Hostname: page-component-848d4c4894-m9kch Total loading time: 0 Render date: 2024-06-06T22:17:12.134Z Has data issue: false hasContentIssue false

Milk fat globule membrane in infant nutrition: a dairy industry perspective

Published online by Cambridge University Press:  16 March 2021

Roberta Claro da Silva*
Affiliation:
Family and Consumer Sciences Department, College of Agriculture and Environmental Sciences (CAES), North Carolina A&T State University, Greensboro, NC27411, USA
Heather L. Colleran
Affiliation:
Family and Consumer Sciences Department, College of Agriculture and Environmental Sciences (CAES), North Carolina A&T State University, Greensboro, NC27411, USA
Salam A. Ibrahim
Affiliation:
Family and Consumer Sciences Department, College of Agriculture and Environmental Sciences (CAES), North Carolina A&T State University, Greensboro, NC27411, USA
*
Author for correspondence: Roberta Claro da Silva, Email: rcsilva@ncat.edu
Rights & Permissions [Opens in a new window]

Abstract

This review provides an overview of the composition, structure, and biological activities of milk fat globule membrane (MFGM) compounds with focus on the future application of this compound as a food ingredient. MFGM is a particular component of mammalian milks and is comprised of a tri-layer of polar lipids, glycolipids and proteins. In recent years, MFGM has been extensively studied for the purpose of enhancing the efficacy of infant nutrition formula. For example, infant formulas supplemented with bovine MFGM have shown promising results with regard to neurodevelopment and defense against infections. Components of MFGM have been shown to present several health benefits as the proteins of the membrane have shown antiviral activity and a reduction in the incidence of diarrhea. Moreover, the presence of sphingomyelin, a phospholipid, implies beneficial effects on human health such as enhanced neuronal development in infants and the protection of neonates from bacterial infections. The development of a lipid that is similar to human milk fat would represent a significant advance for the infant formula industry and would offer high technology formulas for those infants that depend on infant formula. The complexity of the structure of MFGM and its nutritional and technological properties is critically examined in this review with a focus on issues relevant to the dairy industry.

Type
Review Article
Copyright
Copyright © The Author(s), 2021. Published by Cambridge University Press on behalf of Hannah Dairy Research Foundation

Human milk is the primary source of nutrients for newborns, term, and preterm infants. From nutritional, immunological and food safety points of view, human milk is the gold standard as food for children in the first phase of their lives (Jensen, Reference Jensen1999, Reference Jensen2018). Human milk maintains healthy growth and development, improves the gastrointestinal process, favors the mother−child bond (Silva et al., Reference Silva, Cotting, Poltronieri, Balcão, Almeida, Goncalves, Grimaldi and Gioielli2009), and further facilitates the emotional, cognitive and nervous system development of the infant (Calhoun et al., Reference Calhoun, Lunoe, Du and Christensen2000; Bezkorovainy, Reference Bezkorovainy2001). Human milk is rich in biologically active components, such as oligosaccharides, hormones and growth factors that provide the short- and long-term benefits of breastfeeding (Lönnerdal, Reference Lönnerdal2014). These bioactive molecules can affect the microbiota composition, modulate gastrointestinal physiology, enhance intestinal barrier function and stimulate proper development of the immune system (Gila-Diaz et al., Reference Gila-Diaz, Arribas, Algara, Martín-Cabrejas, López de Pablo, Sáenz de Pipaón and Ramiro-Cortijo2019). Moreover, the composition and chemical structure of lipids play a vital role in the superior performance of human milk compared to infant formula.

Despite significant efforts to promote breastfeeding for infants, urbanization and the overall lack of time have reduced the breastfeeding period. Worldwide, only 35% of infants in the newborn to 6-month-old age group are breastfed (Zhao et al., Reference Zhao, Zhao, Du, Binns and Lee2017). The rates of breastfeeding in the US diverge widely along the lines of race, socioeconomic status and ethnicity. According to the National Immunization Survey from the CDC (2017), among children born in 2014, breastfeeding at 6 months after birth, as endorsed by the American Academy of Pediatrics, differed markedly between ethnic groups (Hardison-Moody et al., Reference Hardison-Moody, MacNell, Sa and Bowen2018 and Fig. 1).

Fig. 1. The percentage of breast feed infants in USA (CDC, 2017).

Notwithstanding the strategies to increase breastfeeding, there will always be a demand for the development of formulas for infants who cannot be breastfed due to medical, social or other reasons. Thus, researchers in this field have attempted to produce an infant formula that can mimic the natural beneficial characteristics of human milk as closely as possible. Whilst all of the nutritional components in human milk have value, the particular importance of the lipid fraction in child development has been highlighted in the literature. The development of a new lipid system with properties analogous to the human milk fat globule is an important step toward reducing the gap between formula-fed and breast-fed babies with regard to neurodevelopment, infectious diseases and cholesterol metabolism. At this time, there is still no large-scale manufacturing process that has been adopted by the dairy industry to process MFGM. Thus, it is important to discuss the particularities of each component of the milk fat globule in order to understand its physical, chemical and nutritional characteristics.

Lipids in infant nutrition

Human milk provides more energy in the form of lactose than does milk of dairy species, nevertheless, milk fat remains the major energy source (40–60%) for breastfed infants in the first stage of life (Koletzko et al., Reference Koletzko, Agostoni, Bergmann, Ritzenthaler and Shamir2011; Demmelmair and Koletzko, Reference Demmelmair and Koletzko2018). Human milk contains 3−5% lipids, of which 98% are triacylglycerol with 88% of fatty acids (Jensen, Reference Jensen1999). Human milk fat is a vital source of essential nutrients for infants, such as the polyunsaturated fatty acids (PUFAs) and long-chain polyunsaturated fatty acids (LC-PUFA) of the n-6 and n-3 series (Delplanque et al., Reference Delplanque, Gibson, Koletzko, Lapillonne and Strandvik2015). It is also involved in the transport of vitamins (A, D, E, and K) and liposoluble hormones (Lönnerdal, Reference Lönnerdal1996). The long-chain polyunsaturated fatty acids (LC-PUFA) such as docosahexaenoic (DHA) and arachidonic acids (ARA) shown in Fig. 2 are fundamental to developing visual acuity in infants and help to improve cognitive function (Jensen, Reference Jensen1999). Although human milk contains at least 170 different triacylglycerol structures, only 30 of them represent 70% of the total human milk fat. Importantly, the lipids are present as fat globules mainly constituted of triacylglycerols surrounded by a structural membrane composed of phospholipids, cholesterol, proteins, and glycoproteins termed as milk fat globule membrane.

Fig. 2. Structural formulas of Docosahexaenoic acid (DHA) and Arachidonic acid (ARA) fatty acids.

Fatty acid composition

The fatty acid composition of human milk is variable and is influenced by a variety of factors, such as diet, duration of pregnancy, maternal parity, and stage of lactation. The most important variable to determine fatty acid composition is the maternal diet. Table 1 presents the fatty acid composition of human milk from American mothers; these results were obtained from Yuhas et al. (Reference Yuhas, Pramuk and Lien2006). The main fatty acids are oleic acid (32.8%), palmitic acid (19.3%), linoleic acid (14.8%), myristic acid (4.9%), and stearic acid (6.2%).

Table 1. Fatty acid composition of human milk in the United States

SFA, saturated fatty acid; MUFA, monounsaturated fatty acids; PUFA, polyunsaturated fatty acids.

Adapted from Yuhas et al. (Reference Yuhas, Pramuk and Lien2006).

The saturated fatty acids present in infant diets mostly have 12, 14, 16, and 18 carbon chain lengths (Delplanque et al., Reference Delplanque, Gibson, Koletzko, Lapillonne and Strandvik2015). Besides the nutritional role, the saturated fatty acids may have physiological relevance, such as the occurrence of palmitic acid in pulmonary surfactant (schmidt et al., 2002). In studies in vitro or with animals, capric acid often exhibits bactericidal effects (Bergsson et al., Reference Bergsson, Arnfinnsson, Steingrímsson and Thormar2001) whilst arachidic and behenic acids have immunomodulatory effects (Tollin et al., Reference Tollin, Bergsson, Kai-Larsen, Lengqvist, Sjövall, Griffiths, Skúladóttir, Haraldsson, Jörnvall, Gudmundsson and Agerberth2005), and protein acylation occurs for myristic or palmitic acids (Rioux et al., Reference Rioux, Pédrono and Legrand2011). These saturated fatty acid properties should be worthy of further exploration in humans and infants.

The medium-chain fatty acids (8–10 carbons) also have importance for infant health. Caprylic and capric acids (C8 and C10) are used to facilitate fat absorption in preterm babies through the intestine due to intestinal immaturity; however, there is no evidence demonstrating benefit for energy balance or growth (Lapillonne et al., Reference Lapillonne, Groh-Wargo, Gonzalez and Uauy2013).

Monounsaturated fatty acids (MUFA) are the prevalent group of fatty acids in breast milk and infant formula. The primary monounsaturated fatty acids are oleic acid (C18 : 1n-9) and palmitoleic acid (C16 : 1n-7) (Table 1). Besides the usual functions of fatty acids (source of energy and structural components), oleic acid performs a vital role in the reduction of the melting point of triacylglycerides, thus providing the liquidity required for the formation, transport, and metabolism of milk fat globules (Jensen, Reference Jensen1999).

Polyunsaturated fatty acids (PUFA) such as linoleic acid (LA) (18 : 2n-6) and α-linolenic acid (ALA) (18 : 3n-3) are precursors of long-chain polyunsaturated fatty acids (LCPUFA) such as arachidonic acid (AA; 20 : 4n- 6) and docosahexaenoic acid (DHA; 22 : 6n-3). These fatty acids are necessary for the growth of the brain and the retina during perinatal development and influence metabolic processes, such as decreasing plasma cholesterol (Jensen, Reference Jensen1999). Human milk contents of LA and ALA vary according to the maternal intakes of these fatty acids. The European Food Safety Authority (EFSA) recommends amounts of LA and ALA for infant formula are, respectively, 4.5 and 0.5% of energy content, with high levels set at respectively 10.8 and 0.9% of energy (EFSA, 2014). Due to the limited capacity of term infants to synthesize LCPUFA, such as AA, DHA, and EPA, they depend on the dietary supply during the first months of life (Sahin-Yesilcubuk and Akoh, Reference Sahin-Yesilcubuk and Akoh2017). Supplementation of infant formulas with long-chain LCPUFAs (AA and DHA) at levels resembling human milk is recommended by the Food and Agriculture Organization/World Health Organization since LCPUFAs provide biochemical and functional benefits to the neonate. DHA and ARA-rich oils from single-cell organisms are also potential LCPUFA sources to supplement infant formula (Alvares and Akoh, Reference Alvares and Akoh2016). Single-cell oils are nutraceutical ingredients of non-animal origin produced by oleaginous microorganisms such as algae, fungi, and bacteria currently used in infant formulas to supplement LCPUFAs (Ratledge, Reference Ratledge2004; Papanikolaou and Aggelis, Reference Papanikolaou and Aggelis2011; Ochsenreither et al., Reference Ochsenreither, Glück, Stressler, Fischer and Syldatk2016).

