Next Article in Journal
Is the Behavioural Gender Gap Decreasing? Evidence from Food Consumption in Swiss Single-Person Households
Previous Article in Journal
Tea’s Characteristic Components Eliminate Acrylamide in the Maillard Model System
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Cow’s Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies

by
Emad Yuzbashian
1,
Emily Berg
2,†,
Stepheny C. de Campos Zani
2,† and
Catherine B. Chan
1,2,*
1
Department of Agriculture, Food and Nutritional Science, University of Alberta, Edmonton, AB T6G 2P5, Canada
2
Department of Physiology, University of Alberta, Edmonton, AB T6G 2H7, Canada
*
Author to whom correspondence should be addressed.
These authors contributed equally to this work.
Foods 2024, 13(17), 2837; https://doi.org/10.3390/foods13172837
Submission received: 3 July 2024 / Revised: 26 August 2024 / Accepted: 31 August 2024 / Published: 6 September 2024
(This article belongs to the Section Dairy)

Abstract

:
Obesity disrupts glucose metabolism, leading to insulin resistance (IR) and cardiometabolic diseases. Consumption of cow’s milk and other dairy products may influence glucose metabolism. Within the complex matrix of cow’s milk, various carbohydrates, lipids, and peptides act as bioactive molecules to alter human metabolism. Here, we summarize data from human studies and rodent experiments illustrating how these bioactive molecules regulate insulin and glucose homeostasis, supplemented with in vitro studies of the mechanisms behind their effects. Bioactive carbohydrates, including lactose, galactose, and oligosaccharides, generally reduce hyperglycemia, possibly by preventing gut microbiota dysbiosis. Milk-derived lipids of the milk fat globular membrane improve activation of insulin signaling pathways in animal trials but seem to have little impact on glycemia in human studies. However, other lipids produced by ruminants, including polar lipids, odd-chain, trans-, and branched-chain fatty acids, produce neutral or contradictory effects on glucose metabolism. Bioactive peptides derived from whey and casein may exert their effects both directly through their insulinotropic effects or renin-angiotensin-aldosterone system inhibition and indirectly by the regulation of incretin hormones. Overall, the results bolster many observational studies in humans and suggest that cow’s milk intake reduces the risk of, and can perhaps be used in treating, metabolic disorders. However, the mechanisms of action for most bioactive compounds in milk are still largely undiscovered.

Graphical Abstract

1. Introduction

The regulation of glucose homeostasis within the body depends mainly on the tight regulation of insulin secretion from pancreatic β-cells and its actions on peripheral tissues, including muscle, adipose tissue, and liver [1]. However, impaired secretion of insulin or the existence of insulin resistance (IR) in the peripheral tissues results in chronic hyperglycemia, which leads to the manifestation of impaired glucose homeostasis and the development of metabolic diseases, such as type 2 diabetes mellitus (T2DM). The progression of T2DM and its morbidities causes a significant societal and individual financial burden and is a leading cause of premature mortality and reduced life expectancy [2]. The prevalence of T2DM has risen globally in the last two decades, largely due to obesity, which is attributed to overnutrition and lack of physical activity [3]. Adherence to a healthy diet plan is one of the primary strategies for managing obesity and normalizing glucose homeostasis. Of the dietary components currently under investigation for reducing the risk for T2DM, dairy products have received considerable attention as inducing improvements in insulin sensitivity and a favorable metabolic profile [4,5,6].
Milk is a highly consumed dairy product, but whether it promotes glucose homeostasis or contributes to the risk of T2DM remains a debated topic. A meta-analysis of 35 observational studies including nearly 400,000 participants recently concluded that the highest consumption of both total dairy and milk was associated with a decreased risk of incident metabolic syndrome (comprising pre-diabetes and T2DM) of 20% and 17%, respectively [7]. In a meta-analysis of cohort studies, each 200 g/day increment in consumption of total dairy and milk was associated with a 25% and 7% reduced likelihood of becoming overweight and obese, respectively. A neutral association was found between milk consumption and the risk of T2DM [8]. However, some individual studies revealed adverse [9,10] or null [11,12] effects that may depend on the baseline glycemic state of individuals included in the study. Overall, these epidemiological studies suggest that milk has functional properties that contribute to the regulation of glucose homeostasis, such as increasing insulin sensitivity, enhancing insulin-secreting capacity, or improving glucose metabolism within the liver [13,14,15] but are limited by study designs that cannot determine causation and have inherent residual confounding.
Furthermore, the idea that milk is beneficial is challenged by evidence that milk has strong insulin secretory activity [16], which may cause acute hyperinsulinemia and exacerbate IR [17,18]. On the other hand, by acutely raising insulin, euglycemia may be re-established more rapidly to reduce the demand on the pancreas for insulin [19]. It is essential to consider milk’s complexity, as it contains numerous bioactive compounds that might influence glucose regulation in different directions individually; thus, their resultant effects could be beneficial. Well-designed clinical trials are needed to help resolve these discrepant results, some of which are described in the following sections.
Cow’s milk is a mixture that predominantly consists of water (about 87%). Fats, carbohydrates, protein, vitamins and minerals constitute the remaining 13%, thus making the milk nutritionally dense [20]. Notably, milk contains many bioactive molecules that may elicit metabolic impacts beyond its basic nutritional value [21,22]. The macronutrients are a rich source of diverse bioactive molecules, such as lactose, galactose, and milk oligosaccharides from carbohydrates, the milk fat globule membrane (MFGM), even-, odd- and branched-chain fatty acids, and milk-specific trans fatty acids, along with protein hydrolysates and bioactive peptides derived from milk proteins [23]. Milk also contains other ingredients with bioactivities, including the milk microbiota and microRNAs. As the focus of this review is on the bioactive molecules derived from macronutrients, we refer the reader to several recent reviews covering the milk microbiota and microRNAs [24,25,26,27]. The first objective of the present review is to outline the effects of bioactive compounds found in milk carbohydrates, lipids, and proteins in relevant human clinical trials. The second objective is to discuss how milk bioactive molecules improve glucose regulation using data from animal and cell culture models. This narrative review synthesizes relevant literature from PubMed, Scopus, and Google Scholar, using the appropriate terms and keywords, focusing on human studies, animal experiments, and in vitro studies up to 2024 related to cow’s milk bioactive molecules and glucose homeostasis.

2. Carbohydrates

Lactose is the most abundant carbohydrate in cow’s milk but varies in amount due to many factors, for example the cow’s diet and lactation stage [28]. Cow’s milk also contains small amounts of lactose-derived products such as lactulose, lactitol, lactobionic acid, and galactooligosaccharides [29,30]. Milk has a low glycemic index (GI) and low glycemic load (GL), meaning that its consumption yields a lower blood glucose response compared with an equivalent dose of glucose, which is attributed to slower hydrolysis and absorption of lactose [31,32]. The GI of whole milk varies from 30 to 46 (out of 100), while low-fat or skimmed milk has a GI ranging from 20 to 34 [32]. The commercially available non-sweetened types of milk have a GL ranging between 2 and 5, which classifies them as a low GL food [32]. It should be noted that although milk is considered a low GI and GL food, the rise in blood insulin concentration during the 2 h after consumption, called the insulinemic index, is substantially higher than expected based on the GI of milk [33,34], indicating the presence of an insulinotropic ingredient, which is attributed to amino acids and lipids rather than the carbohydrates in milk products [35]. Persistent hyperinsulinemia potentially results in decreased insulin sensitivity [17], but it is also possible that the hyperinsulinemic response to milk has a positive or even protective impact on blood glucose regulation, especially in those with T2D [36,37]. Overall, the GI of milk means it could be a favorable component of a diet designed to manage blood glucose. To further explain the beneficial effect of milk carbohydrates on glucose metabolism, we will discuss the characteristics of lactose, galactose, and oligosaccharides of milk and their effects on glucose homeostasis in human trials (Table 1) and animal studies (Table 2).

2.1. Lactose

Lactose is a disaccharide of galactose and glucose (β-d-galactopyranosyl-(1→4)-d-glucose). In humans, the capacity to digest milk lactose is provided by lactase-phlorizin hydrolase (LPH aka lactase), a type of β-galactosidase, which is bound to the mucosal membrane of the small intestine. It hydrolyses lactose into glucose and galactose, which are then absorbed and carried to the liver by the portal vein [118]. This enzyme is encoded by the lactase (LCT) gene, located on chromosome 2q21; the activity of this gene is highest in infants. However, after weaning, decreasing abundance of this enzyme causes a gradual reduction in LPH activity in most humans, leading to a state of lactase non-persistence. Consequently, most adults are not able to digest lactose and may experience abdominal discomfort upon its consumption. Those who can digest milk lactose throughout life are considered lactase-persistent [119]. Substantial evidence supports that lactase persistence is influenced by at least five single nucleotide polymorphisms (SNPs) in a regulatory region called MCM6 (minichromosome maintenance complex component 6) upstream of the LCT gene [120]. Lactase persistence is prevalent among individuals of northern and Western European descent, as well as in many African, Middle Eastern, and southern Asian pastoralist communities. However, it is seldom persistent in other regions worldwide [121,122].
In contrast to sucrose, lactose intake is not related to the risk of T2DM development in human observational studies [123,124]. Human clinical trials show that lactose or lactose-derived prebiotics (e.g., lactulose) generally have beneficial or neutral effects on IR-related outcomes [38,39,40,125,126]. Lactose supplementation decreases body weight (BW) and fat accumulation in both healthy rats [64,67] and those with diet-induced obesity (DIO) [65]. These changes coincide with reduced circulating glucose, insulin, and leptin, indicating that lactose regulates metabolic processes [64,66,67].
Evidence supports that impaired glucose metabolism is, to some extent, triggered by elevated circulating bacterial endotoxins due to changes in intestinal permeability [127]. The presence of Bifidobacteria in the gastrointestinal tract is correlated with reduced plasma and intestinal endotoxin levels [128], which can ultimately reduce the detrimental effect of high-fat feeding elicited by endotoxins on glucose metabolism [129]. Furthermore, Bifidobacteria abundance correlates with reduced diabetes symptoms, including improved glucose tolerance [128]. This concept has been discussed in previous reviews [130,131]. In addition to being hydrolyzed and absorbed, lactose acts as a prebiotic [132], stimulating the growth and activity of beneficial bacteria in the gut and contributing to improved gut health [133,134,135]. As indicated earlier, many adults have low expression of LCT and difficulty digesting lactose, but even in lactase-persistent individuals, 5–10% of ingested lactose may escape digestion and pass through the lower intestine, producing digestion symptoms such as flatulence and diarrhea, depending on the degree of intolerance [136,137]. However, frequent lactose consumption promotes the growth of lactose-digesting bacteria, such as Bifidobacteria, as shown in a swine model [138]. These bacteria possess β-galactosidases that hydrolyze lactose into glucose and galactose, which are further converted into short-chain fatty acids, which have beneficial metabolic effects in obesity and other metabolic conditions [139,140,141]. Notably, the enrichment of Bifidobacteria has the added benefit of fermenting lactose without gas production, alleviating gut dysbiosis thus reducing intolerance symptoms [142].
Lactose significantly elevates calcium and magnesium absorption from milk and other foods because its fermentation lowers the pH in the large intestine, which increases the solubility and passive absorption of these minerals [35]. Calcium and magnesium play a role in the regulation of body weight and fat mass by reducing the de novo production of fatty acids, facilitating hydrolysis of stored fats, and creating insoluble complexes with dietary fats in the gastrointestinal tract, which reduce fat absorption [43], all leading to improved glucose homeostasis [35,44,45,46].

