1
|
Wang Q, Wang X, Ding J, Huang L, Wang Z. Structural insight of cell surface sugars in viral infection and human milk glycans as natural antiviral substance. Int J Biol Macromol 2024; 277:133867. [PMID: 39009265 DOI: 10.1016/j.ijbiomac.2024.133867] [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: 04/03/2024] [Revised: 06/24/2024] [Accepted: 07/11/2024] [Indexed: 07/17/2024]
Abstract
Viral infections are caused by the adhesion of viruses to host cell receptors, including sialylated glycans, glycosaminoglycans, and human blood group antigens (HBGAs). Atomic-level structural information on the interactions between viral particles or proteins with glycans can be determined to provide precise targets for designing antiviral drugs. Milk glycans, existing as free oligosaccharides or glycoconjugates, have attracted increasing attention; milk glycans protect infants against infectious diseases, particularly poorly manageable viral infections. Furthermore, several glycans containing structurally distinct sialic acid/fucose/sulfate modifications in human milk acting as a "receptor decoy" and serving as the natural antiviral library, could interrupt virus-receptor interaction in the first line of defense for viral infection. This review highlights the basis of virus-glycan interactions, presents specific glycan receptor binding by gastroenterovirus viruses, including norovirus, enteroviruses, and the breakthroughs in the studies on the antiviral properties of human milk glycans, and also elucidates the role of glycans in respiratory viruses infection. In addition, recent advances in methods for performing virus/viral protein-glycan interactions were reported. Finally, we discuss the prospects and challenges of the studies on the clinical application of human milk glycan for viral interventions.
Collapse
Affiliation(s)
- Qingling Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Xiaoqin Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Jieqiong Ding
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China
| | - Linjuan Huang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| | - Zhongfu Wang
- Shaanxi Natural Carbohydrate Resource Engineering Research Center, College of Food Science and Technology, Northwest University, Xi'an 710069, China.
| |
Collapse
|
2
|
Vassilopoulou E, Agostoni C, Feketea G, Alberti I, Gianni ML, Milani GP. The Role of Breastfeeding in Acute Respiratory Infections in Infancy. Pediatr Infect Dis J 2024:00006454-990000000-00942. [PMID: 38986006 DOI: 10.1097/inf.0000000000004454] [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: 07/12/2024]
Abstract
BACKGROUND Acute respiratory infections (ARIs) affect the respiratory tract, are often caused by viruses such as respiratory syncytial virus and rhinovirus, and present symptoms such as coughing, fever, respiratory distress, and breathing difficulty. The global adherence to exclusive breastfeeding (BF) for the first 6 months of life has reached 44%, supported by the World Health Organization and United Nations International Children's Emergency Fund efforts. BF provides vital nutrients and contributes to infant immune system development, protecting against infections. The role of BF in preventing and reducing complications of ARIs in infants is gaining attention, prompting a review of current data and future research needs. This review aims to summarize the evidence on the role of BF in reducing the risk and severity of ARIs in infants, elucidate the adaptations in breast milk composition during infections, and identify relevant research needs. METHODS AND RESULTS Human milk (HM) is rich in immunoglobulins, antimicrobial peptides, and immunomodulatory factors that protect against various pathogens, including respiratory viruses. Several studies have demonstrated that BF is associated with a significant reduction in hospitalization, oxygen requirements, and mortality in infants with ARIs. The effectiveness of BF varies according to the specific respiratory virus, and a longer duration of exclusive BF appears to enhance its protective effect. It is documented that the composition of HM adjusts dynamically in response to infections, fortifying the infant's immune defenses. Specific immunological components of HM, including leukocytes and immunoglobulins, increase in response to infection in the infant, contributing to the enhancement of the immune defense in infants. Immune-boosting microRNAs enhance immune transfer to the infants and promote early gut maturation, and the HM microbiome along with other factors modifies the infant's gut microbiome and immune system. CONCLUSIONS BF defends infants from respiratory infections, and the investigation of the microRNAs in HM offers new insights into its antiviral properties. The promotion of BF, especially in vulnerable communities, is of paramount importance in alleviating the global burden of ARIs in infancy.
Collapse
Affiliation(s)
- Emilia Vassilopoulou
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Nutritional Sciences and Dietetics, International Hellenic University, Thessaloniki, Greece
| | - Carlo Agostoni
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| | - Gavriela Feketea
- Department of Pharmacology, Toxicology and Clinical Pharmacology, University of Medicine and Pharmacy, Cluj-Napoca, Romania
- Pediatric Allergy Outpatient Clinic, Department of Pediatrics, "Karamandaneio" Children's Hospital of Patra, Patras, Greece
| | - Ilaria Alberti
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
| | - Maria Lorella Gianni
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
- Fondazione I.R.C.C.S. Ca' Granda Ospedale Maggiore Policlinico, Neonatal Intensive Care Unit, Milan, Italy
| | - Gregorio Paolo Milani
- From the Pediatric Unit, Fondazione IRCCS Ca' Granda Ospedale Maggiore Policlinico, Milan, Italy
- Department of Clinical Sciences and Community Health, Università degli Studi di Milano, Milan, Italy
| |
Collapse
|
3
|
Wang Y, Wu J, Zhang H, Yang X, Gu R, Liu Y, Wu R. Comprehensive review of milk fat globule membrane proteins across mammals and lactation periods in health and disease. Crit Rev Food Sci Nutr 2024:1-22. [PMID: 39106211 DOI: 10.1080/10408398.2024.2387763] [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: 08/09/2024]
Abstract
Milk fat globule membrane (MFGM) is a three-layer membrane-like structure encasing natural milk fat globules (MFGs). MFGM holds promise as a nutritional supplement because of the numerous physiological functions of its constituent protein. This review summarizes and compares the differences in MFGM protein composition across various species, including bovines, goats, camels, mares, and donkeys, and different lactation periods, such as colostrum and mature milk, as assessed by techniques such as proteomics and mass spectrometry. We also discuss the health benefits of MFGM proteins throughout life. MFGM proteins promote intestinal development, neurodevelopment, and glucose and lipid metabolism by upregulating tight junction protein expression, brain function-related genes, and glucose and fatty acid biosynthesis processes. We focus on the mechanisms underlying these beneficial effects of MFGM proteins. MFGM proteins activate key substances in in signaling pathways, such as the phosphatidylinositol 3-kinase/protein kinase B, mitogen-activated protein kinase, and myosin light chain kinase signaling pathways. Overall, the consumption of MFGM proteins plays an essential role in conferring health benefits, some of which are important throughout the mammalian life cycle.
Collapse
Affiliation(s)
- Ying Wang
- College of Food Science, Shenyang Agricultural University, Shenyang, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, P.R. China
| | - Junrui Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, P.R. China
| | - Henan Zhang
- College of Food Science, Shenyang Agricultural University, Shenyang, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang, P.R. China
| | - Xujin Yang
- College of Food Science and Engineering, Inner Mongolia Agricultural University, Huhhot, P.R. China
| | - Ruixia Gu
- School of Food Science and Engineering, Yangzhou University, Yangzhou, P.R. China
| | - Yumeng Liu
- College of Food Science, Shenyang Agricultural University, Shenyang, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, P.R. China
| | - Rina Wu
- College of Food Science, Shenyang Agricultural University, Shenyang, P.R. China
- Shenyang Key Laboratory of Microbial Fermentation Technology Innovation, Shenyang, P.R. China
- Liaoning Engineering Research Center of Food Fermentation Technology, Shenyang, P.R. China
| |
Collapse
|
4
|
Francese R, Peila C, Donalisio M, Lamberti C, Cirrincione S, Colombi N, Tonetto P, Cavallarin L, Bertino E, Moro GE, Coscia A, Lembo D. Viruses and Human Milk: Transmission or Protection? Adv Nutr 2023; 14:1389-1415. [PMID: 37604306 PMCID: PMC10721544 DOI: 10.1016/j.advnut.2023.08.007] [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: 05/06/2023] [Revised: 07/14/2023] [Accepted: 08/15/2023] [Indexed: 08/23/2023] Open
Abstract
Human milk (HM) is considered the best source of nutrition for infant growth and health. This nourishment is unique and changes constantly during lactation to adapt to the physiological needs of the developing infant. It is also recognized as a potential route of transmission of some viral pathogens although the presence of a virus in HM rarely leads to a disease in an infant. This intriguing paradox can be explained by considering the intrinsic antiviral properties of HM. In this comprehensive and schematically presented review, we have described what viruses have been detected in HM so far and what their potential transmission risk through breastfeeding is. We have provided a description of all the antiviral compounds of HM, along with an analysis of their demonstrated and hypothesized mechanisms of action. Finally, we have also analyzed the impact of HM pasteurization and storage methods on the detection and transmission of viruses, and on the antiviral compounds of HM. We have highlighted that there is currently a deep knowledge on the potential transmission of viral pathogens through breastfeeding and on the antiviral properties of HM. The current evidence suggests that, in most cases, it is unnecessarily to deprive an infant of this high-quality nourishment and that the continuation of breastfeeding is in the best interest of the infant and the mother.