Triacylglycerol composition

The triacylglycerols (TAG) in human milk fat have a distinct molecular structure (Mazzocchi et al., Reference Mazzocchi, D'Oria, De Cosmi, Bettocchi, Milani, Silano and Agostoni2018). In human milk fat, the sn-2 position of the TAG is predominantly occupied by saturated fatty acids (SFA), while the majority of unsaturated fatty acids (UFA) are distributed at the sn-1,3 positions (Fig. 3). Palmitic acid (16 : 0) is the primary saturated fatty acid (20–25%), and around 60–86% of 16 : 0 is esterified at the sn-2 position of triacylglycerols (β-position) (Innis et al., Reference Innis, Nelson, Lwanga, Rioux and Waslen1996). Several studies over the last three decades have shown that the 16 : 0 distribution at the central position (sn-2) in human milk fat promotes better absorption of lipids as well as calcium in term newborns and preterm infants (Carnielli et al., Reference Carnielli, Luijendijk, van Beek, Boerma, Degenhart and Sauer1995; Delplanque et al., Reference Delplanque, Gibson, Koletzko, Lapillonne and Strandvik2015). During digestion in newborns and infants, the pancreatic and gastric lipases selectively hydrolyze fatty acids at the sn-1,3 positions, producing free fatty acids and a 2-monoacylglycerol. The monoacylglycerol containing palmitic acid (2-palmitoyl glycerol) is more efficiently absorbed than palmitic acid in its free form. This TAG structure is a crucial factor in mineral and fat absorption in the first period of an infant's life and beyond. The palmitic acid in free form has low absorption in the intestinal lumen, whereas short-chain free fatty acids, and 18-carbon free fatty acids (such as oleic and linoleic acid) are well-absorbed (Carnielli et al., Reference Carnielli, Luijendijk, Van Goudoever, Sulkers, Boerlage, Degenhart and Sauer1996; Kennedy et al., Reference Kennedy, Fewtrell, Morley, Abbott, Quinlan, Wells, Bindels and Lucas1999). This is due to the melting point of palmitic acid (61–65°C) being higher than body temperature (Gunstone, Reference Gunstone and Shahidi2009), as well as the strong tendency to form insoluble soaps with divalent cations such as calcium and magnesium at the alkaline pH of the intestine (de Fouw et al., Reference de Fouw, Kivits, Quinlan and van Nielen1994). By contrast, vegetable oils and some mammalian milk fats present SFAs that are primarily located at the sn-1,3 positions of TAG (Mu and Høy, Reference Mu and Høy2004) and unsaturated fatty acids are esterified at sn-2 (Xu, Reference Xu2000) (Fig. 1), which is different from the location in human milk fat, causing a loss of minerals and energy in the infant (Carnielli et al., Reference Carnielli, Luijendijk, Van Goudoever, Sulkers, Boerlage, Degenhart and Sauer1996; Kennedy et al., Reference Kennedy, Fewtrell, Morley, Abbott, Quinlan, Wells, Bindels and Lucas1999; López-López et al., Reference López-López, Castellote-Bargalló, Campoy-Folgoso, Rivero-Urgël, Tormo-Carnicé, Infante-Pina and López-Sabater2001).

Fig. 3. Schematic triacylglycerol structure of human milk fat. (Adapted from Mehrotra et al., Reference Mehrotra, Sehgal and Bangale2018).

Lipid modification technology, particularly interesterification, is used to design lipids with specific TAG structure. Previous research in neonatal nutrition has suggested that developing more biomimetic infant formulas with a TAG structure more similar to that of human milk would lead to positive outcomes in terms of metabolism programming (Oosting et al., Reference Oosting, van Vlies, Kegler, Schipper, Abrahamse-Berkeveld, Ringler, Verkade and van der Beek2014). Many researchers have shown that the lipid structure of human milk can be artificially produced by enzymatic interesterification using as the catalyst the specific lipase sn-1,3 (Quinlan and Chandler, Reference Quinlan and Chandler1992; Mukherjee and Kiewitt, Reference Mukherjee and Kiewitt1998; Yang et al., Reference Yang, Xu, He and Li2003). Several types of oils and fats are used as a substrate for the production of structured lipid substitute for human milk fat, such as tripalmitin, lard fat, bovine milk fat, soybean oil, canola oil, borage oil, sunflower oil, safflower oil. Table 2 presents the main research in lipid modification to produce a structured lipid substitute for human milk fat in the past five years.

Table 2. An updated summary of enzyme-catalyzed synthesis of human milk fat structured lipids

Yaron and collaborators demonstrated an increment in the fecal occurrence of Lactobacillus and Bifidobacterium genera in infants that, for 6 weeks, received a formula supplemented with β-16 : 0 compared to a control group that received formula containing vegetable-sourced 16 : 0 (Yaron et al., Reference Yaron, Shachar, Abramas, Riskin, Bader, Litmanovitz, Bar-Yoseph, Cohen, Levi, Lifshitz, Shamir and Shaoul2013). The mechanisms behind this study need to be better explored but suggest that the position of palmitic acid on the TAG may have an impact on the gut microbiota.

Infant formulas marketed in the US typically include palm, coconut, soybean, and sunflower oils as the primary lipid sources. The development of a structured lipid that is similar to human milk fat using low cost, sustainable oils such as single-cell oils would, therefore, be a significant advance for the infant formula industry and would offer high technology formulas for those infants who do not have access to breastfeeding and must depend on infant formula.

Milk fat globule membrane

The human milk fat globule has aroused the interest of many researchers recently. It is a heterogeneous structure varying in diameter, triglyceride content, membrane, and fatty acid composition. The diameter of these globules generally ranges from 0.2 to 15 μm, with an average diameter between 4 and 5 μm, and milk fat globules in other mammalian species do not show differences in globule diameters ranging between 3 and 5 μm (Lopez and Ménard, Reference Lopez and Ménard2011). The lipid droplets (TAG-rich core) of milk fat globules (including those found in breast milk) are stabilized by a 3-layered membrane called a milk fat globule membrane (MFGM), which maintains triglyceride dispersion in the aqueous phase of the milk (Fig. 4).

Fig. 4. Schematic structure of milk fat globule (Adapted from Koletzko, Reference Koletzko2016).

Preclinical and clinical studies regarding MFGM and its components in relation to resistance to infections, cognitive development, establishment of gut microbiota and infant metabolism was critically examined in a recent review by Brink and Lönnerdal (Reference Brink and Lönnerdal2020). MFGM is highly structured and contains various components, such as specific proteins (mainly glycoproteins), polar lipids (phospholipids and glycosphingolipids), and apolar lipids (cholesterol and cerebrosides) (Dewettinck et al., Reference Dewettinck, Rombaut, Thienpont, Le, Messens and Van Camp2008). This membrane, about 10–20 nmin cross-section, acts as an emulsifier and protects the globules from coalescence and enzymatic degradation.

Although MFGM proteins account for only 1–4% of total milk proteins and 1% of the total globule mass, more than five hundred proteins with diverse functions have been identified by proteomics analysis (Cao et al., Reference Cao, Kang, Yang, Li, Wu, Han, Meng, Wu and Yue2018). Some have been well described, such as mucins, xanthine oxidoreductase, glycosylated butyrophilin, lactadherin, CD proteins, the non-glycosylated adipophilin, and fatty acid binding proteins (Lee et al., Reference Lee, Padhi, Hasegawa, Larke, Parenti, Wang, Hernell, Lönnerdal and Slupsky2018). Several MFGM proteins, such as lactadherin, butyrophilin, and MUC1, have been shown to have antibacterial and antiviral activity in vitro (Lönnerdal, Reference Lönnerdal2016).

Previous studies have supported the concept of the antiviral activity of MFGM proteins. Lactadherin concentration in human milk was associated with activity against rotavirus (Newburg et al., Reference Newburg, Peterson, Ruiz-Palacios, Matson, Morrow, Shults, Guerrero, Chaturvedi, Newburg, Scallan, Taylor, Ceriani and Pickering1998). Kvistgaard et al. (Reference Kvistgaard, Pallesen, Arias, López, Petersen, Heegaard and Rasmussen2004) demonstrated the inhibitory effects of lactadherin and mucin extracted from bovine MFGM in rotavirus infection. MFGM proteins also reduce the incidence of diarrhea in infants that received bovine (MFGM)-enriched protein fraction as a complementary food (Zavaleta et al., Reference Zavaleta, Kvistgaard, Graverholt, Respicio, Guija, Valencia and Lönnerdal2011). Timby et al. (Reference Timby, Domellof, Hernell, Lonnerdal and Domellof2014) added bovine MFGM fraction to infant formula, fed infants with this formula for six months of age and then followed up to 1 year of age. A significant reduction in illness (acute otitis media) and prescriptions of antipyretics in the infants fed the MFGM-supplemented formula was found compared with infants fed regular formula, with no difference compared to breastfed infants. These authors concluded that the addition of MFGM to infant formula had significant health benefits for infants. These observations endorse the role of human milk MFGM proteins in the defense against infection in breastfed infants.

MFGM proteins are influenced by several maternal factors, such as the period of lactation, environmental conditions, and the different species. Not surprisingly, analysis of human colostrum revealed levels of proteins with potential antimicrobial function considerably higher than in mature human milk, which may compensate for immature neonatal immunity (Cao et al., Reference Cao, Kang, Yang, Li, Wu, Han, Meng, Wu and Yue2018).

The polar lipids of the MFGM include phospho- and sphingolipids, that are amphiphilic molecules with a hydrophobic tail and a hydrophilic head group. Phospholipids are complex mixtures of more than 30 molecular species of phosphatidylcholine, phosphatidylethanolamine, phosphatidylserine, phosphatidylinositol, and sphingophospholipids. Phospholipid molecules consist of a glycerol backbone on which two fatty acids are esterified, and a phosphate residue with different organic groups (choline, serine, ethanolamine, among others) may be linked on the third hydroxyl group. Thus, phosphatidylethanolamine, phosphatidylcholine, and sphingomyelin are the major milk phospholipids, representing about 90% of all MFGM polar lipids.

Phosphatidylcholine represents up to 35% of the total of phospholipids, and its primary function is to maintain the permeability of the membrane (Dewettinck et al., Reference Dewettinck, Rombaut, Thienpont, Le, Messens and Van Camp2008; Jiménez-Flores et al., Reference Jiménez-Flores, Higuera-Ciapara and Pouliot2009) due to its flexible morphology. Phosphatidylethanolamine constitutes up to 30% of the total phospholipids in the MFGM, and its mostly bound to unsaturated fatty acids (68%) with high levels of oleic acid (C18 : 1) and linoleic acid (C18 : 2) (Sánchez-Juanes et al., Reference Sánchez-Juanes, Alonso, Zancada and Hueso2009).

