REVIEW

Roles of Milk Fat Globule Membrane on Fat Digestion and Infant Nutrition

Changhoon Chai1https://orcid.org/0000-0003-4320-7311, Sejong Oh2https://orcid.org/0000-0002-5870-3038, Jee-Young Imm3,*https://orcid.org/0000-0003-3152-7051
Author Information & Copyright
1Department of Applied Animal Science, Kangwon National University, Chuncheon 24341, Korea
2Devision of Animal Science, Chonnam National University, Gwangju 61186, Korea
3Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea
*Corresponding author: Jee-Young Imm, Department of Foods and Nutrition, Kookmin University, Seoul 02707, Korea, Tel: +82-2-910-4772, Fax: +82-2-910-5249, E-mail: jyimm@kookmin.ac.kr

© Korean Society for Food Science of Animal Resources. This is an Open-Access article distributed under the terms of the Creative Commons Attribution Non-Commercial License (http://creativecommons.org/licenses/by-nc/3.0) which permits unrestricted non-commercial use, distribution, and reproduction in any medium, provided the original work is properly cited.

Received: Feb 03, 2022 ; Revised: Mar 05, 2022 ; Accepted: Mar 07, 2022

Published Online: May 01, 2022

Abstract

Milk fats are present as globules emulsified in the aqueous phase of milk and stabilized by a delicate membrane architecture called milk fat globule membrane (MFGM). The unique structure and composition of the MFGM play an important role in fat digestion and the metabolic programming of neonates. The objective of this review is to compare the structure, composition, and physicochemical characteristics of fat globules in human milk, bovine milk, and infant formula. It provides an overview of the fat digestion process and enzymes in healthy infants, and describes the possible roles of the MFGM in association with factors affecting fat digestion. Lastly, the health benefits of the MFGM on infant nutrition and future perspectives are discussed with a focus on brain development, metabolic response, and gut health.

Keywords: milk fat globule membrane; human milk; infant formula; digestion; health benefit

Introduction

Milk is the lactic secretion of mammals and contains valuable nutrients and immune components for optimum growth of neonates. The composition of milk varies depending on the lactation stage, energy requirements, and the growth rate of the neonate (Singh and Gallier, 2017). Fat is a major energy source in milk and provides essential fatty acids, phospholipids (PL), and cholesterol required for brain development (He et al., 2020). Fats are present as globules in milk and stabilized by a delicate membrane architecture known as the milk fat globule membrane (MFGM). The physical structure of the milk fat globules (MFG) plays an important role in digestion and postprandial metabolism (Baumgartner et al., 2017; Gallier et al., 2013a; Gallier et al., 2013b).

The composition and integrity of the MFGM are significantly altered in the production of infant formula (IF), and the MFGM is reorganized mainly with caseins and whey proteins during emulsification with vegetable oils. The changes in MFGM critically affect energy acquisition (Turgeon and Brisson, 2020), metabolic consequence (Bourlieu and Michalski, 2015), and gut maturation in infants (Li et al., 2018a).

Milk-based IF is commonly used for infants who cannot access human milk (HM). IF-fed infants showed greater body fats and body weight gain compared to HM-fed infants (Lönnerdal, 2014). The increased serum levels of amino acids, insulin, and urea observed in IF-fed infants are due to the increased utilization of protein rather than fat (He et al., 2019). Yuan et al. (2020) reported that fat is preferentially utilized in HM, and HM displays a faster fat digestion rate compared to IF. This suggests that the bioavailability of nutrients can be changed during the production of IF, and both quantitative and qualitative standardization should take into account for humanization of IF.

Whey and cream are two major sources for the production of MFGM and the composition of the MFGM varies depending on sources. Anhydrous milk fat contains only a trace amount of sphingomyelin (SM), whereas buttermilk (12–22 g/100 g PL) and butter serum (24–29 g/100 g PL) have a high level of SM (Rombaut et al., 2007). In addition, the PL content in dairy ingredients varies significantly depending on the analytical methods, season, and lactation period (Anto et al., 2020). In the present review, we would like to share up-to-date information about the role of the MFGM on fat digestion and infant nutrition.

Structure of Milk Fat Globule Membrane (MFGM)

The MFGM is a triple-layered membrane (2%–6% of the globule mass) with 10–50 nm thickness. The MFGM is mainly composed of polar lipids (PL, glycolipids) and membrane-specific proteins (El-Salam and El-Shibiny, 2020). As shown in Fig. 1, the inner triacylglycerol (TAG) core is surrounded by glycerophospholipids and membrane proteins derived from the endoplasmic reticulum and external bilayers derived from the apical membrane and containing SM and ganglioside (GA) (Singh and Gallier, 2017). These MFGM components have lateral and asymmetrical organization. There are two distinct lipid domains in the lipid portion of MFGM. SM is closely associated with cholesterol and forms a densely packed liquid-ordered phase. The SM-rich domain is surrounded by a loosely packed glycerophospholipid matrix containing a high proportion of unsaturated PL (Simons and Vaz, 2004). SM and glycerophospholipids have different structural characteristics in terms of head group structure, hydrocarbon tail length, and degree of unsaturation. The predominant fatty acids of SM are long-chain saturated fatty acids and are esterified via an amide bond to the amino group of sphingosine, whereas two unsaturated fatty acids are bound to a glycerol backbone in glycerophospholipid such as phosphatidylcholine (PC; Li et al., 2015). The polarity of SM is significantly different from PC because of its asymmetric molecular structure and high hydrogen-bonding potential (Slotte, 2016). This segregated PL distribution influences the stabilization and digestion of MFG. The ordered domain contains low-digestible PL, namely SM and glycosphingolipid, whereas the highly unsaturated glycerophospholipid such as PC, phosphatidylethanolamine (PE), phosphatidylinositol, and phosphatidylserine, are readily hydrolyzed in the gut by the phospholipases (Lopez et al., 2019). Moreover, self-assembled SM-cholesterol clusters called lipid rafts act as cell signaling molecules (Rajendran and Simons, 2005). There are strong associations between the protein and lipid components in MFGM. Some fatty acids, including palmitic, stearic, and oleic acids, are strongly bound to the protein components of the membrane and possibly affects the membrane shape and plasticity (Keenan et al., 1982).

kosfa-42-3-351-g1
Fig. 1. Three-dimensional structure of milk fat globule.
Download Original Figure

Composition of Milk Fat Globule Membrane (MFGM)

Lipid composition of MFGM

The lipid fraction of MFGM is mainly composed of TAG (56%–62%), polar lipids (26%–46%), and a minor lipid fraction [diacylglycerol, free fatty acids (FFA) and sterols] (Smoczyński et al., 2012). PL in the MFGM acts as an essential nutrient for organ growth. PL is a major constituent of the brain and provides choline, which is required for various fundamental biological metabolisms (Sánchez et al., 2021). The overall PL compositions of human MFGM and bovine MFGM are similar, but the PL derived from plants has a quite different composition, as shown in Fig. 2. Human and bovine MFGM accounts for approximately 40 and 26 wt% of the total polar lipids, respectively, and HM contains more SM but less PE compared to bovine milk (Mathiassen et al., 2015).

kosfa-42-3-351-g2
Fig. 2. Relative proportion of PL species (%) from different sources. Mature human and bovine milk (Zou et al., 2013), egg (Lordan et al., 2017), soy (Choe et al., 2014), sunflower (Carelli et al., 1997). PC, phosphatidylcholine; PE, phosphatidylethanolamine; PS, phosphatidylserine; PA, phosphatidic acid (in case of sunflower); PL, phospholipids.
Download Original Figure

SM accounts for 25% of the total milk polar lipids (MPL) and is complexed with cholesterol in a mass ratio of 3:1 (Dewettinck et al., 2008). The SM content in milk generally increases with fat content but does not have a clear relationship with the size of the MFG (Graves et al., 2007). HM-fed infants obtain about 150 mg SM/day (Nilsson, 2016). Classification of GA is based on the number of sialic acids. GM (monosialylated GAs) and GD (disialylated GAs) are major GA species in HM, and GD3 (Neu5Aca2-8Neu5Aca2-3Galb1-4Glc-Cer) is the most abundant GD in HM (Fig. 3; Ali et al., 2021).

kosfa-42-3-351-g3
Fig. 3. Major gangliosides (GM3 and GD3) contents in goat, bovine, and human milk. Goat milk (Puente et al., 1994), bovine milk (Lee et al., 2013), and human milk (Giuffrida et al., 2014). GM3, monosiasylated ganglioside 3; GD3, disiasylated ganglioside 3.
Download Original Figure
Protein composition of milk fat globule membrane (MFGM)

MFGM proteins account for 25%–75% of the total mass of the MFGM, and more than 500 MFGM proteins have been identified by proteomic analysis (Reinhardt and Lippolis, 2006). Major MFGM proteins are bound to the membrane structure with different binding strengths. For instance, xanthine dehydrogenase, xanthine oxidase, and mucin-1 are loosely bound and associated with protection against bacterial infection, whereas butyrophilin and adipophilin have relatively high affinities to the membrane structure and are related to protection from multiple sclerosis and fatty acid metabolism (Affolter et al., 2010; Mather, 2000). The affinity of MFGM proteins to the membrane structure also has practical implications for MFGM isolation and extraction (Zheng et al., 2013). Covalently cross-linked butyrophilin-xanthine oxidoreductase may support the physical structure of MFGs by connecting the inner and outer polar lipid layers.