Collapse
Affiliation(s)
- Rachele Francese
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy
| | - Chiara Peila
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Manuela Donalisio
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy
| | - Cristina Lamberti
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Simona Cirrincione
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Nicoletta Colombi
- Biblioteca Federata di Medicina "Ferdinando Rossi", University of Turin, Turin, Italy
| | - Paola Tonetto
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Laura Cavallarin
- Institute of the Science of Food Production - National Research Council, Grugliasco, TO, Italy
| | - Enrico Bertino
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy
| | - Guido E Moro
- Italian Association of Human Milk Banks (AIBLUD), Milan, Italy.
| | - Alessandra Coscia
- Department of Public Health and Pediatrics, Neonatal Intensive Care Unit, University of Turin, Turin, Italy.
| | - David Lembo
- Department of Clinical and Biological Sciences, Laboratory of Molecular Virology and Antiviral Research, University of Turin, Orbassano (TO), Italy.
| |
Collapse
|
5
|
Umar M, Ruktanonchai U, Makararpong D, Anal AK. Enhancing Immunity Against Pathogens Through Glycosylated Bovine Colostrum Proteins. FOOD REVIEWS INTERNATIONAL 2023. [DOI: 10.1080/87559129.2023.2169866] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Affiliation(s)
- Muhammad Umar
- Department of Food, Agriculture and Bioresources, Asian Institute of Technology, Pathumthani, Thailand
| | - Uracha Ruktanonchai
- NANOTEC, National Science and Technology Development Agency, Pathumthani, Thailand
| | | | - Anil Kumar Anal
- Department of Food, Agriculture and Bioresources, Asian Institute of Technology, Pathumthani, Thailand
| |
Collapse
|
6
|
Kaplan M, Şahutoğlu AS, Sarıtaş S, Duman H, Arslan A, Pekdemir B, Karav S. Role of milk glycome in prevention, treatment, and recovery of COVID-19. Front Nutr 2022; 9:1033779. [PMID: 36424926 PMCID: PMC9680090 DOI: 10.3389/fnut.2022.1033779] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2022] [Accepted: 10/10/2022] [Indexed: 08/23/2023] Open
Abstract
Milk contains all essential macro and micro-nutrients for the development of the newborn. Its high therapeutic and antimicrobial content provides an important function for the prevention, treatment, and recovery of certain diseases throughout life. The bioactive components found in milk are mostly decorated with glycans, which provide proper formation and modulate the biological functions of glycosylated compounds. The glycome of milk consists of free glycans, glycolipids, and N- and O- glycosylated proteins. Recent studies have shown that both free glycans and glycan-containing molecules have antiviral characteristics based on different mechanisms such as signaling, microbiome modulation, natural decoy strategy, and immunomodulatory action. In this review, we discuss the recent clinical studies and potential mechanisms of free and conjugated glycans' role in the prevention, treatment, and recovery of COVID-19.
Collapse
Affiliation(s)
- Merve Kaplan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | | | - Sümeyye Sarıtaş
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Hatice Duman
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Ayşenur Arslan
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Burcu Pekdemir
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| | - Sercan Karav
- Department of Molecular Biology and Genetics, Çanakkale Onsekiz Mart University, Çanakkale, Turkey
| |
Collapse
|
7
|
Membrane-Tethered Mucin 1 Is Stimulated by Interferon and Virus Infection in Multiple Cell Types and Inhibits Influenza A Virus Infection in Human Airway Epithelium. mBio 2022; 13:e0105522. [PMID: 35699372 PMCID: PMC9426523 DOI: 10.1128/mbio.01055-22] [Citation(s) in RCA: 8] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Influenza A virus (IAV) causes significant morbidity and mortality in the human population. Tethered mucin 1 (MUC1) is highly expressed in airway epithelium, the primary site of IAV replication, and also by other cell types that influence IAV infection, including macrophages. MUC1 has the potential to influence infection dynamics through physical interactions and/or signaling activity, yet MUC1 modulation and its impact during viral pathogenesis remain unclear. Thus, we investigated MUC1-IAV interactions in an in vitro model of human airway epithelium (HAE). Our data indicate that a recombinant IAV hemagglutinin (H3) and H3N2 virus can bind endogenous HAE MUC1. Notably, infection of HAE with H1N1 or H3N2 IAV strains does not trigger MUC1 shedding but instead stimulates an increase in cell-associated MUC1 protein. We observed a similar increase after type I or III interferon (IFN) stimulation; however, inhibition of IFN signaling during H1N1 infection only partially abrogated this increase, indicating that multiple soluble factors contribute to MUC1 upregulation during the antiviral response. In addition to HAE, primary human monocyte-derived macrophages also upregulated MUC1 protein in response to IFN treatment and conditioned media from IAV-infected HAE. Then, to determine the impact of MUC1 on IAV pathogenesis, we developed HAE genetically depleted of MUC1 and found that MUC1 knockout cultures exhibited enhanced viral growth compared to control cultures for several IAV strains. Together, our data support a model whereby MUC1 inhibits productive uptake of IAV in HAE. Infection then stimulates MUC1 expression on multiple cell types through IFN-dependent and -independent mechanisms that further impact infection dynamics.
Collapse
|
8
|
Lillehoj EP, Luzina IG, Atamas SP. Mammalian Neuraminidases in Immune-Mediated Diseases: Mucins and Beyond. Front Immunol 2022; 13:883079. [PMID: 35479093 PMCID: PMC9035539 DOI: 10.3389/fimmu.2022.883079] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2022] [Accepted: 03/21/2022] [Indexed: 12/28/2022] Open
Abstract
Mammalian neuraminidases (NEUs), also known as sialidases, are enzymes that cleave off the terminal neuraminic, or sialic, acid resides from the carbohydrate moieties of glycolipids and glycoproteins. A rapidly growing body of literature indicates that in addition to their metabolic functions, NEUs also regulate the activity of their glycoprotein targets. The simple post-translational modification of NEU protein targets-removal of the highly electronegative sialic acid-affects protein folding, alters protein interactions with their ligands, and exposes or covers proteolytic sites. Through such effects, NEUs regulate the downstream processes in which their glycoprotein targets participate. A major target of desialylation by NEUs are mucins (MUCs), and such post-translational modification contributes to regulation of disease processes. In this review, we focus on the regulatory roles of NEU-modified MUCs as coordinators of disease pathogenesis in fibrotic, inflammatory, infectious, and autoimmune diseases. Special attention is placed on the most abundant and best studied NEU1, and its recently discovered important target, mucin-1 (MUC1). The role of the NEU1 - MUC1 axis in disease pathogenesis is discussed, along with regulatory contributions from other MUCs and other pathophysiologically important NEU targets.
Collapse
Affiliation(s)
- Erik P. Lillehoj
- Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD, United States
| | - Irina G. Luzina
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
- Research Service, Baltimore Veterans Affairs (VA) Medical Center, Baltimore, MD, United States
| | - Sergei P. Atamas
- Department of Medicine, University of Maryland School of Medicine, Baltimore, MD, United States
| |
Collapse
|
9
|
Yadav M, Kapoor A, Verma A, Ambatipudi K. Functional Significance of Different Milk Constituents in Modulating the Gut Microbiome and Infant Health. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2022; 70:3929-3947. [PMID: 35324181 DOI: 10.1021/acs.jafc.2c00335] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/14/2023]
Abstract
Human milk, the gold standard for optimal nourishment, controls the microbial composition of infants by either enhancing or limiting bacterial growth. The milk fat globule membrane has gained interest in gut-related functions and cognitive development. The membrane proteins can directly interact with probiotic bacteria, influencing their survival and adhesion through gastrointestinal transit, whereas membrane phospholipids increase the residence time of probiotic bacteria in the gut. The commensal bacteria in milk act as the initial inoculum in building up the gut colonization of an infant, whereas oligosaccharides promote proliferation of beneficial microorganisms. Interestingly, milk extracellular vesicles are also involved in influencing the microbiota composition but are not well-explored. This review highlights the contribution of different milk components in modulating the infant gut microbiota, particularly the fat globule membrane, and the complex interplay between host- and brain-gut microbiota signaling affecting infant and adult health positively.
Collapse
Affiliation(s)
- Monica Yadav
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Ayushi Kapoor
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Aparna Verma
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| | - Kiran Ambatipudi
- Department of Biosciences and Bioengineering, Indian Institute of Technology Roorkee, Roorkee 247667, India
| |
Collapse
|
10
|
Kim YJ. Immunomodulatory Effects of Human Colostrum and Milk. Pediatr Gastroenterol Hepatol Nutr 2021; 24:337-345. [PMID: 34316468 PMCID: PMC8279828 DOI: 10.5223/pghn.2021.24.4.337] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/02/2021] [Revised: 04/27/2021] [Accepted: 05/17/2021] [Indexed: 01/03/2023] Open
Abstract
The immune system is not fully developed in human neonates and infants; breastfeeding is important in this stage as the bioactive components of human breast milk are known to have anti-microbial, anti-inflammatory, and immunomodulatory effects, and can therefore contribute to an infant's immunity against allergies, asthma, autoimmune diseases, and inflammatory bowel disease. Herein, the positive effect on the immune system by human colostrum and milk are reviewed.
Collapse
Affiliation(s)
- Yong Joo Kim
- Department of Pediatrics, Hanyang University College of Medicine, Seoul, Korea
| |
Collapse
|
11
|
Thum C, Roy NC, Everett DW, McNabb WC. Variation in milk fat globule size and composition: A source of bioactives for human health. Crit Rev Food Sci Nutr 2021; 63:87-113. [PMID: 34190660 DOI: 10.1080/10408398.2021.1944049] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Milk fat globules (MFGs) are secreted from the mammalian gland and are composed of a triacylglycerol core surrounded by a triple membrane structure, the milk fat globule membrane (MFGM). The MFGM contains complex lipids and proteins reported to have nutritional, immunological, neurological and digestive functions. Human and ruminant milk are shown to share a similar MFG structure but with different size, profile and abundance of protein and polar lipids. This review summarizes the reported data on human, bovine, caprine and ovine MFG composition and concentration of bioactive components in different MFG-size fractions. A comprehensive understanding of compositional variations between milk from different species and MFG size fractions may help promote various milk sources as targeted supplements to improve human development and health. MFG size and MFGM composition are species-specific and affected by lactation, diet and breed (or maternal origin). Purification and enrichment methods for some bioactive proteins and lipids present in the MFGM have yet to be established or are not scaled sufficiently to be used to supplement human diets. To overcome this problem, MFG size selection through fractionation or herd selection may provide a convenient way to pre-enrich the MFG fraction with specific protein and lipid components to fulfill human dietary and health requirements.
