitis (NEC), or death in the first 8 weeks of life in newborns with a birth weight <2,000 g. METHODS A retrospective cohort study was conducted, with the exposure being the consumption of mother's own LF and mother's own milk in the first 10 days of life, and the outcome being LOS, NEC, or death during days 11 and 56 of life, analyzed by Cox regression. RESULTS Two hundred and ninety-nine infants were enrolled, including 240 with human LF intake information. The average daily human LF intake over days 4-10 of life was 283 mg/kg/day (IQR 114-606 mg/kg/day). The hazard ratio (HR) of mother's own milk LF intake ≥100 mg/kg/day in days 4-10 for LOS, NEC, or death was 0.297 (95% CI 0.156-0.568, p < 0.001); the adjusted HR was 0.752 (95% CI 0.301-1.877, p = 0.541). The adjusted HR of mother's own milk cumulative intake (days 4-10) of 54-344 mL/kg (25-75 quartiles) for LOS, NEC, or death was 0.414 (95% CI 0.196-0.873, p = 0.02). Infants who developed an event (LOS, NEC, or death) had significantly less median daily human LF intake than those that did not (89 vs. 334 mg/kg/day, respectively, p < 0.0001). CONCLUSION Consumption of higher amounts of mother's own milk in the first days of life is associated with less infection, NEC, and death. Early human milk intake should be strongly encouraged in all newborns.
Collapse
Affiliation(s)
- Theresa J Ochoa
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru, .,School of Public Health, University of Texas Health Science Center at Houston, Houston, Texas, USA, .,Doctoral School of Biomedical Sciences, KU Leuven, Leuven, Belgium,
| | - Karina Mendoza
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Cesar Carcamo
- Facultad de Salud Pública y Administración, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jaime Zegarra
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Sicilia Bellomo
- Facultad de Medicina, Universidad Peruana Cayetano Heredia, Lima, Peru
| | - Jan Jacobs
- Department of Microbiology and Immunology, KU Leuven, Leuven, Belgium.,Department of Clinical Science, Institute of Tropical Medicine, Antwerp, Belgium
| | - Veerle Cossey
- Department of Development and Regeneration, KU Leuven, Leuven, Belgium
| |
Collapse
|
39
|
Recent Advances in Prevention and Therapies for Clinical or Experimental Necrotizing Enterocolitis. Dig Dis Sci 2019; 64:3078-3085. [PMID: 30989465 DOI: 10.1007/s10620-019-05618-2] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/30/2018] [Accepted: 04/08/2019] [Indexed: 01/09/2023]
Abstract
Necrotizing enterocolitis (NEC) is one of the most severe diseases of preterm neonates and has a high mortality rate. With the development of inspection techniques and new biomarkers, the diagnostic accuracy of NEC is constantly improving. The most recognized potential risk factors include prematurity, formula-feeding, infection, and microbial dysbiosis. With further understanding of the pathogenesis, more effective prevention and therapies will be applied to clinical or experimental NEC. At present, such new potential prevention and therapies for NEC are mainly focused on the Toll-like receptor 4 inflammatory signaling pathway, the repair of intestinal barrier function, probiotics, antioxidative stress, breast-feeding, and immunomodulatory agents. Many new studies have changed our understanding of the pathogenesis of NEC and improve our approaches for preventing and treating of NEC each year. This review provides an overview of the recent researches focused on clinical or experimental NEC and highlights the advances made within the past 5 years toward the development of new potential preventive approaches and therapies for this disease.
Collapse
|
40
|
Hård A, Nilsson AK, Lund A, Hansen‐Pupp I, Smith LEH, Hellström A. Review shows that donor milk does not promote the growth and development of preterm infants as well as maternal milk. Acta Paediatr 2019; 108:998-1007. [PMID: 30565323 PMCID: PMC6520191 DOI: 10.1111/apa.14702] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 10/09/2018] [Accepted: 12/12/2018] [Indexed: 12/28/2022]
Abstract
Aim This nonsystematic review examined differences in the composition of raw maternal breastmilk and pasteurised donor milk and possible health effects on preterm infants. Methods We searched PubMed up to July 2018 for studies published in English that focused on four comparisons as follows: raw maternal milk versus donor milk, human milk before and after Holder pasteurisation, milk from mothers who delivered preterm and at term and milk collected during early and late lactation. We also searched for possible effects of the milk components, as well as the effects of maternal and donor milk on preterm infants’ health. Results Raw maternal milk contained factors involved in antioxidant and anti‐inflammatory defence, gut microbiome establishment and the maturation of immune defences, food tolerability and metabolism. Many of these factors were reduced or abolished in processed donor milk. Both maternal milk and donor milk have been associated with a reduced incidence of necrotising enterocolitis. High‐dose feeding with maternal milk during the neonatal period reportedly reduced the risk of other morbidities and promoted growth and neurodevelopment. Conclusion Many of the components in raw maternal breastmilk were lacking in pasteurised donor milk, which was inferior in promoting the growth and development of very preterm infants.