Peroxidases and SM provide protection against oxidation of glycerophospholipids containing PUFA (Coliva et al., 2020; Fong et al., 2007; Oborina and Yappert, 2003). The tight hydrogen bonding between the amide portion of SM and interfacial water molecules interferes with the penetration of oxidizing species in the SM bilayer, resulting in better protection than PC (Oborina and Yappert, 2003). Thus, the extent of lipid peroxidation was inversely correlated with the SM fraction in liposomes (Coliva et al., 2020). Lipases may not have access to the lipid core of MFGs with intact outer polar lipid bilayers in the gut. Non-covalent binding between the outer polar lipid bilayer and inner polar lipid monolayer can be detached by enzymatic reactions in the stomach (Ye et al., 2011). In terms of mechanical property, the HM membrane extract showed greater compressibility than the bovine milk membrane extract due to a greater unsaturated fatty acids content. The greater content of anionic PL in HM means that the HM has superior lipase adsorption activity than bovine milk (Bourlieu et al., 2020).

Fat Digestion in Infants

Fat digestion is an interfacial process, and lipolytic enzymes acting on the surface of emulsified fat droplets determine the rate of fat digestion (Golding and Wooster, 2010). There are significant differences in lipid digestive physiology between infants and adults because of the small size of digestive organs and the differences in the activity of digestive enzymes, gastric pH, gut maturity, and dietary pattern (Abrahamse et al., 2012). In contrast to adults, the role of pancreatic TAG lipase on lipid digestion is not significant in infants while gastric lipase (GL), bile salt-stimulated lipase (BSSL), and pancreatic-lipase related protein 2 (PLRP2) compensate for the insufficient bile salts and pancreatic lipases (Lindquist and Hernell, 2010).

Milk fat digestion in infants is a sequential and balanced process, as shown in Fig. 4. The secretion and role of lingual lipase in fat digestion are uncertain, and actual fat digestion in infants is initiated in the stomach. As the pH drops below 5.5, the MFGM structure becomes less stable, and leads to coagulation of the fat globules (Lopez et al., 2017). Lipid digestion by GL in the stomach facilitates colipase adsorption at the interface and stimulates the action of BSSL and PLRP2 (Bernbäck et al., 1989; Johnson et al., 2013). GL has high activity over a broad pH range (optimum pH: 5.4–5.8) and hydrolyzes ester bonds at sn-3 of TAG. The extent of TAG hydrolysis by GL is up to 10%–30% and 60% in adults and infants, respectively (Abrahamse et al., 2012). The accumulation of liberated fatty acids at the emulsion surface inhibits GL (Pafumi et al., 2002). The 1,2-diacylglycerols produced by GL are further digested by PLRP2 and BSSL. BSSL shows high activity in the neonatal period and hydrolyzes TAG without positional specificity. BSSL is able to hydrolyze TAG, diacylglycerols, and PL and plays an important role in PL digestion in infants with low bile acid content (He et al., 2020). BSSL is an endogenous abundant lipase present in HM but has not been detected in bovine milk. BSSL readily hydrolyzes cholesterol ester, PL, and ceramide (Hernell and Bläckberg, 1994). BSSL is activated upon contact with bile salt but loses its activity upon pasteurization of HM. Alternatively, high-pressure treatment has been suggested as a way to minimize the heat-induced inactivation of BSSL (Singh and Gallier, 2017). Supplementation of recombinant BSSL to pasteurized HM and IF improved the absorption of long-chain fatty acids and the mean growth velocity of preterm infants (n=63) (Casper et al., 2014). BSSL has greater hydrolytic activity on medium and long TAG compared to PLRP2 (Xiao et al., 2011). These two enzymes are also important in PL digestion because PL is barely digested in the infant’s stomach. Dietary SM is hydrolyzed in the intestinal mucosa by alkaline sphingomyelinase (optimum pH: 9.0) to produce phosphocholine and ceramide. In a subsequent step, neutral ceramidase (optimum pH: 7.2) hydrolyzes ceramide to fatty acid and sphingosine. Released sphingosine is adsorbed, phosphorylated to sphingosine-1-phosphate (S1P) and converted to palmitic acid via S1P-lyase in the gut mucosa (Nilsson, 2016). Daily consumption of HM provides approximately 13 and 62 mg SM for preterm (170 mL) and full-term infants (800 mL) infants, respectively (Garcia et al., 2012). Intact MFGM and the presence of BSSL in HM are probably responsible for the difference in lipid absorption between HM-fed and IF-fed infants (Singh and Gallier, 2017).

kosfa-42-3-351-g4
Fig. 4. Sequential digestion of human milk fat globules versus bovine milk fat globules in the stomach (A) and small intestine (B) of infants. (A) Major event: Digestion of MFGM protein by pepsin; MFGM becomes unstable at pH<5.5; GL digestion of fat globules stimulates BSSL and PLRP2 activity; Accumulation of fatty acids and diacylglycerols on fat globule surface. (B) Major event: Secretion of bile salts and formation of micelles; Mixed with lipolysis products produced from the stomach; PLRP2 and BSSL hydrolyze MFGM (BSSL is only expressed in HM); SM can be digested to ceramide by alkaline sphingomyelinase; Ceramide is further hydrolyzed to fatty acids and sphingosine by neutral ceramidase and BSSL. MFGM, milk fat globule membrane; GL, gastric lipase; BSSL, bile-salt-stimulated lipase; PLRP2, pancreatic-lipase-related protein 2; HM, human milk; SM, sphingomyelin.
Download Original Figure

Gut microbiota can also contribute to fat digestion and absorption by modulation of the gene expression of lipid digestion enzymes. Martinez-Guryn et al. (2018) demonstrated that small intestinal microbiota regulates host adaptation in response to dietary lipids. This might suggest a possible synergic interaction of probiotics with the MFGM in IF for efficient lipid digestion.

Factors Affecting Fat Digestion and Absorption

Particle size and interfacial composition

MFGM components stabilize MFG from coalescence and flocculation by providing electrostatic and steric repulsive forces (Singh, 2019). The physicochemical characteristics of fat globules in HM and bovine milk (or IF) are indicated in Table 1. The average fat globule size of HM (~6 μm) is more than 10 times greater than that of IF (~0.6 μm). IF shows a much higher ζ-potential (–36) than HM (–8) because the membrane PL is mostly replaced by casein and whey proteins during homogenization (Yuan et al., 2020).

Table 1. Physicochemical characteristics of fat globules in human and bovine milk (or IF)
Characteristics Human milk Bovine milk/IF References
Fat globules size (μm) 0.35–13 1.0–10 (milk) 0.3–0.8 (IF) Lopez and Ménard (2011), Ménard et al. (2010), Lopez et al. (2015)
Surface area (m2/g fat) 2 2.6±0.2 (milk) 26.4±2.7 (UHT milk) 20–40 (IF) Lopez et al. (2015)
Zeta potential (mV) −7.85 −9.4 (milk) −34 to −38 (IF) Ménard et al. (2010) Yuan et al. (2020)
Total polar lipids (mg/100 mL) 20.4±2.8 19.2±0.8 (milk) Zou et al. (2013)
Concentration of polar lipids (mg/100 mL)
SM 3–14 2.3±0.2 Zheng et al. (2019) Sánchez-Juanes et al. (2009)
PC 2–11 3.3±0.2
PE 0.2–9 2.9±0.2
PI 0.2–3.3 1.4±0.1 (PS/PI)
PS 0.8–4.5
Cholesterol (mg/L) 90–150 300 (milk) 0–4 (IF) Koletzko (2016) Jensen et al. (1990)

IF, infant formula; SM, sphingomyelin; PC, phosphatidylcholine; PE, phosphatidyl-ethanolamine; PI, phosphatidylinositol; PS, phosphatidylserine.