Collapse
Affiliation(s)
- Caroline Thum
- AgResearch, Grasslands Research Centre, Palmerston North, New Zealand.,Riddet Institute, Palmerston North, New Zealand
| | - Nicole C Roy
- AgResearch, Grasslands Research Centre, Palmerston North, New Zealand.,Riddet Institute, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, The University of Auckland, Auckland, New Zealand.,Department of Human Nutrition, University of Otago, Dunedin, New Zealand
| | - David W Everett
- AgResearch, Grasslands Research Centre, Palmerston North, New Zealand.,Riddet Institute, Palmerston North, New Zealand
| | - Warren C McNabb
- Riddet Institute, Palmerston North, New Zealand.,High-Value Nutrition National Science Challenge, The University of Auckland, Auckland, New Zealand
| |
Collapse
|
12
|
Morrin ST, Buck RH, Farrow M, Hickey RM. Milk-derived anti-infectives and their potential to combat bacterial and viral infection. J Funct Foods 2021. [DOI: 10.1016/j.jff.2021.104442] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022] Open
|
13
|
Boudry G, Charton E, Le Huerou-Luron I, Ferret-Bernard S, Le Gall S, Even S, Blat S. The Relationship Between Breast Milk Components and the Infant Gut Microbiota. Front Nutr 2021; 8:629740. [PMID: 33829032 PMCID: PMC8019723 DOI: 10.3389/fnut.2021.629740] [Citation(s) in RCA: 63] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2020] [Accepted: 02/16/2021] [Indexed: 12/12/2022] Open
Abstract
The assembly of the newborn's gut microbiota during the first months of life is an orchestrated process resulting in specialized microbial ecosystems in the different gut compartments. This process is highly dependent upon environmental factors, and many evidences suggest that early bacterial gut colonization has long-term consequences on host digestive and immune homeostasis but also metabolism and behavior. The early life period is therefore a "window of opportunity" to program health through microbiota modulation. However, the implementation of this promising strategy requires an in-depth understanding of the mechanisms governing gut microbiota assembly. Breastfeeding has been associated with a healthy microbiota in infants. Human milk is a complex food matrix, with numerous components that potentially influence the infant microbiota composition, either by enhancing specific bacteria growth or by limiting the growth of others. The objective of this review is to describe human milk composition and to discuss the established or purported roles of human milk components upon gut microbiota establishment. Finally, the impact of maternal diet on human milk composition is reviewed to assess how maternal diet could be a simple and efficient approach to shape the infant gut microbiota.
Collapse
Affiliation(s)
- Gaëlle Boudry
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| | - Elise Charton
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
- UMR STLO INRAE, Institut Agro, Rennes, France
| | | | | | - Sophie Le Gall
- INRAE, UR BIA, Nantes, France
- INRAE, BIBS facility, Nantes, France
| | | | - Sophie Blat
- Institut NuMeCan, INRAE, INSERM, Univ Rennes, Saint-Gilles, France
| |
Collapse
|
14
|
Abstract
This review highlights clinical outcomes of human milk from infancy through adulthood. Human milk outcomes of both preterm and term infants, including critically ill term infants (such as infants with congenital heart disease and those requiring therapeutic hypothermia) are summarized. Several human milk diets are identified to reduce the risk of specific diseases. Emerging research of newly discovered components of human milk are also reviewed. Human milk has significant effects on the gut microbiome, somatic growth, and neurocognitive outcomes. Continued research promises to improve donor human milk and donor milk derived products to achieve better outcomes for infants who do not receive their own mother's milk. The promotion of human milk is well-founded on evidence from the previous half century.
Collapse
Affiliation(s)
- Katherine E Chetta
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, United States.
| | - Elizabeth V Schulz
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Uniformed Services University, United States
| | - Carol L Wagner
- Department of Pediatrics, Division of Neonatal-Perinatal Medicine, Medical University of South Carolina, Shawn Jenkins Children's Hospital, 10 McClennan Banks Drive, MSC 915, Charleston, SC 29425, United States
| |
Collapse
|
15
|
van Leeuwen SS, te Poele EM, Chatziioannou AC, Benjamins E, Haandrikman A, Dijkhuizen L. Goat Milk Oligosaccharides: Their Diversity, Quantity, and Functional Properties in Comparison to Human Milk Oligosaccharides. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2020; 68:13469-13485. [PMID: 33141570 PMCID: PMC7705968 DOI: 10.1021/acs.jafc.0c03766] [Citation(s) in RCA: 51] [Impact Index Per Article: 12.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Human milk is considered the golden standard in infant nutrition. Free oligosaccharides in human milk provide important health benefits. These oligosaccharides function as prebiotics, immune modulators, and pathogen inhibitors and were found to improve barrier function in the gut. Infant formulas nowadays often contain prebiotics but lack the specific functions of human milk oligosaccharides (hMOS). Milk from domesticated animals also contains milk oligosaccharides but at much lower levels and with less diversity. Goat milk contains significantly more oligosaccharides (gMOS) than bovine (bMOS) or sheep (sMOS) milk and also has a larger diversity of structures. This review summarizes structural studies, revealing a diversity of up to 77 annotated gMOS structures with almost 40 structures fully characterized. Quantitative studies of goat milk oligosaccharides range from 60 to 350 mg/L in mature milk and from 200 to 650 mg/L in colostrum. These levels are clearly lower than in human milk (5-20 g/L) but higher than in other domesticated dairy animals, e.g., bovine (30-60 mg/L) and sheep (20-40 mg/L). Finally, the review focuses on demonstrated and potential functionalities of gMOS. Some studies have shown anti-inflammatory effects of mixtures enriched in gMOS. Goat MOS also display prebiotic potential, particularly in stimulating growth of bifidobacteria preferentially. Although functional studies of gMOS are still limited, several structures are also found in human milk and have known functions as immune modulators and pathogen inhibitors. In conclusion, goat milk constitutes a promising alternative source for milk oligosaccharides, which can be used in infant formula.
Collapse
Affiliation(s)
- Sander S. van Leeuwen
- Department
of Laboratory Medicine, Cluster Human Nutrition and Health, University Medical Center Groningen (UMCG), Hanzeplein 1, 9713 GZ Groningen, Netherlands
- E-mail:
| | | | | | | | | | - Lubbert Dijkhuizen
- CarbExplore
Research BV, 9747 AN Groningen, Netherlands
- Department
of Microbial Physiology, Groningen Biomolecular Sciences and Biotechnology
Institute (GBB), University of Groningen, 9700 AB Groningen, Netherlands
| |
Collapse
|
16
|
Valverde P, Martínez JD, Cañada FJ, Ardá A, Jiménez-Barbero J. Molecular Recognition in C-Type Lectins: The Cases of DC-SIGN, Langerin, MGL, and L-Sectin. Chembiochem 2020; 21:2999-3025. [PMID: 32426893 PMCID: PMC7276794 DOI: 10.1002/cbic.202000238] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Revised: 05/19/2020] [Indexed: 12/16/2022]
Abstract
Carbohydrates play a pivotal role in intercellular communication processes. In particular, glycan antigens are key for sustaining homeostasis, helping leukocytes to distinguish damaged tissues and invading pathogens from healthy tissues. From a structural perspective, this cross-talk is fairly complex, and multiple membrane proteins guide these recognition processes, including lectins and Toll-like receptors. Since the beginning of this century, lectins have become potential targets for therapeutics for controlling and/or avoiding the progression of pathologies derived from an incorrect immune outcome, including infectious processes, cancer, or autoimmune diseases. Therefore, a detailed knowledge of these receptors is mandatory for the development of specific treatments. In this review, we summarize the current knowledge about four key C-type lectins whose importance has been steadily growing in recent years, focusing in particular on how glycan recognition takes place at the molecular level, but also looking at recent progresses in the quest for therapeutics.
Collapse
Affiliation(s)
- Pablo Valverde
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - J Daniel Martínez
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - F Javier Cañada
- Centro de Investigaciones Biológicas Margarita Salas, CSIC, Ramiro de Maeztu 9, 28040, Madrid, Spain
- CIBER de Enfermedades Respiratorias (CIBERES), Avda Monforte de Lemos 3-5, 28029, Madrid, Spain
| | - Ana Ardá
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
| | - Jesús Jiménez-Barbero
- CIC bioGUNE, Basque Research Technology Alliance, BRTA, Bizkaia Technology park, Building 800, 48160, Derio, Spain
- Ikerbasque, Basque Foundation for Science, 48009, Bilbao, Spain
- Department of Organic Chemistry II, Faculty of Science and Technology, UPV-EHU, 48940, Leioa, Spain
| |
Collapse
|
17
|
Identification and Detection of Bioactive Peptides in Milk and Dairy Products: Remarks about Agro-Foods. Molecules 2020; 25:molecules25153328. [PMID: 32707993 PMCID: PMC7435915 DOI: 10.3390/molecules25153328] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Revised: 07/13/2020] [Accepted: 07/17/2020] [Indexed: 02/07/2023] Open
Abstract
Food-based components represent major sources of functional bioactive compounds. Milk is a rich source of multiple bioactive peptides that not only help to fulfill consumers 'nutritional requirements but also play a significant role in preventing several health disorders. Understanding the chemical composition of milk and its products is critical for producing consistent and high-quality dairy products and functional dairy ingredients. Over the last two decades, peptides have gained significant attention by scientific evidence for its beneficial health impacts besides their established nutrient value. Increasing awareness of essential milk proteins has facilitated the development of novel milk protein products that are progressively required for nutritional benefits. The need to better understand the beneficial effects of milk-protein derived peptides has, therefore, led to the development of analytical approaches for the isolation, separation and identification of bioactive peptides in complex dairy products. Continuous emphasis is on the biological function and nutritional characteristics of milk constituents using several powerful techniques, namely omics, model cell lines, gut microbiome analysis and imaging techniques. This review briefly describes the state-of-the-art approach of peptidomics and lipidomics profiling approaches for the identification and detection of milk-derived bioactive peptides while taking into account recent progress in their analysis and emphasizing the difficulty of analysis of these functional and endogenous peptides.
Collapse
|
18
|
Donalisio M, Cirrincione S, Rittà M, Lamberti C, Civra A, Francese R, Tonetto P, Sottemano S, Manfredi M, Lorenzato A, Moro GE, Giribaldi M, Cavallarin L, Giuffrida MG, Bertino E, Coscia A, Lembo D. Extracellular Vesicles in Human Preterm Colostrum Inhibit Infection by Human Cytomegalovirus In Vitro. Microorganisms 2020; 8:microorganisms8071087. [PMID: 32708203 PMCID: PMC7409124 DOI: 10.3390/microorganisms8071087] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2020] [Revised: 06/08/2020] [Accepted: 07/17/2020] [Indexed: 11/16/2022] Open
Abstract
Breast milk is a complex biofluid that nourishes infants, supports their growth and protects them from diseases. However, at the same time, breastfeeding is a transmission route for human cytomegalovirus (HCMV), with preterm infants being at a great risk of congenital disease. The discrepancy between high HCMV transmission rates and the few reported cases of infants with severe clinical illness is likely due to the protective effect of breast milk. The aim of this study was to investigate the anti-HCMV activity of human preterm colostrum and clarify the role of colostrum-derived extracellular vesicles (EVs). Preterm colostrum samples were collected and the EVs were purified and characterized. The in vitro anti-HCMV activity of both colostrum and EVs was tested against HCMV, and the viral replication step inhibited by colostrum-purified EVs was examined. We investigated the putative role EV surface proteins play in impairing HCMV infection using shaving experiments and proteomic analysis. The obtained results confirmed the antiviral action of colostrum against HCMV and demonstrated a remarkable antiviral activity of colostrum-derived EVs. Furthermore, we demonstrated that EVs impair the attachment of HCMV to cells, with EV surface proteins playing a role in mediating this action. These findings contribute to clarifying the mechanisms that underlie the protective role of human colostrum against HCMV infection.