2.2. Galactose

Galactose, once hydrolyzed from lactose, is rapidly absorbed as a monosaccharide. The total galactose content per 100 g of milk, considering both the free galactose and that derived from lactose, ranges from 2.37–2.52 g [143]. Data on the average galactose intake in human populations are limited, with estimates ranging from 1.5–3.6 g per day in an Iranian female population [144] to below 0.5 g per day in a sample of Japanese women [145].
In human trials, galactose has drawn attention as a low-GI sugar because it causes very modest rises in plasma glucose and insulin concentrations in normal and T2DM participants [44,45], and also increases the secretion of incretins, glucagon-like peptide-1 (GLP-1) and glucose-independent insulinotropic peptide (GIP), which are produced in the small intestine [43,146], and promote satiety [46,147]. Both short- and long-term randomized controlled trials (RCT) in women who consumed galactose- rather than glucose-sweetened drinks report increased fat mobilization and oxidation [41], consistent with its smaller insulinemic effect [42,45].
Recent rodent studies shed light on the potential mechanism by which galactose improves glucose homeostasis. Providing galactose at 15% of dry matter for 9 weeks in non-diabetic, healthy rats increases hepatic insulin sensitivity and hepatic glycogen storage in the fed state, improves insulin capacity to decrease glycogen synthase phosphorylation, and reduces Firmicutes bacteria in the gut. Neither basal glucose nor insulin are modified by the dietary intervention [69]. Partial substitution of glucose with galactose in high-fat diet (HFD)-challenged female mice reduces BW, obesity, homeostatic model assessment of IR (HOMA-IR), and induces genes involved in insulin signaling in white adipose tissue depots, which could explain improved insulin sensitivity [68].
Galactose may also benefit liver carbohydrate metabolism. Nearly 90% of absorbed galactose is retained in the liver as galactose-1-phosphate (Gal-1-P), which serves as a substrate for glycogen production [148]. It can also be converted to alternative substrates (glucose, lactate, or fatty acids (FA)) that extrahepatic cells can easily utilize. Due to the delayed release of galactose into the peripheral circulation as glucose, it could be a preferred energy source compared with other monosaccharides because of its low GI and low insulinemic response [149].
Another mechanism by which galactose improves glucose homeostasis in rodents, as it does in humans, is by stimulating GLP-1 and GIP secretion into the circulation after a meal, which promotes insulin secretion to help maintain glucose homeostasis [70,146]. In addition, chronic galactose administration also restores a normal GIP and GLP-1 physiology in diabetes models [145,150]. Well-described IR- and T2DM-mitigating effects of GLP-1 are reported elsewhere [151,152]. The antioxidant and anti-inflammatory properties of galactose may also account for improvement in IR and obesity-related outcomes. Oral galactose supplementation reduces oxidative stress and inflammation markers in the liver of low-dose streptozotocin (STZ)-induced diabetes in rats [70].

2.3. Oligosaccharides

Cow’s milk oligosaccharides (CMOs) are complex carbohydrates consisting of 3 to 20 monosaccharides that are not digested but rather serve as prebiotics. Cow’s colostrum contains 1–2 g/L of CMOs; however, typical cow’s milk has significantly less CMOs (about 0.01–0.05 g/L). More than 100 different oligosaccharides are present in cow’s milk [153,154]. These include neutral oligosaccharides composed of glucose, galactose, and N-acetylglucosamine. Acidic oligosaccharides (sialyloligosaccharides) are attached to sialic and uronic acids. Lacto-N-biose, lacto-N-neotetraose (LNnT), lacto-N-triose (LNT), 3′-sialyllactose, 6′-sialyllactose, and disialyllactose are just a few of the oligosaccharides present in cow’s milk [21,155].
While human studies investigating CMO supplementation on glucose homeostasis are rare, animal studies provide promising results [71,72]. In DIO mice, CMOs improve glucose tolerance [72], reduce BW gain [71], decrease gut permeability [67,68] and systemic inflammation [72], and correct microbial dysbiosis [71,72]. Rats treated with HFD plus sialyloligosaccharides have improved insulin sensitivity and lower glycemic response in an oral glucose tolerance test (OGTT), along with higher adiponectin and lower leptin concentrations than the HFD controls [73]. Sialyloligosaccharides upregulate several genes involved in insulin signaling in both the liver and white adipose tissues, including Gck, Kcnj11, Mtor, Pi3k, and Prckz, while downregulating inflammatory markers such as Iκbkβ and Mapk1 in the liver [73]. A mixture of CMOs improves the health and growth of undernourished mice pups and piglets by influencing their gut bacteria and metabolism, suggesting that CMOs, via the gut microbiota, have a significant impact on the development and regulation of metabolic pathways [75]. CMOs including tetrasaccharides LNT and LNnT promote the growth of short-chain fatty acid-generating bacteria [156] such as Bifidobacterium, Lactobacillus, and Bacteroides [157,158]. Supplementation with CMOs increases the expression of butyrate-generating bacterial genes in Western diet-fed mouse models, and butyrate has anti-inflammatory effects in the liver and colon [159].

3. Lipids

The fat content of whole cow’s milk varies from about 3.0 to 6.0%, but is typically in the range of 3.5 to 4.7% and is comprised of more than 400 distinct FA, making milk the most complex and diverse dietary fat source. The composition of milk fat is influenced by the cow’s diet and the ruminal biohydrogenation process, as well as the breed, lactation stage, age of the cow, and the season and geographical location [160].
Milk fat consists of FA with carbon chain lengths ranging from 4 to 24. Palmitic acid (C16:0) and stearic acid (C18:0) make up the majority of saturated fatty acids (SFA) in milk fat, constituting around two-thirds of the total milk fat content [161]. Oleic acid (C18:1) is the predominant unsaturated FA. Milk fat also includes distinct FA produced by rumen microorganisms, such as trans isomers of octadecenoic acid ranging from 18:1 t4 to t16. Ruminant-derived milk and meat are the only sources of non-industrial trans-fatty acids (TFAs) in the human diet. Furthermore, the presence of branched-chain fatty acids (BCFAs) and odd-chain FAs, namely pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0), in the unique FA composition of milk fat is noteworthy. Approximately 14% of the FA found in milk fat is classified as unique dairy-derived FAs and knowledge of their bioactivity is emerging [162].
The major lipids in cow’s milk include monoglycerides, diglycerides, triglycerides (TG), free fatty acids (FFA), phospholipids (PL), glycolipids, and sterols [162]. Milk fat is packaged inside a tri-layer milk fat globule membrane (MFGM), consisting of proteins, cholesterol, and polar lipids, which emulsifies and stabilizes the milk fat. Epidemiological evidence suggests that consuming dairy fat is neutral or beneficial in preventing chronic diseases [163]. This section focuses on bioactive lipids identified in cow’s milk and their effects on glucose metabolism (Table 1 and Table 2).