Collapse
Affiliation(s)
- Anna‐Lena Hård
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Anders K. Nilsson
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| | - Anna‐My Lund
- Department of Pediatrics Institute of Clinical Sciences Lund Lund University and Skane University Hospital Lund Sweden
| | - Ingrid Hansen‐Pupp
- Department of Pediatrics Institute of Clinical Sciences Lund Lund University and Skane University Hospital Lund Sweden
| | - Lois E. H. Smith
- Department of Ophthalmology Boston Children's Hospital Harvard Medical School Boston MA USA
| | - Ann Hellström
- Department of Ophthalmology Institute of Neuroscience and Physiology Sahlgrenska Academy University of Gothenburg Gothenburg Sweden
| |
Collapse
|
41
|
Griffiths J, Jenkins P, Vargova M, Bowler U, Juszczak E, King A, Linsell L, Murray D, Partlett C, Patel M, Berrington J, Embleton N, Dorling J, Heath PT, McGuire W, Oddie S. Enteral lactoferrin to prevent infection for very preterm infants: the ELFIN RCT. Health Technol Assess 2019; 22:1-60. [PMID: 30574860 DOI: 10.3310/hta22740] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Infections acquired in hospital are an important cause of morbidity and mortality in very preterm infants. Several small trials have suggested that supplementing the enteral diet of very preterm infants with lactoferrin, an antimicrobial protein processed from cow's milk, prevents infections and associated complications. OBJECTIVE To determine whether or not enteral supplementation with bovine lactoferrin (The Tatua Cooperative Dairy Company Ltd, Morrinsville, New Zealand) reduces the risk of late-onset infection (acquired > 72 hours after birth) and other morbidity and mortality in very preterm infants. DESIGN Randomised, placebo-controlled, parallel-group trial. Randomisation was via a web-based portal and used an algorithm that minimised for recruitment site, weeks of gestation, sex and single versus multiple births. SETTING UK neonatal units between May 2014 and September 2017. PARTICIPANTS Infants born at < 32 weeks' gestation and aged < 72 hours at trial enrolment. INTERVENTIONS Eligible infants were allocated individually (1 : 1 ratio) to receive enteral bovine lactoferrin (150 mg/kg/day; maximum 300 mg/day) or sucrose (British Sugar, Peterborough, UK) placebo (same dose) once daily from trial entry until a postmenstrual age of 34 weeks. Parents, caregivers and outcome assessors were unaware of group assignment. OUTCOMES Primary outcome - microbiologically confirmed or clinically suspected late-onset infection. Secondary outcomes - microbiologically confirmed infection; all-cause mortality; severe necrotising enterocolitis (NEC); retinopathy of prematurity (ROP); bronchopulmonary dysplasia (BPD); a composite of infection, NEC, ROP, BPD and mortality; days of receipt of antimicrobials until 34 weeks' postmenstrual age; length of stay in hospital; and length of stay in intensive care, high-dependency and special-care settings. RESULTS Of 2203 enrolled infants, primary outcome data were available for 2182 infants (99%). In the intervention group, 316 out of 1093 (28.9%) infants acquired a late-onset infection versus 334 out of 1089 (30.7%) infants in the control group [adjusted risk ratio (RR) 0.95, 95% confidence interval (CI) 0.86 to 1.04]. There were no significant differences in any secondary outcomes: microbiologically confirmed infection (RR 1.05, 99% CI 0.87 to 1.26), mortality (RR 1.05, 99% CI 0.66 to 1.68), NEC (RR 1.13, 99% CI 0.68 to 1.89), ROP (RR 0.89, 99% CI 0.62 to 1.28), BPD (RR 1.01, 99% CI 0.90 to 1.13), or a composite of infection, NEC, ROP, BPD and mortality (RR 1.01, 99% CI 0.94 to 1.08). There were no differences in the number of days of receipt of antimicrobials, length of stay in hospital, or length of stay in intensive care, high-dependency or special-care settings. There were 16 reports of serious adverse events for infants in the lactoferrin group and 10 for infants in the sucrose group. CONCLUSIONS Enteral supplementation with bovine lactoferrin does not reduce the incidence of infection, mortality or other morbidity in very preterm infants. FUTURE WORK Increase the precision of the estimates of effect on rarer secondary outcomes by combining the data in a meta-analysis with data from other trials. A mechanistic study is being conducted in a subgroup of trial participants to explore whether or not lactoferrin supplementation affects the intestinal microbiome and metabolite profile of very preterm infants. TRIAL REGISTRATION Current Controlled Trials ISRCTN88261002. FUNDING This project was funded by the National Institute for Health Research (NIHR) Health Technology Assessment programme and will be published in full in Health Technology Assessment; Vol. 22, No. 74. See the NIHR Journals Library website for further project information. This trial was also sponsored by the University of Oxford, Oxford, UK. The funder provided advice and support and monitored study progress but did not have a role in study design or data collection, analysis and interpretation.