Collapse
Affiliation(s)
- Manuela Donalisio
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
- Correspondence: (M.D.); (A.C.); Tel.: +39-011-6705427 (M.D.); +39-011-3134437 (A.C.)
| | - Simona Cirrincione
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Massimo Rittà
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Cristina Lamberti
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Andrea Civra
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Rachele Francese
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| | - Paola Tonetto
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Stefano Sottemano
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Marcello Manfredi
- Center for Translational Research on Autoimmune and Allergic Disease (CAAD), University of Piemonte Orientale, 28100 Novara, Italy;
| | - Annalisa Lorenzato
- Candiolo Cancer Institute, FPO-IRCCS, 10060 Candiolo (TO), Italy;
- Department of Oncology, University of Turin, 10060 Candiolo (TO), Italy
| | - Guido E. Moro
- Italian Association of Human Milk Banks, 20126 Milano, Italy;
| | - Marzia Giribaldi
- Research Centre for Engineering and Agro-food Processing (CREA), 10135 Torino, Italy;
| | - Laura Cavallarin
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Maria Gabriella Giuffrida
- Consiglio Nazionale delle Ricerche-Istituto di Scienze delle Produzioni Alimentari, 10095 Grugliasco (TO), Italy; (S.C.); (C.L.); (L.C.); (M.G.G.)
| | - Enrico Bertino
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
| | - Alessandra Coscia
- Neonatal Intensive Care Unit, Department of Public Health and Pediatrics, University of Turin, 10126 Torino, Italy; (P.T.); (S.S.); (E.B.)
- Correspondence: (M.D.); (A.C.); Tel.: +39-011-6705427 (M.D.); +39-011-3134437 (A.C.)
| | - David Lembo
- Laboratory of Molecular Virology, Department of Clinical and Biological Sciences, University of Turin, 10043 Orbassano, Italy; (M.R.); (A.C.); (R.F.); (D.L.)
| |
Collapse
|
19
|
Sanwlani R, Fonseka P, Chitti SV, Mathivanan S. Milk-Derived Extracellular Vesicles in Inter-Organism, Cross-Species Communication and Drug Delivery. Proteomes 2020; 8:11. [PMID: 32414045 PMCID: PMC7356197 DOI: 10.3390/proteomes8020011] [Citation(s) in RCA: 82] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 05/07/2020] [Accepted: 05/11/2020] [Indexed: 12/13/2022] Open
Abstract
Milk is considered as more than a source of nutrition for infants and is a vector involved in the transfer of bioactive compounds and cells. Milk contains abundant quantities of extracellular vesicles (EVs) that may originate from multiple cellular sources. These nanosized vesicles have been well characterized and are known to carry a diverse cargo of proteins, nucleic acids, lipids and other biomolecules. Milk-derived EVs have been demonstrated to survive harsh and degrading conditions in gut, taken up by various cell types, cross biological barriers and reach peripheral tissues. The cargo carried by these dietary EVs has been suggested to have a role in cell growth, development, immune modulation and regulation. Hence, there is considerable interest in understanding the role of milk-derived EVs in mediating inter-organismal and cross-species communication. Furthermore, various attributes such as it being a natural source, as well as its abundance, scalability, economic viability and lack of unwarranted immunologic reactions, has generated significant interest in deploying milk-derived EVs for clinical applications such as drug delivery and disease therapy. In this review, the role of milk-derived EVs in inter-organismal, cross-species communication and in drug delivery is discussed.
Collapse
Affiliation(s)
| | | | | | - Suresh Mathivanan
- Department of Biochemistry and Genetics, La Trobe Institute for Molecular Science, La Trobe University, Bundoora, VIC 3083, Australia; (R.S.); (P.F.); (S.V.C.)
| |
Collapse
|
20
|
Orczyk-Pawiłowicz M, Lis-Kuberka J. The Impact of Dietary Fucosylated Oligosaccharides and Glycoproteins of Human Milk on Infant Well-Being. Nutrients 2020; 12:nu12041105. [PMID: 32316160 PMCID: PMC7230487 DOI: 10.3390/nu12041105] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2020] [Revised: 04/05/2020] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
Apart from optimal nutritional value, human milk is the feeding strategy to support the immature immunological system of developing newborns and infants. The most beneficial dietary carbohydrate components of breast milk are human milk oligosaccharides (HMOs) and glycoproteins (HMGs), involved in both specific and nonspecific immunity. Fucosylated oligosaccharides represent the largest fraction of human milk oligosaccharides, with the simplest and the most abundant being 2'-fucosyllactose (2'FL). Fucosylated oligosaccharides, as well as glycans of glycoproteins, as beneficial dietary sugars, elicit anti-adhesive properties against fucose-dependent pathogens, and on the other hand are crucial for growth and metabolism of beneficial bacteria, and in this aspect participate in shaping a healthy microbiome. Well-documented secretor status related differences in the fucosylation profile of HMOs and HMGs may play a key but underestimated role in assessment of susceptibility to fucose-dependent pathogen infections, with a potential impact on applied clinical procedures. Nevertheless, due to genetic factors, about 20% of mothers do not provide their infants with beneficial dietary carbohydrates such as 2'-FL and other α1,2-fucosylated oligosaccharides and glycans of glycoproteins, despite breastfeeding them. The lack of such structures may have important implications for a wide range of aspects of infant well-being and healthcare. In light of the above, some artificial mixtures used in infant nutrition are supplemented with 2'-FL to more closely approximate the unique composition of maternal milk, including dietary-derived fucosylated oligosaccharides and glycoproteins.
Collapse
Affiliation(s)
| | - Jolanta Lis-Kuberka
- Correspondence: (M.O.-P.); (J.L.-K.); Tel.: +48-71-770-30-64 (M.O.-P.); +48-71-770-32-17 (J.L.-K.)
| |
Collapse
|
21
|
Polyclonal HIV envelope-specific breast milk antibodies limit founder SHIV acquisition and cell-associated virus loads in infant rhesus monkeys. Mucosal Immunol 2018; 11:1716-1726. [PMID: 30115994 PMCID: PMC6420805 DOI: 10.1038/s41385-018-0067-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 06/18/2018] [Accepted: 06/23/2018] [Indexed: 02/04/2023]
Abstract
Breast milk HIV-1 transmission is currently the predominant contributor to pediatric HIV infections. Yet, only ~10% of breastfeeding infants born to untreated HIV-infected mothers become infected. This study assessed the protective capacity of natural HIV envelope-specific antibodies isolated from the milk of HIV-infected women in an infant rhesus monkey (RM), tier 2 SHIV oral challenge model. To mimic placental and milk maternal antibody transfer, infant RMs were i.v. infused and orally treated at the time of challenge with a single weakly neutralizing milk monoclonal antibody (mAb), a tri-mAb cocktail with weakly neutralizing and ADCC functionalities, or an anti-influenza control mAb. Of these groups, the fewest tri-mAb-treated infants had SHIV detectable in plasma or tissues (2/6, 5/6, and 7/8 animals infected in tri-mAb, single-mAb, and control-mAb groups, respectively). Tri-mAb-treated infants demonstrated significantly fewer plasma transmitted/founder variants and reduced peripheral CD4+ T cell proviral loads at 8 weeks post-challenge compared to control mAb-treated infants. Abortive infection was observed as detectable CD4+ T cell provirus in non-viremic control mAb- and single mAb-, but not in tri-mAb-treated animals. These results suggest that polyfunctional milk antibodies contribute to the natural inefficiency of HIV-1 transmission through breastfeeding and infant vaccinations eliciting non-neutralizing antibody responses could reduce postnatal HIV transmission.
Collapse
|
22
|
Henrick BM, Yao XD, Nasser L, Roozrogousheh A, Rosenthal KL. Breastfeeding Behaviors and the Innate Immune System of Human Milk: Working Together to Protect Infants against Inflammation, HIV-1, and Other Infections. Front Immunol 2017; 8:1631. [PMID: 29238342 PMCID: PMC5712557 DOI: 10.3389/fimmu.2017.01631] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 11/09/2017] [Indexed: 12/21/2022] Open
Abstract
The majority of infants’ breastfeeding from their HIV-infected mothers do not acquire HIV-1 infection despite exposure to cell-free virus and cell-associated virus in HIV-infected breast milk. Paradoxically, exclusive breastfeeding regardless of the HIV status of the mother has led to a significant decrease in mother-to-child transmission (MTCT) compared with non-exclusive breastfeeding. Although it remains unclear how these HIV-exposed infants remain uninfected despite repeated and prolonged exposure to HIV-1, the low rate of transmission is suggestive of a multitude of protective, short-lived bioactive innate immune factors in breast milk. Indeed, recent studies of soluble factors in breast milk shed new light on mechanisms of neonatal HIV-1 protection. This review highlights the role and significance of innate immune factors in HIV-1 susceptibility and infection. Prevention of MTCT of HIV-1 is likely due to multiple factors, including innate immune factors such as lactoferrin and elafin among many others. In pursuing this field, our lab was the first to show that soluble toll-like receptor 2 (sTLR2) directly inhibits HIV infection, integration, and inflammation. More recently, we demonstrated that sTLR2 directly binds to selective HIV-1 proteins, including p17, gp41, and p24, leading to significantly reduced NFκB activation, interleukin-8 production, CCR5 expression, and HIV infection in a dose-dependent manner. Thus, a clearer understanding of soluble milk-derived innate factors with known antiviral functions may provide new therapeutic insights to reduce vertical HIV-1 transmission and will have important implications for protection against HIV-1 infection at other mucosal sites. Furthermore, innate bioactive factors identified in human milk may serve not only in protecting infants against infections and inflammation but also the elderly; thus, opening the door for novel innate immune therapeutics to protect newborns, infants, adults, and the elderly.