3.1. Milk Fat Globule Membrane

MFGM contains both polar and neutral lipids and surrounds a TG core with an average diameter of 4  μm [164]. The polar lipids in MFGM included glycerophospholipids (e.g., phosphatidylcholine (PC)) and sphingolipids (e.g., sphingomyelin (SM)), which comprise about 1% of milk fat but about 25% of the components of MFGM. These polar lipids emulsify and stabilize TG within the aqueous phase of milk. Like other fats, milk TG undergoes hydrolysis by lipases to a mixture of FFA, mono- and di-glycerides in the intestinal lumen prior to its absorption by enterocytes, where it is repackaged into chylomicrons to be delivered to other metabolic tissues for subsequent uptake, storage or use [164,165,166,167]. At the same time, the PL of the MFGM, such as SM and PC, are also hydrolyzed by phospholipases. In addition, the polar lipids from the MFGM may become incorporated into enterocytes and influence gut physiology [168]. The industrialized processing of milk to remove fat and produce low-fat and fat-free dairy products leads to the loss of MFGM and a 40% decrease in total milk polar lipid. Both homogenization and pasteurization affect the rate of digestion, the size of MFGM, and its microstructure. Homogenized MFGM is digested more rapidly and releases more FA than native globules [169,170]. The intricate composition, segregation, and characteristics of MFGM have been thoroughly reviewed elsewhere [167].
Several RCTs show that MFGM positively impacts indicators of metabolic health and glucose homeostasis [47,48,49,171]. The inclusion of whipping cream enriched with intact MFGM in isoenergetic test meals high in saturated fat lowers the postprandial insulin response in a group of non-diabetic overweight and obese adults compared with the control meal [49]. Consistent with this finding, adding MFGM in palm oil reduces circulating insulin and glucose concentrations compared with palm oil alone [48]. In another RCT of isoenergetic diets, providing whipping cream (enriched with MFGM) for 8 weeks results in a slight increase in body mass index (BMI) among overweight adults compared with butter oil. However, there were no significant differences in fasting plasma glucose, insulin, and HOMA-IR between the groups [47]. In an RCT comparing MFGM-enriched milk beverage to a comparator beverage containing soy phospholipid and palm/coconut oil for 2 weeks, individuals with metabolic syndrome experienced no significant changes in fasting glucose, insulin, GLP-1, and HOMA-IR within or between the two treatment groups [50].
Multiple animal studies show consistent beneficial effects of MFGM feeding and illuminate potential mechanisms. Supplementation of MFGM for 8 weeks reduced glucose intolerance in a T2DM mouse model with the mechanism attributed to enhanced PI3K-AKT pathway and inhibited c-Jun N-terminal kinase (JNK) signaling in the insulin-resistant liver and skeletal muscle [80]. The JNK pathway plays an important role in insulin regulation because it inhibits AKT phosphorylation in skeletal muscle to accentuate IR [172,173]. Inhibition of JNK alleviates diabetes symptoms and improves insulin sensitivity in T2DM rats [174]. The further beneficial impact of MFGM on glucose regulation is elucidated in the early exposure of offspring to MFGM [79]. The administration of MFGM HFD-induced obese rats during pregnancy and lactation resulted in the amelioration of HFD-induced ectopic fat accumulation, in addition to an improvement in insulin resistance by inducing p-AKT and reducing p-IRS in adult offspring [78]. In another study, Ye et al. showed an improvement in glucose tolerance without significantly impacting the weight gain in C57BL/6 mice offspring exposed to maternal HFD when supplemented with MFGM [81]. They attributed this effect to the presence of beneficial bacteria in the gut.
Regarding body composition and weight gain in the presence of IR, MFGM treatment elicits promising results in animal studies. Obese, pregnant female rats fed MFGM have reduced BW, enhanced glucose tolerance and insulin sensitivity, and restored expression of genes involved in insulin signaling in multiple tissues [77]. MFGM-supplemented animals have reduced BW gain and obesity [77], possibly explained by upregulated thermogenic genes such as Ucp1 and Cidea to increase brown adipose activity and browning of white adipose tissue [76]. In cultured human HepG2 cells, MFGM exposure mitigates the accumulation of lipid droplets and maintains higher glucose uptake when compared to control cells, concomitant with an increased abundance of glucoregulatory enzymes such as glucose-6-phosphatase, glyceraldehyde-3-phosphate dehydrogenase, glycogen phosphorylase, and hexokinase, and consistent with the improvement in insulin signaling and glucose metabolism [175].
The presence of glycerophospholipids and sphingolipids in MFGM may mediate its effects on metabolism [14]. Subclasses of glycerophospholipids include PC, phosphatidylethanolamine, phosphatidylserine, and phosphatidylinositol. The sphingolipids include SM and glycosphingolipids. SM, PC, and phosphatidylethanolamine are the most abundant polar lipids in the MFGM, constituting approximately 30%, 30%, and 20% of milk polar lipids, respectively. Phosphatidylserine and phosphatidylinositol are less abundant, each making up less than 10% of milk polar lipids [176].
In a human trial, an RCT of 62 overweight or obese men demonstrated the impact of daily intake of 2 g of PL-enriched milk to elicit reduced waist circumference compared to unenriched milk fat for 8 weeks. However, 57 male participants receiving milk 3 g of PL-enriched milk for 7 weeks showed no significant differences in fasting glucose, insulin, or the insulin sensitivity index compared to those receiving 2.8 g soy PL [51].
Several research studies indicate that milk polar lipids improve metabolic diseases through regulation of the gut microbiota [52,77,83,84,88,176]. Adding 1.6% milk PL (0.38% SM) to an HFD resulted in a decrease in fecal Lactobacillus in C57BL/6 mice, whereas 1.1% milk PL (0.25% SM) increased Bifidobacterium compared to the control HFD group [82]. Akkermansia muciniphila increased in mice fed with milk PL, enhancing insulin sensitivity and protecting against inflammation caused by metabolic endotoxemia [177,178]. The source of SM may determine its ability to influence metabolism, as two studies using milk SM found no effects on BW, adiposity [87], blood glucose [86,87] or HOMA-IR [86] in mouse models. However, egg-derived SM did lower fasting glucose levels [86].
As mentioned previously, conditions such as obesity and IR are interconnected with gut health and the ability of the intestinal barrier to prevent the translocation of endotoxins that trigger inflammation; these actions are mediated via the Toll-like receptor (TLR)-4/nuclear factor (NF)-κB pathway [129,179,180]. In neonatal rodents, milk polar lipids derived from buttermilk enhance gut barrier function by reducing intestinal inflammation typically induced by the TLR-4-NF-κB pathway [181]. Additionally, milk polar lipids protect mice against impairments in gut barrier integrity caused by lipopolysaccharides [182]. In cell-based assays, PL- or ganglioside-containing fractions of the MFGM reduce inflammatory responses such as neutrophil elastase activity, superoxide production, and IL-1β release [183]. These findings highlight the potential role of milk-derived polar lipids in modulating gut health and inflammation, which often has implications for glucose homeostasis.
Nagasawa et al. identified interesting effects of dihydrosphingosine, a product of SM hydrolysis, on the activation of GPR120, a receptor expressed by enteroendocrine cells that facilitates the release of the incretin GLP-1 [184]. In vitro, dihydrosphingosine in conjunction with phytosphingosine exhibited potent activation of GPR120, although sphingosine did not [185]. Further research is necessary to explore the potential of milk polar lipids in modulating incretin production.

3.2. Fatty Acids

Milk is a source of FAs of varied length and saturation, which individually may have contrasting effects on metabolic health. FA species are classified as saturated and unsaturated odd-chain (OCFA), even-chain (ECFA), monounsaturated, polyunsaturated, branched-chain (BCFA), and TFA [186]. Except for the trans species, which include conjugated linoleic acid (CLA), rumenic acid (RA) and trans-11 vaccenic acid (VA), these FAs are not uniquely present in cow’s milk. This section will briefly summarize the general effects of cow’s milk FAs on glucose homeostasis, focusing on OCFA and TFA with a brief summary of the other classes.

3.2.1. Even-Chain Fatty Acids

The major saturated ECFA in whole milk includes myristic, palmitic, and stearic acid, respectively C14:0 (0.3 g/100 g), C16:0 (0.8 g/100 g), and C18:0 (0.4 g/100 g), with C10:0 and C12:0 contributing less than 0.2 g/100 g. ECFA makes up approximately 68% of FA present in cow’s milk [90,186].
Numerous studies report associations between consumption of ECFAs C14:0, C16:0, and C18:0 (not specific to milk) and increased risk of T2DM, obesity, IR, and metabolic dysfunction [89,187,188]. C16:0 and C14:0 feeding elicits increases in BW, reduced insulin secretion, hepatic steatosis, and increased endoplasmic reticulum stress in mouse adipose tissue and insulin-secreting INS-1E cells [89,189]. Clarifying whether ECFAs from dairy products confer different effects because of the unique dairy matrix compared to other dietary sources could help to allay concerns regarding whole milk [5,6]. Indeed, recent comprehensive systematic reviews [4] and large-scale observational studies [190,191] find, at minimum, no harm and potentially a benefit of consuming dairy fat on cardiovascular risk factors.

3.2.2. Odd-Chain Fatty Acids

The saturated OCFAs are pentadecanoic acid (C15:0) and heptadecanoic acid (C17:0), which comprise only 1.5% of FA in milk [90]. They are produced by rumen microbial fermentation and microbial de novo lipogenesis. The synthesis of these FAs by human enzymes is minute, rendering them dependable biomarkers of dairy fat consumption [192]. C15:0 and C17:0 FA concentrations in milk are approximately 0.9 g/100 g and 0.5 g/100 g, respectively. Their concentration in the blood is proposed as an indicator of the fat content of the consumed dairy products to distinguish between the consumption of low-fat or fat-free dairy products versus whole-milk dairy products [193].
The negative association of higher circulating concentrations of OCFAs with lower risk of cardiometabolic disorders indicates potential health advantages that extend beyond their application as biomarkers [194]. Observational data in humans demonstrates a lower risk of T2DM associated with higher proportions of C15:0 and C17:0 in erythrocyte membranes [195]. Furthermore, studies report a negative correlation between metabolic disease risk and circulating C15:0 and C17:0 [91,196]. Although the inverse associations of C15:0 with metabolic disease risk are consistently supported [197,198,199], the evidence for C17:0 is more variable [197]. Experiments to determine if the benefits of OCFA counterbalance ECFA effects when consumed within the dairy matrix would help clarify why dairy products do not appear to have the same detrimental effects as other foods rich in long-chain ECFA.
A recent 12-week RCT of 88 women with metabolic dysfunction-associated steatotic liver disease (MASLD) compares the effects of a modified Mediterranean diet with and without 300 mg C15:0 supplementation to a control group maintaining a habitual diet. BMI, liver fat content, and various metabolic risk factors are improved in both diet intervention groups compared to the control group and are more pronounced in the diet with C15:0 supplementation [53]. The primary outcomes do not directly measure specific effects of C15:0 supplementation on glucose homeostasis. However, given the overall metabolic improvements and potential gut microbiome alterations induced by C15:0 supplementation, such as increased abundance of B. adolescentis [53], it is plausible that these changes could indirectly influence glucose regulation.
In preclinical investigations utilizing diverse human cell lines and rodent models, consistent beneficial outcomes on metabolic parameters are observed, particularly for C15:0. In cultured mouse myotubes, C15:0 treatment increases glucose uptake and glucose transporter 4 (GLUT4) translocation to the cell membrane, especially when administered with insulin, suggesting a significant enhancement of insulin sensitivity [200]. GLUT4 is the major glucose transporter responsible for glucose uptake in muscle and adipose cells, and its translocation to the cell surface is associated with increased glucose uptake from the circulation, reflected in normalized blood glucose levels and, probably, improved insulin sensitivity [201]. The mechanism behind increased GLUT4 translocation involves the activation of 5′ AMP-activated protein kinase (AMPK) and AKT substrate 160 (AS160) [200]. In human cell culture studies, 10 to 50 μM of C15:0 is a partial agonist of peroxisome proliferator-activated receptors (PPAR) α/δ. It also lowers mitochondrial reactive oxygen species production in HepG2 cells [90], underscoring its capacity to enhance mitochondrial function. This is vital for maintaining hepatocyte function and metabolic homeostasis, including glucose regulation. Furthermore, C15:0 activates anti-inflammatory and antifibrotic pathways in cell cultures representing multiple tissues, as shown by reduced pro-inflammatory markers such as monocyte chemoattractant protein 1 MCP-1 and secreted immunoglobulin G, alongside decreased fibrosis indicators like collagen I and plasminogen activator inhibitor 1. In an HFD rat model, C15:0 supplementation reduces proinflammatory cytokines and lowers fasting blood glucose and BW gain [90]. These results highlight its potential in mitigating chronic inflammation and fibrosis, conditions often associated with metabolic disorders including IR and impaired glucose homeostasis [90].
Overall, dairy-derived C15:0 mediates improved glucoregulation by reducing dyslipidemia, promoting mitochondrial repair, and decreasing inflammation. Indeed, some researchers argue that C15:0 could be an essential FA [90,202]. In contrast, in HFD-fed male rats, C17:0 elicits no phenotypic changes [90]. Likewise, in HFD-fed male mice supplemented with either dairy fat or C17:0, exacerbated liver inflammation is detected with no improvement in insulin sensitivity [92]. However, for both OCFAs, more in vivo data are required.