Download Excel Table

Smaller-sized fat globules have greater digestion rates than large-sized ones because of increased surface area for lipase adsorption; however, the composition and structure of the interfacial layer surrounding the fat globules also influence the lipid digestion rate (Bourlieu and Michalski, 2015). In accordance with the relationship between surface area and digestion rate, the GL digestion of bovine milk (smaller-sized fat globules) is faster than that of yak milk. However, this trend is reversed when the fat globules in yak milk are adjusted to sizes similar to that observed in bovine milk due to the structural characteristics of yak milk, such as its higher proportion of short-chain fatty acids and SM-rich ordered domain (Luo et al., 2018; Luo et al., 2020). The presence of MPL improves the hydrolytic action of GL by 2.5-fold compared to soy lecithin (Mathiassen et al., 2015). Similarly, the interfacial composition of an emulsion critically influences the rate and extent of lipolysis by pancreatic lipase. The presence of emulsifiers, such as proteins, PL, and surfactant (Tween 20), interact with lipase or released fatty acids and promote/inhibit adsorption of lipase on the emulsion surface (Mun et al., 2007).

Zhao et al. (2019) observed the changes in the size and interfacial composition of MFG following homogenization and combined homogenization-pasteurization (HTST vs. UHT). Homogenization recruited milk proteins (caseins and whey proteins) to the surface of MFG but expelled glycosylated proteins due to disintegration of the MFGM. These process-induced changes modulate digestion rate. Ultimately, thermal processing of homogenized milk slowed the initial rate of lipid digestion and the overall extent of intestinal digestion in vitro (Liang et al., 2017). The thermal treatment was suggested to limit the ability of lipase to adsorb to the fat globules by promoting increased fat globule flocculation or changes in the structure of the interfacial layer surrounding the fat globules, such as protein crosslinking and denaturation. Although these results were consistent with Tunick et al. (2016), they contrasted with those of Bourlieu and Michalski (2015) that thermal processing could increase the extent of lipid digestion under simulated small intestinal conditions. These discrepancies across studies may be caused by differences in the characteristics of the milk samples, processing conditions, or simulated digestion models used. It is important to consider these factors in the interpretation of the findings. Nonetheless, manipulation of the interfacial structure or aggregation state of MFG would be expected to affect milk fat digestion.

In an independent study, mature HM showed a greater in vitro gastrointestinal rate than commercial IF despite a larger fat globule size, perhaps, in part, due to thinner MFGM interfacial layer surrounding the fat globules, which regulates lipase-mediated lipolysis (Cheong et al., 2018). According to Pan et al. (2022), IF prepared from MFGM lacks interface PL associated with the fat globule structure, has a small particle size (0.38 μm) and a thicker interface layer (and interacts with casein micelles), most of the MFGM exists in the aqueous phase in free-form, and IF does not form a fat globule structure like HM. The homogenization-mediated thick interfacial protein layer formed on the fat globule surface of IF possibly impairs the hydrolytic action of lipase. A novel IF concept mimicking the HM fat globule structure has been suggested (Gallier et al., 2015).

Phospholipids (PL) content

Bläckberg et al. (1981) reported that pancreatic lipase hardly hydrolyzes fat globules covered with MFGM in HM but fat globules are readily hydrolyzed with the aid of colipase and phospholipase A2. The result of that study implied a protective function of the PL surface against the digestive action of pancreatic lipase. Recently, Lu et al. (2021) demonstrated that the addition of PL (soy lecithin; 0.5%, w/v) to milk before homogenization delayed liberation of FFA from fat globules during simulated in vitro intestinal digestion. In addition, the stability of fat globules during storage was increased in PL-added milk by suppressing the adsorption of whey proteins on the fat globule surface.

The effect of PL on fat digestion varies depending on digestion stage (stomach vs small intestine) and type of PL. The activity of GL on emulsions coated with milk PL increased noticeably compared to a soy lecithin-stabilized emulsion with similar size (Mathiassen et al., 2015). The major difference between milk and soy PL is SM. The lipid-ordered SM-cholesterol domain in MFGM might facilitate lipase adsorption to the emulsion surface. Conversely, the opposite effect was observed in the rate of pancreatic lipase hydrolysis, but only when the milk PL-stabilized emulsion was pretreated with GL the emulsion has greater pancreatic lipase activity compared to its soy PL counterpart, a result that was confirmed in vivo in mice (Mathiassen et al., 2015). IF does not contain BSSL, which accounts for about 1/3 of the lipase activity in infants. Thus, the strategic design of the lipid droplet surface layer in IF to maximize GL activity could improve the total fatty acid absorption, benefiting IF-fed infants especially pre-term infants.

An MPL-stabilized emulsion showed enhanced in vitro intestinal digestion and postprandial lipid metabolism in mice compared to an emulsion stabilized by soy PL (Lecomte et al., 2015). The proposed reason for these results was that both gastric emptying and intestinal absorption were promoted in the MPL emulsion. More in-depth clinical research is required to confirm whether postprandial lipid metabolism can be modulated by the selection of the type of PL or lipid emulsifiers. The estimated SM intake of HM-fed infants (4 wks old) is about 18–84 mg/day, whereas that of standard IF-fed infants is 9.6–20.4 mg/day (assumed 600 mL consumption/day; Zheng et al., 2019). This difference possibly causes critical difference in fat digestion and the subsequent utilization of fat. Generally, the SM content of commercial milk-based IF is less than that in HM and even less in soy-based IF because soy-based IF does not have SM (Cilla et al., 2016).

Liang et al. (2018) examined the effect of emulsifier type (sodium caseinate, lactoferrin, whey protein isolate, and MPL) and emulsion droplet size on gastrointestinal digestion in vitro. They could not find any significant difference on overall lipid digestion profiles depending on emulsifier type. However, the MPL-stabilized emulsion did not show the transient lag phase observed during digestion of the protein-stabilized emulsions. In a recent study by Liu et al. (2021), the fatty acid release was inversely correlated with the particle size of MPL-stabilized emulsions. Furthermore, the overall gastrointestinal FFA release from MPL-stabilized emulsions was comparable to that of HM. This implies that the particle size of IF does not have to be close to HM as long as an appropriate amount of MPL is incorporated into IF.

Regio-distribution of fatty acid in triacylglycerol (TAG)

The fatty acids of the TAG in HM have preferential region-distribution. Palmitic acid (C16:0), the primary saturated fatty acid (20%–25%), is esterified at the sn-2 position mainly (>70%), whereas the sn-1,3 positions of TAG are occupied by unsaturated fatty acids (Innis, 2011). Fatty acid region-distribution affects fat digestion kinetics because GL shows stereospecificity for the sn-3. Medium chain fatty acids (C8–C12) are mainly located in the sn-3 position of TAG and are rapidly utilized in the liver after absorption via the portal vein (Gómez-Cortés et al., 2018). Fatty acids esterified at the sn-2 position of TAG are regarded as a hot spot because about 70% of the fatty acids absorbed as sn-2 monoacylglycerols are absorbed across the enterocytes and conserved in the original position during re-esterification into TAGs for secretion into the plasma as chylomicrons (Zhang et al., 2018). The improved absorption of at sn-2 palmitate compared to the one esterified to TAG 1,3 positions in HM and IF is well known (Innis et al., 1994; Tomarelli et al., 1968). The absorption of fatty acids decreases as the fatty acid chain length or unsaturation degree increases in HM. The decreased long chain saturated fatty acid and fatty acid-calcium soap formation are associated with constipation and decreased mineral bioavailability (Li et al., 2010). Moreover, the accumulation of long chain fatty acids at the interface limits lipase access to other TAG (Guo et al., 2017).

Hageman et al. (2019) used an in vitro digestion assay to compare the FFA release from IF prepared from only vegetable fat with a milk and vegetable fat combination (67:33). There was no significant difference in the total fatty acid release, but greater amounts of short (C4:0) and medium chain fatty acid (C6:0, C8:0, and C10:0) were released from the IF containing combined fat compared to IF containing vegetable fat alone. The delivery of butyric acid (C4:0) possibly contributes to the development of gut maturation. Differences in TAG composition effect the self-assembly behavior of the lipid species during gastrointestinal digestion, with increasing evidence suggesting that the more copious fatty acid digestion products guide the self-assembly process. Pham et al. (2020) demonstrated that not all IF form the same micellar cubic phase that HM forms during lipid digestion. Inverse micellar cubic phases were formed from milk liberating more long chain unsaturated fatty acids (C18:1 and C18:2) by contrast to hexagonal and inverse bicontinuous cubic phases of lower interfacial curvature from milk liberating more long chain saturated fatty acids (C14:0, C16:0, and C18:0). Although the actual function of specific structures is still uncertain, it could modulate lipase adsorption at the interface, with interesting possibilities for designing IF that effectively mimic the nanoscale structures seen in HM and, in turn, improve the nutritional outcomes of IF-fed infants.

Matrix effects

The food matrix refers to the specific organization of food constituents in space or the presence of nutrients that critically influence the release and absorption of nutrients during digestion in the gastrointestinal tract (Ubbink et al., 2008). Lamothe et al. (2017) compared the rate of fatty acid release from various dairy matrices (milk, yogurt, and cheese). Fatty acid release from the solid matrix was delayed compared to the liquid and semi-solid dairy matrices.