Collapse
Affiliation(s)
- Bethany M Henrick
- Department of Food Science and Technology, University of California, Davis, Davis, CA, United States.,Foods for Health Institute, University of California, Davis, Davis, CA, United States
| | - Xiao-Dan Yao
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Laila Nasser
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Ava Roozrogousheh
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| | - Kenneth L Rosenthal
- McMaster Immunology Research Centre, Department of Pathology and Molecular Medicine, Michael G. DeGroote Institute for Infectious Disease Research, McMaster University, Hamilton, ON, Canada
| |
Collapse
|
23
|
Mall AS, Habte H, Mthembu Y, Peacocke J, de Beer C. Mucus and Mucins: do they have a role in the inhibition of the human immunodeficiency virus? Virol J 2017; 14:192. [PMID: 28985745 PMCID: PMC5639604 DOI: 10.1186/s12985-017-0855-9] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2017] [Accepted: 09/22/2017] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Mucins are large O-linked glycosylated proteins which give mucus their gel-forming properties. There are indications that mucus and mucins in saliva, breast milk and in the cervical plug inhibit the human immunodeficiency virus (HIV-1) in an in vitro assay. Crude mucus gels form continuous layers on the epithelial surfaces of the major internal tracts of the body and protect these epithelial surfaces against aggressive luminal factors such as hydrochloric acid and pepsin proteolysis in the stomach lumen, the movement of hard faecal pellets in the colon at high pressure, the effects of shear against the vaginal epithelium during intercourse and the presence of foreign substances in the respiratory airways. Tumour-associated epitopes on mucins make them suitable as immune-targets on malignant epithelial cells, rendering mucins important as diagnostic and prognostic markers for various diseases, even influencing the design of mucin-based vaccines. Sub-Saharan Africa has the highest prevalence of HIV-AIDS in the world. The main points of viral transmission are via the vaginal epithelium during sexual intercourse and mother-to-child transmission during breast-feeding. There have been many studies showing that several body fluids have components that prevent the transmission of HIV-1 from infected to non-infected persons through various forms of contact. Crude saliva and its purified mucins, MUC5B and MUC7, and the purified mucins from breast milk, MUC1 and MUC4 and pregnancy plug cervical mucus (MUC2, MUC5AC, MUC5B and MUC6), inhibit HIV-1 in an in vitro assay. There are conflicting reports of whether crude breast-milk inhibits HIV-1 in an in vitro assay. However studies with a humanised BLT mouse show that breast-milk does inhibit HIV and that breast-feeding is still advisable even amongst HIV-positive women in under-resourced areas, preferably in conjunction with anti-retroviral treatment. CONCLUSION These findings raise questions of how such a naturally occurring biological substance such as mucus, with remarkable protective properties of epithelial surfaces against aggressive luminal factors in delicate locations, could be used as a tool in the fight against HIV-AIDS, which has reached epidemic proportions in sub-Saharan Africa.
Collapse
Affiliation(s)
- Anwar Suleman Mall
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Habtom Habte
- Discipline of Medical Virology, University of Stellenbosch & Tygerberg Hospital, Parow, South Africa
| | - Yolanda Mthembu
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Julia Peacocke
- Division of General Surgery, University of Cape Town and Immune Modulation and Biotherapeutics Discovery, Boehringer- Ingelheim, Danbury, USA
| | - Corena de Beer
- Department of Surgery, Division of General Surgery, University of Cape Town, Observatory Cape, 7925 South Africa
| |
Collapse
|
24
|
Benefits of Lactoferrin, Osteopontin and Milk Fat Globule Membranes for Infants. Nutrients 2017; 9:nu9080817. [PMID: 28788066 PMCID: PMC5579611 DOI: 10.3390/nu9080817] [Citation(s) in RCA: 86] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 07/23/2017] [Accepted: 07/24/2017] [Indexed: 12/11/2022] Open
Abstract
The provision of essential and non-essential amino acids for breast-fed infants is the major function of milk proteins. In addition, breast-fed infants might benefit from bioactivities of milk proteins, which are exhibited in the intestine during the digestive phase and by absorption of intact proteins or derived peptides. For lactoferrin, osteopontin and milk fat globule membrane proteins/lipids, which have not until recently been included in substantial amounts in infant formulas, in vitro experiments and animal models provide a convincing base of evidence for bioactivities, which contribute to the protection of the infant from pathogens, improve nutrient absorption, support the development of the immune system and provide components for optimal neurodevelopment. Technologies have become available to obtain these compounds from cow´s milk and the bovine compounds also exhibit bioactivities in humans. Randomized clinical trials with experimental infant formulas incorporating lactoferrin, osteopontin, or milk fat globule membranes have already provided some evidence for clinical benefits. This review aims to compare findings from laboratory and animal experiments with outcomes of clinical studies. There is good justification from basic science and there are promising results from clinical studies for beneficial effects of lactoferrin, osteopontin and the milk fat globule membrane complex of proteins and lipids. Further studies should ideally be adequately powered to investigate effects on clinically relevant endpoints in healthy term infants.
Collapse
|
25
|
Sims B, Farrow AL, Williams SD, Bansal A, Krendelchtchikov A, Gu L, Matthews QL. Role of TIM-4 in exosome-dependent entry of HIV-1 into human immune cells. Int J Nanomedicine 2017; 12:4823-4833. [PMID: 28740388 PMCID: PMC5505621 DOI: 10.2147/ijn.s132762] [Citation(s) in RCA: 50] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Exosomes, 30–200 nm nanostructures secreted from donor cells and internalized by recipient cells, can play an important role in the cellular entry of some viruses. These microvesicles are actively secreted into various body fluids, including blood, urine, saliva, cerebrospinal fluid, and breast milk. We successfully isolated exosomes from human breast milk and plasma. The size and concentration of purified exosomes were measured by nanoparticle tracking, while Western blotting confirmed the presence of the exosomal-associated proteins CD9 and CD63, clathrin, and T cell immunoglobulin and mucin proteins (TIMs). Through viral infection assays, we determined that HIV-1 utilizes an exosome-dependent mechanism for entry into human immune cells. The virus contains high amounts of phosphatidylserine (PtdSer) and may bind PtdSer receptors, such as TIMs. This mechanism is supported by our findings that exosomes from multiple sources increased HIV-1 entry into T cells and macrophages, and viral entry was potently blocked with anti-TIM-4 antibodies.
Collapse
Affiliation(s)
- Brian Sims
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology.,Center for AIDS Research
| | | | - Sparkle D Williams
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology
| | | | - Alexandre Krendelchtchikov
- Division of Neonatology, Department of Pediatrics.,Department of Cell, Developmental and Integrative Biology.,Division of Infectious Diseases
| | - Linlin Gu
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, University of Alabama at Birmingham, Birmingham
| | - Qiana L Matthews
- Center for AIDS Research.,Division of Infectious Diseases.,Microbiology Program, Department of Biological Sciences, College of Science, Technology, Engineering and Mathematics, Alabama State University, Montgomery, AL, USA
| |
Collapse
|
26
|
He S, Tang H, Yi H, Xu W, Ma Y, Wang R. Properties of emulsions from milk fat globule membrane and its components. INTERNATIONAL JOURNAL OF FOOD PROPERTIES 2017. [DOI: 10.1080/10942912.2017.1343348] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
- Shenghua He
- Department of Food Science and Engineering, School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, P. R. China
| | - Haishan Tang
- Department of Food Science and Engineering, School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, P. R. China
| | - Huaxi Yi
- College of Food Science and Engineering, Ocean University of China, Qingdao, Shandong Province, P. R. China
| | - Weili Xu
- Department of Food Science and Engineering, School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, P. R. China
| | - Ying Ma
- Department of Food Science and Engineering, School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, P. R. China
| | - Rongchun Wang
- Department of Food Science and Engineering, School of Chemical Engineering and Technology, Harbin Institute of Technology, Harbin, P. R. China
| |
Collapse
|
27
|
Brijesha N, Nishimura SI, Aparna HS. Comparative Glycomics of Fat Globule Membrane Glycoconjugates from Buffalo (Bubalus bubalis) Milk and Colostrum. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2017; 65:1496-1506. [PMID: 28145111 DOI: 10.1021/acs.jafc.6b03330] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/06/2023]
Abstract
The health-promoting effects of milk fat globule membrane (MFGM) glycoconjugates has attracted curiosity especially with regard to the challenges encountered to unravel the glycan complexities of MFGM glycoproteins and glycosphingolipids. In this context, we characterized glycans present in buffalo milk and colostrum fat globule membranes by matrix-assisted laser desorption/ionization time-of-flight (MALDI-TOF) MS analysis by adopting chemoselective glycoblotting technique. Unlike human and bovine MFGM glycoproteins, the variations were obvious with respect to their number, size, heterogeneity, and abundance among the samples analyzed. Among N-linked glycans, mono-, di-, and trisialyl glycans were apparent in colostrum, while MFGM predominantly contained mono- and disialyl glycans, in addition to neutral and high-mannose glycoforms. The structural assignments of major glycans were confirmed by TOF/TOF analysis. Core 1 O-glycans were more common in both samples, and the major glycosphingolipids were GM3 and GD3 irrespective of the samples analyzed. The colostrum N-glycans, being effective antibacterials against human pathogens, established the structure-function relationship of oligosaccharides in early milk in providing innate protection to the newborn.
Collapse
Affiliation(s)
- Nagaraju Brijesha
- Department of Biotechnology, University of Mysore , Manasagangotri, Mysore 570 006, Karnataka, India
| | | | | |
Collapse
|
28
|
Yang M, Peng X, Wu J, Wu RN, Liu B, Ye W, Xu X, Yue X. Differential proteomic analysis of milk fat globule membrane proteins in human and bovine colostrum by iTRAQ-coupled LC-MS/MS. Eur Food Res Technol 2016. [DOI: 10.1007/s00217-016-2798-6] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
29
|
|
30
|
Boks MA, Gunput STG, Kosten I, Gibbs S, van Vliet SJ, Ligtenberg AJM, van Kooyk Y. The Human Glycoprotein Salivary Agglutinin Inhibits the Interaction of DC-SIGN and Langerin with Oral Micro-Organisms. J Innate Immun 2016; 8:350-61. [PMID: 27082983 DOI: 10.1159/000443016] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2015] [Accepted: 12/03/2015] [Indexed: 11/19/2022] Open
Abstract
Salivary agglutinin (SAG), also known as gp340 or SALSA, is a glycoprotein encoded by the Deleted in Malignant Brain Tumours 1 gene and is abundantly present in human saliva. SAG aggregates bacteria and viruses, thereby promoting their clearance from the oral cavity. The mucosa lining the oral cavity contains dendritic cells (DC) and Langerhans cells (LC), which express the C-type lectin receptors (CLR) DC-SIGN and Langerin, respectively. Both DC-SIGN and Langerin recognise mannose and fucose carbohydrate structures on pathogens and self-glycoproteins to regulate immunity and homeostasis. The purpose of this study was to investigate whether SAG interacts with these CLR and whether this interferes with the binding to oral pathogens. We show that whole parotid saliva and SAG, when coated to microplates, strongly interact with DC-SIGN and Langerin, probably via mannose and fucose structures. Also, primary human DC and LC bind parotid saliva and SAG via DC-SIGN and Langerin, respectively. Furthermore, SAG binding to DC-SIGN or Langerin prevented binding to the micro-organisms Candida albicans and Escherichia coli which express mannose and fucose-containing glycan structures. Thus, binding of saliva glycoprotein SAG to DC-SIGN and Langerin may inhibit pathogen-DC/LC interactions, and could prove to be a new immunomodulatory mechanism of SAG.