Sphingomyelin is an important constituent of the milk phospholipid fraction, due to its high concentration in the MFGM (up to 35% of phospholipids) and its implication in many beneficial effects on human health, such as neuronal development in infants and protection of neonates from bacterial infections (Bourlieu et al., Reference Bourlieu, Bouzerzour, Ferret-Bernard, Bourgot, Chever, Ménard, Deglaire, Cuinet, Ruyet, Bonhomme, Dupont and Huërou-Luron2015). Interestingly, about 97% of all MFGM sphingomyelins are composed of medium-chain or long-chain saturated fatty acids (C16 : 0, C22 : 0, C23 : 0, and C24 : 0) with high melting temperatures. More rarely, 17% or more of milk sphingomyelins contain C23 : 0 fatty acids (Dewettinck et al., Reference Dewettinck, Rombaut, Thienpont, Le, Messens and Van Camp2008). This unique characteristic of milk sphingomyelins suggests a specific, probably biological, function well beyond its simple structural role in the MFGM. Feeding rats pups with bovine milk sphingomyelins has been shown to improve markers of gut maturation (Motouri et al., Reference Motouri, Matsuyama, Yamamura, Tanaka, Aoe, Iwanaga and Kawakami2003), providing evidence of the importance of dietary sphingomyelin for newborn gut development. Studies have shown a heterogeneous distribution of polar lipids in the outer bilayer of the human MFGM, with the lateral segregation of sphingomyelin in domains that could have functional roles (Lopez and Ménard, Reference Lopez and Ménard2011; Zou et al., Reference Zou, Guo, Huang, Jin, Cheong, Wang and Xu2012).

MFGM fragments are considered functional ingredients as natural emulsifiers (Schmelz, Reference Schmelz2000) due to the amphiphilic nature of phospholipids, their effectiveness at low concentrations and their original function in stabilizing the fat globules in whole milk (Corredig and Dalgleish, Reference Corredig and Dalgleish1998). Kanno (Reference Kanno1989) investigated the emulsifying properties (foam and emulsion stability, emulsion capability, and whip ability) of MFGM isolates by reconstituting the milk fat globules and concluded that the amounts of MFGM material (20–80 mg g/1fat) significantly affected the properties of the reconstituted emulsions.

MFGM also provides considerable amounts of cholesterol. Cholesterol is a crucial building block for all cell membranes. It affects the development of myelin in the central and peripheral nervous systems, assists as the substratum for the synthesis of bile acids, lipoproteins, vitamin D, hormones, and oxysterols that modulate cholesterol, lipid, and glucose homeostasis (Koletzko, Reference Koletzko2016). The consumption of cholesterol by breastfeeding is associated with higher plasma concentrations of total and low-density lipoprotein cholesterol (LDL-cholesterol) compared to formula-fed infants (Hernell et al., Reference Hernell, Fewtrell, Georgieff, Krebs and Lönnerdal2015). However, this difference in serum cholesterol disappears in childhood (Fomon et al., Reference Fomon, Rogers, Ziegler, Nelson and Thomas1984) and adolescence and, after that, previously breastfed infants have slightly lower serum cholesterol than their formula-fed counterparts (Owen et al., Reference Owen, Whincup, Odoki, Gilg and Cook2002).

Although MFGM is only some nanometers thick, and the diameters of the milk fat globules are in the micrometer range, MFGM seems to be essential for the physiological properties of human milk. MFGM might contribute to the differences in outcome between breastfed and formula-fed infants, which continue to exist even though the difference has become smaller with improved formula qualities (Koletzko and Shamir, Reference Koletzko and Shamir2016).

Human vs. bovine MFGM

MFGM is a component of bovine milk that historically has been discarded with the milk fat during the production of infant formula. Commercially available MFGM isolates are predominantly bovine sourced, and the polar lipids and proteins should be better explored as an emulsifier or stabilizer, combining technological and nutritional functionality (Dewettinck et al., Reference Dewettinck, Rombaut, Thienpont, Le, Messens and Van Camp2008). Human and bovine MFGM have similar polar lipid assemblies, with hundreds of different molecules of polar lipids, varying the polar heads and acyl chains. However, they differ on two major points: human MFGM contains, on average, more sphingomyelin than bovine, and human MFGM acyl chains are, on average, more unsaturated than bovine (Lopez and Ménard, Reference Lopez and Ménard2011). Human MFGM is rich in LC-PUFA of the omega 3 series, such as docosahexaenoic acid (DHA), which is considered as essential for cerebral and visual development during the neonatal period (Yehuda et al., Reference Yehuda, Rabinovitz and Mostofsky2005). As stated above, the capacity of bioconversion of linolenic acid and alpha-linolenic acid into long-chain polyunsaturated fatty acids is limited during the neonatal period, inducing dependence on the dietary intakes from human milk lipids (Lonnerdal, 2016).

There has been little study of non-cattle MFGM, so it is of note that Ji et al. (Reference Ji, Xu, Cui, Ma and Zhou2019) recently compared the MFGM of five different species (yak, camel, bovine, buffalo, and goat). These kinds of milk differ in the MFGM content as follows: yak milk presented the highest amount (0.23%) followed by bovine milk (0.18%), buffalo milk (0.15%), camel milk (0.13%) and goat milk with the lowest amount of MFGM (0.068%). The protein content of yak milk was the highest (425.1 mg/g) followed by buffalo MFGM (416.5 mg/g), bovine MFGM (378.7 mg/g), camel MFGM (348.1 mg/g) and goat MFGM (302.6 mg/g) (Ji et al., Reference Ji, Xu, Cui, Ma and Zhou2019). Lamothe et al. (Reference Lamothe, Robitaille, St-Gelais and Britten2008) studied the effects of washing treatment on phospholipids and milk fat globule membrane but did not find differences in the sphyngomielin contents of bovine and caprine milk.

Advances in proteomics technology, especially quantitative proteomics, have allowed an increased knowledge and characterization of MFGM proteins (Sui et al., Reference Sui, Zhao, Wang, Zhang, Guo, Yu and Li2014). Thus, proteomics has been successfully applied to studies of human, bovine, and goat MFGM proteins (Bianchi et al., Reference Bianchi, Puglia, Landi, Matteoni, Perini, Armini, Verani, Trombetta, Soldani, Roncada, Greppi, Pallini and Bini2009; Spertino et al., Reference Spertino, Cipriani, De Angelis, Giuffrida, Marsano and Cavaletto2012). Most of the published studies on MFGM proteins have been dedicated to the bovine species. Major MFGM bovine proteins include mucin 1, xanthine oxidase, CD36, butyrophilin, adipophilin, PAS6/7 (lactadherin), and fatty acid-binding protein (Cavalleto et al., Reference Cavalleto, Giuffrida, Conti and Bösze2008). Ye et al. (Reference Ye, Singh, Taylor and Anema2002) characterized bovine MFGM using one- and two-dimensional SDS-PAGE to determine the effect of heat treatment on MFGM proteins from early, mid-, and late-season and they found that xanthine oxidase and butyrophilin formed aggregates via intermolecular disulfide bonds after the heating process. The aggregation reduces the physical stability of the protein in question, not only leading to a loss of activity but also other critical problems such as toxicity and immunogenicity (Wang et al., Reference Wang, Singh, Li, Toler, King and Nema2012).

In addition to the typical MFGM proteins such as xanthine oxidase, butyrophilin and fatty acid-binding protein, other proteins have been mapped in human MFGM. These include the following: the IgM m chain, the IgA a chain, the HLA class I heavy chain, immunoglobulin light chains, secretory piece, J chain, actin, a acid glycoprotein, albumin and casein, with particular attention given to the pattern of apolipoproteins E, A-I, A-II, and H (Reinhardt and Lippolis, Reference Reinhardt and Lippolis2006; Cavalleto et al., Reference Cavalleto, Giuffrida, Conti and Bösze2008). Higher concentrations of four proteins involved in the mucosal defense system (IgA, CD14, LTF, and LYZ) were found more in human milk than in bovine milk. These differences in concentration between species may be related to differences in the development of the immune system of babies and calves (Cavalleto et al., Reference Cavalleto, Giuffrida, Conti and Bösze2008).

MFGM in infant formulas

Infant formula development has undergone many modifications in previous decades, with the goal of improving nutritional performance in order to be more similar to human milk. These changes, some of which are relatively recent, include modifications of whey and casein as well as addition of taurine, MFGM, nucleotides, docosahexaenoic acid, arachidonic acid, prebiotics (fructooligosaccharides/galactooligosaccharides) and lutein (Koletzko, 2016). Despite these improvements, differences remain between breastfed infants and formula-fed infants with regard to short-term (e.g., illnesses, cognitive development) and long-term (e.g., obesity, diabetes, cardiovascular disease) outcomes.

When nonfat milk is used as the base for infant formula, and animal fat is replaced with vegetable oils, the levels of MFGM are usually very low (Brink and Lönnerdal, Reference Brink and Lönnerdal2020) with a lack of essential bioactive molecules. Such commercial infant formulas contain lipid droplets that have distinct interfacial compositions (such as citrate glycerides, monoglycerides, or diglycerides) compared to human milk. The droplets in infant formulas are typically much smaller than breast milk fat globules, with particles diameter range around 0.3–0.8 μm and a higher surface area (from about 20−40 m2 g1 fat) with an interfacial layer composed primarily of dairy proteins or surfactants (Lopez and Ménard, Reference Lopez and Ménard2011). The smaller lipid droplets found in infant milk formulas can be attributed to homogenization, a stage involved in the production of these processed products (Wei et al., Reference Wei, Jin and Wang2019a, Reference Wei, Yang, Yang, Wang, Yang, Jin, Wang, Lai and Wang2019b).

The infant formula lipid macrostructure, such as lipid droplets and the interfacial composition, has recently been reported to impact on metabolic programming (Oosting et al., Reference Oosting, van Vlies, Kegler, Schipper, Abrahamse-Berkeveld, Ringler, Verkade and van der Beek2014). The addition of MFGM fragments and/or phospholipids from bovine milk at the surface of processed lipid droplets is an efficient conveyor of energy, and also conveys some low-digestible bioactive complex lipids from the MFGM. Over the last decade, several research groups and manufacturers have investigated the addition of ingredients rich in MFGM or complex lipids to infant formula in order to improve growth and development outcomes in infants (Oosting et al., Reference Oosting, Kegler, Wopereis, Teller, van de Heijning, Verkade and van der Beek2012, Reference Oosting, van Vlies, Kegler, Schipper, Abrahamse-Berkeveld, Ringler, Verkade and van der Beek2014; Billeaud et al., Reference Billeaud, Puccio, Saliba, Guillois, Vaysse, Pecquet and Steenhout2014; Timby et al., Reference Timby, Domellof, Hernell, Lonnerdal and Domellof2014, Reference Timby, Hernell, Vaarala, Melin, Lönnerdal and Domellöf2015; Guan et al., Reference Guan, MacGibbon, Fong, Zhang, Liu, Rowan and McJarrow2015; Hernell et al., Reference Hernell, Timby, Domellöf and Lönnerdal2016; Reis et al., Reference Reis, Bermingham, Reis, Deb-Choudhury, MacGibbon, Fong, McJarrow, Bibiloni, Bassett and Roy2016).