3.2.3. Trans Fatty Acids

TFAs naturally occurring in milk constitute 4–6% of the overall fat content (~0.1 g per 100 mL of whole milk). VA (18:1, n-7) is the most abundant TFA in dairy milk fat, with relative amounts of total TFA varying depending on the cows’ diet [161,203].
Several in vivo and in vitro studies demonstrate the beneficial impacts of VA on metabolic endpoints. In T2DM male Sprague Dawley rats, 1.0% w/w VA supplementation in a diet containing butter oil as the dairy background improves fasting blood glucose and glucose-stimulated insulin secretion in vivo and from isolated islets [94], which is associated with increased β-cell area and up-regulation of the FA receptor GPR40 in islets [94]. VA supplementation also elicits greater total energy expenditure accompanied by an enhanced respiratory exchange ratio, indicating improvement in substrate utilization in a rat model of MASLD and metabolic syndrome [93]. Supplementing 1.0% VA in the diet reduces the mRNA of Fas and Acaca in the liver, which may contribute to the observed decrease in MASLD symptoms and also reduce the amount of visceral adipose tissue in obese JCR:LA-cp rats [93]. Similar results occur in fa/fa Zucker rats fed with 1.5% (w/w) VA [96]. The observed impact on visceral adipose tissue may be via VA acting as a ligand for both PPAR-α and PPAR-γ [204]. Despite these encouraging findings, in human studies, providing diets enriched in VA for 4–5 weeks had no significant effect on insulinemia and glycemia in either healthy men or overweight women [205,206].
CLA has garnered significant scientific and general public attention because of its putative modulation of cardiometabolic risk factors, including obesity, inflammatory indicators, IR, glucose metabolism, and diabetes [207]. The primary source of CLA in cow’s milk is biohydrogenation of linoleic acid by microorganisms in the rumen [208]. Pasture-based diets enhance the concentration of CLA in milk because of the linoleic acid precursor present in the forage [208,209]. In addition, a substantial proportion of CLA present in milk is produced in the mammary gland of the cow from VA [209].
Despite the attention, results in human studies are mixed. A meta-analysis of 13 RCTs reveals a notable rise in fasting blood glucose after administering CLA to human participants with a high cardiovascular disease risk, but the results are highly heterogeneous. In addition, no significant impact is seen on hemoglobin A1c (HbA1c) or HOMA-IR compared to the control groups [56]. Other meta-analyses examining the effects of CLA on IR and risk factors associated with diabetes show its potential to decrease key metabolic health indicators, such as inflammatory markers interleukin-6 and tumor necrosis factor-α [210], BMI and body composition [54,55], and oxidative stress markers [211]. Thus, CLA supplementation, as part of a strategy to mitigate risk factors linked to IR and diabetes, may be beneficial [212] but effects on overall cardiovascular risk are less impressive. One reason for the divergent results may be because CLA consists of a mixture of isomers.
Limited clinical studies of the isomer RA on human health outcomes, particularly in relation to glucose homeostasis, demonstrate mixed results. A cross-over design RCT of healthy individuals, providing meals substituting 3% VA, 3% industrial CLA isomers, or 1% RA for stearic acid each for 24 days finds no significant differences among the dietary treatments on blood glucose, insulin, or HOMA-IR [57]. In contrast, RA supplementation in males with obesity (approximately 1% of energy intake for 12 weeks) reduces insulin sensitivity compared to placebo [213], thus raising concerns about the impact of RA on metabolic health.
Most of the evidence for the health benefits of CLA or RA, including their hypothesized antidiabetic capabilities, is from studies in animal and in vitro models. In rodents, CLA supplementation has inconsistent IR-related outcomes, yielding worsened insulin sensitivity in a lactating mouse study [98] but improving OGTT in a HFD-rat model [99]. In the latter study, recovery of Ucp2 and Ucp3 mRNA expression in muscle suggests increased energy expenditure as a mechanism. RA-enriched butter significantly increases fasting serum insulin in male Wistar rats fed an HFD [101]. In female C57Bl/6J mice, 6 months of supplementation with RA (0.5% of total fat) increases lean mass and decreases fat mass, and notably decreases glucose, HOMA-IR, and improves insulin sensitivity, consistent with reduced IR [97]. Similarly, ob/ob mice provided RA have reduced plasma insulin, glucose and HOMA-IR and an improvement in insulin sensitivity [100].
RA mimics the ability of insulin to upregulate GLUT4 trafficking to the plasma membrane and increase glucose uptake in L6 myotubes upon acute exposure to RA [214]. Also, the exposure of HepG2 liver cells to RA inhibits gluconeogenesis by reducing key enzymes Pck1 and G6pc1 expression [215].

3.2.4. Branched-Chain Fatty Acids

BCFAs in milk are typically SFAs with one or multiple methyl branches along their carbon chains. The structural forms of BCFA are characterized by a branch point on the second-to-last carbon atom in iso BCFAs and a branch on the third-to-last carbon in ante-iso BCFAs. Microorganisms in the rumen utilize the dietary branched-chain amino acids (BCAA) valine, leucine, and isoleucine to produce BCFA. BCFAs contribute 1.7–3.4% of the total fatty acids in milk and consuming three servings of whole milk per day provides 367–763 mg of BCFA [216]. Monomethyl BCFAs with chain lengths of 14–17 carbons predominate, featuring either iso or ante-iso configurations. The most abundant milk BCFA is ante-iso C15:0, followed by iso C17:0, iso C15:0, ante-iso C17:0, and iso C16:0 [217]. Multimethyl BCFAs, specifically phytanic acid (0.1–0.5%, or 7–37 mg per serving of whole milk) and its derivative pristanic acid (0.04–0.06% or 3–4 mg per serving of whole milk), are minor components of milk fat [218,219]. The origin and metabolism of BCFAs has been reviewed and elaborated elsewhere [216].
Recent observational studies highlight a significant association between BCFA intake and energy and glucose homeostasis. Total BCFAs in serum are higher in non-obese compared to obese women, with iso-BCFAs displaying an inverse association with BMI [220]. Circulating BCFAs are associated with higher weight loss and possibly less body fat accumulation after gastric bypass surgery in people with obesity [221]. Furthermore, the proportions of total and individual BCFAs in the adipose tissue of lean subjects are higher than those of obese individuals [222], associated with reduced prevalence of dysglycemia [223].
The liver content of BCFA of female C57BL/6J mice is inversely correlated with total hepatic fat accumulation, suggesting that BCFAs may play a role in lipid metabolism regulation [224]. A recent in vitro study using a human fatty liver cell line (L02 cells) supports this observation, showing that iso-15:0 and iso-18:0 BCFAs reduce cellular TG and upregulate genes involved in lipid catabolism [225]. In the rat insulinoma INS-1 β-cell line (a well-established model for studies of pancreatic islet β-cell function) BCFA iso-17:0 upregulates critical transcription factors for optimal insulin secretion like PDX1 and PPAR-γ [102].
Dietary BCFAs are hypothesized to decrease inflammation, which is an underlying cause of glucose dysregulation. In humans, an inverse association is observed between serum iso-BCFAs (e.g., iso-15:0, iso-16:0, iso-17:0, and anteiso-15:0) and C-reactive protein, an important inflammatory marker associated with the risk of T2DM [220]. Sprague Dawley rat pups that consume a diet containing a mix of BCFAs have enhanced expression of interleukin-10, an anti-inflammatory cytokine [226]. Also, BCFA reduces the mRNA of key pro-inflammatory mediators such as Cxcl8, Tlr4, and Nfkb1 in a model of lipopolysaccharide-induced inflammation in gastrointestinal Caco-2 cells [227]. While most studies to date intervened with a combination of BCFAs, thoroughly investigating milk-derived BCFAs and specific milk BCFA species, such as iso-15:0 and iso-17:0, is needed in both in vivo and in vitro models to elucidate their effects on glucose homeostasis to help in developing targeted dietary recommendations and therapeutic interventions involving BCFA.