Collapse
Affiliation(s)
- Martine A Boks
- Department of Molecular Cell Biology and Immunology, VU University Medical Center (VUmc), Amsterdam, The Netherlands
| | | | | | | | | | | | | |
Collapse
|
31
|
Exosomes in Human Immunodeficiency Virus Type I Pathogenesis: Threat or Opportunity? Adv Virol 2016; 2016:9852494. [PMID: 26981123 PMCID: PMC4766318 DOI: 10.1155/2016/9852494] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2015] [Revised: 12/07/2015] [Accepted: 12/20/2015] [Indexed: 12/22/2022] Open
Abstract
Nanometre-sized vesicles, also known as exosomes, are derived from endosomes of diverse cell types and present in multiple biological fluids. Depending on their cellular origins, the membrane-bound exosomes packed a variety of functional proteins and RNA species. These microvesicles are secreted into the extracellular space to facilitate intercellular communication. Collective findings demonstrated that exosomes from HIV-infected subjects share many commonalities with Human Immunodeficiency Virus Type I (HIV-1) particles in terms of proteomics and lipid profiles. These observations postulated that HIV-resembled exosomes may contribute to HIV pathogenesis. Interestingly, recent reports illustrated that exosomes from body fluids could inhibit HIV infection, which then bring up a new paradigm for HIV/AIDS therapy. Accumulative findings suggested that the cellular origin of exosomes may define their effects towards HIV-1. This review summarizes the two distinctive roles of exosomes in regulating HIV pathogenesis. We also highlighted several additional factors that govern the exosomal functions. Deeper understanding on how exosomes promote or abate HIV infection can significantly contribute to the development of new and potent antiviral therapeutic strategy and vaccine designs.
Collapse
|
32
|
Shen R, Achenbach J, Shen Y, Palaia J, Rahkola JT, Nick HJ, Smythies LE, McConnell M, Fowler MG, Smith PD, Janoff EN. Mother-to-Child HIV-1 Transmission Events Are Differentially Impacted by Breast Milk and Its Components from HIV-1-Infected Women. PLoS One 2015; 10:e0145150. [PMID: 26680219 PMCID: PMC4683070 DOI: 10.1371/journal.pone.0145150] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2015] [Accepted: 12/01/2015] [Indexed: 11/21/2022] Open
Abstract
Breast milk is a vehicle of infection and source of protection in post-natal mother-to-child HIV-1 transmission (MTCT). Understanding the mechanism by which breast milk limits vertical transmission will provide critical insight into the design of preventive and therapeutic approaches to interrupt HIV-1 mucosal transmission. However, characterization of the inhibitory activity of breast milk in human intestinal mucosa, the portal of entry in postnatal MTCT, has been constrained by the limited availability of primary mucosal target cells and tissues to recapitulate mucosal transmission ex vivo. Here, we characterized the impact of skimmed breast milk, breast milk antibodies (Igs) and non-Ig components from HIV-1-infected Ugandan women on the major events of HIV-1 mucosal transmission using primary human intestinal cells and tissues. HIV-1-specific IgG antibodies and non-Ig components in breast milk inhibited the uptake of Ugandan HIV-1 isolates by primary human intestinal epithelial cells, viral replication in and transport of HIV-1- bearing dendritic cells through the human intestinal mucosa. Breast milk HIV-1-specific IgG and IgA, as well as innate factors, blocked the uptake and transport of HIV-1 through intestinal mucosa. Thus, breast milk components have distinct and complementary effects in reducing HIV-1 uptake, transport through and replication in the intestinal mucosa and, therefore, likely contribute to preventing postnatal HIV-1 transmission. Our data suggests that a successful preventive or therapeutic approach would require multiple immune factors acting at multiple steps in the HIV-1 mucosal transmission process.
Collapse
Affiliation(s)
- Ruizhong Shen
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (ENJ); (RS)
| | - Jenna Achenbach
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
| | - Yue Shen
- Department of Biological Sciences, Auburn University, Auburn, Alabama United States of America
| | - Jana Palaia
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
| | - Jeremy T. Rahkola
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
| | - Heidi J. Nick
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Lesley E. Smythies
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Michelle McConnell
- Centers for Disease Control and Prevention, Atlanta, Georgia, United States of America
| | - Mary G. Fowler
- The Makerere University-Johns Hopkins University Research Collaboration, Kampala, Uganda
| | - Phillip D. Smith
- Department of Medicine (Division of Gastroenterology), University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Veterans Affairs Medical Center, Birmingham, Alabama, United States of America
| | - Edward N. Janoff
- Mucosal and Vaccine Research Program Colorado (MAVRC), University of Colorado Denver, Aurora, Colorado, United States of America
- Denver Veterans Affairs Medical Center, Denver, Colorado, United States of America
- * E-mail: (ENJ); (RS)
| |
Collapse
|
33
|
Wahl A, Baker C, Spagnuolo RA, Stamper LW, Fouda GG, Permar SR, Hinde K, Kuhn L, Bode L, Aldrovandi GM, Garcia JV. Breast Milk of HIV-Positive Mothers Has Potent and Species-Specific In Vivo HIV-Inhibitory Activity. J Virol 2015; 89:10868-78. [PMID: 26292320 PMCID: PMC4621099 DOI: 10.1128/jvi.01702-15] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2015] [Accepted: 08/10/2015] [Indexed: 01/17/2023] Open
Abstract
UNLABELLED Despite the nutritional and health benefits of breast milk, breast milk can serve as a vector for mother-to-child HIV transmission. Most HIV-infected infants acquire HIV through breastfeeding. Paradoxically, most infants breastfed by HIV-positive women do not become infected. This is potentially attributed to anti-HIV factors in breast milk. Breast milk of HIV-negative women can inhibit HIV infection. However, the HIV-inhibitory activity of breast milk from HIV-positive mothers has not been evaluated. In addition, while significant differences in breast milk composition between transmitting and nontransmitting HIV-positive mothers have been correlated with transmission risk, the HIV-inhibitory activity of their breast milk has not been compared. This knowledge may significantly impact the design of prevention approaches in resource-limited settings that do not deny infants of HIV-positive women the health benefits of breast milk. Here, we utilized bone marrow/liver/thymus humanized mice to evaluate the in vivo HIV-inhibitory activity of breast milk obtained from HIV-positive transmitting and nontransmitting mothers. We also assessed the species specificity and biochemical characteristics of milk's in vivo HIV-inhibitory activity and its ability to inhibit other modes of HIV infection. Our results demonstrate that breast milk of HIV-positive mothers has potent HIV-inhibitory activity and indicate that breast milk can prevent multiple routes of infection. Most importantly, this activity is unique to human milk. Our results also suggest multiple factors in breast milk may contribute to its HIV-inhibitory activity. Collectively, our results support current recommendations that HIV-positive mothers in resource-limited settings exclusively breastfeed in combination with antiretroviral therapy. IMPORTANCE Approximately 240,000 children become infected with HIV annually, the majority via breastfeeding. Despite daily exposure to virus in breast milk, most infants breastfed by HIV-positive women do not acquire HIV. The low risk of breastfeeding-associated HIV transmission is likely due to antiviral factors in breast milk. It is well documented that breast milk of HIV-negative women can inhibit HIV infection. Here, we demonstrate, for the first time, that breast milk of HIV-positive mothers (nontransmitters and transmitters) inhibits HIV transmission. We also demonstrate that breast milk can prevent multiple routes of HIV acquisition and that this activity is unique to human milk. Collectively, our results support current guidelines which recommend that HIV-positive women in resource-limited settings exclusively breastfeed in combination with infant or maternal antiretroviral therapy.
Collapse
Affiliation(s)
- Angela Wahl
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Caroline Baker
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Rae Ann Spagnuolo
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| | - Lisa W Stamper
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Genevieve G Fouda
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Sallie R Permar
- Duke Human Vaccine Institute, Duke University Medical Center, Durham, North Carolina, USA
| | - Katie Hinde
- Department of Human Evolutionary Biology, Harvard University, Cambridge, Massachusetts, USA
| | - Louise Kuhn
- Gertrude H. Sergievsky Center and Department of Epidemiology, Mailman School of Public Health, Columbia University, New York, New York, USA
| | - Lars Bode
- Division of Neonatal Medicine and Division of Pediatric Gastroenterology and Nutrition, Department of Pediatrics, University of California, San Diego, San Diego, California, USA
| | - Grace M Aldrovandi
- Department of Pediatrics, Children's Hospital Los Angeles, University of Southern California, Los Angeles, California, USA
| | - J Victor Garcia
- Division of Infectious Diseases, Center for AIDS Research, University of North Carolina at Chapel Hill School of Medicine, Chapel Hill, North Carolina, USA
| |
Collapse
|
34
|
Lis-Kuberka J, Kątnik-Prastowska I, Berghausen-Mazur M, Orczyk-Pawiłowicz M. Lectin-based analysis of fucosylated glycoproteins of human skim milk during 47 days of lactation. Glycoconj J 2015; 32:665-74. [PMID: 26318738 PMCID: PMC4651984 DOI: 10.1007/s10719-015-9615-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2015] [Revised: 08/06/2015] [Accepted: 08/10/2015] [Indexed: 01/01/2023]
Abstract
Glycoproteins of human milk are multifunctional molecules, and their fucosylated variants are potentially active molecules in immunological events ensuring breastfed infants optimal development and protection against infection diseases. The expression of fucosylated glycotopes may correspond to milk maturation stages. The relative amounts of fucosylated glycotopes of human skim milk glycoproteins over the course of lactation from the 2nd day to the 47th day were analyzed in colostrums, transitional and mature milk samples of 43 healthy mothers by lectin-blotting using α1-2-, α1-6-, and α1-3-fucose specific biotinylated Ulex europaeus (UEA), Lens culinaris (LCA), and Lotus tetragonolobus (LTA) lectins, respectively. The reactivities of UEA and LCA with the milk glycoproteins showed the highest expression of α1-2- and α1-6-fucosylated glycotopes on colostrum glycoproteins. The level of UEA-reactive glycoproteins from the beginning of lactation to the 14th day was high and relatively stable in contrast to LCA-reactive glycoproteins, the level of which significantly decreased from 2–3 to 7–8 days then remained almost unchanged until the 12th–14th days. Next, during the progression of lactation the reactivities with both lectins declined significantly. Eighty percent of α1-2- and/or α1-6-fucosylated glycoproteins showed a high negative correlation with milk maturation. In contrast, most of the analyzed milk glycoproteins were not recognized or weakly recognized by LTA and remained at a low unchanged level over lactation. Only a 30-kDa milk glycoprotein was evidently LTA-reactive, showing a negative correlation with milk maturation. The gradual decline of high expression of α1-2- and α1-6-, but not α1-3-, fucoses on human milk glycoproteins of healthy mothers over lactation was associated with milk maturation.