Oosting et al. (Reference Oosting, Kegler, Wopereis, Teller, van de Heijning, Verkade and van der Beek2012, Reference Oosting, van Vlies, Kegler, Schipper, Abrahamse-Berkeveld, Ringler, Verkade and van der Beek2014) in animal studies observed that infant formula containing TAG from vegetable origin and covered by polar lipids from the MFGM (a concept infant formula) reduced fat accumulation as well as fasting plasma leptin, resistin, glucose, and lipids (TAG and total cholesterol) in adult animals fed with a western diet, compared to the group fed with standard infant formula. The same group (Baars et al., Reference Baars, Oosting, Engels, Kegler, Kodde, Schipper, Verkade and van der Beek2016) studied the potential role played by large lipid droplets only, of polar lipid-coating only or the combination of both in the protective effect against obesity in later life. They concluded that a diet containing a combination of large lipid droplets with a coating of MFGM mimicking the lipid droplet architecture as present in mammalian milk prevents body fat accumulation in aniumal models when challenged with a moderate western diet during adolescence and adulthood. Although the mechanisms remain unclear, these animal studies showed that early nutrition with MFGM is associated with sustained effects on later life obesity.

The supplementation of infant formula with MFGM has shown several positive results in clinical studies, such as behavioral improvement and regulation in preschool children (Veereman-Wauters et al., Reference Veereman-Wauters, Staelens, Rombaut, Dewettinck, Deboutte, Brummer, Boone and Le Ruyet2012). A randomized clinical trial with infants below 2 months of age presented an improvement in the neurocognitive development when they received MFGM-supplemented, low-energy, low-protein experimental infant formula compared to standard formula. The cognitive score at 12 months of age was significantly higher in the MFGM-supplemented experimental infant formula group than in the standard infant formula group and, furthermore, was not significantly different from that of the breastfed group (Timby et al., Reference Timby, Domellof, Hernell, Lonnerdal and Domellof2014).

There are several pieces of evidence that MGFM components influence brain development, with tremendous differences compared with formula-fed newborns (Gurnida et al., Reference Gurnida, Rowan, Idjradinata, Muchtadi and Sekarwana2012; Oshida et al., Reference Oshida, Shimizu, Takase, Tamura, Shimizu and Yamashiro2003; Tanaka et al., Reference Tanaka, Hosozawa, Kudo, Yoshikawa, Hisata, Shoji, Shinohara and Shimizu2013). Polar lipid supplementation could reduce the gap in neuronal functions such as cognitive performance of infant fed newborns, behavioral development, and myelination-promoting markers between breastfed and formula-fed infants. All these findings even pose the idea that MFGM supplementation has a beneficial effect on neural functions throughout life. These benefits to neurodevelopment may be associated with the ganglioside content of the MFGM, considering the high ganglioside content in nervous tissue, the high requirement in the perinatal period due to rapid brain growth and the demonstrated uptake of dietary gangliosides (Palmano et al., Reference Palmano, Rowan, Guillermo, Guan and McJarrow2015). In a clinical trial study Gurnida et al. (Reference Gurnida, Rowan, Idjradinata, Muchtadi and Sekarwana2012) compared infants fed with an experimental formula enriched with gangliosides to a group fed a standard formula. The authors observed an improvement in hand and eye coordination in those infants receiving the formula containing the gangliosides, and this improvement was correlated with an increase in serum ganglioside levels. In a recent large study, multiple neurodevelopmental tests were conducted in order to investigat the performance of infants receiving infant formula enriched with MFGM-10 and lactoferrin (Li et al., Reference Li, Zhang, Fu, Wang and Zhang2019). It was observed that infants receiving formula with added bovine MFGM and bovine lactoferrin had an accelerated neurodevelopmental profile at day 365 and improved language subcategories at day 545.

Another recent study revealed a significant difference in the fat globule size and membrane structure between the human milk and infant formulas supplied with the MFGM supplemented products (Bourlieu et al., Reference Bourlieu, Deglaire, De Oliveira, Ménard, Le Gouar, Carrière and Dupont2017). The size and structure of fat globule in the infant formula supplemented with MFGM is interesting yet rarely reported (Wei et al., Reference Wei, Jin and Wang2019a, Reference Wei, Yang, Yang, Wang, Yang, Jin, Wang, Lai and Wang2019b). Given the aforementioned protective effects of MFGM, new studies have shown that the supplementation of MFGM formula might prove beneficial within the developing intestine. The supplementation of infant formulas with MFGM also resulted in a bactericidal effect against various foodborne diseases (Bhinder et al., Reference Bhinder, Allaire, Garcia, Lau, Chan, Ryz, Bosman, Graef, Crowley, Celiberto, Berkmann, Dyer, Jacobson, Surette, Innis and Vallance2017). Studies in rats showed enrichment of Gram-negative Bifidobacterium in feces of animals that received supplementation with bovine MFGM (Lee et al., Reference Lee, Padhi, Hasegawa, Larke, Parenti, Wang, Hernell, Lönnerdal and Slupsky2018). Fragments of MFGM that escape digestion in the lumen may proceed to the large intestine and support the colonization of microbial communities (Bourlieu et al., Reference Bourlieu, Bouzerzour, Ferret-Bernard, Bourgot, Chever, Ménard, Deglaire, Cuinet, Ruyet, Bonhomme, Dupont and Huërou-Luron2015). Evaluation of the colonic bacteria of germ-free mice fed two different types of emulsifiers, soy lecithin vs. MFGM phospholipids (Lacprodan® PL-20, Arla, Denmark), revealed that MFGM phospholipids tended to enrich Porphyromonadaceae, while soy lecithin tended to enrich Enterobacteriaceae and Enterococcaceae. In a distinctive study, rat pups fed a formula supplemented with this same bovine MFGM presented an increased gut microbial species richness and uniformity compared with rat pups fed a formula containing vegetable fat.

Bacteria (including Lactobacillus) present in milk have been shown to preferentially associate with the MFGM utilizing glycan adhesion factors that enable bacteria to bind to mucin (Guerin et al., Reference Guerin, Burgain, Gomand, Scher and Gaiani2019). Besides, bacteria with higher surface hydrophobicity, such as L. reuteri, are better able to adhere to the MFGM (Brisson et al., Reference Brisson, Payken, Sharpe and Jiménez-Flores2010), a phenomenon that is related to properties of the bacterial cell surface. In this context, several patents for utilizing the MFGM as a probiotic carrier have been published and recently reviewed with a focus on lactic acid bacteria (Guerin et al., Reference Guerin, Burgain, Gomand, Scher and Gaiani2019).

Infants fed with formula showed deficits in intestinal development compared with breastfed infants, however, this deficiency could be overcome with the addition of MFGM to the infant formula (Bhinder et al., Reference Bhinder, Allaire, Garcia, Lau, Chan, Ryz, Bosman, Graef, Crowley, Celiberto, Berkmann, Dyer, Jacobson, Surette, Innis and Vallance2017). In a recent clinical trial study, He et al. (Reference He, Parenti, Grip, Lönnerdal, Timby, Domellöf, Hernell and Slupsky2019) characterized the fecal microbiome and metabolome of infants fed a bovine MFGM supplemented formula and compared it with the standard formula used with a breastfed reference group. Their results showed an influence of MFGM on the fecal metabolome that may be associated with gut microbial activity and function.

While researchers are still making efforts to comprehend the functionalities of MFGM, manufacturers have already supplemented formulas with fragments/extracts of bovine MFGM and/or phospholipids-enriched materials (Ramiro-Cortijo et al., Reference Ramiro-Cortijo, Singh, Liu, Medina-Morales, Yakah, Freedman and Martin2020). Studies on infant formulas supplemented with bovine MFGM have shown promising results on neurodevelopment (Timby et al., Reference Timby, Domellof, Hernell, Lonnerdal and Domellof2014) and defense against infections (Timby et al., Reference Timby, Hernell, Vaarala, Melin, Lönnerdal and Domellöf2015). Despite nutritional advances in infant formula with the addition of fragments of MFGM, commercial formulas still have shortcomings compared to human milk. For example, the configuration of the fat globule and the core triglyceride structure should be addressed in an integrated manner. The creation of a novel lipid system closer to human milk fat globule is thus an crucial step toward narrowing the gap between formula-fed and breastfed infants with regard to neurodevelopment, infectious diseases, and cholesterol metabolism.

Advances in infant nutrition

Research and innovation in the field of formulas for children aim not only to approximate the nutritional composition of breast milk, the aforementioned gold standard. The development of new infant formula will also provide benefits at a functional level, especially concerning protection against infectious diseases or metabolic disorders and the stimulation of immunological or neurocognitive development. Adding to molecular structure, the supramolecular structure of dietary lipids in human milk may also influence the bioavailability of PUFA for the developing brain. More recently, with the enhancement of knowledge regarding supramolecular structure's influence on the digestion and absorption of lipids (Gallier et al., Reference Gallier, Vocking, Post, Van De Heijning, Acton, Van Der Beek and Van Baalen2015), interest in simulating the globule structure of human milk fat in infant formula has increased (Wei et al., Reference Wei, Jin and Wang2019a).

The strategy of creating a droplet-based emulsion stable biomimetic human milk is a real technological change. The patented nutrition innovation Nuturis® is an infant formula developed with added milk fat globule membrane to a concept formula. The infant formula has large phospholipid coated lipid droplets (mode diameter 3–5 μm) that mimic the structure of lipids in human milk (Gallier et al., Reference Gallier, Vocking, Post, Van De Heijning, Acton, Van Der Beek and Van Baalen2015). The MFGM fractions are isolated from the by-products of the butter industry (buttermilk and butterserums: Conway et al., Reference Conway, Gauthier and Pouliot2014) using micro and ultrafiltration technologies. Another patented product is the Lacprodan® MFGM-10 designed with a unique protein and lipid profile, including compounds such as lactoferrin, IgG, sialic acid, phospholipids, and gangliosides. According to the manufacturer, Lacprodan® MFGM-10 results in significantly improved cognitive development among formula-fed infants at year one compared to a standard formula. Infants receiving formula supplemented with Lacprodan® MFGM-10 demonstrated similar cognitive performance as breastfed infants.

More than 3500 patents about infant formula were registered in an open database (espacenet patent) in the past five years. Among them, 105 targeted the use of MFGM isolates, 32 involved supplementation with bioactive components such as oligosaccharides and 141 invovled the use of the emulsion process using polar lipids (Espacenet Search, 2020). Besides the use of ingredients closer to human milk, different technological processes to produce new infant formula are also extensively mentioned in this database.

In order to mimic the TAG structure of human milk fat, lipid modification has been used and substitutes such as Betapol® and INFAT® were developed. These substitutes have the regiospecific distribution of the fatty acids similar to the positional distribution of the palmitic acid found in human milk. Both lipids are obtained after interesterification (enzymatic or chemical) (Innis, Reference Innis2011; Zou et al., Reference Zou, Pande and Akoh2016). These substitutes still are not yet seen as being used commonly in infant formulas, probably due to their cost.

The success of the human genome analysis and the initiative on precision medicine has accelerated progress towards personalized healthcare (Collins and Varmus, Reference Collins and Varmus2015). The use of omics technologies such as genomics, epigenomics, transcriptomics, proteomics, and metabolomics in health research has not only advanced the field of medicine but also revolutionized nutritional sciences (German, et al., Reference German, Zivkovic, Dallas and Smilowitz2011; Kellogg et al., Reference Kellogg, Dunn and Snyder2018). Food engineers, pediatricians, and nutritionists have more information to design infant formula for tailored needs and individuals to make personalized dietary decisions for health management.