Collapse
Affiliation(s)
- James Griffiths
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Paula Jenkins
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Monika Vargova
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Ursula Bowler
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Edmund Juszczak
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Andrew King
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - Louise Linsell
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | - David Murray
- National Perinatal Epidemiology Unit, University of Oxford, Oxford, UK
| | | | | | - Janet Berrington
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | - Nicholas Embleton
- Newcastle Neonatal Service, Royal Victoria Infirmary, Newcastle upon Tyne, UK
| | | | - Paul T Heath
- St George's, University of London and St George's University Hospitals NHS Trust, London, UK
| | - William McGuire
- Centre for Reviews and Dissemination, University of York, York, UK
| | - Sam Oddie
- Bradford Institute for Health Research, Bradford, UK
| |
Collapse
|
42
|
Affiliation(s)
- Paolo Manzoni
- Division of Pediatrics and Neonatology, Degli Infermi Hospital, Biella, Italy
| |
Collapse
|
43
|
Bazacliu C, Neu J. Pathophysiology of Necrotizing Enterocolitis: An Update. Curr Pediatr Rev 2019; 15:68-87. [PMID: 30387398 DOI: 10.2174/1573396314666181102123030] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2018] [Revised: 09/12/2018] [Accepted: 09/15/2018] [Indexed: 12/12/2022]
Abstract
NEC is a devastating disease that, once present, is very difficult to treat. In the absence of an etiologic treatment, preventive measures are required. Advances in decoding the pathophysiology of NEC are being made but a more comprehensive understanding is needed for the targeting of preventative strategies. A better definition of the disease as well as diagnostic criteria are needed to be able to specifically label a disease as NEC. Multiple environmental factors combined with host susceptibility appear to contribute to enhanced risks for developing this disease. Several different proximal pathways are involved, all leading to a common undesired outcome: Intestinal necrosis. The most common form of this disease appears to involve inflammatory pathways that are closely meshed with the intestinal microbiota, where a dysbiosis may result in dysregulated inflammation. The organisms present in the intestinal tract prior to the onset of NEC along with their diversity and functional capabilities are just beginning to be understood. Fulfillment of postulates that support causality for particular microorganisms is needed if bacteriotherapies are to be intelligently applied for the prevention of NEC. Identification of molecular effector pathways that propagate inflammation, understanding of, even incipient role of genetic predisposition and of miRNAs may help solve the puzzle of this disease and may bring the researchers closer to finding a treatment. Despite recent progress, multiple limitations of the current animal models, difficulties related to studies in humans, along with the lack of a "clear" definition will continue to make it a very challenging disease to decipher.
Collapse
Affiliation(s)
- Catalina Bazacliu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| | - Josef Neu
- Department of Pediatrics, Division of Neonatology, University of Florida, FL, United States
| |
Collapse
|
44
|
Abstract
Enteral feeding and composition play a chief role in the prevention and treatment of necrotizing enterocolitis (NEC). In the face of decades of research on this fatal disease, the exact mechanism of disease is still poorly understood. There is established evidence that providing mother's own breast milk and standardization of feeding regimens leads to a decreased risk for NEC. More recent studies have focused on the provision of donor human milk or an exclusive human milk diet in the endeavor to prevent NEC while still maintaining adequate nutrition to the premature infant. There is growing literature on the provision of specific human milk components and its effect on the incidence of NEC.