The supramolecular structure of the MFG influences the rate of lipid digestion (Michalski et al., 2006). In a dietary intervention study involving 49 men and women, cheese consumption lowered LDL-cholesterol compared with butter intake of equal fat content (Hjerpsted et al., 2011). This indicates that the integrity of the MFGM matrix in the products possibly delivers different health consequences. In addition, the high calcium content of dairy products, such as cheese, is involved in reduced fat absorption by the formation of calcium-fatty acid complexes (Lamothe et al., 2017).

In randomized human clinical trial (n=57), consumption of an equal amount (40 g milk fat/day, 8 wks) of whipping cream (no MFGM) or butter oil (MFGM) led to significant differences in the lipoprotein profile and cholesterol metabolism (Rosqvist et al., 2015). The exact mechanisms for this result still remain unclear. The one possibility is that the SM in the MFGM suppressed intestinal cholesterol uptake via decreasing the thermodynamic activity of cholesterol monomers (Eckhardt et al., 2002). These findings suggest that the manipulation of lipid structure poses different health consequences.

The homogenization-induced increased casein content on the fat globules surface results in gastric coagulation and inhibitory effect on GL. The physical state of milk in the stomach and the protein integrity also affect milk lipids digestibility (Lopez et al., 2015). Lipid digestion in IF was increased when intact proteins were replaced with hydrolyzed proteins (Nguyen et al., 2018). Fondaco et al. (2015) compared the physicochemical properties and rate of in vitro lipid digestion in HM, bovine milk, and IF (4 types). A positive linear relationship between the rate of lipolysis and fat globule surface area was only found in IF. This implies that other matrix components are possibly involved in the rate of lipolysis in HM. In addition, the pH value for maximum viscosity differed (pH 3.0 for HM vs. pH 4.0–5.0 for IF), which could influence the rate of gastric emptying and possibly satiety, but there was no correlation found between viscosity and lipid bioaccessibility.

The physical state of fat (solid fat vs liquid oil) critically affects its digestibility. Whey protein-stabilized emulsions containing increasing levels of solid fat (hydrogenated soybean oil, melting point>37°C) showed decreasing liberation of FFA during intestinal digestion in vitro (Guo et al., 2017).

The Role of Milk Fat Globule Membrane (MFGM) in Infant Nutrition

Brain development

There is a growing body of evidence regarding the beneficial health effects of MFGM in infants. MFGM supplementation to IF (0.5 g/L) accelerated neurodevelopment and promoted cognitive function in healthy full-term infants with a low incidence of pathogen-associated adverse effects (Li et al., 2019). Infants fed formula enriched with SM (28–71 mg/mL) displayed increased developmental myelination in the brains and later verbal development in the first 2 years of life (O’Muircheartaigh et al., 2014; Schneider et al., 2019). Comparing the effect of SM-fortified milk (20% SM vs. 13% SM) on the neurobehavioral development of low-birth-weight infants revealed a markedly higher serum SM level and neurodevelopment test score in the SM-enriched group than the control group. Dietary SM and its metabolites such as cerebroside, are able to cross the blood-brain barrier and promote myelination (Tanaka et al., 2013). Although the long-term effect of SM fortification is still not clear, SM has great potential in brain development in preterm or low-birth-weight infants.

Healthy 6-month-old infants fed IF fortified with MPL showed an increased serum GD level and improved hand-eye coordination, and general IQ compared to the control group fed standard IF but did not differ in cognitive development score or GD serum level from the reference group of healthy exclusively breastfed infants (Gurnida et al., 2012). On other work, MFGM supplementation improved insulin signaling in hippocampus and cerebral cortex of aged rats, which suggests that MPL possibly improve age-related cognitive decline (Tomé-Carneiro et al., 2018). However, these positive effects should be confirmed in clinical studies.

The effect of MFGM or MFGM/prebiotic combination on stress (maternal separation)-induced microbiota change, visceral hypersensitivity, and brain function was evaluated in rats (O’Mahony et al., 2020). The MFGM supplementation significantly increased the beta-diversity of the cecal microbiome and improved spatial learning in the stress group (O’Mahony et al., 2020). A similar phenomenon might be expected between MFGM and probiotics on cognitive development (Kosmerl et al., 2021). Further studies are needed to examine this possibility and elucidate mechanism of interaction. It may be that the MFGM interacts with the bacterial surface to facilitate the delivery of probiotics to their target site of action or that the MFGM creates a favorable environment in the gastrointestinal tract for the probiotic species, alters their metabolism, and, ultimately, influences the gut microbiome. According to these studies, the MFGM-mediated improved brain function may be associated with modulation of the gut microbiota via the gut-brain axis, and supplementing an MFGM concentrate may modify gut microbial composition and by-products to a profile more comparable to an exclusively breastfed reference group.

Metabolic responses

Supplementation of IF with MFGM altered metabolic outcomes due to a shift in the preference for protein utilization (e.g., increased level of lactate, succinate and amino acids) to a preference for fat utilization and converted the fecal microbiome to more like that of HM-fed infants (He et al., 2019). The high sensitivity to lipolysis and β-oxidation in early life can help inhibit excessive weight gain in later life and thus decrease the risk of obesity (Lee et al., 2021). MFGM supplementation significantly reduced adipogenesis and body weight gain by promoting brown fat formation in white adipose tissues in rats (Li et al., 2018b). A subsequent study found that administration of MFGM to high-fat diet-fed rats during pregnancy and lactation stimulated brown fat development in male offspring (Li et al., 2020). Recently, Zhang et al. (2021) demonstrated that MFGM supplementation during suckling reduced the risk of maternal high-fat diet-induced nonalcoholic fatty liver disease in mice due, in part, to decreased oxidative stress, and restoration of mitochondrial dysfunction. The same research group evaluated the effect of MFGM supplementation during pregnancy and lactation in obese rats on the skeletal outcomes of male offspring. Maternal MFGM supplementation (400 mg/kg body weight) ameliorated the stunted skeletal growth of male offspring at weaning and protected against high-fat diet-induced bone microstructure degeneration and insulin resistance in adulthood offspring (Han et al., 2021). The enhanced insulin-like growth factor-I activity was suggested as one of the possible reasons for the positive skeletal outcomes. Based on these results, MFGM has a protective role against diet-induced obesity development and obesity-related complications.

SM, the major MFGM constituent, was evaluated for its effect on lipid metabolism. Narita et al. (2016) found that long chain bases of SM were effectively transported into cells via acyl-CoA synthetases and competitively inhibited the uptake of long chain fatty acids. Other research demonstrated the anti-inflammatory activity of SM. Dietary SM reduced systematic inflammation in a diet-induced obese mice model, and milk SM downregulated pro-inflammatory gene expressions, such as tumor necrosis factor-alpha (TNF-α) and C-C motif chemokine ligand 2 (CCL2) in LPS-stimulated RAW 264.7 macrophages (Norris et al., 2017). Interestingly, the long chain sphingosine bases (C16-ceramide and C24-ceramide), exerted a similar anti-inflammatory effect to SM, but dihydroceramide species (sphinganine base) did not display anti-inflammatory activity. Human clinical studies should be undertaken to confirm the beneficial effects of SM on lipid metabolism and immune modulation.

Richard et al. (2017) reported that the administration of a choline mixture (50% PC, 25% free cholines, and 25% glycerophosphocholines) to rat dams during lactation improved the immune system development in offspring and elicited a more effective maternal inflammatory response following mitogenic immune challenge. Similar effects were confirmed when butter milk was used as a choline source (Azarcoya-Barrera et al., 2021).

Gut health

It is well known that SM and its metabolites present in MFGM involved in the regulation of cell growth, intestinal lipid uptake, and gastrointestinal immune responses (Rohrhofer et al., 2021). They act as signaling molecules and mediates cell proliferation and apoptosis. For example, S1P improves endothelial cell survival and migration of immune cells such as lymphocytes, dendriatic cells and macrophages which are closely related to gut homeostasis (Nilsson, 2016). Moreover, metabolites of dietary sphingolipids influence gut microbiota by modulation of cell attachment of commensal bacteria or pathogens (Rohrhofer et al., 2021). In addition to sphingolipids, GA in MFGM also modulate proinflammatory signaling in the gut. Miklavcic et al. (2012) reported that the decreased GM3 stimulates synthesis of proinflammatory signals and increases susceptibility to pathogens. Conversely, dietary GA consumption alleviate inflammatory symptoms by blocking inflammatory cascade.

According to Bezirtzoglou et al. (2011), HM-fed infants had doubled the fecal number of Bifidobacterium cells of IF-fed infants. IF feeding increased the Atopobium level and decreased the numbers of Bifidobacterium. In an independent study, supplementation of IF with dairy lipid and MFGM influenced protein digestibility and microbiota composition. Incorporation of milk lipid and MFGM fragments in IF accelerated mucosal immune development, reduced protein digestion, and altered the fecal microbiota composition in neonatal piglets (Le Huёrou-Luron et al., 2018).