Technological developments of MFGM

Several authors have reviewed the potential application of MFGM in dairy products and infant formula over the last decade (Singh, Reference Singh2006; Dewettinck et al., Reference Dewettinck, Rombaut, Thienpont, Le, Messens and Van Camp2008; Jiménez-Flores and Brisson, Reference Jiménez-Flores and Brisson2008; Spence et al., Reference Spence, Jimenez-Flores, Qian and Goddik2009). Although the physical-chemical characteristics and functions of the MFGM structure have not been completely elucidated, several technological methods designed to isolate MFGM or generate MFGM ingredients have been brought to the commercial-scale. As a result, the application of these ingredients as supplements for infant formula or other products is becoming increasingly practical.

There are many reasons why the dairy industry has not yet adopted a standard large-scale manufacturing process for MFGM ingredients. Native MFGM is very unstable during processing as handling of raw milk on farms, in transportation, in silos at manufacturing plants and during cream separation results in shear and fluid turbulence causing MFGM damage. Damage to MFGM may cause associated materials, including milk phospholipids, to be depleted from the native milk fat globules in the aqueous milk phase (Huang et al., Reference Huang, Zheng, Brennan, Mohan, Stipkovits, Li and Kulasiri2020). Consequently, more than half of the milk phospholipids in raw milk remain in skim milk (Rombaut et al., Reference Rombaut, Camp and Dewettinck2006; Britten et al., Reference Britten, Lamothe and Robitaille2008). Other limitations are the uncertainties and variables involved in the milk phospholipid fractionation processes.

The processes of isolating and purifying buttermilk MFGM components have been examined in several studies over the past decade (Astaire et al., Reference Astaire, Ward, German and Jiménez-Flores2003; Calvo et al., Reference Calvo, Martín-Hernández, García-Serrano, Castro-Gómez, Alonso-Miravalles, García-Martín, Megino-Tello, Alonso and Fontecha2020; Morin et al., Reference Morin, Pouliot and Jiménez-Flores2006; Reference Morin, Britten, Jiménez-Flores and Pouliot2007; Rombaut et al., Reference Rombaut, Camp and Dewettinck2006). For example, MFGM can be separated from fresh milk or dairy by-products such as buttermilk and butter serum, which are particularly rich in MFGM materials. Consequently, the isolation and proper use of MFGM properties can significantly increase the value-added element of dairy products. Depending on the raw cream's processing history, the nutritional and functional properties of the resulting buttermilk may vary considerably. Moreover, the origin (whey buttermilk, serum-butter or sweet buttermilk) of MFGM components and the matrix surrounding them both seem to affect the resultant biological activity. The MFGM fraction isolated from buttermilk can be utilized as a functional ingredient to stabilize oil-in-water emulsions. Recently, milk phospholipids isolated from MFGM fractions have become commercially available, opening up new possibilities based on phospholipid liposomes' ability to entrap bioactive compounds.

MFGM extracts can be obtained by combining physical processes, whereas with the extraction of specific phospholipids from MFGM-enriched fractions, solvent-extraction is the chosen method (Lee et al., Reference Lee, Kim, Hong, Suh and Jo2020; Price et al., Reference Price, Fei, Clark and Wang2018). Destabilization of the natural emulsion of the milk fat globule is the starting point of most commercial processes to produce MFGM-enriched ingredients (Huang et al., Reference Huang, Zheng, Brennan, Mohan, Stipkovits, Li and Kulasiri2020). For example, as illustrated in Fig. 5, the cream obtained by skimming whole milk constitutes the raw material for butter manufacturing. The churning of milk cream produces butter grains composed primarily of triacylglycerol (TAG) and buttermilk, as well as an aqueous phase containing MFGM constituents (Fig. 5). Buttermilk processing can involve evaporation and spray drying, however, in order to increase the protein and MFGM content of the powder, membrane separation processes can be applied. In cheesemaking processes, the cheese-whey is a by-product that is also considered to be a potential source of MFGM-enriched ingredients.

Fig. 5. Schematic representation of the different processing steps used in the manufacturing of butter and buttermilk.

MFGM-enriched compounds can also be concentrated by employing two other procedures, with the first one being the anhydrous milk fat production process. This process produces a >99.5% fat phase and an aqueous phase called beta serum that contains all MFGM components (Fontecha et al., Reference Fontecha, Brink, Wu, Pouliot, Visioli and Jiménez-Flores2020). The second method requires melting and holding butter at 60°C for 30 min, followed by centrifugal separation leading to a >99.5% fat oil phase and an aqueous buttermilk phase containing MFGM constituents (Fontecha et al., Reference Fontecha, Brink, Wu, Pouliot, Visioli and Jiménez-Flores2020).

Several attempts have been made to separate and isolate MFGM material from commercial buttermilk, with much of the focus on the separation of MFGM material from the proteins of the skim milk. The similarities in the size of casein micelles and MFGM components is the first major obstacle to overcome in order to achieve effective fractionation of buttermilk (Singh, Reference Singh2006). The extraction of MFGM components from buttermilk may include ultrafiltration, followed by supercritical fluid extraction to obtain an ingredient significantly enriched in phospholipids (Costa et al., Reference Costa, Elias-Argote, Jiménez-Flores and Gigante2010). By using micro-and ultrafiltration, the transmission of components through the membrane was found to depend on filtration conditions such as temperature, pH, pore size and type of membrane material and the type of buttermilk (Morin et al., Reference Morin, Pouliot and Jiménez-Flores2006; Rombaut et al., Reference Rombaut, Dejonckheere and Dewettinck2007). Sachdeva and Buchheim (Reference Sachdeva and Buchheim1997) proposed using rennet or the acidification to pH 4.6 following microfiltration in order to remove caseins before the concentration of MFGM. Using another approach, Corredig et al. (Reference Corredig, Roesch and Dalgleish2003) and Roesch et al. (Reference Roesch, Rincon and Corredig2004) used citrate to dissociate casein micelles, followed by microfiltration to collect MFGM material retentate. However, this application also causes loss of MFGM material (Rombaut et al., Reference Rombaut, Camp and Dewettinck2006). Moreover, the addition of a sodium citrate agent results in dispersion of not only casein micelles but also MFGM fragments (Rombaut et al., Reference Rombaut, Camp and Dewettinck2006). More recently Spitsberg et al. (Reference Spitsberg, Ivanov and Shritz2019) obtained considerable success in isolating MFGM from buttermilk using Ca-binding salts, but this was at a research level.

Price et al. (Reference Price, Fei, Clark and Wang2018) obtained different fractions of MFGM from whey protein concentrate using solvent extraction. Their study showed that different classes of phospholipids responded to ethanol concentration and extraction temperature differently. The optimum processing condition is dependent on the desired phospholipid content and recovery yield, and the highest phospholipid content was obtained using 70% ethanol at 70°C. For instance, sphingomyelin was favored during 70% ethanol extraction at 60°C, whereas phosphatidylserine enrichment required first removing other lipids.

The development of methods for the extraction of MFGM from buttermilk through microfiltration may increase the opportunity to produce this ingredient on a commercial scale. The conditions to which fat globules are exposed during processing affect the native structure and composition of MFGM and are likely to have consequences on its functionality. Hence, greater exploitation of the value of MFGM may require redesigning some of the dairy processes to which the globules are subjected.

In conclusion, the best nutrient source for infants continues to be breast milk from a healthy mother. However, under certain conditions, infant formulas must serve as either a sole or partial source of nutrition for at least the first year of life. Over the past few decades there has been a tremendous improvement in the nutritional quality of infant formulas. New ingredients are being developed that meet not only the infant's nutritional requirements but also provide other benefits such as immunity and enhanced cognitive development. Several additional health-related properties have been reported for MFGM, including anticholesterolemic effects, inhibition of cancer cell growth, and bactericidal properties. The MFGM structure is highly complex and composed of an assortment of lipids and proteins with specific nutritional end technological properties. Despite nutritional advances in infant formula with the addition of fragments of MFMG, commercial formulas still have shortcomings compared to human milk. The development of methods to isolate MFGM compounds from buttermilk should be expanded to produce this ingredient on a commercial scale. However, before the economics of such processes can be appreciated, the exceptional functionality of MFGM compounds needs to be further explored in order to be better understood.

Acknowledgements

Authors would like to acknowledge the support of the Agricultural Research Station at North Carolina Agricultural and Technical State University (Greensboro, NC 27411, USA). This research was funded, in part, by Grants (project Number NC.X337-5-21-170-1 and NC.X341-5-21-170-1) from the National Institute of Food and Agriculture (NIFA). Its contents are solely the responsibility of the authors and do not necessarily represent the official views of NIFA.