Collapse
Affiliation(s)
- Diomel de la Cruz
- University of Florida, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA.
| | - Catalina Bazacliu
- University of Florida, Department of Pediatrics, Division of Neonatology, Gainesville, FL, USA
| |
Collapse
|
45
|
Telang S. Lactoferrin: A Critical Player in Neonatal Host Defense. Nutrients 2018; 10:nu10091228. [PMID: 30181493 PMCID: PMC6165050 DOI: 10.3390/nu10091228] [Citation(s) in RCA: 72] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 08/30/2018] [Accepted: 08/30/2018] [Indexed: 12/15/2022] Open
Abstract
Newborn infants are at a high risk for infection due to an under-developed immune system, and human milk has been shown to exhibit substantial anti-infective properties that serve to bolster neonatal defenses against multiple infections. Lactoferrin is the dominant whey protein in human milk and has been demonstrated to perform a wide array of antimicrobial and immunomodulatory functions and play a critical role in protecting the newborn infant from infection. This review summarizes data describing the structure and important functions performed by lactoferrin in protecting the neonate from infection and contributing to the maturation of the newborn innate and adaptive immune systems. We also briefly discuss clinical trials examining the utility of lactoferrin supplementation in the prevention of sepsis and necrotizing enterocolitis in newborn infants. The data reviewed provide rationale for the continuation of studies to examine the effects of lactoferrin administration on the prevention of sepsis in the neonate.
Collapse
Affiliation(s)
- Sucheta Telang
- Division of Neonatology, Department of Pediatrics, University of Louisville, Louisville, KY 40202, USA.
- Division of Hematology/Oncology, Department of Medicine, James Graham Brown Cancer Center, University of Louisville, Louisville, KY 40202, USA.
| |
Collapse
|
46
|
He Y, Cao L, Yu J. Prophylactic lactoferrin for preventing late-onset sepsis and necrotizing enterocolitis in preterm infants: A PRISMA-compliant systematic review and meta-analysis. Medicine (Baltimore) 2018; 97:e11976. [PMID: 30170397 PMCID: PMC6392939 DOI: 10.1097/md.0000000000011976] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND Currently, prophylactic use of drugs to promote a healthy gut microbiota and immune system in preterm infants is hot debated, among which lactoferrin is a promising supplementation. However, the effect and safety of lactoferrin to prevent late-onset sepsis (LOS) and necrotizing enterocolitis (NEC) in preterm infants remains controversial. METHODS Databases including Medline, Ovid-Embase, The Cochrane Library, CBM, CNKI, and VIP database of Chinese Journal were searched to collect randomized controlled trials (RCTs) about lactoferrin for preventing LOS and NEC in preterm infants. Languages of included RCTs were restricted to English and Chinese. Meta-analysis was conducted by Rev Man 5.3 software. The Mantel-Haenszel method with random-effects model was used to calculate pooled relative risks (RRs) and 95% confidence intervals (CIs). RESULTS A total of 9 RCTs, involving 1834 patients, were included. Pooled analysis showed that prophylactic lactoferrin could significantly reduce the incidence all culture-proven LOS (41/629 [6.5%] vs 96/659 [15.3%]; RR 0.47; 95% CI 0.33-0.67; P < .01) and NEC (stage II or more) (9/448 [2.0%] vs 26/462 [5.6%]; RR 0.40; 95% CI 0.18-0.86; P < .01). Lactoferrin was also associated with a significantly decreased hospital-acquired infection (16/139 [11.5%] vs 35/140 [25%]; RR 0.47; 95% CI 0.27-0.80; P < .01); and infection-related mortality (4/474 [0.8%] vs 25/505 [4.9%]; RR 0.24; 95% CI 0.04-1.32; P < .01, I = 53%). Lactoferrin could shorten time to reach full enteral feeding (weighted mean difference [WMD] = -2.11, 95% CI -3.12 to -1.10; P < .01) and showed a decreasing trend of duration of hospitalization (WMD = -1.69, 95% CI -6.87 to 3.50; P < .01; I = 95%). Lactoferrin did not have a significant effect on all-cause mortality (22/625 [3.5%] vs 35/647 [5.4%]; RR 0.70; 95% CI 0.38-1.30; P = .16; I = 13%). None of the included trials reported any confirmed adverse effects caused by the supplemented lactoferrin or probiotics. CONCLUSION Current evidence indicates that lactoferrin could significantly reduce the incidence of NEC and LOS, and decrease the risk of hospital-acquired infection and infection-related mortality in premature infants without obvious adverse effects.