4. Protein Hydrolysates and Peptides

Milk proteins are nutritionally rich because they have an adequate balance of essential amino acids. Cow’s milk has a total protein content of about 3.5% by weight yielding 21% of the calories in whole milk. Caseins (specifically α-, β-, γ- and κ-casein) and wheys (several globulins, including α- and β-lactoglobulin) are the primary classes of protein found in milk, in a ratio of 4:1. Membrane globular proteins, glycoproteins, and lipoproteins mainly constitute the outer layer of MFGM. These proteins constitute less than 2% of the overall protein content [228,229].
Ingestion of intact whey has acute and chronic effects on glycemia. Indeed, a meta-analysis showed that whey consumption reduces the area under the curve of a glucose tolerance test concurrent with enhanced insulinemic response in both healthy people and those with T2D [19]. Chronically, whey reduced fasting insulin, FBG, and HOMA-IR in a sub-analysis pooling the results of 36 RCTs [230], consistent with three other recent meta-analyses [231,232,233,234,235]. The sub-analysis of Mohammadi suggested casein was less effective than whey in modulating glycemic parameters [230].
Milk is an excellent source of bioactive peptides. Bioactive peptides are usually 2–20 amino acids in length and, besides their nutritional value, exert beneficial physiological effects [236]. Bioactive peptides derived from milk are initially inactive within the primary structure of milk protein. The bioactive forms are primarily generated through proteolysis of casein and whey proteins [237,238,239], but β-lactoglobulin hydrolysis also generates bioactive peptides, such as wheylin [240,241].
The most well-researched milk-derived bioactive peptides are valine-proline-proline (VPP) and isoleucine-proline-proline (IPP) [242]. Although mostly tested for their anti-hypertensive effects [243,244,245,246], milk-derived hydrolysates can improve IR and modulate glucose tolerance in humans [63,247,248]. However, a meta-analysis of human trials found no consistent effect of casein hydrolysate on cardiovascular risk factors, showing that despite significantly lowering blood pressure, there was no significant effect of FBG [249]. Another meta-analysis included the combination of protein hydrolysates from different sources, indicating a significant reduction in postprandial blood glucose response among adults with normal glycemia as well as those with hyperglycemia [250]. Herein, we review clinical and preclinical studies that utilized milk hydrolysates and peptides and their effect on IR and glucose-related measurements but do not consider total protein from milk, such as whey isolates, casein isolates, or whey proteins.
In general, dietary supplementation with milk-derived bioactive peptides/hydrolysates does not affect food intake in animals. However, one study shows that acute, but not chronic, diet supplementation with a casein-derived peptide (CHM-273S–sequence: SKDIGSESTEDQAME) reduces food intake in C57BL/6 mice [113] and others find that whey hydrolysate decreases food intake compared to a chow diet in rats [104].

4.1. Whey Hydrolysate

Whey hydrolysates comprised of smaller peptides and free amino acids are prepared by enzyme digestion at acidic or alkaline pH [251,252]. In a randomized, cross-over trial in patients with prediabetes, acute provision of 1400 mg of whey hydrolysate before a carbohydrate-rich challenge meal significantly decreased the postprandial elevation of blood glucose as compared to the placebo, indicating better management of blood glucose. Long-term usage markedly decreased HbA1c but did not further improve OGTT. Twice the dose did not improve any outcome [63]. In an RCT conducted on overweight/obese women, the group that received a combination of an energy-restricted diet and whey hydrolysate (20 g) for 12 weeks had reduced BMI and body fat when compared to energy restriction alone [59]. In contrast, a 12-week intervention with 60 g/day of whey hydrolysate in adults with abdominal obesity did not result in changes to the HOMA-IR [62].
Among the animal studies using whey hydrolysate and investigating insulin sensitivity, signaling, or glucose tolerance [103,104,105,106,107,108,110], consistent improvements in glucose tolerance are demonstrated; however, the mechanisms identified are different in each study, depending on the pathways investigated. Zucker diabetic fatty (ZDF) rats fed whey hydrolysate (13 weeks) on a background of chow diet exhibit improved OGTT and lowered HbA1c versus controls. The hydrolysate has no effect on these parameters in lean Wistar rats or on fasting glucose or insulin in either ZDF or Wistar rats [104]. The authors hypothesize that the effects observed in ZDF rats could be due to lower circulating glucagon, which would lead to decreased hepatic gluconeogenesis [104]. In ob/ob mice, whey hydrolysate supplementation for 8 weeks improves OGTT and decreases fasting insulin and HOMA-IR. These effects are attributed to higher insulin-secreting capacity, as measured from isolated pancreatic islets [106], perhaps mediated by increased plasma concentration of insulinotropic amino acids following enhanced intestinal absorption. Lean, insulin-sensitive mice exhibited improved OGTT but no other changes [106]. A short-term treatment with whey hydrolysate and exercise intervention in Wistar rats elicits no changes in fasting insulin compared to controls, but the hydrolysate-treated animals have increased liver and skeletal muscle glycogen content. In addition, up-regulated skeletal muscle GLUT4 translocation to the plasma membrane and p-AKT (indicating insulin signaling) is more pronounced in whey + exercise rats [103]. The combined results of these studies indicate that whey hydrolysates affect both insulin secretion and signaling to improve glucose homeostasis. However, a minority of studies do not find benefit. For example, in C57BL/6 mice, whey hydrolysate feeding for 13 weeks exacerbates the effects of HFD, including increased BW, impaired glucose tolerance, and increased HOMA-IR, while having a null effect on insulin sensitivity [105]. These effects are accompanied by ectopic lipid accumulation and lower mitochondrial oxidative phosphorylation in skeletal muscle, and PPAR−α suppression in WAT, which can contribute to IR [105].
Whey hydrolysate may influence glucose homeostasis by modulating intestinal hormones. In vitro studies simulating gastrointestinal digestion demonstrate that whey hydrolysate enhances the synthesis and processing of GLP-1 more than intact whey isolate. Furthermore, whey hydrolysate reduces dipeptidyl peptidase-IV (DPP-IV) activity in a cell model [253]. GLP-1, an incretin hormone involved in glucose control, has a relatively short half-life due to the activity of DPP-IV, which is present in both cell-associated and circulatory states and is highly expressed in blood and enterocytes. The involvement of DPP-IV in maintaining glucose homeostasis entails the deactivation of incretins, GIP, and GLP-1 [247].

4.2. Casein Hydrolysate

In adults with T2DM, casein hydrolysate interventions consistently improve glucoregulation by increasing insulin secretion. Compared with placebo, consumption of casein hydrolysate (0.3 g/kg) in conjunction with leucine after a main meal by people with T2DM results in a substantial decrease in the mean 24-h blood glucose and increased circulating insulin [60]. Another study demonstrates enhanced de novo insulin production in participants with T2DM supplemented with casein hydrolysate (0.35 g/kg) in comparison to the consumption of a control carbohydrate diet [254], effects that are maintained even in the absence of supplementary amino acids [248]. In another RCT, including patients with T2DM, an OGTT was conducted after acute administration of casein hydrolysate at a dose of 0, 6, or 12 g. The highest dose resulted in elevated insulin and decreased glucose levels post-challenge [61].
Casein hydrolysate supplementation also demonstrates beneficial effects on insulin signaling and glucose handling in rodents. Long-term casein hydrolysate supplementation in an HFD-induced obese mouse model improved glucose tolerance [110]. Although no changes are seen in an insulin tolerance test (ITT), insulin signaling is enhanced in skeletal muscle, liver, and WAT via increased AKT activation. Less pronounced or null changes in vivo are seen in chow diet-supplemented animals [110]. Casein hydrolysates (4%) administered to mice with HFD-induced obesity decreased systemic inflammation induced by the HFD and improved ITT (i.e., IR), reduced fasting insulin and HOMA-IR, although FBG was unchanged compared with the HFD group [111]. Conversely, in a T2DM mouse model, 8 weeks of casein hydrolysate recovered BW lost after STZ treatment without affecting food intake [107]. FBG and OGTT were decreased, but no changes in fasting insulin were observed. The casein hydrolysate increases skeletal muscle glycogen content and enhances muscle synthesis protein activation via phosphorylation of glycogen synthase kinase-3β. Moreover, insulin signaling is enhanced with increased phosphorylation of IRS-1, PI3K, and AKT and GLUT4 translocation to the plasma membrane [107]. Of note, the authors report casein hydrolysate modifies the gut microbiome, which may contribute to the benefits. In DIO C57Bl/6 mice, casein hydrolysate decreased BW, FBG, insulin, glucagon, and leptin, with no changes in the OGTT after 7–8 weeks of supplementation [108]. This is accompanied by decreased adipocyte size, higher ex vivo FA oxidation in subcutaneous WAT, and increased expression of browning markers such as Ucp1, Cox8b, and Mpzl-2 in this model, which might increase energy expenditure [108]. Finally, β-lactoglobulin hydrolysate provided acutely to KK-Ay mice yielded improvements in both OGTT and ITT accompanied by increased p-AKT in the liver and skeletal muscle [109], but more studies of this hydrolysate are required.
The bioactivity of protein hydrolysates depends on the combinations of enzymes used. Oral administration of casein hydrolysate produced using a combination of papain and Flavourzyme resulted in a drop in FBG and HbA1c, and improvement in OGTT, in rats with T2DM caused by STZ and an HFD compared with hydrolysates prepared with papain or Flavourzyme alone. All treatment groups experienced an increase in circulating insulin and HOMA-β (a measure of β-cell function) compared with the control group [255]. Furthermore, the administration of Flavourzyme-papain hydrolysate increases phosphorylation of AMPK and the expression of Glut2, along with inhibition of phosphoenolpyruvate carboxylase kinase activity and an elevation in glycogen content in the liver [255]. Others report that Flavourzyme-papain casein hydrolysates reduce liver oxidative damage by increasing the transcription of Nrf2, which upregulates antioxidant activity. It was proposed that such treatment could be a viable option for managing liver damage in individuals with T2DM [112].
In cell culture experiments, casein hydrolysates dose-dependently inhibit mitogen-activated protein kinase (MAPK)-c-JNK phosphorylation and increase the phosphorylation of ERK in TNF-α-induced insulin-resistant 3T3-L1 adipocytes, consistent with the potential to improve chronic inflammation in WAT [111]. In HepG2 liver cells, glucose uptake was enhanced by Flavourzyme-papain casein hydrolysates in the absence of insulin, an activity attenuated by the application of inhibitors targeting both the AKT and AMPK pathways, whereas the AMPK signaling route was exclusively activated by Flavourzyme-papain hydrolysates in T2DM rats (see above) [255]. C2C12 myotubes, a model of skeletal muscle incubated with casein hydrolysate, have enhanced 2-deoxy-glucose uptake compared with untreated cells, explained by increased p-AMPK and liver kinase B1 [256].
Overall, casein hydrolysate consistently improves endpoints related to glucose homeostasis in both human and animal studies. The mechanisms may include enhanced insulin secretion and insulin signaling in the liver, adipose, and skeletal muscle. Hydrolysates prepared with papain and Flavourzyme in combination show efficacy in activating cell pathways that regulate glucose uptake in multiple trials.