References

Alvares, CA and Akoh, CC (2016) Enzymatic synthesis of high sn-2 DHA and ARA modified oils for the formulation of infant formula fat analogues. Journal of American Oil Chemists Society 93, 383395.CrossRefGoogle Scholar
Astaire, JC, Ward, R, German, JB and Jiménez-Flores, R (2003) Concentration of polar MFGM lipids from buttermilk by microfiltration and supercritical fluid extraction. Journal of Dairy Science 86, 22972307.CrossRefGoogle ScholarPubMed
Baars, A, Oosting, A, Engels, E, Kegler, D, Kodde, A, Schipper, L, Verkade, HJ and van der Beek, EM (2016) Milk fat globule membrane coating of large lipid droplets in the diet of young mice prevents body fat accumulation in adulthood. British Journal of Nutrition 115, 19301937.CrossRefGoogle ScholarPubMed
Bergsson, G, Arnfinnsson, J, Steingrímsson, O and Thormar, H (2001) Killing of gram-positive cocci by fatty acids and monoglycerides. APMIS Journal of Pathology, Microbiology and Immunology 10, 670678.Google Scholar
Bezkorovainy, A (2001) Probiotics: determinants of survival and growth in the gut. American Journal of Clinical Nutrition 73(2 Suppl.), 399S405S.CrossRefGoogle Scholar
Bhinder, G, Allaire, JM, Garcia, C, Lau, JT, Chan, JM, Ryz, NR, Bosman, ES, Graef, FA, Crowley, SM, Celiberto, LS, Berkmann, JC, Dyer, RA, Jacobson, K, Surette, MG, Innis, SM and Vallance, BA (2017) Milk fat globule membrane supplementation in formula modulates the neonatal gut microbiome and normalizes intestinal development. Scientific Reports 7, 45274.CrossRefGoogle ScholarPubMed
Bianchi, L, Puglia, M, Landi, C, Matteoni, S, Perini, D, Armini, A, Verani, M, Trombetta, C, Soldani, P, Roncada, P, Greppi, G, Pallini, V and Bini, L (2009) Solubilization methods and reference 2-DE map of cow milk fat globules. Journal of Proteomics 72, 853864.CrossRefGoogle ScholarPubMed
Billeaud, C, Puccio, G, Saliba, E, Guillois, B, Vaysse, C, Pecquet, S and Steenhout, P (2014) Safety and tolerance evaluation of milk fat globule membrane-enriched infant formulas: a randomized controlled multicenter non-inferiority trial in healthy term infants. Clinical Medicine Insights Pediatrics 8, 5160.Google ScholarPubMed
Bourlieu, C, Bouzerzour, K, Ferret-Bernard, S, Bourgot, CL, Chever, S, Ménard, O, Deglaire, A, Cuinet, I, Ruyet, PL, Bonhomme, C, Dupont, D and Huërou-Luron, IL (2015) Infant formula interface and fat source impact on neonatal digestion and gut microbiota. European Journal of Lipid Science and Technology 117, 15001512.CrossRefGoogle Scholar
Bourlieu, C, Deglaire, A, De Oliveira, SC, Ménard, O, Le Gouar, Y, Carrière, F and Dupont, D (2017) Towards infant formula biomimetic of human milk structure and digestive behaviour. OCL 24, D206.CrossRefGoogle Scholar
Brink, LR and Lönnerdal, B (2020) Milk fat globule membrane: the role of its various components in infant health and development. Journal of Nutrition Biochemistry 85, 108465.CrossRefGoogle ScholarPubMed
Brisson, G, Payken, HF, Sharpe, JP and Jiménez-Flores, R (2010) Characterization of Lactobacillus reuteri interaction with milk fat globule membrane components in dairy products. Journal of Agricultural and Food Chemistry 58(9), 56125619.CrossRefGoogle ScholarPubMed
Britten, M, Lamothe, S and Robitaille, G (2008) Effect of cream treatment on phospholipids and protein recovery in butter-making process. International Journal of Food Science Technology 43, 651657.CrossRefGoogle Scholar
Bryś, J, Flores, L, Górska, A, Ostrowska-Ligęza, E, Bryś, A, Niemiec, T and Koczoń, P (2019) The synthesis followed by spectral and calorimetric evaluation of stability of human milk fat substitutes obtained from thistle milk and lard. International Journal of Analytical Chemistry 2019, 5417962.Google Scholar
Calhoun, DA, Lunoe, M, Du, Y and Christensen, RD (2000) Granulocyte colony-stimulating factor is present in human milk and its receptor is present in human fetal intestine. Pediatrics 105(1), e7.CrossRefGoogle ScholarPubMed
Calvo, MV, Martín-Hernández, MC, García-Serrano, A, Castro-Gómez, MP, Alonso-Miravalles, L, García-Martín, R, Megino-Tello, J, Alonso, L and Fontecha, J (2020) Comprehensive characterization of neutral and polar lipids of buttermilk from different sources and its milk fat globule membrane isolates. Journal of Food Composition and Analysis 86, 103386.Google Scholar
Cao, X, Kang, S, Yang, M, Li, W, Wu, S, Han, H, Meng, L, Wu, R and Yue, X (2018) Quantitative N-glycoproteomics of milk fat globule membrane in human colostrum and mature milk reveals changes in protein glycosylation during lactation. Food & Function 21, 11631172.CrossRefGoogle Scholar
Carnielli, VP, Luijendijk, IH, van Beek, RH, Boerma, GJ, Degenhart, HJ and Sauer, PJ (1995) Effect of dietary triacylglycerol fatty acid positional distribution on plasma lipid classes and their fatty acid composition in preterm infants. American Journal of Clinical Nutrition 62, 776781.CrossRefGoogle ScholarPubMed
Carnielli, VP, Luijendijk, IH, Van Goudoever, JB, Sulkers, EJ, Boerlage, AA, Degenhart, HJ and Sauer, PJ (1996) Structural position and amount of palmitic acid in infant formulas: effects on fat, fatty acid, and mineral balance. Journal Pediatrics Gastroenteroogy Nutrition 23, 553560.CrossRefGoogle ScholarPubMed
Cavalleto, M, Giuffrida, MG and Conti, A (2008) Milk fat globule membrane components–a proteomic approach. In Bösze, Z (ed.), Bioactive Components of Milk. Advances in Experimental Medicine and Biology, vol. 606. New York, NY: Springer, pp. 129141.CrossRefGoogle Scholar
CDC (2017) Breastfeeding Among U.S. Children Born 2010–2017, New York, USA. Entre for Disease Control National Immunization Survey.Google Scholar
Collins, FS and Varmus, H (2015) A new initiative on precision medicine. New England Journal of Medicine 26(372), 793795.CrossRefGoogle Scholar
Conway, V, Gauthier, SF and Pouliot, Y (2014) Buttermilk: much more than a source of milk phospholipids. Animal Frontiers 4, 4451.CrossRefGoogle Scholar
Corredig, M and Dalgleish, D (1998) Characterization of the interface of an oil-in-water emulsion stabilized by milk fat globule membrane material. Journal of Dairy Research 65, 465477.CrossRefGoogle Scholar
Corredig, M, Roesch, RR and Dalgleish, DG (2003) Production of a novel ingredient from buttermilk. Journal of Dairy Science 86, 27442750.CrossRefGoogle ScholarPubMed
Costa, MR, Elias-Argote, XE, Jiménez-Flores, R and Gigante, ML (2010) Use of ultrafiltration and supercritical fluid extraction to obtain a whey buttermilk powder enriched in milk fat globule membrane phospholipids. International Dairy Journal 20, 598602.CrossRefGoogle Scholar
de Fouw, NJ, Kivits, GA, Quinlan, PT and van Nielen, WG (1994) Absorption of isomeric, palmitic acid-containing triacylglycerols resembling human milk fat in the adult rat. Lipids 29, 765770.CrossRefGoogle ScholarPubMed
Delplanque, B, Gibson, R, Koletzko, B, Lapillonne, A and Strandvik, B (2015) Lipid quality in infant nutrition: current knowledge and future opportunities. Journal of Pediatrics Gastroenterology Nutrition 61, 817.CrossRefGoogle ScholarPubMed
Demmelmair, H and Koletzko, B (2018) Lipids in human milk. Best Practice & Research Clinical Endocrinology & Metabolism 32, 5768.CrossRefGoogle ScholarPubMed
Dewettinck, K, Rombaut, R, Thienpont, N, Le, TT, Messens, K and Van Camp, J (2008) Nutritional and technological aspects of milk fat globule membrane material. International Dairy Journal 18, 436457.Google Scholar
EFSA NDA (2014) Panel scientific opinion on the essential composition of infant and follow-on formulae. EFSA Journal 12, 3760.CrossRefGoogle Scholar
Faustino, AR, Osório, NM, Tecelão, C, Canet, A, Valero, F and Ferreira-Dias, S (2016) Camelina oil as a source of polyunsaturated fatty acids for the production of human milk fat substitutes catalyzed by a heterologous Rhizopus oryzae lipase. European Journal of Lipid Science and Technology 118, 532544.CrossRefGoogle Scholar
Fomon, SJ, Rogers, RR, Ziegler, EE, Nelson, SE and Thomas, LN (1984) Indices of fatness and serum cholesterol at age eight years in relation to feeding and growth during early infancy. Pediatrics Research 18, 12331238.CrossRefGoogle ScholarPubMed
Fontecha, J, Brink, L, Wu, S, Pouliot, Y, Visioli, F and Jiménez-Flores, R (2020) Sources, production, and clinical treatments of milk fat globule membrane for infant nutrition and well-being. Nutrients 12, 1607.CrossRefGoogle ScholarPubMed
Gallier, S, Vocking, K, Post, JA, Van De Heijning, B, Acton, D, Van Der Beek, EM and Van Baalen, T (2015) A novel infant milk formula concept: mimicking the human milk fat globule structure. Colloids Surfaces B Biointerfaces 136, 329339.CrossRefGoogle ScholarPubMed
German, JB, Zivkovic, AM, Dallas, DC and Smilowitz, JT (2011) Nutrigenomics and personalized diets: what will they mean for food? Annual Review Food Science Technology 2, 97123.Google Scholar
Ghosh, M, Sengupta, A, Bhattacharyya, D and Ghosh, M (2016) Preparation of human milk fat analogue by enzymatic interesterification reaction using palm stearin and fish oil. Journal of Food Science and Technology-Mysore 53, 20172024.CrossRefGoogle Scholar
Gila-Diaz, A, Arribas, SM, Algara, A, Martín-Cabrejas, MA, López de Pablo, ÁL, Sáenz de Pipaón, M and Ramiro-Cortijo, D (2019) A review of bioactive factors in human breastmilk: a focus on prematurits. Nutrients 10, 1307.CrossRefGoogle Scholar
Guan, J, MacGibbon, A, Fong, B, Zhang, R, Liu, K, Rowan, A and McJarrow, P (2015) Long-term supplementation with beta serum concentrate (BSC), a complex of milk lipids, during post-natal brain development improves memory in rats. Nutrients 7, 45264541.CrossRefGoogle Scholar
Guerin, J, Burgain, J, Gomand, F, Scher, J and Gaiani, C (2019) Milk fat globule membrane glycoproteins: valuable ingredients for lactic acid bacteria encapsulation? Critical Reviews in Food Science and Nutrition 59, 639651.CrossRefGoogle ScholarPubMed
Gunstone, FD (2009) Vegetable oils. In Shahidi, F (ed.), Bailey's Industrial Oil and Fat Products. Edible Oil and Fat Products: Chemistry, Chemical Properties, and Health Effects, 6th Edn, vol. 2. Hoboken, New Jersey: John Wiley & Sons, pp. 53–101.Google Scholar
Gurnida, DA, Rowan, AM, Idjradinata, P, Muchtadi, D and Sekarwana, N (2012) Association of complex lipids containing gangliosides with cognitive development of 6-month-old infants. Early Human Development 88, 595601.Google ScholarPubMed