MFGM supplementation in IF stimulates epithelial cell proliferation and gut barrier function in rats by strengthening tight junction proteins and modulating the neonatal gut microbiome (Bhinder et al., 2017). MFGM supplementation also improved obesity-associated gut dysbiosis and increased the Bacteroidetes/Firmicutes ratio in high-fat diet-fed mice study (Li et al., 2018a). Following the analysis of fecal samples, Zhao et al. (2022) reported that MFGM components especially lactadherin (milk fat globule-epidermal grow factor-8), sialic acid, and PL, promoted growth of Bifidobacterium and suppressed Veillonella, Escherichia and Shigella in healthy full-term newborns. By contrast, supplementation of MFGM and lactoferrin to IF led to only subtle changes in the stool microbiome and metabolome of healthy full-term infants at 4 mo of age (Chichlowski et al., 2021). The authors of that study reported that the abundance of the microbial community varies primarily depending on infant age rather than the diet.

The MFGM enhanced the gut barrier function and lowered the intestinal permeability in a rat model of short bowel syndrome (Yu et al., 2021). Regulation of the NLRP6 inflammasome pathway contributed to the improvement of gut dysbiosis. According to Levy et al. (2015), microbiota-modulated metabolites (taurine, histamine, and spermine) regulate NLRP6 inflammasome, intestinal IL-18, and downstream antimicrobial peptide (AMP) patterns. Inflammasome deficiency distorts this harmonious AMP environment. Restoration of the metabolite-inflammasome-AMP axis reestablishes a normal microbiota and ameliorates colitis. Zhang et al. (2020) reported that MFGM improved viability of Lactobacillus rhamnosus GG from bile stress both in vitro and in the murine GI tract. The promotion of probiotic survival by MFGM might provide a beneficial effect on gut health.

Safety of Milk Fat Globule Membrane (MFGM) Fortification

GD supplementation to IF at 1.43 mg/100 kcal significantly reduced Escherichia coli in feces of preterm newborn infants at 7 days after birth and substantially increased the level of bifidobacteria in feces at 30 days without any hint of adverse effects (Rueda et al., 1998). Billeaud et al. (2014) reported that the MFGM might have different nutritional and safety consequences depending on the actual composition of the MFGM ingredient. Lipid-rich MFGM supplementation (n=70, age 14 days) for 14 weeks did not show any major safety concerns such as weight gain, morbidity, and metabolic markers, whereas protein-rich MFGM enrichment (n=72) was associated with a higher eczema rate compared to the controls (n=57). However, caution is needed in the interpretation of results because eczema rate assessment was not standardized and was mainly based on the parental report. Moreover, more large-scale clinical data are required to draw solid conclusions about the health benefits and safety of MFGM supplementation (Fontecha et al., 2020).

Conclusion

To date, the importance of the fat globule organization and interfacial composition on infant nutrition has been overlooked compared to the chemical composition of lipid in IF. The lipid species of milk exosomes are close to that of the MFGM, and some of the bioactivity of exosomes is derived from their lipid content (Subra et al., 2010). Although evidence supporting MFGM or its fraction in the formulation of IF are increasing, several issues outlined below should be clarified to proceed to the next clinical stage.

Standardization of the MFGM ingredient is a priority. Currently, MFGM is produced from dairy side streams, such as serum or cream concentrate. Substantial variations in the types and amounts of PL, sialic acid, and membrane protein components are observed depending on the source and processing methods (Brink et al., 2020; Qu et al., 2019). Heterogeneity in the MFGM composition is probably a major reason for the discrepancy in the beneficial effects of MFGM supplementation in children (Ambrożej et al., 2021). The concentration of individual MFGM components still shows large variation, and underlying the molecular mechanisms of MFGM for infant health are still ambiguous. Moreover, the exact and synergistic roles of individual components in MFGM on specific health effects should be clarified to establish a nutritional recommendation. In addition the effective target population (pregnant mothers or infants) should be carefully selected to optimize the health benefits. The final future challenge will be the modulation of lipid digestion and subsequent fat absorption via the formation of a composite structure. Furthermore, a multi-composite emulsion structure that elicits an improved sensory quality will be possible in the future.

Conflicts of Interest

The authors declare no potential conflicts of interest.

Acknowledgments

This research was supported by Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Science, ICT and Future Planning (2020R1A2C1011548). The funder had no role in study design, data collection and analysis, decision to publish, or preparation of the manuscript.

Author Contributions

Conceptualization: Imm JY. Data curation: Chai C, Oh S, Imm JY. Investigation: Chai C, Oh S, Imm JY. Writing - original draft: Chai C, Oh S, Imm JY. Writing - review & editing: Chai C, Oh S, Imm JY.

Ethics Approval

This article does not require IRB/IACUC approval because there are no human and animal participants.

References

1.

Abrahamse E, Minekus M, van Aken GA, van de Heijning B, Knol J, Bartke N, Oozeer R, van der Beek EM, Ludwig T. 2012; Development of the digestive system—experimental challenges and approaches of infant lipid digestion. Food Dig. 3:63-77

2.

Affolter M, Grass L, Vanrobaeys F, Casado B, Kussmann M. 2010; Qualitative and quantitative profiling of the bovine milk fat globule membrane proteome. J Proteomics. 73:1079-1088

3.

Ali AH, Wei W, Wang X. 2021; A review of milk gangliosides: Occurrence, biosynthesis, identification, and nutritional and functional significance. Int J Dairy Technol. 75:21-45

4.

Ambrożej D, Dumycz K, Dziechciarz P, Ruszczyński M. 2021; Milk fat globule membrane supplementation in children: Systematic review with meta-analysis. Nutrients. 13:714

5.

Anto L, Warykas SW, Torres-Gonzalez M, Blesso CN. 2020; Milk polar lipids: Underapprecoated lipids with emerging health benefits. Nutrients. 12:1001

6.

Azarcoya-Barrera J, Field CJ, Goruk S, Makarowski A, Curtis JM, Pouliot Y, Jacobs RL, Richard C. 2021; Buttermilk: An important source of lipid soluble forms of choline that influences the immune system development in Spargue-Dawley rat offspring. Eur J Nutr. 60:2807-2818

7.

Baumgartner S, van de Heijning BJM, Acton D, Mensink RP. 2017; Infant milk fat droplet size and coating affect postprandial responses in healthy adult men: A proof-of-concept study. Eur J Clin Nutr. 71:1108-1113

8.

Bernbäck S, Bläckberg L, Hernell O. 1989; Fatty acids generated by gastric lipase promote human milk triacylglycerol digestion by pancreatic colipase-dependent lipase. Biochim Biophys Acta. 1001:286-293

9.

Bezirtzoglou E, Tsiotsias A, Welling GW. 2011; Microbiota profile in feces of breast- and formula-fed newborns by using fluorescence in situ hybridization (FISH). Anaerobe. 17:478-482

10.

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, Vallance BA. 2017; Milk fat globule membrane supplementation in formula modulates the neonatal gut microbiome and normalizes intestinal development. Sci Rep. 7:45274

11.

Billeaud C, Puccio G, Saliba E, Guillois B, Vaysse C, Pecquet S, 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. Clin Med Insights Pediatr. 8:51-60

12.

Bläckberg L, Hernell O, Olivecrona T. 1981; Hydrolysis of human milk fat globules by pancreatic lipase: Role of colipase, phospholipase A2, and bile salts. J Clin Invest. 67:1748-1752

13.

Bourlieu C, Mahdoueni W, Paboeuf G, Gicquel E, Ménard O, Pezennec S, Bouhallab S, Deglaire A, Dupont D, Carrière F, Vié V. 2020; Physico-chemical behaviors of human and bovine milk membrane extracts and their influence on gastric lipase adsorption. Biochimie. 169:95-105

14.

Bourlieu C, Ménard O, De La Chevasnerie A, Sams L, Rousseau F, Madec MN, Robert B, Deglaire A, Pezennec S, Bouhallab S, Carrière F, Dupont D. 2015; The structure of infant formulas impacts their lipolysis, proteolysis and disintegration during in vitro gastric digestion. Food Chem. 182:224-235

15.

Bourlieu C, Michalski MC. 2015; Structure-function relationship of the milk fat globule. Curr Opin Clin Nutr Metab Care. 18:118-127

16.

Brink LR, Herren AW, McMillen S, Fraser K, Agnew M, Roy N, Lönnerdal B. 2020; Omics analysis reveals variations among commercial sources of bovine milk fat globule membrane. J Dairy Sci. 103:3002-3016

17.

Carelli AA, Brevedan MIV, Crapiste GH. 1997; Quantitative determination of phospholipids in sunflower oil. J Am Oil Chem Soc. 74:511-514

18.

Casper C, Carnielli VP, Hascoet JM, Lapillonne A, Maggio L, Timdahl K, Olsson B, Vågerö M, Hernell O. 2014; rhBSSL improves growth and LCPUFA absorption in preterm infants fed formula or pasteurized breast milk. J Pediatr Gastroenterol Nutr. 59:61-69

19.