Collapse
Affiliation(s)
- Yi He
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
| | - Luying Cao
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
| | - Jialin Yu
- Department of Neonatology, Children's Hospital of Chongqing Medical University, Ministry of Education Key Laboratory of Child Development and Disorders, Yuzhong
- China International Science and Technology Cooperation Base of Child Development and Critical Disorders
- Chongqing Key Laboratory of Pediatrics, Chongqing, China
| |
Collapse
|
47
|
Bovine Lactoferrin Modulates Dendritic Cell Differentiation and Function. Nutrients 2018; 10:nu10070848. [PMID: 29966271 PMCID: PMC6073808 DOI: 10.3390/nu10070848] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2018] [Revised: 06/19/2018] [Accepted: 06/26/2018] [Indexed: 11/16/2022] Open
Abstract
Lactoferrin is an abundant glycoprotein in bovine milk that has immunomodulatory effects on human cells. Bovine lactoferrin (LF) binds lipopolysaccharides (LPS) with high affinity and is postulated to act via TLR4-dependent and -independent mechanisms. It has been shown that LF modulates differentiation of human monocytes into tolerogenic dendritic cells. However, in a previous study, we showed that LPS also mediates differentiation into tolerogenic dendritic cells (DC). Since LF binds LPS with high affinity, it remains to be investigated whether LF or LPS is mediating these effects. We, therefore, further investigated the LPS-independent effect of LF on differentiation of human monocytes into dendritic cells (DC). Human monocytes were isolated by magnetic cell sorting from freshly isolated PBMCs and cultured for six days in the presence of IL-4 and GM-CSF with or without LF or proteinase K treated LF to generate DC. These immature DC were stimulated for 48 h with LPS or Poly I:C + R848. Cell surface marker expression and cytokine production were measured by flow cytometry. DC differentiated in the presence of LF produced higher IL-6 and IL-8 levels during differentiation and showed a lower expression of CD1a and HLA-DR. These LFDCs showed to be hyporesponsive towards TLR ligands as shown by their semi-mature phenotype and reduced cytokine production. The effect of LF was abrogated by proteinase K treatment, showing that the functional effects of LF were not mediated by LPS contamination. Thus, LF alters DC differentiation and dampens responsiveness towards TLR ligands. This study indicates that LF can play a role in immune homeostasis in the human GI tract.
Collapse
|
48
|
Lanik WE, Xu L, Luke CJ, Hu EZ, Agrawal P, Liu VS, Kumar R, Bolock AM, Ma C, Good M. Breast Milk Enhances Growth of Enteroids: An Ex Vivo Model of Cell Proliferation. J Vis Exp 2018. [PMID: 29553558 PMCID: PMC5912412 DOI: 10.3791/56921] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
Human small intestinal enteroids are derived from the crypts and when grown in a stem cell niche contain all of the epithelial cell types. The ability to establish human enteroid ex vivo culture systems are important to model intestinal pathophysiology and to study the particular cellular responses involved. In recent years, enteroids from mice and humans are being cultured, passaged, and banked away for future use in several laboratories across the world. This enteroid platform can be used to test the effects of various treatments and drugs and what effects are exerted on different cell types in the intestine. Here, a protocol for establishing primary stem cell-derived small intestinal enteroids derived from neonatal mice and premature human intestine is provided. Moreover, this enteroid culture system was utilized to test the effects of species-specific breast milk. Mouse breast milk can be obtained efficiently using a modified human breast pump and expressed mouse milk can then be used for further research experiments. We now demonstrate the effects of expressed mouse, human, and donor breast milk on the growth and proliferation of enteroids derived from neonatal mice or premature human small intestine.