Hardison-Moody, A, MacNell, L, Sa, E and Bowen, S (2018) How social, cultural, and economic environments shape infant feeding for low-income women: a qualitative study in North Carolina. Journal of Academic Nutrition Diet 118, 18861894.CrossRefGoogle ScholarPubMed
He, Y, Li, J, Kodali, S, Balle, T, Chen, B and Guo, Z (2017) Liquid lipases for enzymatic concentration of n-3 polyunsaturated fatty acids in monoacylglycerols via ethanolysis: catalytic specificity and parameterization. Bioresource Technology 224, 445456.CrossRefGoogle ScholarPubMed
He, X, Parenti, M, Grip, T, Lönnerdal, B, Timby, N, Domellöf, M, Hernell, O and Slupsky, CM (2019) Fecal microbiome and metabolome of infants fed bovine MFGM supplemented formula or standard formula with breast-fed infants as reference: a randomized controlled trial. Science Reports 12, 11589.CrossRefGoogle Scholar
Hernell, O, Fewtrell, MS, Georgieff, MK, Krebs, NF and Lönnerdal, B (2015) Summary of current recommendations on iron provision and monitoring of iron status for breastfed and formula-fed infants in resource-rich and resource-constrained countries. Journal of Pediatrics 167, S40S47.CrossRefGoogle ScholarPubMed
Hernell, O, Timby, N, Domellöf, M and Lönnerdal, B (2016) Clinical benefits of milk fat globule membranes for infants and children. Journal of Pediatrics 173, S60S65.CrossRefGoogle ScholarPubMed
Huang, Z, Zheng, H, Brennan, CS, Mohan, MS, Stipkovits, L, Li, L and Kulasiri, D (2020) Production of milk phospholipid-enriched dairy ingredients. Foods (basel, Switzerland) 9, 263.Google ScholarPubMed
Innis, SM (2011) Dietary triacylglycerol structure and its role in infant nutrition. Advanced Nutrition 2, 275283.CrossRefGoogle ScholarPubMed
Innis, SM, Nelson, CM, Lwanga, D, Rioux, FM and Waslen, P (1996) Feeding formula without arachidonic acid and docosahexaenoic acid has no effect on preferential looking acuity or recognition memory in healthy full-term infants at 9 mo of age. American Journal of Clinical Nutrition 64, 4046.CrossRefGoogle ScholarPubMed
Jensen, RG (1999) Lipids in human milk. Lipids 34, 12431271.Google ScholarPubMed
Jensen, RG (2018) The Lipids in Human Milk. Boca Ratton, FL, USA: CRC Press, 224p.CrossRefGoogle Scholar
Ji, X, Xu, W, Cui, J, Ma, Y and Zhou, S (2019) Goat and buffalo milk fat globule membranes exhibit better effects at inducing apoptosis and reduction the viability of HT-29 cells. Scientific Reports 9, 2577.Google ScholarPubMed
Jiménez-Flores, R and Brisson, G (2008) The milk fat globule membrane as an ingredient: why, how, when? Dairy Science and Technology 88, 518.CrossRefGoogle Scholar
Jiménez-Flores, R, Higuera-Ciapara, I and Pouliot, Y (2009) 11 – Beverages based on milk fat globule membrane (MFGM) and other novel concepts for dairy-based functional beverages. Functional and Speciality Beverage Technology Woodhead Publishing Series in Food Science, Technology and Nutrition, pp. 281296.CrossRefGoogle Scholar
Kanno, C (1989) Emulsifying properties of bovine milk fat globule membrane in milk fat emulsion – conditions for the reconstitution of milk-fat globules. Journal of Food Science 54, 15341539.CrossRefGoogle Scholar
Kellogg, RA, Dunn, J and Snyder, MP (2018) Personal omics for precision health. Circulation Research 27, 11691171.Google Scholar
Kennedy, K, Fewtrell, MS, Morley, R, Abbott, R, Quinlan, PT, Wells, JCK, Bindels, JC and Lucas, A (1999) Double-blind, randomized trial of a synthetic triacylglycerol in formula-fed term infants: effects on stool biochemistry, stool characteristics, and bone mineralization. The American Journal of Clinical Nutrition 70, 920927.CrossRefGoogle ScholarPubMed
Koletzko, B (2016) Human milk lipids. Annals of Nutrition and Metabolism 69 (Suppl 2), 2840.Google Scholar
Koletzko, B and Shamir, R (2016) Infant formula: does one size fit all? Current Opinion of Clinical Nutrition Metabolic Care 19, 205207.Google ScholarPubMed
Koletzko, B, Agostoni, C, Bergmann, R, Ritzenthaler, K and Shamir, R (2011) Physiological aspects of human milk lipids and implications for infant feeding: a workshop report. Acta Paediatrica 100, 14051415.CrossRefGoogle ScholarPubMed
Kvistgaard, AS, Pallesen, LT, Arias, CF, López, S, Petersen, TE, Heegaard, CW and Rasmussen, JT (2004) Inhibitory effects of human and bovine milk constituents on rotavirus infections. Journal Dairy Science 87, 40884096.CrossRefGoogle ScholarPubMed
Lamothe, S, Robitaille, G, St-Gelais, D and Britten, M (2008) Butter making from caprine creams: effect of washing treatment on phospholipids and milk fat globule membrane proteins distribution. Journal of Dairy Research 75, 439443.CrossRefGoogle ScholarPubMed
Lapillonne, A, Groh-Wargo, S, Gonzalez, CH and Uauy, R (2013) Lipid needs of preterm infants: updated recommendations. Journal of Pediatrics 162, S37S47.CrossRefGoogle ScholarPubMed
Lee, H, Padhi, E, Hasegawa, Y, Larke, J, Parenti, M, Wang, A, Hernell, O, Lönnerdal, B and Slupsky, C (2018) Compositional dynamics of the milk fat globule and Its role in infant development. Frontiers in Pediatrics 24(6), 313.CrossRefGoogle Scholar
Lee, K, Kim, A, Hong, KB, Suh, HJ and Jo, K (2020) Preparation and characterization of a polar milk lipid-enriched component from whey powder. Food Sciences of Animal Resource 40, 209220.CrossRefGoogle ScholarPubMed
Li, F, Zhang, S, Fu, C, Wang, A and Zhang, D (2019) Comparative transcriptional analysis and RNA interference reveal immunoregulatory pathways involved in growth of the oriental river prawn Macrobrachium nipponense. Comparative Biochemistry and Physiology Part D Genomics Proteomics 29, 2431.CrossRefGoogle ScholarPubMed
Lönnerdal, B (1996) Recombinant human milk proteins – an opportunity and a challenge. American Journal Clinical Nutrition 63, 622S626S.Google ScholarPubMed
Lönnerdal, B (2014) Infant formula and infant nutrition: bioactive proteins of human milk and implications for composition of infant formulas. American Journal Clinical Nutrition 99, 712S717S.Google ScholarPubMed
Lönnerdal, B (2016) Bioactive proteins in human milk: Health, nutrition and implications for infant formulas. Journal of Pediatrics 173 Suppl, S4S9.CrossRefGoogle ScholarPubMed
Lopez, C and Ménard, O (2011) Human milk fat globules: polar lipid composition and in situ structural investigations revealing the heterogeneous distribution of proteins and the lateral segregation of sphingomyelin in the biological membrane. Colloids Surfaces B Biointerfaces 83, 2941.CrossRefGoogle ScholarPubMed
López-López, A, Castellote-Bargalló, AI, Campoy-Folgoso, C, Rivero-Urgël, M, Tormo-Carnicé, R, Infante-Pina, D and López-Sabater, MC (2001) The influence of dietary palmitic acid triacylglyceride position on the fatty acid, calcium and magnesium contents of at term newborn faeces. Early Human Development 65, S83S94.CrossRefGoogle ScholarPubMed
Mazzocchi, A, D'Oria, V, De Cosmi, V, Bettocchi, S, Milani, GP, Silano, M and Agostoni, C (2018) The role of lipids in human milk and infant formulae. Nutrients 4, 10567.Google Scholar
Mehrotra, V, Sehgal, SK and Bangale, NR (2018) Fat structure and composition in human milk and infant formulas: implications in infant health. Clinical Epidemiology Global Health 7, 153159.CrossRefGoogle Scholar
Morin, P, Pouliot, Y and Jiménez-Flores, R (2006) A comparative study of the fractionation of regular buttermilk and whey buttermilk by microfiltration. Journal of Food Engineering 77, 521528.CrossRefGoogle Scholar
Morin, P, Britten, M, Jiménez-Flores, R and Pouliot, Y (2007) Microfiltration of buttermilk and washed cream buttermilk for concentration of milk fat globule membrane components. Journal of Dairy Science 90, 21322140.Google ScholarPubMed
Motouri, M, Matsuyama, H, Yamamura, J, Tanaka, M, Aoe, S, Iwanaga, T and Kawakami, H (2003) Milk sphingomyelin accelerates enzymatic and morphological maturation of the intestine in artificially reared rats. Journal of Pediatrics Gastroenterology and Nutrition 36, 241247.CrossRefGoogle ScholarPubMed
Mu, H and Høy, CE (2004) The digestion of dietary triacylglycerols. Progress Lipid Research 43, 105133.CrossRefGoogle ScholarPubMed
Mukherjee, K and Kiewitt, I (1998) Structured triacylglycerols resembling human milk fat by transesterification catalyzed by papaya (Carica papaya) latex. Biotechnology Letters 20, 613616.CrossRefGoogle Scholar
Newburg, DS, Peterson, JA, Ruiz-Palacios, GM, Matson, DO, Morrow, AL, Shults, J, Guerrero, ML, Chaturvedi, P, Newburg, SO, Scallan, CD, Taylor, MR, Ceriani, RL and Pickering, LK (1998) Role of human milk lactadherin in protection against symptomatic rotavirus infection. Lancet (London, England) 18, 11601164.CrossRefGoogle Scholar
Ochsenreither, K, Glück, C, Stressler, T, Fischer, L and Syldatk, C (2016) Production strategies and applications of microbial single cell oils. Frontiers in Microbiology 5(7), 1539.Google Scholar
Oosting, A, Kegler, D, Wopereis, HJ, Teller, IC, van de Heijning, BJ, Verkade, HJ and van der Beek, EM (2012) Size and phospholipid coating of lipid droplets in the diet of young mice modify body fat accumulation in adulthood. Pediatric Research 72, 362369.CrossRefGoogle ScholarPubMed
Oosting, A, van Vlies, N, Kegler, D, Schipper, L, Abrahamse-Berkeveld, M, Ringler, S, Verkade, HJ and van der Beek, EM (2014) Effect of dietary lipid structure in early postnatal life on mouse adipose tissue development and function in adulthood. Brazilian Journal of Nutrition 28, 215226.CrossRefGoogle Scholar
Oshida, K, Shimizu, T, Takase, M, Tamura, Y, Shimizu, T and Yamashiro, Y (2003) Effects of dietary sphingomyelin on central nervous system myelination in developing rats. Pediatric Research 53, 589593.CrossRefGoogle ScholarPubMed
Owen, CG, Whincup, PH, Odoki, K, Gilg, JA and Cook, DG (2002) Infant feeding and blood cholesterol: a study in adolescents and a systematic review. Pediatrics 110, 597608.CrossRefGoogle ScholarPubMed
Palmano, K, Rowan, A, Guillermo, R, Guan, J and McJarrow, P (2015) The role of gangliosides in neurodevelopment. Nutrient 22, 38913913.Google Scholar
Papanikolaou, S and Aggelis, G (2011) Lipids of oleaginous yeasts. Part II: technology and potential applications. European Journal of Lipid Science and Technology 113, 10521073.Google Scholar
Price, N, Fei, T, Clark, S and Wang, T (2018) Extraction of phospholipids from a dairy by-product (whey protein phospholipid concentrate) using ethanol. Journal of Dairy Science 101, 87788787.CrossRefGoogle ScholarPubMed