Cheong LZ, Jiang C, He X, Song S, Lai OM. 2018; Lipid profiling, particle size determination, and in vitro simulated gastrointestinal lipolysis of mature human milk and infant formula. J Agric Food Chem. 66:12042-12050

20.

Chichlowski M, Bokulich N, Harris CL, Wampler JL, Li F, Berseth CL, Rudolph C, Wu SS. 2021; Effect of bovine milk fat globule membrane and lactoferrin in infant formula on gut microbiome and metabolome at 4 months of age. Curr Dev Nutr. 5nzab027

21.

Choe J, Oh B, Choe E. 2014; Effect of soybean lecithin on iron-catalyzed or chlorophyll-photosensitized oxidation of canola oil emulsion. J Food Sci. 79:C2203-C2208

22.

Cilla A, Diego Quintaes K, Barberá R, Alegría A. 2016; Phospholipids in human milk and infant formulas: Benefits and needs for correct infant nutrition. Crit Rev Food Sci Nutr. 56:1880-1892

23.

Coliva G, Lange M, Colombo S, Chervet JP, Domingues MR, Fedorova M. 2020; Sphingomyelins prevent propagation of lipid peroxidation—LC-MS/MS evaluation of inhibition mechanisms. Molecules. 25:1925

24.

Dewettinck K, Rombaut R, Thienpont N, Le TT, Messens K, Van Camp J. 2008; Nutritional and technological aspects of milk fat globule membrane material. Int Dairy J. 18:436-457

25.

Eckhardt ERM, Wang DQH, Donovan JM, Carey MC. 2002; Dietary sphingomyelin suppresses intestinal cholesterol absorption by decreasing thermodynamic activity of cholesterol monomers. Gastroenterology. 122:948-956

26.

El-Salam M, El-Shibiny S. 2020; Milk fat globule membrane: An overview with particular emphasis on its nutritional and health benefits. Int J Dairy Technol. 73:639-655

27.

Fondaco D, AlHasawi F, Lan Y, Ben-Elazar S, Connolly K, Rogers MA. 2015; Biophysical aspects of lipid digestion in human breast milk and SimilacTM infant formulas. Food Biophys. 10:282-291

28.

Fong BY, Norris CS, MacGibbon AKH. 2007; Protein and lipid composition of bovine milk-fat-globule membrane. Int Dairy J. 17:275-288

29.

Fontecha J, Brink L, Wu S, Pouliot Y, Visioli F, Jiménez-Flores R. 2020; Sources, production, and clinical treatments of milk fat globule membrane for infant nutrition and well-being. Nutrients. 12:1607

30.

Gallier S, Cui J, Olson TD, Rutherfurd SM, Ye A, Moughan PJ, Singh H. 2013a; In vivo digestion of bovine milk fat globules: Effect of processing and interfacial structural changes I. Gastric digestion. Food Chem. 141:3273-3281

31.

Gallier S, Vocking K, Post JA, Van De Heijning B, Acton D, Van Der Beek EM, Van Baalen T. 2015; A novel infant milk formula concept: Mimicking the human milk fat globule structure. Colloids Surf B Biointerfaces. 136:329-339

32.

Gallier S, Zhu XQ, Rutherfurd SM, Ye A, Moughan PJ, Singh H. 2013b; In vivo digestion of bovine milk fat globules: Effect of processing and interfacial structural changes. II. Upper digestive tract digestion. Food Chem. 141:3215-3223

33.

Garcia C, Lutz NW, Confort-Gouny S, Cozzone PJ, Armand M, Bernard M. 2012; Phospholipid fingerprints of milk from different mammalians determined by 31P NMR: Towards specific interest in human health. Food Chem. 135:1777-1783

34.

Giuffrida F, Elmelegy IM, Thakkar SK, Marmet C, Destaillats F. 2014; Longitudinal evolution of the concentration of gangliosides GM3 and GD3 in human milk. Lipids. 49:997-1004

35.

Golding M, Wooster TJ. 2010; The influence of emulsion structure and stability on lipid digestion. Curr Opin Colloid Interface Sci. 15:90-101

36.

Gómez-Cortés P, Juárez M, de la Fuente MA. 2018; Milk fatty acids and potential health benefits: An updated vision. Trends Food Sci Technol. 81:1-9

37.

Graves ELF, Beaulieu AD, Drackley JK. 2007; Factors affecting the concentration of sphingomyelin in bovine milk. J Dairy Sci. 90:706-715

38.

Guo Q, Bellissimo N, Rousseau D. 2017; The physical state of emulsified edible oil modulates its in vitro digestion. J Agric Food Chem. 65:9120-9127

39.

Gurnida DA, Rowan AM, Idjradinata P, Muchtadi D, Sekarwana N. 2012; Association of complex lipids containing gangliosides with cognitive development of 6-month-old infants. Early Hum Dev. 88:595-601

40.

Hageman JHJ, Keijer J, Dalsgaard TK, Zeper LW, Carrière F, Feitsma AL, Nieuwenhuizen AG. 2019; Free fatty acid release from vegetable and bovine milk fat-based infant formulas and human milk during two-phase in vitro digestion. Food Funct. 10:2102-2113

41.

Han L, Du M, Ren F, Mao X. 2021; Milk polar lipids supplementation to obese rats during pregnancy and lactation benefited skeletal outcomes of male offspring. Mol Nutr Food Res. 65:2001208

42.

He X, McClorry S, Hernell O, Lönnerdal B, Slupsky CM. 2020; Digestion of human milk fat in healthy infants. Nutr Res. 83:15-29

43.

He X, Parenti M, Grip T, Lönnerdal B, Timby N, Domellöf M, Hernell O, 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. Sci Rep. 9:11589

44.

Hernell O, Bläckberg L. 1994; Human milk bile salt-stimulated lipase: Functional and molecular aspects. J Pediatr. 125:S56-S61

45.

Hjerpsted J, Leedo E, Tholstrup T. 2011; Cheese intake in large amounts lowers LDL-cholesterol concentrations compared with butter intake of equal fat content. Am J Clin Nutr. 94:1479-1484

46.

Innis SM. 2011; Dietary triacylglycerol structure and its role in infant nutrition. Adv Nutr. 2:275-283

47.

Innis SM, Dyer R, Nelson CM. 1994; Evidence that palmitic acid is absorbed as sn-2 monoacylglycerol from human milk by breast-fed infants. Lipids. 29:541-545

48.

Jensen RG, Ferris AM, Lammi-Keefe CJ, Henderson RA. 1990; Lipids of bovine and human milks: A comparison. J Dairy Sci. 73:223-240

49.

Johnson K, Ross L, Miller R, Xiao X, Lowe ME. 2013; Pancreatic lipase-related protein 2 digests fats in human milk and formula in concert with gastric lipase and carboxyl ester lipase. Pediatr Res. 74:127-132

50.

Keenan TW, Heid HW, Stadler J, Jarasch ED, Franke WW. 1982; Tight attachment of fatty acids to proteins associated with milk lipid globule membrane. Eur J Cell Biol. 26:270-276.

51.

Koletzko B. 2016; Human milk lipids. Annu Nutr Metab. 69(Suppl)2:28-40

52.

Kosmerl E, Rocha-Mendoza D, Ortega-Anaya J, Jiménez-Flores R, García-Cano I. 2021; Improving human health with milk fat globule membrane, lactic acid bacteria, and bifidobacteria. Microorganisms. 9:341

53.

Lamothe S, Rémillard N, Tremblay J, Britten M. 2017; Influence of dairy matrices on nutrient release in a simulated gastrointestinal environment. Food Res Int. 92:138-146

54.

Le Huёrou-Luron I, Bouzerzour K, Ferret-Bernard S, Ménard O, Le Normand L, Perrier C, Le Bourgot C, Jardin J, Bourlieu C, Carton T, Le Ruyet P, Cuinet I, Bonhomme C, Dupont D. 2018; A mixture of milk and vegetable lipids in infant formula changes gut digestion, mucosal immunity and microbiota composition in neonatal piglets. Eur J Nutr. 57:463-476

55.

Lecomte M, Bourlieu C, Meugnier E, Penhoat A, Cheillan D, Pineau G, Loizon E, Trauchessec M, Claude M, Ménard O, Géloёn A, Laugerette F, Michalski MC. 2015; Milk polar lipids affect in vitro digestive lipolysis and postprandial lipid metabolism in mice. J Nutr. 145:1770-1777

56.

Lee H, German JB, Kjelden R, Lebrilla CB, Barile D. 2013; Quantitative analysis of gangliosides in bovine milk and colostrum-based dairy products by ultrahigh performance liquid chromatography-tandem mass spectrometry. J Agric Food Chem. 61:9689-9696

57.