Collapse
Affiliation(s)
- Jolanta Lis-Kuberka
- Department of Chemistry and Immunochemistry, Wrocław Medical University, Bujwida 44a, 50-345, Wrocław, Poland
| | - Iwona Kątnik-Prastowska
- Department of Chemistry and Immunochemistry, Wrocław Medical University, Bujwida 44a, 50-345, Wrocław, Poland
| | - Marta Berghausen-Mazur
- 1st Department and Clinic of Gynaecology and Obstetrics, Wrocław Medical University, T. Chałubińskiego 3, 50-368, Wrocław, Poland
| | - Magdalena Orczyk-Pawiłowicz
- Department of Chemistry and Immunochemistry, Wrocław Medical University, Bujwida 44a, 50-345, Wrocław, Poland.
| |
Collapse
|
35
|
Colorectal mucus binds DC-SIGN and inhibits HIV-1 trans-infection of CD4+ T-lymphocytes. PLoS One 2015; 10:e0122020. [PMID: 25793526 PMCID: PMC4368515 DOI: 10.1371/journal.pone.0122020] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2014] [Accepted: 02/09/2015] [Indexed: 12/20/2022] Open
Abstract
Bodily secretions, including breast milk and semen, contain factors that modulate HIV-1 infection. Since anal intercourse caries one of the highest risks for HIV-1 transmission, our aim was to determine whether colorectal mucus (CM) also contains factors interfering with HIV-1 infection and replication. CM from a number of individuals was collected and tested for the capacity to bind DC-SIGN and inhibit HIV-1 cis- or trans-infection of CD4+ T-lymphocytes. To this end, a DC-SIGN binding ELISA, a gp140 trimer competition ELISA and HIV-1 capture/ transfer assays were utilized. Subsequently we aimed to identify the DC-SIGN binding component through biochemical characterization and mass spectrometry analysis. CM was shown to bind DC-SIGN and competes with HIV-1 gp140 trimer for binding. Pre-incubation of Raji-DC-SIGN cells or immature dendritic cells (iDCs) with CM potently inhibits DC-SIGN mediated trans-infection of CD4+ T-lymphocytes with CCR5 and CXCR4 using HIV-1 strains, while no effect on direct infection is observed. Preliminary biochemical characterization demonstrates that the component seems to be large (>100kDa), heat and proteinase K resistant, binds in a α1–3 mannose independent manner and is highly variant between individuals. Immunoprecipitation using DC-SIGN-Fc coated agarose beads followed by mass spectrometry indicated lactoferrin (fragments) and its receptor (intelectin-1) as candidates. Using ELISA we showed that lactoferrin levels within CM correlate with DC-SIGN binding capacity. In conclusion, CM can bind the C-type lectin DC-SIGN and block HIV-1 trans-infection of both CCR5 and CXCR4 using HIV-1 strains. Furthermore, our data indicate that lactoferrin is a DC-SIGN binding component of CM. These results indicate that CM has the potential to interfere with pathogen transmission and modulate immune responses at the colorectal mucosa.
Collapse
|
36
|
Koning N, Kessen SFM, Van Der Voorn JP, Appelmelk BJ, Jeurink PV, Knippels LMJ, Garssen J, Van Kooyk Y. Human Milk Blocks DC-SIGN-Pathogen Interaction via MUC1. Front Immunol 2015; 6:112. [PMID: 25821450 PMCID: PMC4358221 DOI: 10.3389/fimmu.2015.00112] [Citation(s) in RCA: 36] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2014] [Accepted: 02/26/2015] [Indexed: 12/23/2022] Open
Abstract
Beneficial effects of breastfeeding are well-recognized and include both immediate neonatal protection against pathogens and long-term protection against allergies and autoimmune diseases. Although several proteins have been identified to have anti-viral or anti-bacterial effects like secretory IgA or lactoferrin, the mechanisms of immune modulation are not fully understood. Recent studies identified important beneficial effects of glycans in human milk, such as those expressed in oligosaccharides or on glycoproteins. Glycans are recognized by the carbohydrate receptors C-type lectins on dendritic cell (DC) and specific tissue macrophages, which exert important functions in immune modulation and immune homeostasis. A well-characterized C-type lectin is dendritic cell-specific intercellular adhesion molecule-3-grabbing non-integrin (DC-SIGN), which binds terminal fucose. The present study shows that in human milk, MUC1 is the major milk glycoprotein that binds to the lectin domain of DC-SIGN and prevents pathogen interaction through the presence of Lewis x-type oligosaccharides. Surprisingly, this was specific for human milk, as formula, bovine or camel milk did not show any presence of proteins that interacted with DC-SIGN. The expression of DC-SIGN is found in young infants along the entire gastrointestinal tract. Our data thus suggest the importance of human milk glycoproteins for blocking pathogen interaction to DC in young children. Moreover, a potential benefit of human milk later in life in shaping the infants immune system through DC-SIGN cannot be ruled out.
Collapse
Affiliation(s)
- Nathalie Koning
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | - Sabine F M Kessen
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| | | | - Ben J Appelmelk
- Department of Medical Microbiology and Infection Control, VU University Medical Center , Amsterdam , Netherlands
| | - Prescilla V Jeurink
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Leon M J Knippels
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Johan Garssen
- Immunology, Danone Research - Centre for Specialised Nutrition , Wageningen , Netherlands ; Faculty of Science, Utrecht Institute for Pharmaceutical Sciences, University Utrecht , Utrecht , Netherlands
| | - Yvette Van Kooyk
- Department of Molecular Cell Biology and Immunology, VU University Medical Center , Amsterdam , Netherlands
| |
Collapse
|
37
|
The evolution of HIV-1 interactions with coreceptors and mannose C-type lectin receptors. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 129:109-40. [PMID: 25595802 DOI: 10.1016/bs.pmbts.2014.10.004] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
The phenotype of human immunodeficiency virus type 1 (HIV-1) commonly evolves between and within infected individuals, at virus transmission, and during disease progression. This evolution includes altered interactions between the virus and its coreceptors, i.e., chemokine receptors, as well as mannose C-type lectin receptors (CLRs). Transmitted/founder viruses are predominantly restricted to CCR5, whereas the subsequent intrapatient evolution of HIV-1 coreceptor use during progressive disease can be subdivided into two distinct pathways. Accordingly, the CCR5-restricted virus population is either gradually replaced by virus variants able to use CXCR4 or evolves toward an altered, more flexible use of CCR5. Despite a strong dependency on these coreceptors for host cell entry, HIV-1 also interacts with other cell surface molecules during target cell attachment, including the CLRs. The virus interaction with the CLRs may result either in the efficient transfer of virus to CD4(+) T cells or in the degradation of the virus in endosomal compartments. The determinants of the diverse outcomes depend on which CLR is engaged and also on the glycan makeup of the envelope glycoproteins, which may evolve with the strength of the immune pressure during the disease course. With the current clinical introduction of CCR5 antagonists and the development of additional entry inhibitors, knowledge on the evolution and baseline characteristics of HIV-1 interactions with coreceptor and CLR interactions may play important roles for individualized and optimized treatment strategies. This review summarizes our current understanding of the evolution of HIV-1 interactions with these receptors.
Collapse
|
38
|
Abstract
Human milk is a complete source of nourishment for the infant. Exclusive breastfeeding not only sustains the infant's development but also guides the proliferation of a protective intestinal microbiota. Among the many components of milk that modulate the infant gut microbiota, the milk glycans, which comprise free oligosaccharides, glycoproteins, and glycolipids, are increasingly recognized as drivers of microbiota development and overall gut health. These glycans may display pleiotropic functions, conferring protection against infectious diseases and also acting as prebiotics, selecting for the growth of beneficial intestinal bacteria. The prebiotic effect of milk glycans has direct application to prevention of diseases such as necrotizing enterocolitis, a common and devastating disease of preterm infants. In this article, we review the impact of the human (and bovine) milk glycome on gut health through establishment of a milk-oriented microbiota in the neonate.
Collapse
Affiliation(s)
- Alline R. Pacheco
- Department of Viticulture and Enology, University of California, Davis, California 95616
- Foods for Health Institute, University of California, Davis, California 95616
| | - Daniela Barile
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Food Science and Technology, University of California, Davis, California 95616
| | - Mark A. Underwood
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Pediatrics, University of California, Davis, California 95616
| | - David A. Mills
- Department of Viticulture and Enology, University of California, Davis, California 95616
- Foods for Health Institute, University of California, Davis, California 95616
- Department of Food Science and Technology, University of California, Davis, California 95616
| |
Collapse
|
39
|
Soluble toll-like receptor 2 is significantly elevated in HIV-1 infected breast milk and inhibits HIV-1 induced cellular activation, inflammation and infection. AIDS 2014; 28:2023-32. [PMID: 25265071 DOI: 10.1097/qad.0000000000000381] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES We previously demonstrated that immunodepletion of soluble Toll-like receptor 2 (sTLR2) from human breast milk significantly increased HIV infection in vitro. The aims of this study were to characterize sTLR2 levels in breast milk from HIV-infected and uninfected women, and identify a mechanism by which sTLR2 inhibits HIV-induced cellular activation and infection. DESIGN Blinded studies of breast milk from HIV-infected and uninfected Nigerian and Canadian women were evaluated for levels of sTLR2, proinflammatory cytokines and viral antigenemia. In-vitro experiments were conducted using cell lines to assess sTLR2 function in innate responses and effect on HIV infection. RESULTS Breast milk from HIV-infected women showed significantly higher levels of sTLR2 than uninfected breast milk. Further, sTLR2 levels correlated with HIV-1 p24 and interleukin (IL)-15, thus suggesting a local innate compensatory response in the HIV-infected breast. Given the significantly higher levels of sTLR2 in breast milk from HIV-infected women, we next demonstrated that mammary epithelial cells and macrophages, which are prevalent in milk, produced significantly increased levels of sTLR2 following exposure to HIV-1 proteins p17, p24 and gp41 or the TLR2 ligand, Pam3CSK4. Our results also demonstrated that sTLR2 physically interacts with p17, p24 and gp41 and inhibits HIV-induced nuclear factor kappa-light-chain-enhancer of activated B cells activation, and inflammation. Importantly, binding of sTLR2 to HIV-1 proteins inhibited a TLR2-dependent increase in chemokine receptor 5 expression, thus resulting in significantly reduced HIV-1 infection. CONCLUSION These results indicate novel mechanisms by which sTLR2 plays a critical role in inhibiting mother-to-child HIV transmission.