Quinlan, PT and Chandler, IC (1992) Human milk fat substitutes. Australian Patent Au-A-10344/92.Google Scholar
Ramiro-Cortijo, D, Singh, P, Liu, Y, Medina-Morales, E, Yakah, W, Freedman, SD and Martin, CR (2020) Breast milk lipids and fatty acids in regulating neonatal intestinal development and protecting against intestinal injury. Nutrients 19, 534.CrossRefGoogle Scholar
Ratledge, C (2004) Fatty acid biosynthesis in microorganisms being used for single cell oil production. Biochimie 86, 807815.CrossRefGoogle ScholarPubMed
Reinhardt, T and Lippolis, J (2006) Bovine milk fat globule membrane proteome. Journal of Dairy Research 73, 406416.CrossRefGoogle ScholarPubMed
Reis, MM, Bermingham, EN, Reis, MG, Deb-Choudhury, S, MacGibbon, A, Fong, B, McJarrow, P, Bibiloni, R, Bassett, SA and Roy, NC (2016) Effect of dietary complex lipids on the biosynthesis of piglet brain gangliosides. Journal of Agriculture and Food Chemistry 17, 12451255.CrossRefGoogle Scholar
Rioux, V, Pédrono, F and Legrand, P (2011) Regulation of mammalian desaturases by myristic acid: N-terminal myristoylation and other modulations. Biochimica Biophysica Acta 1811, 18.CrossRefGoogle ScholarPubMed
Roesch, RR, Rincon, A and Corredig, M (2004) Emulsifying properties of fractions prepared from commercial buttermilk by microfiltration. Journal of Dairy Science 87, 40804087.CrossRefGoogle ScholarPubMed
Rombaut, R, Camp, JV and Dewettinck, K (2006) Phospho- and sphingolipid distribution during processing of milk,butter and whey. International Journal of Food Science and Technology 41, 435443.CrossRefGoogle Scholar
Rombaut, R, Dejonckheere, V and Dewettinck, K (2007) Filtration of milk fat globule membrane fragments from acid buttermilk cheese whey. Journal of Dairy Science 90, 16621673.CrossRefGoogle ScholarPubMed
Sachdeva, S and Buchheim, W (1997) Recovery of phospholipids from buttermilk using membrane processing. Kieler Milchwirtschaftliche Forschungsberichte 49, 4768.Google Scholar
Sahin-Yesilcubuk, N and Akoh, CC (2017) Biotechnological and novel approaches for designing structured lipids intended for infant nutrition. Journal of the American Oil Chemists Society 94, 10051034.CrossRefGoogle Scholar
Sánchez-Juanes, F, Alonso, JM, Zancada, L and Hueso, P (2009) Glycosphingolipids from bovine milk and milk fat globule membranes: a comparative study. Adhesion to enterotoxigenic Escherichia coli strains. Biological Chemistry 390, 3140.CrossRefGoogle ScholarPubMed
Schmelz, EM (2000) Dietary sphingomyelin and other sphingolipids in health and disease. Britsh Nutrition Foundation Bulletin 25, 135139.CrossRefGoogle Scholar
Schmidt, R, Meier, U, Markart, P, Grimminger, F, Velcovsky, HG, Morr, H, Seeger, W and Günther, A (2002) Altered fatty acid composition of lung surfactant phospholipids in interstitial lung disease. American Journal Physiology Lung Cellular Molecular Physiology 283, L1079L1085.CrossRefGoogle ScholarPubMed
Silva, RC, Cotting, LN, Poltronieri, TP, Balcão, VM, Almeida, DB, Goncalves, LA, Grimaldi, R and Gioielli, LA (2009) The effects of enzymatic interesterification on the physical-chemical properties of blends of lard and soybean oil. Food Science and Technology/Lebensmittel-Wissenschaft+Technologie 42, 12751282.Google Scholar
Singh, H (2006) The milk fat globule membrane − A biophysical system for food applications. Current Opinion in Colloid Interface and Science 11, 154163.CrossRefGoogle Scholar
Spence, AJ, Jimenez-Flores, R, Qian, M and Goddik, L (2009) The influence of temperature and pressure factors in supercritical fluid extraction for optimizing nonpolar lipid extraction from buttermilk powder. Journal of Dairy Science 92, 458468.CrossRefGoogle ScholarPubMed
Spertino, S, Cipriani, V, De Angelis, C, Giuffrida, MG, Marsano, F and Cavaletto, M (2012) Proteome profile and biological activity of caprine, bovine and human milk fat globules. Molecular Biosystems 8, 967974.CrossRefGoogle ScholarPubMed
Spitsberg, VL, Ivanov, L and Shritz, V (2019) Recovery of milk fat globule membrane (MFGM) from buttermilk: effect of Ca-binding salts. Journal of Dairy Research 86, 374376.CrossRefGoogle ScholarPubMed
Sui, S, Zhao, J, Wang, J, Zhang, R, Guo, C, Yu, T and Li, N (2014) Comparative proteomics of milk fat globule membrane proteins from transgenic cloned cattle. PLoS One 18, e105378.CrossRefGoogle Scholar
Tanaka, K, Hosozawa, M, Kudo, N, Yoshikawa, N, Hisata, K, Shoji, H, Shinohara, K and Shimizu, T (2013) The pilot study: sphingomyelin-fortified milk has a positive association with the neurobehavioural development of very low birth weight infants during infancy, randomized control trial. Brain Development 35, 4552.CrossRefGoogle Scholar
Tecelão, C, Perrier, V, Dubreucq, E and Ferreira-Dias, S (2019) Production of human milk fat substitutes by interesterification of tripalmitin with ethyl oleate catalyzed by Candida parapsilosis lipase/acyltransferase. Journal of the American Oil Chemists' Society 96, 777787.CrossRefGoogle Scholar
Timby, K, Domellof, E, Hernell, O, Lonnerdal, B and Domellof, M (2014) Neurodevelopment, nutrition, and growth until 12 mo of age in infants fed a low-energy, low-protein formula supplemented with bovine milkfat globule membranes: a randomized controlled trial. American Journal of Clinical Nutrition 99, 860868.CrossRefGoogle Scholar
Timby, N, Hernell, O, Vaarala, O, Melin, M, Lönnerdal, B and Domellöf, M (2015) Infections in infants fed formula supplemented with bovine milk fat globule membranes. Journal Pediatrics Gastroenterology and Nutrition 60, 384389.Google ScholarPubMed
Tollin, M, Bergsson, G, Kai-Larsen, Y, Lengqvist, J, Sjövall, J, Griffiths, W, Skúladóttir, GV, Haraldsson, A, Jörnvall, H, Gudmundsson, GH and Agerberth, B (2005) Vernix caseosa as a multi-component defence system based on polypeptides, lipids and their interactions. Cellular Molecular Life Science 62, 23902399.Google ScholarPubMed
Veereman-Wauters, G, Staelens, S, Rombaut, R, Dewettinck, K, Deboutte, D, Brummer, RJ, Boone, M and Le Ruyet, P (2012) Milk fat globule membrane (INPULSE) enriched formula milk decreases febrile episodes and may improve behavioral regulation in young children. Nutrition (Burbank, Los Angeles County, Calif.) 28, 749752.CrossRefGoogle ScholarPubMed
Wang, W, Singh, SK, Li, N, Toler, MR, King, KR and Nema, S (2012) Immunogenicity of protein aggregates − concerns and realities. International Journal of Pharmaceutics 431, 111.Google ScholarPubMed
Wang, X, Zou, S, Miu, Z, Jin, Q and Wang, X (2019) Enzymatic preparation of structured triacylglycerols with arachidonic and palmitic acids at the sn-2 position for infant formula use. Food Chemistry 283, 331337.CrossRefGoogle ScholarPubMed
Wei, W, Jin, Q and Wang, X (2019 a) Human milk fat substitutes: past achievements and current trends. Progress Lipid Research 74, 6986.CrossRefGoogle ScholarPubMed
Wei, W, Yang, J, Yang, D, Wang, X, Yang, Z, Jin, Q, Wang, M, Lai, J and Wang, X (2019 b) Phospholipid composition and fat globule structure I: comparison of human milk fat from different gestational ages. Lactation Stages, and Infant Formulas. Journal Agriculture Food Chemistry 18, 1392213928.CrossRefGoogle Scholar
Xu, X (2000) Enzymatic production of structured lipids: process reactions and acyl migration. Inform (silver Spring, Md ) 11, 11211129.Google Scholar
Yang, TK, Xu, XB, He, C and Li, LT (2003) Lipase-catalyzed modification of lard to produce human milk fat substitutes. Food Chemistry 80, 473481.CrossRefGoogle Scholar
Yaron, S, Shachar, D, Abramas, L, Riskin, A, Bader, D, Litmanovitz, I, Bar-Yoseph, F, Cohen, T, Levi, L, Lifshitz, Y, Shamir, R and Shaoul, R (2013) Effect of high β-palmitate content in infant formula on the intestinal microbiota of term infants. Journal Pediatrics Gastroenterology and Nutrition 56, 376381.CrossRefGoogle ScholarPubMed
Ye, A, Singh, H, Taylor, MW and Anema, S (2002) Characterization of protein components of natural and heat-treated milk fat globule membranes. International Dairy Journal 12, 393402.CrossRefGoogle Scholar
Yehuda, S, Rabinovitz, S and Mostofsky, DI (2005) Essential fatty acids and the brain: from infancy to aging. Neurobiological Aging 26, 98102.CrossRefGoogle ScholarPubMed
Yuhas, R, Pramuk, K and Lien, EL (2006) Human milk fatty acid composition from nine countries varies most in DHA. Lipids 41, 851858.Google ScholarPubMed
Zavaleta, N, Kvistgaard, AS, Graverholt, G, Respicio, G, Guija, H, Valencia, N and Lönnerdal, B (2011) Efficacy of an mfgm-enriched complementary food in diarrhea, anemia, and micronutrient status in infants. Journal of Pediatric Gastroenterology and Nutrition 53, 561568.CrossRefGoogle ScholarPubMed
Zhao, J, Zhao, Y, Du, M, Binns, CW and Lee, AH (2017) Maternal education and breastfeeding practices in China: a systematic review and meta-analysis. Midwifery 50, 6271.Google ScholarPubMed
Zou, XQ, Guo, Z, Huang, JH, Jin, QZ, Cheong, LZ, Wang, XG and Xu, XB (2012) Human milk fat globules from different stages of lactation: a lipid composition analysis and microstructure characterization. Journal Agriculture Food Chemistry 25, 71587167.CrossRefGoogle Scholar
Zou, L, Pande, G and Akoh, CC (2016) Infant formula fat analogs and human milk fat: new focus on infant developmental needs. Annual Review of Food Science and Technology 7, 139165.CrossRefGoogle ScholarPubMed
Zou, X, Ye, L, He, X, Wu, S, Zhang, H and Jin, Q (2020) Preparation of DHA-rich medium- and long-chain triacylglycerols by lipase-catalyzed acidolysis of microbial oil from Schizochytrium sp. with medium-chain fatty acids. Applied Biochemistry and Biotechnology 191, 12941314.CrossRefGoogle ScholarPubMed
Figure 0

Fig. 1. The percentage of breast feed infants in USA (CDC, 2017).

Figure 1

Fig. 2. Structural formulas of Docosahexaenoic acid (DHA) and Arachidonic acid (ARA) fatty acids.

Figure 2

Table 1. Fatty acid composition of human milk in the United States

Figure 3

Fig. 3. Schematic triacylglycerol structure of human milk fat. (Adapted from Mehrotra et al., 2018).

Figure 4

Table 2. An updated summary of enzyme-catalyzed synthesis of human milk fat structured lipids

Figure 5

Fig. 4. Schematic structure of milk fat globule (Adapted from Koletzko, 2016).

Figure 6

Fig. 5. Schematic representation of the different processing steps used in the manufacturing of butter and buttermilk.