4.3. Bioactive Peptides

The significance of milk-derived peptides in human physiology and health remains relatively uncertain because our understanding of them is mostly based on preclinical studies. Studies investigating specific peptides derived from milk in rodents [109,113,114,115,116,117] exhibit consistent improvement in glucose tolerance, insulin signaling, and insulin sensitivity. For example, dietary supplementation with a glycomacropeptide derived from casein for 12 weeks decreased insulin and HOMA-IR but did not protect against high-fat/high-sugar diet-induced obesity or improve OGTT in C57BL/6 mice [115]. This casein-derived glycomacropeptide increases liver insulin signaling through phospho-AKT, modulates the abundance of proteins involved in lipid metabolism and gluconeogenesis via increased phospho-acetyl CoA carboxylase, PPAR-α, PPAR-γ coactivator-1α and carnitine palmitoyl transferase-1α, and decreases fatty acid synthase, phosphoenolpyruvate carboxykinase and glucose-6-phosphatase [115]. A different casein-derived peptide (CHM-273S–sequence: SKDIGSESTEDQAME) acutely elicited improved glucose tolerance in Sprague Dawley rats associated with enhanced liver insulin signaling through the AKT pathway [113]. In C57BL/6 mice, the same peptide provided chronically in the diet reduced BW, visceral fat mass, adipocyte size, fasting glucose, and HOMA-IR despite no changes in fasting insulin. Its acute administration to mice does not elicit any improvements in glucose tolerance, which suggests its effects are mediated by metabolic adaptations [113].
VPP, a well-studied milk-derived peptide with anti-hypertensive properties, showed no effects on BW, fasting glucose, or fasting insulin but improved ITT and white adipose tissue inflammation after 16 weeks of supplementation in HFD-fed C57BL/6 mice. These effects are attributed to the renin-angiotensin system inhibitory activity of the peptide [116]. Another casein-derived peptide supplemented prior to exercise training led to decreased BW gain, fat mass, FBG and HOMA-IR after 4 weeks in HFD mice. OGTT was improved in treated animals, along with increased GLUT4 protein content in skeletal muscle [114]. A bioactive dipeptide derived from β-lactoglobulin called wheylin-1 (sequence: MH) improved ITT and enhances p-AKT in the liver of KK-Ay mice [109]. In normal mice, acute administration of a casein-derived peptide (VPYPQ) improved OGTT, an effect attributed to DPP-IV inhibitory activity seen in vitro [117].
Individual amino acids found in milk proteins may also influence glycemic parameters. Compiled evidence indicates that higher circulating branched-chain amino acids (BCAAs), which include leucine, isoleucine, and valine, may increase the risk of T2D [257], whereas reduced circulating BCAAs demonstrate alleviation of insulin resistance [258]. BCAAs may influence both insulin sensitivity and insulin secretion [259,260,261]. However, the impact of dietary intake of BCAAs is an ongoing debate that has been comprehensively evaluated in current reviews [262,263,264]. Their effects in isolation may be distinct from the ingestion of intact proteins, hydrolysates, or peptides. Indeed, the n-terminal amino acid of both VPP and IPP is a BCAA. Another example is isoleucine-arginine-tryptophan, an egg white-derived tripeptide, which improves glucose tolerance and insulin sensitivity in rats and mice despite one of its constituents being a BCAA [265,266].
Overall, milk-derived hydrolysates and peptides induce relatively consistent improvements in IR, insulin signaling, and glucose tolerance in rodents. Variability in response to hydrolysates likely stems from the variety of tested doses, ranging from 1 mg/mL of water [105] or 100–200 mg/Kg BW [106,107] in the case of casein hydrolysates. Also, differences in species/strains of rodents and background diet (HFD or low-fat/chow diet) confound results. Studies utilizing specific peptides have demonstrated more consistent results, particularly in the enhancement of insulin signaling at the tissue level. However, single peptides in vivo could be less effective than hydrolysates if the peptides within the mixture provide synergistic effects. A summary of the actions of the peptides and hydrolysates leading to improvements observed in the studies outlined is illustrated in Table 2.

5. Summary and Conclusions

This review highlights the multifaceted role of cow’s milk components in modulating glucose homeostasis and insulin sensitivity, which could be utilized to prevent and manage T2DM. The primary carbohydrate in milk, lactose, has a low GI and does not raise blood glucose compared to other simple sugars, which is beneficial for maintaining stable blood glucose. Furthermore, the prebiotic properties of lactose and oligosaccharides promote the growth of beneficial gut bacteria, contributing to improved gut health and potentially enhancing glucose metabolism through mechanisms that reduce inflammation. The FA profile of milk includes a complex array of saturated and unsaturated fats that have diverse effects on health. Notably, the MFGM and certain FAs, such as odd-chain C15:0 and VA, are associated with positive metabolic outcomes. These components may improve insulin sensitivity and modulate lipid metabolism and inflammation, but animal results are not consistently replicated in human trials. The bioactive peptides derived from casein and whey influence insulin secretion and sensitivity, which are essential mechanisms for maintaining normal blood glucose. They may also enhance incretin activity by inhibiting DPP-IV. An overall summary is provided in Figure 1.
Although cow’s milk is the most popular milk consumed by the majority of people, the health impacts of milk from other ruminant species, including goats, sheep, and camels, has received much attention because their consumption in a particular region or country may be high [267,268]. Similar to cow’s milk, other ruminants’ milk is also rich in various bioactive molecules that may overlap with cow’s milk or be specific to the species [269,270]. For example, bioactive molecules in sheep’s milk, including CLA and lactoferrin, can enhance insulin sensitivity and reduce inflammation, similar to the effects observed with cow’s milk [271]. Given the particular composition of its medium-chain fatty acids, goat’s milk holds promise for improving lipid metabolism and reducing hyperglycemia [272]. Furthermore, camel’s milk, rich in insulin-like proteins, was shown to decrease glycemia while improving insulin secretion, thus possibly having therapeutic benefits in the management of T2DM [273]. These comparative insights underline the potential of the bioactive molecules in the milk of different species in therapeutic interventions, specifically in modulating glucose homeostasis [15,274,275,276,277].
In conclusion, it is evident that milk-derived bioactive molecules are capable of modulating glucose homeostasis. This modulation is achieved through direct effects on insulin signaling pathways and indirect effects via improving obesity phenotypes. The studies included in this review administered a wide range of milk-derived bioactive compounds, mainly in isolated form. Although most of the studies we found report benefits of carbohydrate- and protein-derived molecules, experiments using lipid-derived bioactives were more equivocal and more research is needed to reach stronger conclusions. It is worth noting that the administered doses of these bioactive molecules may be difficult to achieve through the consumption of milk. Furthermore, milk contains other functional constituents, including BCAAs and exosomal cargos (like microRNAs), whose impact on glucose homeostasis was not elaborated on in this review. Future research should focus on identifying the optimal types and amounts of milk-derived components for preventing and managing metabolic diseases and on understanding the complex interactions between these components and the human body. This will enable the development of targeted dietary recommendations and therapeutic interventions that leverage the full spectrum of benefits offered by milk and its derivatives.

Author Contributions

Conceptualization, E.Y., E.B., S.C.d.C.Z. and C.B.C.; writing—original draft preparation, E.Y., E.B. and S.C.d.C.Z.; writing—review and editing, C.B.C.; visualization, E.Y., E.B. and S.C.d.C.Z.; supervision, C.B.C.; project administration, C.B.C. All authors have read and agreed to the published version of the manuscript.

Funding

No funding was provided for the creation of this review article. Graduate student stipends have been provided as follows: E.Y.—Alberta Diabetes Institute & International Helmholtz Research School for Diabetes, Alberta Graduate Excellence Scholarship; E.B.—Alberta Diabetes Institute and the Canadian Institutes of Health Research; S.C.d.C.Z.—Canadian Institutes of Health Research.

Data Availability Statement

No new data were created or analyzed in this study. Data sharing is not applicable to this article.

Conflicts of Interest

C.B.C. has received research funding from the Dairy Farmers of Canada. The funders had no role in in the writing of the manuscript or in the decision to publish this review. E.B., E.Y. and S.C.d.C.Z. declare no conflicts of interest.