Lee H, Slupsky CM, Heckmann AB, Christensen B, Peng Y, Li X, Hernell O, Lönnerdal B, Li Z. 2021; Milk fat globule membrane as a modulator of infant metabolsim and gut microbiota: A formula supplement narrowing the metabolic differences between breastfed and formula-fed infants. Mol Nutr Food Res. 65:2000603

58.

Levy M, Thaiss CA, Zeevi D, Dohnalová L, Zilberman-Schapira G, Mahdi JA, David E, Savidor A, Korem T, Herzig Y, Pevsner-Fischer M, Shapiro H, Christ A, Harmelin A, Halpern Z, Latz E, Flavell RA, Amit I, Segal E, Elinav E. 2015; Microbiota-modulated metabolites shape the intestinal microenvironment by regulating NLRP6 inflammasome signaling. Cell. 163:1428-1443

59.

Li F, Wu SS, Berseth CL, Harris CL, Richards JD, Wampler JL, Zhuang W, Cleghorn G, Rudolph CD, Liu B, Shaddy DJ, Colombo J. 2019; Improved neurodevelopmental outcomes associated with bovine milk fat globule membrane and lactoferrin in infant formula: A randomized, controlled trial. J Pediatr. 215:24-31.e8

60.

Li J, Wang X, Zhang T, Wang C, Huang Z, Luo X, Deng Y. 2015; A review on phospholipids and their main applications in drug delivery systems. Asian J Pharm Sci. 10:81-98

61.

Li T, Gao J, Du M, Mao X. 2018a; Milk fat globule membrane supplementation modulates the gut microbiota and attenuates metabolic endotoxemia in high-fat diet-fed mice. J Funct Foods. 47:56-65

62.

Li T, Gao J, Du M, Song J, Mao X. 2018b; Milk fat globule membrane attenuates high-fat diet-induced obesity by inhibiting adipogenesis and increasing uncoupling protein 1 expression in white adipose tissue of mice. Nutrients. 10:331

63.

Li T, Gong H, Yuan Q, Du M, Ren F, Mao X. 2020; Supplementation of polar lipids-enriched milk fat globule membrane in high-fat diet-fed rats during pregnancy and lactation promotes brown/beige adipocyte development and prevents obesity in male offspring. FASEB J. 34:4619-4634

64.

Li Y, Mu H, Andersen JET, Xu X, Meyer O, Ørngreen A. 2010; New human milk fat substitutes from butterfat to improve fat absorption. Food Res Int. 43:739-744

65.

Liang L, Qi C, Wang X, Jin Q, McClements DJ. 2017; Influence of homogenization and thermal processing on the gastrointestinal fate of bovine milk fat: In vitro digestion study. J Agric Food Chem. 65:11109-11117

66.

Liang L, Zhang X, Wang X, Jin Q, McClements DJ. 2018; Influence of dairy emulsifier type and lipid droplet size on gastrointestinal fate of model emulsions: In vitro digestion study. J Agric Food Chem. 66:9761-9769

67.

Lindquist S, Hernell O. 2010; Lipid digestion and absorption in early life: An update. Curr Opin Clin Nutr Metab Care. 13:314-320

68.

Liu L, Pan Y, Zhang X, Zhang Y, Li X. 2021; Effect of particle size and interface composition on the lipid digestion of droplets covered with membrane phospholipids. J Agric Food Chem. 69:159-169

69.

Lönnerdal B. 2014; Infant formula and infant nutrition: Bioactive proteins of human milk and implications for composition of infant formulas. Am J Clin Nutr. 99:712S-717S

70.

Lopez C, Cauty C, Guyomarc’h F. 2015; Organization of lipids in milks, infant milk formulas and various dairy products: Role of technological processes and potential impacts. Dairy Sci Technol. 95:863-893

71.

Lopez C, Cauty C, Guyomarc’h F. 2019; Unraveling the complexity of milk fat globules to tailor bioinspired emulsions providing health benefits: The key role played by the biological membrane. Eur J Lipid Sci Technol. 121:1800201

72.

Lopez C, Cauty C, Rousseau F, Blot M, Margolis A, Famelart MH. 2017; Lipid droplets coated with milk fat globule membrane fragments: Microstructure and functional properties as a function of pH. Food Res Int. 91:26-37

73.

Lopez C, 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 Surf B Biointerfaces. 83:29-41

74.

Lordan R, Tsoupras A, Zabetakis I. 2017; Phospholipids of animal and marine origin: Structure, function, and anti-inflammatory properties. Molecules. 22:1964

75.

Lu N, Wang J, Chen Z, Zhang X, Chen C, Wang S. 2021; The effect of adding phospholipids before homogenization on the properties of milk fat globules. LWT-Food Sci Technol. 146:111659

76.

Luo J, Huang Z, Liu H, Zhang Y, Ren F. 2018; Yak milk fat globules from the Qinghai-Tibetan Plateau: Membrane lipid composition and morphological properties. Food Chem. 245:731-737

77.

Luo J, Liu L, Liu T, Shen Q, Liu C, Zhou H, Ren F. 2020; Simulated in vitro infant gastrointestinal digestion of yak milk fat globules: A comparison with cow milk fat globules. Food Chem. 314:126160

78.

Martinez-Guryn K, Hubert N, Frazier K, Urlass S, Musch MW, Ojeda P, Pierre JF, Miyoshi J, Sontag TJ, Cham CM, Reardon CA, Leone V, Chang EB. 2018; Small intestine microbiota regulate host digestive and absorptive adaptive responses to dietary lipids. Cell Host Microbe. 23:458-469.e5

79.

Mather IH. 2000; A review and proposed nomenclature for major proteins of the milk-fat globule membrane. J Dairy Sci. 83:203-247

80.

Mathiassen JH, Nejrup RG, Frøkiær H, Nilsson Å, Ohlsson L, Hellgren LI. 2015; Emulsifying triglycerides with dairy phospholipids instead of soy lecithin modulates gut lipase activity. Eur J Lipid Sci Technol. 117:1522-1539

81.

Ménard O, Ahmad S, Rousseau F, Briard-Bion V, Gaucheron F, Lopez C. 2010; Buffalo vs. cow milk fat globules: Size distribution, zeta-potential, compositions in total fatty acids and in polar lipids from the milk fat globule membrane. Food Chem. 120:544-551

82.

Michalski MC, Soares AF, Lopez C, Leconte N, Briard V, Geloen A. 2006; The supramolecular structure of milk fat influences plasma triacylglycerols and fatty acid profile in the rat. Eur J Nutr. 45:215-224

83.

Miklavcic JJ, Schnabl KL, Mazurak VC, Thomson ABR, Clandinin MT. 2012; Dietary ganglioside reduces proinflammatory signaling in the intestine. J Nutr Metab. 2012:280286

84.

Mun S, Decker EA, McClements DJ. 2007; Influence of emulsifier type on in vitro digestibility of lipid droplets by pancreatic lipase. Food Res Int. 40:770-781

85.

Narita T, Naganuma T, Sase Y, Kihara A. 2016; Long-chain bases of sphingolipids are transported into cells via the acyl-CoA synthetases. Sci Rep. 6:25469

86.

Nguyen TTP, Bhandari B, Cichero J, Prakash S. 2018; In vitro lipolysis of dairy and soy based infant formula. Food Res Int. 106:696-705

87.

Nilsson Å. 2016; Role of sphingolipids in infant gut health and immunity. J Pediatr. 173:S53-S59

88.

Norris GH, Porter CM, Jiang C, Blesso CN. 2017; Dietary milk sphingomyelin reduces systemic inflammation in diet-induced obese mice and inhibits LPS activity in macrophages. Beverages. 3:37

89.

Oborina EM, Yappert MC. 2003; Effect of sphingomyelin versus dipalmitoylphosphatidylcholine on the extent of lipid oxidation. Chem Phys Lipids. 123:223-232

90.

O’Mahony SM, McVey Neufeld KA, Waworuntu RV, Pusceddu MM, Manurung S, Murphy K, Strain C, Laguna MC, Peterson VL, Stanton C, Berg BM, Dinan TG, Cryan JF. 2020; The enduring effects of early-life stress on the microbiota-gut-brain axis are buffered by dietary supplementation with milk fat globule membrane and a prebiotic blend. Eur J Neurosci. 51:1042-1058

91.

O’Muircheartaigh J, Dean DC III, Ginestet CE, Walker L, Waskiewicz N, Lehman K, Dirks H, Piryatinsky I, Deoni SCL. 2014; White matter development and early cognition in babies and toddlers. Hum Brain Mapp. 35:4475-4487

92.

Pafumi Y, Lairon D, de la Porte PL, Juhel C, Storch J, Hamosh M, Armand M. 2002; Mechanisms of inhibition of triacylglycerol hydrolysis by human gastric lipase. J Biol Chem. 277:28070-28079

93.