Collapse
|
40
|
Wood LF, Chahroudi A, Chen HL, Jaspan HB, Sodora DL. The oral mucosa immune environment and oral transmission of HIV/SIV. Immunol Rev 2014; 254:34-53. [PMID: 23772613 DOI: 10.1111/imr.12078] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
The global spread of human immunodeficiency virus (HIV) is dependent on the ability of this virus to efficiently cross from one host to the next by traversing a mucosal membrane. Unraveling how mucosal exposure of HIV results in systemic infection is critical for the development of effective therapeutic strategies. This review focuses on understanding the immune events associated with the oral route of transmission (via breastfeeding or sexual oral intercourse), which occurs across the oral and/or gastrointestinal mucosa. Studies in both humans and simian immunodeficiency virus (SIV) monkey models have identified viral changes and immune events associated with oral HIV/SIV exposure. This review covers our current knowledge of HIV oral transmission in both infants and adults, the use of SIV models in understanding early immune events, oral immune factors that modulate HIV/SIV susceptibility (including mucosal inflammation), and interventions that may impact oral HIV transmission rates. Understanding the factors that influence oral HIV transmission will provide the foundation for developing immune therapeutic and vaccine strategies that can protect both infants and adults from oral HIV transmission.
Collapse
Affiliation(s)
- Lianna F Wood
- Seattle Biomedical Research Institute, Seattle, WA, USA
| | | | | | | | | |
Collapse
|
41
|
Abstract
Most infants born to human immunodeficiency virus (HIV)-infected women escape HIV infection. Infants evade infection despite an immature immune system and, in the case of breastfeeding, prolonged repetitive exposure. If infants become infected, the course of their infection and response to treatment differs dramatically depending upon the timing (in utero, intrapartum, or during breastfeeding) and potentially the route of their infection. Perinatally acquired HIV infection occurs during a critical window of immune development. HIV's perturbation of this dynamic process may account for the striking age-dependent differences in HIV disease progression. HIV infection also profoundly disrupts the maternal immune system upon which infants rely for protection and immune instruction. Therefore, it is not surprising that infants who escape HIV infection still suffer adverse effects. In this review, we highlight the unique aspects of pediatric HIV transmission and pathogenesis with a focus on mechanisms by which HIV infection during immune ontogeny may allow discovery of key elements for protection and control from HIV.
Collapse
|
42
|
Exosomes from breast milk inhibit HIV-1 infection of dendritic cells and subsequent viral transfer to CD4+ T cells. AIDS 2014; 28:171-80. [PMID: 24413309 DOI: 10.1097/qad.0000000000000159] [Citation(s) in RCA: 125] [Impact Index Per Article: 12.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
OBJECTIVE To investigate whether exosomes derived from human breast milk or plasma confer protection against HIV-1 infection of monocyte-derived dendritic cells (MDDCs) and subsequent viral transfer to CD4 T cells. DESIGN MDDCs were generated and milk and plasma-derived exosomes were isolated from healthy donors. To determine the capacity of exosomes to inhibit HIV-1 infection, MDDCs were preincubated with exosomes before exposure to HIV-1BaL. To investigate transfer of HIV-1 from MDDCs to CD4 T cells, MDDCs preincubated with exosomes and HIV-1BaL were cocultured with allogeneic CD4 T cells. To explore receptors used by MDDCs for binding of exosomes, blocking experiments were performed. METHODS Productive HIV-1 infection was analysed in MDDCs and CD4 T cells by determining p24 expression by flow cytometry. Confocal microscopy and flow cytometry was used to investigate uptake of fluorescently labelled exosomes by MDDCs. RESULTS Milk exosomes, but not plasma exosomes, bind MDDCs via DC-SIGN inhibiting HIV-1 infection of MDDCs and subsequent viral transfer to CD4 T cells. CONCLUSION We propose that milk exosomes act as a novel protective factor against vertical transmission of HIV-1 by competing with HIV-1 for binding to DC-SIGN on MDDCs.
Collapse
|
43
|
O'Riordan N, Kane M, Joshi L, Hickey RM. Structural and functional characteristics of bovine milk protein glycosylation. Glycobiology 2014; 24:220-36. [PMID: 24398766 DOI: 10.1093/glycob/cwt162] [Citation(s) in RCA: 116] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Most secreted and cell membrane proteins in mammals are glycosylated. Many of these glycoproteins are also prevalent in milk and play key roles in the biomodulatory properties of milk and ultimately in determining milk's nutritional quality. Although a significant amount of information exists on the types and roles of free oligosaccharides in milk, very little is known about the glycans associated with milk glycoproteins, in particular, the biological properties that are linked to their presence. The main glycoproteins found in bovine milk are lactoferrin, the immunoglobulins, glycomacropeptide, a glycopeptide derived from κ-casein, and the glycoproteins of the milk fat globule membrane. Here, we review the glycoproteins present in bovine milk, the information currently available on their glycosylation and the biological significance of their oligosaccharide chains.
Collapse
Affiliation(s)
- Noelle O'Riordan
- Teagasc Food Research Centre, Moorepark, Fermoy, County Cork, Ireland
| | | | | | | |
Collapse
|
44
|
Le TT, Van Camp J, Dewettinck K. Milk Fat Globule Membrane Material. STUDIES IN NATURAL PRODUCTS CHEMISTRY 2014. [DOI: 10.1016/b978-0-444-63294-4.00012-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/10/2023]
|
45
|
Le TT, Debyser G, Gilbert W, Struijs K, Van Camp J, Van de Wiele T, Devreese B, Dewettinck K. Distribution and isolation of milk fat globule membrane proteins during dairy processing as revealed by proteomic analysis. Int Dairy J 2013. [DOI: 10.1016/j.idairyj.2013.05.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
46
|
Struijs K, Van de Wiele T, Le TT, Debyser G, Dewettinck K, Devreese B, Van Camp J. Milk fat globule membrane glycoproteins prevent adhesion of the colonic microbiota and result in increased bacterial butyrate production. Int Dairy J 2013. [DOI: 10.1016/j.idairyj.2013.05.001] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
47
|
Peterson R, Cheah WY, Grinyer J, Packer N. Glycoconjugates in human milk: Protecting infants from disease. Glycobiology 2013; 23:1425-38. [DOI: 10.1093/glycob/cwt072] [Citation(s) in RCA: 77] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
|
48
|
Abstract
Breastfeeding protects the neonate against pathogen infection. Major mechanisms of protection include human milk glycoconjugates functioning as soluble receptor mimetics that inhibit pathogen binding to the mucosal cell surface, prebiotic stimulation of gut colonization by favorable microbiota, immunomodulation, and as a substrate for bacterial fermentation products in the gut. Human milk proteins are predominantly glycosylated, and some biological functions of these human milk glycoproteins (HMGPs) have been reported. HMGPs range in size from 14 kDa to 2,000 kDa and include mucins, secretory immunoglobulin A, bile salt-stimulated lipase, lactoferrin, butyrophilin, lactadherin, leptin, and adiponectin. This review summarizes known biological roles of HMGPs that may contribute to the ability of human milk to protect neonates from disease.
Collapse
Affiliation(s)
- Bo Liu
- Department of Biology, Boston College, Chestnut Hill, MA 02467, USA
| | | |
Collapse
|
49
|
Chatterton DE, Nguyen DN, Bering SB, Sangild PT. Anti-inflammatory mechanisms of bioactive milk proteins in the intestine of newborns. Int J Biochem Cell Biol 2013; 45:1730-47. [DOI: 10.1016/j.biocel.2013.04.028] [Citation(s) in RCA: 251] [Impact Index Per Article: 22.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 04/25/2013] [Accepted: 04/28/2013] [Indexed: 01/04/2023]
|
50
|
Oncolytic vesicular stomatitis virus in an immunocompetent model of MUC1-positive or MUC1-null pancreatic ductal adenocarcinoma. J Virol 2013; 87:10283-94. [PMID: 23864625 DOI: 10.1128/jvi.01412-13] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Vesicular stomatitis virus (VSV) is a promising oncolytic agent against various malignancies. Here, for the first time, we tested VSV in vitro and in vivo in a clinically relevant, immunocompetent mouse model of pancreatic ductal adenocarcinoma (PDA). Our system allows the study of virotherapy against PDA in the context of overexpression (80% of PDA patients) or no expression of human mucin 1 (MUC1), a major marker for poor prognosis in patients. In vitro, we tested three VSV recombinants, wild-type VSV, VSV-green fluorescent protein (VSV-GFP), and a safe oncolytic VSV-ΔM51-GFP, against five mouse PDA cell lines that either expressed human MUC1 or were MUC1 null. All viruses demonstrated significant oncolytic abilities independent of MUC1 expression, although VSV-ΔM51-GFP was somewhat less effective in two PDA cell lines. In vivo administration of VSV-ΔM51-GFP resulted in significant reduction of tumor growth for tested mouse PDA xenografts (+MUC1 or MUC1 null), and antitumor efficacy was further improved when the virus was combined with the chemotherapeutic drug gemcitabine. The antitumor effect was transient in all tested groups. The developed system can be used to study therapies involving various oncolytic viruses and chemotherapeutics, with the goal of inducing tumor-specific immunity while preventing premature virus clearance.
Collapse
|