References

  1. DeFronzo, R.A.; Ferrannini, E. Insulin resistance: A multi-faceted syndrome responsible for NIDDM, obesity, hypertension, dyslipidemia and atherosclerotic cardiovascular disease. Diabetes Care 1991, 14, 173–194. [Google Scholar] [CrossRef] [PubMed]
  2. DeFronzo, R.A.; Ferrannini, E.; Groop, L.; Henry, R.R.; Herman, W.H.; Holst, J.J.; Hu, F.B.; Kahn, C.R.; Raz, I.; Shulman, G.I.; et al. Type 2 diabetes mellitus. Nat. Rev. Dis. Prim. 2015, 1, 15019. [Google Scholar] [CrossRef] [PubMed]
  3. Ruze, R.; Liu, T.; Zou, X.; Song, J.; Chen, Y.; Xu, R.; Yin, X.; Xu, Q. Obesity and type 2 diabetes mellitus: Connections in epidemiology, pathogenesis, and treatments. Front Endocrinol 2023, 14, 1161521. [Google Scholar] [CrossRef]
  4. Drouin-Chartier, J.; Cote, J.A.; Me, L.; Brassard, D.; Tessier-Grenier, M.; Desroches, S.; Couture, P.; Lamarche, L. Comprehensive review of the impact of dairy foods and dairy fat on cardiometabolic risk. Adv. Nutr. 2016, 7, 1041–1051. [Google Scholar] [CrossRef] [PubMed]
  5. Mozaffarian, D. Dairy foods, obesity, and metabolic health: The role of the food matrix compared with single nutrients. Adv. Nutr. 2019, 10, 917S–923S. [Google Scholar] [CrossRef]
  6. Poppitt, S.D. Cow’s milk and dairy consumption: Is there now consensus for cardiometabolic health? Front. Nutr. 2020, 7, 574725. [Google Scholar] [CrossRef]
  7. Jin, S.; Je, Y. Dairy consumption and risk of metabolic syndrome: Results from Korean population and meta-analysis. Nutrients 2021, 13, 1574. [Google Scholar] [CrossRef]
  8. Feng, Y.; Zhao, Y.; Liu, J.; Huang, Z.; Yang, X.; Qin, P.; Chen, C.; Luo, X.; Li, Y.; Wu, Y.; et al. Consumption of Dairy Products and the Risk of Overweight or Obesity, Hypertension, and Type 2 Diabetes Mellitus: A Dose-Response Meta-Analysis and Systematic Review of Cohort Studies. Adv. Nutr. 2022, 13, 2165–2179. [Google Scholar] [CrossRef] [PubMed]
  9. Brouwer-Brolsma, E.M.; Sluik, D.; Singh-Povel, C.M.; Feskens, E.J.M. Dairy product consumption is associated with pre-diabetes and newly diagnosed type 2 diabetes in the Lifelines Cohort Study. Br. J. Nutr. 2018, 119, 442–455. [Google Scholar] [CrossRef]
  10. Slurink, I.A.; Corpeleijn, E.; Bakker, S.J.; Jongerling, J.; Kupper, N.; Smeets, T.; Soedamah-Muthu, S.S. Dairy consumption and incident prediabetes: Prospective associations and network models in the large population-based Lifelines Study. Am. J. Clin. Nutr. 2023, 118, 1077–1090. [Google Scholar] [CrossRef]
  11. Sluijs, I.; Forouhi, N.; Beulens, J.; van der Schouw, Y.; Agnoli, C.; Arriola, L.; Balkau, B.; Barricarte, A.; Boeing, H.; Bueno-de-Mesquita, H.; et al. The amount and type of dairy product intake and incidence of type 2 diabetes: Results from the EPIC-InterAct Study. Am. J. Clin. Nutr. 2012, 96, 382–390. [Google Scholar] [CrossRef] [PubMed]
  12. Hruby, A.; Ma, J.; Rogers, G.; Meigs, J.B.; Jacques, P.F. Associations of dairy intake with incident prediabetes or diabetes in middle-aged adults vary by both diary type and glycemic status. J. Nutr. 2017, 247, 1764–1775. [Google Scholar] [CrossRef] [PubMed]
  13. Yuzbashian, E.; Fernando, D.N.; Pakseresht, M.; Eurich, D.T.; Chan, C.B. Dairy product consumption and risk of non-alcoholic fatty liver disease: A systematic review and meta-analysis of observational studies. Nutr. Metab. Cardiovasc. Dis. 2023, 33, 1461–1471. [Google Scholar] [CrossRef]
  14. Yuzbashian, E.; Moftah, S.; Chan, C.B. Graduate Student Literature Review: A scoping review on the impact of consumption of dairy products on phosphatidylcholine and lysophosphatidylcholine in circulation and the liver in human studies and animal models. J. Dairy Sci. 2023, 106, 24–38. [Google Scholar] [CrossRef]
  15. Althnaibat, R.M.; Bruce, H.L.; Wu, J.; Gänzle, M.G. Bioactive peptides in hydrolysates of bovine and camel milk proteins: A review of studies on peptides that reduce blood pressure, improve glucose homeostasis, and inhibit pathogen adhesion. Food Res. Int. 2024, 175, 113748. [Google Scholar] [CrossRef]
  16. Liljeberg Elmståhl, H.; Bjorck, I. Milk as a supplement to mixed meals may elevate postprandial insulinaemia. Eur. J. Clin. Nutr. 2001, 55, 994–999. [Google Scholar] [CrossRef]
  17. Del Prato, S.; Leonetti, F.; Simonson, D.C.; Sheehan, P.; Matsuda, M.; Ra, D. Effect of sustained physiologic hyperinsulinaemia and hyperglycaemia on insulin secretion and insulin sensitivity in man. Diabetologia 1994, 37, 1025–1035. [Google Scholar] [CrossRef] [PubMed]
  18. Juan, C.C.; Fang, V.S.; Kwok, C.F.; Perng, J.C.; Chou, Y.C.; Ho, L.T. Exogenous hyperinsulinemia causes insulin resistance, hyperendothelinemia, and subsequent hypertension in rats. Metabolism 1999, 48, 465–471. [Google Scholar] [CrossRef]
  19. Wolever, T.M.; Zurbau, A.; Koecher, K.; Au-Yeung, F. The Effect of Adding Protein to a Carbohydrate Meal on Postprandial Glucose and Insulin Responses: A Systematic Review and Meta-Analysis of Acute Controlled Feeding Trials. J. Nutr. 2024, in press. [Google Scholar] [CrossRef]
  20. Pfeuffer, M.; Schrezenmeir, J. Milk and the metabolic syndrome. Obes. Res. 2007, 8, 109–118. [Google Scholar] [CrossRef]
  21. Robinson, R.C. Structures and metabolic properties of bovine milk oligosaccharides and their potential in the development of novel therapeutics. Front. Nutr. 2019, 6, 50. [Google Scholar] [CrossRef] [PubMed]
  22. Park, Y.W. The impact of plant-based non-dairy alternative milk on the dairy industry. Food Sci. Anim. Resour. 2021, 41, 8–15. [Google Scholar] [CrossRef]
  23. Korhonen, H.J. 20-Bioactive milk proteins, peptides and lipids and other functional components derived from milk and bovine colostrum. In Functional Foods, 2nd ed.; Saarela, M., Ed.; Woodhead Publishing: Sawston, UK, 2011; pp. 471–511. [Google Scholar]
  24. Zempleni, J.; Sukreet, S.; Zhou, F.; Wu, D.; Mutai, E. Milk-Derived Exosomes and Metabolic Regulation. Annu. Rev. Anim. Biosci. 2019, 7, 245–262. [Google Scholar] [CrossRef] [PubMed]
  25. Bolyen, E.; Rideout, J.R.; Dillon, M.R.; Bokulich, N.A.; Abnet, C.C.; Al-Ghalith, G.A.; Alexander, H.; Alm, E.J.; Arumugam, M.; Asnicar, F.; et al. Reproducible, interactive, scalable and extensible microbiome data science using QIIME 2. Nat. Biotechnol. 2019, 37, 852–857. [Google Scholar] [CrossRef] [PubMed]
  26. Melnik, B.C. Lifetime Impact of Cow’s Milk on Overactivation of mTORC1: From Fetal to Childhood Overgrowth, Acne, Diabetes, Cancers, and Neurodegeneration. Biomolecules 2021, 11, 404. [Google Scholar] [CrossRef]
  27. Taponen, S.; McGuinness, D.; Hiitiö, H.; Simojoki, H.; Zadoks, R.; Pyörälä, S. Bovine milk microbiome: A more complex issue than expected. Vet. Res. 2019, 50, 44. [Google Scholar] [CrossRef]
  28. National Research Council. Factors Affecting the Composition of Milk from Dairy Cows. In Designing Foods: Animal Product Options in the Marketplace; National Academies Press: Washington, DC, USA, 1988. [Google Scholar]
  29. Taylor, S.L.; Kabourek, J. Food Intolerance|Milk Allergy. In Encyclopedia of Food Sciences and Nutrition; Caballero, B., Ed.; Academic Press: Oxford, UK, 2003; pp. 2631–2634. [Google Scholar]
  30. Zivkovic, A.; Barile, D. Bovine milk as a source of functional oligosaccharides for improving human health. Adv. Nutr. 2011, 2, 284–289. [Google Scholar] [CrossRef]
  31. Wolever, T.M.; Miller, J.B. Sugars and blood glucose control. Am. J. Clin. Nutr. 1995, 62, 212S–221S; discussion 221S–227S. [Google Scholar] [CrossRef]
  32. Atkinson, F.S.; Brand-Miller, J.C.; Foster-Powell, K.; Buyken, A.E.; Goletzke, J. International tables of glycemic index and glycemic load values 2021: A systematic review. Am. J. Clin. Nutr. 2021, 114, 1625–1632. [Google Scholar] [CrossRef] [PubMed]
  33. Hoyt, G.; Hickey, M.S.; Cordain, L. Dissociation of the glycaemic and insulinaemic responses to whole and skimmed milk. Br. J. Nutr. 2005, 93, 175–177. [Google Scholar] [CrossRef]
  34. Hoppe, C.; Mølgaard, C.; Vaag, A.; Barkholt, V.; Michaelsen, K.F. High intakes of milk, but not meat, increase s-insulin and insulin resistance in 8-year-old boys. Eur. J. Clin. Nutr. 2005, 59, 393–398. [Google Scholar] [CrossRef] [PubMed]
  35. Ostman, E.M.; Liljeberg Elmståhl, H.G.; Björck, I.M. Inconsistency between glycemic and insulinemic responses to regular and fermented milk products. Am. J. Clin. Nutr. 2001, 74, 96–100. [Google Scholar] [CrossRef] [PubMed]