Collapse
Affiliation(s)
- Wyatt E Lanik
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine
| | | | - Cliff J Luke
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine
| | | | | | | | - Rajesh Kumar
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine
| | - Alexa M Bolock
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine
| | - Congrong Ma
- Division of Newborn Medicine, Department of Pediatrics, University of Pittsburgh School of Medicine
| | - Misty Good
- Division of Newborn Medicine, Department of Pediatrics, Washington University School of Medicine;
| |
Collapse
|
49
|
MacManus CF, Collins CB, Nguyen TT, Alfano RW, Jedlicka P, de Zoeten EF. VEN-120, a Recombinant Human Lactoferrin, Promotes a Regulatory T Cell [Treg] Phenotype and Drives Resolution of Inflammation in Distinct Murine Models of Inflammatory Bowel Disease. J Crohns Colitis 2017; 11:1101-1112. [PMID: 28472424 PMCID: PMC5881664 DOI: 10.1093/ecco-jcc/jjx056] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/26/2017] [Accepted: 04/20/2017] [Indexed: 12/13/2022]
Abstract
BACKGROUND AND AIMS Inflammatory bowel disease [IBD] is characterised by a disruption of immune homeostasis, which is tightly regulated to protect against harmful pathogens yet not react to commensal antigens. Animal studies indicate that regulatory T cells [Treg] modulate the immune response to prevent IBD development. Lactoferrin [LF] is an endogenous anti-inflammatory pleiotropic protein secreted at high concentrations in colostrum and at mucosal sites. However, the effect of LF on specific T lymphocyte populations has not been studied. Here, we identify a novel mechanism by which a recombinant human LF, VEN-120, regulates T cell populations in health and disease. METHODS Two murine models of intestinal inflammation, the dextran sodium sulphate colitis model and the TNFΔARE/+ model of ileitis, were used to study the anti-inflammatory and T cell modulating ability of VEN-120. Flow cytometry was used to evaluate T cell populations within the lamina propria and mesenteric lymph nodes, and to evaluate the effect of VEN-120 on CD4+ T cells in vitro. RESULTS VEN-120 reduced inflammation in both models of IBD, accompanied by increased Tregs in the intestinal lamina propria. Treatment of CD4+ T cells in vitro resulted in an upregulation of Treg genes and skewing towards a Treg population. This in vitro T cell skewing translated to an increase of Treg homing to the intestinal lamina propria and associated lymph tissue in healthy mice. CONCLUSIONS These data provide a novel immunological mechanism by which VEN-120 modulates T cells to restrict inflammatory T cell-driven disease.
Collapse
Affiliation(s)
- Christopher F MacManus
- Ventria Biosciences, Fort Collins, CO, USA,Corresponding author: Christopher MacManus, Ventria Bioscience, 320 E. Vine Dr., Fort Collins, CO 80524, USA. Tel.: 970-407-1239;
| | - Colm B Collins
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | - Tom T Nguyen
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| | | | - Paul Jedlicka
- Department of Pathology, University of Colorado School of Medicine, Aurora, Colorado, USA
| | - Edwin F de Zoeten
- Department of Pediatrics, University of Colorado School of Medicine, Aurora, CO, USA
| |
Collapse
|
50
|
Pammi M, Suresh G. Enteral lactoferrin supplementation for prevention of sepsis and necrotizing enterocolitis in preterm infants. Cochrane Database Syst Rev 2017; 6:CD007137. [PMID: 28658720 PMCID: PMC6481465 DOI: 10.1002/14651858.cd007137.pub5] [Citation(s) in RCA: 81] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
BACKGROUND Lactoferrin, a normal component of human colostrum and milk, can enhance host defenses and may be effective for prevention of sepsis and necrotizing enterocolitis (NEC) in preterm neonates. OBJECTIVES Primary objective 1. To assess the safety and effectiveness of lactoferrin supplementation to enteral feeds for prevention of sepsis and NEC in preterm neonates Secondary objectives 1. To determine the effects of lactoferrin supplementation to enteral feeds to prevent neonatal sepsis and/or NEC on duration of positive-pressure ventilation, development of chronic lung disease (CLD) or periventricular leukomalacia (PVL), length of hospital stay to