Pan Y, Liu L, Tian S, Li X, Hussain M, Li C, Zhang L, Zhang Q, Leng Y, Jiang S, Liang S. 2022; Comparative analysis of interfacial composition and structure of fat globules in human milk and infant formulas. Food Hydrocoll. 124:107290

94.

Pham AC, Peng KY, Salim M, Ramirez G, Hawley A, Clulow AJ, Boyd BJ. 2020; Correlating digestion-driven self-assembly in milk and infant formulas with changes in lipid composition. ACS Appl Bio Mater. 3:3087-3098

95.

Puente R, Garcia-Pardo LA, Rueda R, Gil A, Hueso P. 1994; Changes in ganglioside and sialic acid contents of goat milk during lactation. J Dairy Sci. 77:39-44

96.

Qu X, Hu H, Wang Y, Cao C, Li H, Liu X, Yu J. 2019; Proteomics analysis of milk fat globule membrane enriched materials derived from by-products during different stages of milk-fat processing. LWT-Food Sci Technol. 116:108531

97.

Rajendran L, Simons K. 2005; Lipid rafts and membrane dynamics. J Cell Sci. 118:1099-1102

98.

Reinhardt TA, Lippolis JD. 2006; Bovine milk fat globule membrane proteome. J Dairy Res. 73:406-416

99.

Richard C, Lewis ED, Goruk S, Wadge E, Curtis JM, Jacobs RL, Field CJ. 2017; Feeding a mixture of choline forms to lactating dams improves the development of the immune system in Sprague-Dawley rat offspring. Nutrients. 9:567

100.

Rohrhofer J, Zwirzitz B, Selberherr E, Untersmayr E. 2021; The impact of dietary sphingolipids on intestinal microbiota and gastrointestinal immune homeostasis. Front Immunol. 12:635704

101.

Rombaut R, Dewettinck K, Van Camp J. 2007; Phospho- and sphingolipid content of selected dairy products as determined by HPLC coupled to an evaporative light scattering detector (HPLC–ELSD). J Food Compos Anal. 20:308-312

102.

Rosqvist F, Smedman A, Lindmark-Månsson H, Paulsson M, Petrus P, Straniero S, Rudling M, Dahlman I, Risérus U. 2015; Potential role of milk fat globule membrane in modulating plasma lipoproteins, gene expression, and cholesterol metabolism in humans: A randomized study. Am J Clin Nutr. 102:20-30

103.

Rueda R, Sabatel JL, Maldonado J, Molina-Font JA, Gil A. 1998; Addition of gangliosides to an adapted milk formula modifies levels of fecal Escherichia coli in preterm newborn infants. J Pediatr. 133:90-94

104.

Sánchez C, Franco L, Regal P, Lamas A, Cepeda A, Fente C. 2021; Breast milk: A source of functional compounds with potential application in nutrition and therapy. Nutrients. 13:1026

105.

Sánchez-Juanes F, Alonso JM, Zancada L, Hueso P. 2009; Distribution and fatty acid content of phospholipids from bovine milk and bovine milk fat globule membranes. Int Dairy J. 19:273-278

106.

Schneider N, Hauser J, Oliveira M, Cazaubon E, Mottaz SC, O'Neill BV, Steiner P, Deoni SCL. 2019; Sphingomyelin in brain and cognitive development: Preliminary data. Eneuro 6:ENEURO. :0421-18.2019

107.

Simons K, Vaz WLC. 2004; Model systems, lipid rafts, and cell membranes. Annu Rev Biophys Biomol Struct. 33:269-295

108.

Singh H. 2019; Symposium review: Fat globules in milk and their structural modifications during gastrointestinal digestion. J Dairy Sci. 102:2749-2759

109.

Singh H, Gallier S. 2017; Nature's complex emulsion: The fat globules of milk. Food Hydrocoll. 68:81-89

110.

Slotte JP. 2016; The importance of hydrogen bonding in sphingomyelin's membrane interactions with co-lipids. Biochim Biophys Acta. 1858:304-310

111.

Smoczyński M, Staniewski B, Kiełczewska K. 2012; Composition and structure of the bovine milk fat globule membrane—some nutritional and technological implications. Food Rev Int. 28:188-202

112.

Subra C, Grand D, Laulagnier K, Stella A, Lambeau G, Paillasse M, De Medina P, Monsarrat B, Perret B, Silvente-Poirot S, Poirot M, Record M. 2010; Exosomes account for vesicle-mediated transcellular transport of activatable phospholipases and prostaglandins. J Lipid Res. 51:2105-2120

113.

Tanaka K, Hosozawa M, Kudo N, Yoshikawa N, Hisata K, Shoji H, Shinohara K, 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 Dev. 35:45-52

114.

Tomarelli RM, Meyer BJ, Weaber JR, Bernhart FW. 1968; Effect of positional distribution on the absorption of the fatty acids of human milk and infant formulas. J Nutr. 95:583-590

115.

Tomé-Carneiro J, Carmen Crespo M, Burgos-Ramos E, Tomas-Zapico C, García-Serrano A, Castro-Gómez P, Venero C, Pereda-Pérez I, Baliyan S, Valencia A, Fontecha J, Dávalos A, Visioli F. 2018; Buttermilk and krill oil phospholipids improve hippocampal insulin resistance and synaptic signaling in aged rats. Mol Neurobiol. 55:7285-7296

116.

Tunick MH, Ren DX, Van Hekken DL, Bonnaillie L, Paul M, Kwoczak R, Tomasula PM. 2016; Effect of heat and homogenization on in vitro digestion of milk. J Dairy Sci. 99:4124-4139

117.

Turgeon SL, Brisson G. 2020; Symposium review: The dairy matrix—Bioaccessibility and bioavailability of nutrients and physiological effects. J Dairy Sci. 103:6727-6736

118.

Ubbink J, Burbidge A, Mezzenga R. 2008; Food structure and functionality: A soft matter perspective. Soft Matter. 4:1569-1581

119.

Xiao X, Mukherjee A, Ross LE, Lowe ME. 2011; Pancreatic lipase-related protein-2 (PLRP2) can contribute to dietary fat digestion in human newborns. J Biol Chem. 286:26353-26363

120.

Ye A, Cui J, Singh H. 2011; Proteolysis of milk fat globule membrane proteins during in vitro gastric digestion of milk. J Dairy Sci. 94:2762-2770

121.

Yu Z, Li Y, Niu Y, Tang Q, Wu J. 2021; Milk fat globule membrane enhances colonic–mucus-barrier function in a rat model of short-bowel syndrome. JPEN J Parenter Enteral Nutr. 45:916-925

122.

Yuan T, Zhu X, Mu G, Qian F, Xiong H. 2020; Effects of chemical composition and microstructure in human milk and infant formulas on lipid digestion. J Agric Food Chem. 68:5462-5470

123.

Zhang H, Zhao H, Zhang Y, Shen Y, Su H, Jin J, Jin Q, Wang X. 2018; Characterization of positional distribution of fatty acids and triacylglycerol molecular compositions of marine fish oils rich in omega-3 polyunsaturated fatty acids. BioMed Res Int. 2018:3529682

124.

Zhang L, Chichlowski M, Gross G, Holle MJ, Lbarra-Sánchez LA, Wang S, Miller MJ. 2020; Milk fat globule membrane protects Lactobacillus rhamnosus GG from bile stress by regulating exopolysaccharide production and biofilm formation. J Agric Food Chem. 68:6646-6655

125.

Zhang Q, Ye L, Xin F, Zhou J, Cao B, Dong Y, Qian L. 2021; Milk fat globule membrane supplementation during suckling ameliorates maternal high fat diet–induced hepatic steatosis in adult male offspring of mice. J Nutr. 151:1487-1496

126.

Zhao J, Yi W, Liu B, Dai Y, Jiang T, Chen S, Wang J, Feng B, Qiao W, Liu Y, Zhou H, He J, Hou J, Chen L. 2022; MFGM components promote gut Bifidobacterium growth in infant and in vitro. Eur J Nutr. 61:277-288

127.

Zhao L, Du M, Mao X. 2019; Change in interfacial properties of milk fat globules by homogenization and thermal processing plays a key role in their in vitro gastrointestinal digestion. Food Hydrocoll. 96:331-342

128.

Zheng H, Jiménez-Flores R, Everett DW. 2013; Bovine milk fat globule membrane proteins are affected by centrifugal washing processes. J Agric Food Chem. 61:8403-8411

129.

Zheng L, Fleith M, Giuffrida F, O'Neill BV, Schneider N. 2019; Dietary polar lipids and cognitive development: A narrative review. Adv Nutr. 10:1163-1176

130.

Zou X, Huang J, Jin Q, Guo Z, Liu Y, Cheong L, Xu X, Wang X. 2013; Lipid composition analysis of milk fats from different mammalian species: Potential for use as human milk fat substitutes. J Agric Food Chem. 61:7070-7080