discharge among survivors, and adverse neurological outcomes at two years of age or later2. To determine the adverse effects of lactoferrin supplementation for prophylaxis of neonatal sepsis and/or NECWhen data were available, we analyzed the following subgroups.1. Gestational age < 32 weeks and 32 to 36 weeks2. Birth weight < 1000 g (extremely low birth weight (ELBW) infants) and birth weight < 1500 g (very low birth weight (VLBW) infants)3. Type of feeding: breast milk versus formula milk SEARCH METHODS: We used the search strategy of the Cochrane Neonatal Review Group (CNRG) to update our search in December 2016. We searched the databases Cochrane Central Register of Controlled Trials (CENTRAL), MEDLINE, PREMEDLINE, Embase, and the Cumulative Index to Nursing and Allied Health Literature (CINAHL), as well as trial registries and conference proceedings. SELECTION CRITERIA Randomized controlled trials (RCTs) evaluating oral lactoferrin at any dose or duration to prevent sepsis or NEC in preterm neonates. DATA COLLECTION AND ANALYSIS Review authors used standard methods of the CNRG. MAIN RESULTS This review includes six RCTs. Trial results show that lactoferrin supplementation to enteral feeds decreased late-onset sepsis (typical risk ratio (RR) 0.59, 95% confidence interval (CI) 0.40 to 0.87; typical risk difference (RD) -0.06, 95% CI -0.10 to -0.02; number needed to treat for an additional beneficial outcome (NNTB) 17, 95% CI 10 to 50; six trials, 886 participants; low-quality evidence) and NEC stage II or III (typical RR 0.40, 95% CI 0.18 to 0.86; typical RD -0.04, 95% CI -0.06 to -0.01; NNTB 25, 95% CI 17 to 100; four studies, 750 participants; low-quality evidence). Lactoferrin supplementation did not have an effect on "all-cause mortality" (typical RR 0.65, 95% CI 0.37 to 1.11; typical RD -0.02, 95% CI -0.05 to 0; six studies, 1041 participants; low-quality evidence).Lactoferrin supplementation to enteral feeds with probiotics decreased late-onset sepsis (RR 0.27, 95% CI 0.12 to 0.60; RD -0.13, 95% CI -0.19 to -0.06; NNTB 8, 95% CI 5 to 17; one study, 321 participants; low-quality evidence) and NEC stage II or III (RR 0.04, 95% CI 0.00 to 0.62; RD -0.05, 95% CI -0.08 to -0.03; NNTB 20, 95% CI 12.5 to 33.3; one study, 496 participants; low-quality evidence), but not "all-cause mortality" (low-quality evidence).Lactoferrin supplementation to enteral feeds with or without probiotics decreased bacterial and fungal sepsis but not CLD or length of hospital stay (low-quality evidence). Investigators reported no adverse effects and did not evaluate long-term neurological outcomes and PVL. AUTHORS' CONCLUSIONS Evidence of low quality suggests that lactoferrin supplementation to enteral feeds with or without probiotics decreases late-onset sepsis and NEC stage II or III in preterm infants without adverse effects. Completed ongoing trials will provide data from more than 6000 preterm neonates, which may enhance the quality of the evidence. Clarification regarding optimal dosing regimens, types of lactoferrin (human or bovine), and long-term outcomes is needed.
Collapse
Key Words
- humans
- infant, newborn
- enteral nutrition
- administration, oral
- bacterial infections
- bacterial infections/epidemiology
- bacterial infections/prevention & control
- cause of death
- chronic disease
- enterocolitis, necrotizing
- enterocolitis, necrotizing/epidemiology
- enterocolitis, necrotizing/prevention & control
- infant, premature
- infant, premature, diseases
- infant, premature, diseases/prevention & control
- lactobacillus rhamnosus
- lactoferrin
- lactoferrin/administration & dosage
- lung diseases
- lung diseases/epidemiology
- mycoses
- mycoses/epidemiology
- mycoses/prevention & control
- numbers needed to treat
- probiotics
- probiotics/administration & dosage
- randomized controlled trials as topic
- retinopathy of prematurity
- retinopathy of prematurity/epidemiology
- sepsis
- sepsis/prevention & control
Collapse
Affiliation(s)
- Mohan Pammi
- Baylor College of MedicineSection of Neonatology, Department of Pediatrics6621, Fannin, MC.WT 6‐104HoustonUSA77030
| | - Gautham Suresh
- Baylor College of MedicineSection of Neonatology, Department of Pediatrics6621, Fannin, MC.WT 6‐104HoustonUSA77030
| |
Collapse
|