  36. Gao, R.; Rapin, N.; Elnajmi, A.M.; Gordon, J.; Zello, G.A.; Chilibeck, P.D. Skim milk as a recovery beverage after exercise is superior to a sports drink for reducing next-day postprandial blood glucose and increasing postprandial fat oxidation. Nutr. Res. 2020, 82, 58–66. [Google Scholar] [CrossRef] [PubMed]
  37. Shkembi, B.; Huppertz, T. Glycemic Responses of Milk and Plant-Based Drinks: Food Matrix Effects. Foods 2023, 12, 453. [Google Scholar] [CrossRef]
  38. Pathak, K.; Zhao, Y.; Calton, E.K.; James, A.P.; Newsholme, P.; Sherriff, J.; Soares, M.J. The impact of leucine supplementation on body composition and glucose tolerance following energy restriction: An 8-week RCT in adults at risk of the metabolic syndrome. Eur. J. Clin. Nutr. 2024, 78, 155–162. [Google Scholar] [CrossRef]
  39. Mohsenpour, M.A.; Kaseb, F.; Nazemian, R.; Mozaffari-Khosravi, H.; Fallahzadeh, H.; Salehi-Abargouei, A. The effect of a new mixture of sugar and sugar-alcohols compared to sucrose and glucose on blood glucose increase and the possible adverse reactions: A phase I double-blind, three-way randomized cross-over clinical trial. Endocrinol Diabetes Nutr 2019, 66, 647–653. [Google Scholar] [CrossRef]
  40. Nilsson, M.; Stenberg, M.; Frid, A.H.; Holst, J.J.; Bjorck, I.M. Glycemia and insulinemia in healthy subjects after lactose-equivalent meals of milk and other food proteins: The role of plasma amino acids and incretins. Am. J. Clin. Nutr. 2004, 80, 1246–1253. [Google Scholar] [CrossRef]
  41. Mohammad, M.A.; Sunehag, A.L.; Rodriguez, L.A.; Haymond, M.W. Galactose promotes fat mobilization in obese lactating and nonlactating women. Am. J. Clin. Nutr. 2011, 93, 374–381. [Google Scholar] [CrossRef]
  42. Sunehag, A.L.; Haymond, M.W. Splanchnic galactose extraction is regulated by coingestion of glucose in humans. Metabolism 2002, 51, 827–832. [Google Scholar] [CrossRef]
  43. Morgan, L.M.; Wright, J.W.; Marks, V. The effect of oral galactose on GIP and insulin secretion in man. Diabetologia 1979, 16, 235–239. [Google Scholar] [CrossRef]
  44. Ercan, N.; Nuttall, F.Q.; Gannon, M.C.; Redmon, J.B.; Sheridan, K.J. Effects of glucose, galactose, and lactose ingestion on the plasma glucose and insulin response in persons with non-insulin-dependent diabetes mellitus. Metabolism 1993, 42, 1560–1567. [Google Scholar] [CrossRef] [PubMed]
  45. Coss-Bu, J.A.; Sunehag, A.L.; Haymond, M.W. Contribution of galactose and fructose to glucose homeostasis. Metabolism 2009, 58, 1050–1058. [Google Scholar] [CrossRef] [PubMed]
  46. Duckworth, L.C.; Backhouse, S.H.; O’Hara, J.P.; Stevenson, E.J. Effect of galactose ingestion before and during exercise on substrate oxidation, postexercise satiety, and subsequent energy intake in females. J. Am. Coll. Nutr. 2016, 35, 1–12. [Google Scholar] [CrossRef] [PubMed]
  47. Rosqvist, F.; Smedman, A.; Lindmark-Månsson, H.; Paulsson, M.; Petrus, P.; Straniero, S.; Rudling, M.; Dahlman, I.; Risérus, U. 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. 2015, 102, 20–30. [Google Scholar] [CrossRef]
  48. Demmer, E.; Van Loan, M.D.; Rivera, N.; Rogers, T.S.; Gertz, E.R.; German, J.B.; Smilowitz, J.T.; Zivkovic, A.M. Addition of a dairy fraction rich in milk fat globule membrane to a high-saturated fat meal reduces the postprandial insulinaemic and inflammatory response in overweight and obese adults. J. Nutr. Sci. 2016, 5, e14. [Google Scholar] [CrossRef]
  49. Beals, E.; Kamita, S.G.; Sacchi, R.; Demmer, E.; Rivera, N.; Rogers-Soeder, T.S.; Gertz, E.R.; Van Loan, M.D.; German, J.B.; Hammock, B.D.; et al. Addition of milk fat globule membrane-enriched supplement to a high-fat meal attenuates insulin secretion and induction of soluble epoxide hydrolase gene expression in the postprandial state in overweight and obese subjects. J. Nutr. Sci. 2019, 8, e16. [Google Scholar] [CrossRef]
  50. Pokala, A.; Quarles, W.R.; Ortega-Anaya, J.; Jimenez-Flores, R.; Cao, S.; Zeng, M.; Hodges, J.K.; Bruno, R.S. Milk-Fat-Globule-Membrane-Enriched Dairy Milk Compared with a Soy-Lecithin-Enriched Beverage Did Not Adversely Affect Endotoxemia or Biomarkers of Gut Barrier Function and Cardiometabolic Risk in Adults with Metabolic Syndrome: A Randomized Controlled Crossover Trial. Nutrients 2023, 15, 3259. [Google Scholar] [CrossRef]
  51. Weiland, A.; Bub, A.; Barth, S.W.; Schrezenmeir, J.; Pfeuffer, M. Effects of dietary milk- and soya-phospholipids on lipid-parameters and other risk indicators for cardiovascular diseases in overweight or obese men-two double-blind, randomised, controlled, clinical trials. J. Nutr. Sci. 2016, 5, e21. [Google Scholar] [CrossRef]
  52. Vors, C.; Joumard-Cubizolles, L.; Lecomte, M.; Combe, E.; Ouchchane, L.; Drai, J.; Raynal, K.; Joffre, F.; Meiller, L.; Le Barz, M.; et al. Milk polar lipids reduce lipid cardiovascular risk factors in overweight postmenopausal women: Towards a gut sphingomyelin-cholesterol interplay. Gut 2020, 69, 487–501. [Google Scholar] [CrossRef]
  53. Chooi, Y.C.; Zhang, Q.A.; Magkos, F.; Ng, M.; Michael, N.; Wu, X.; Volchanskaya, V.S.B.; Lai, X.; Wanjaya, E.R.; Elejalde, U.; et al. Effect of an Asian-adapted Mediterranean diet and pentadecanoic acid on fatty liver disease: The TANGO randomized controlled trial. Am. J. Clin. Nutr. 2024, 119, 788–799. [Google Scholar] [CrossRef]
  54. Asbaghi, O.; Shimi, G.; Hosseini Oskouie, F.; Naseri, K.; Bagheri, R.; Ashtary-Larky, D.; Nordvall, M.; Rastgoo, S.; Zamani, M.; Wong, A. The effects of conjugated linoleic acid supplementation on anthropometrics and body composition indices in adults: A systematic review and dose-response meta-analysis. Br. J. Nutr. 2024, 131, 406–428. [Google Scholar] [CrossRef] [PubMed]
  55. Onakpoya, I.J.; Posadzki, P.P.; Watson, L.K.; Davies, L.A.; Ernst, E. The efficacy of long-term conjugated linoleic acid (CLA) supplementation on body composition in overweight and obese individuals: A systematic review and meta-analysis of randomized clinical trials. Eur. J. Nutr. 2012, 51, 127–134. [Google Scholar] [CrossRef]
  56. Ghodoosi, N.; Rasaei, N.; Goudarzi, K.; Hashemzadeh, M.; Dolatshahi, S.; Omran, H.S.; Amirani, N.; Ashtary-Larky, D.; Shimi, G.; Asbaghi, O. The effects of conjugated linoleic acid supplementation on glycemic control, adipokines, cytokines, malondialdehyde and liver function enzymes in patients at risk of cardiovascular disease: A GRADE-assessed systematic review and dose-response meta-analysis. Nutr. J. 2023, 22, 47. [Google Scholar] [CrossRef] [PubMed]
  57. Gebauer, S.K.; Destaillats, F.; Dionisi, F.; Krauss, R.M.; Baer, D.J. Vaccenic acid and trans fatty acid isomers from partially hydrogenated oil both adversely affect LDL cholesterol: A double-blind, randomized controlled trial. Am. J. Clin. Nutr. 2015, 102, 1339–1346. [Google Scholar] [CrossRef]
  58. Riserus, U. Trans fatty acids and insulin resistance. Atheroscl Suppl. 2006, 7, 37–39. [Google Scholar] [CrossRef]
  59. Sun, Y.; Ling, C.; Liu, L.; Zhang, J.; Wang, J.; Tong, X.; Hidayat, K.; Chen, M.; Chen, X.; Zhou, H.; et al. Effects of Whey Protein or Its Hydrolysate Supplements Combined with an Energy-Restricted Diet on Weight Loss: A Randomized Controlled Trial in Older Women. Nutrients 2022, 14, 4540. [Google Scholar] [CrossRef]
  60. Manders, R.J.; Praet, S.F.; Meex, R.C.; Koopman, R.; de Roos, A.L.; Wagenmakers, A.J.; Saris, W.H.; van Loon, L.J. Protein hydrolysate/leucine co-ingestion reduces the prevalence of hyperglycemia in type 2 diabetic patients. Diabetes Care 2006, 29, 2721–2722. [Google Scholar] [CrossRef]
  61. Jonker, J.T.; Wijngaarden, M.A.; Kloek, J.; Groeneveld, Y.; Gerhardt, C.; Brand, R.; Kies, A.K.; Romijn, J.A.; Smit, J.W. Effects of low doses of casein hydrolysate on post-challenge glucose and insulin levels. Eur. J. Intern. Med. 2011, 22, 245–248. [Google Scholar] [CrossRef] [PubMed]
  62. Fuglsang-Nielsen, R.; Rakvaag, E.; Vestergaard, P.; Hermansen, K.; Gregersen, S.; Starup-Linde, J. The Effects of 12-Weeks Whey Protein Supplements on Markers of Bone Turnover in Adults with Abdominal Obesity—A Post Hoc Analysis. Front. Endocrinol 2022, 13, 832897. [Google Scholar] [CrossRef]
  63. Sartorius, T.; Weidner, A.; Dharsono, T.; Boulier, A.; Wilhelm, M.; Schon, C. Postprandial effects of a proprietary milk protein hydrolysate containing bioactive peptides in prediabetic subjects. Nutrients 2019, 11, 1700. [Google Scholar] [CrossRef]
  64. Liu, G.; Hughes, C.L.; Mathur, R.; Foster, W.G.; Davis, V.L.; Magoffin, D.A. Metabolic effects of dietary lactose in adult female rats. Reprod. Nutr. Dev. 2003, 43, 567–576. [Google Scholar] [CrossRef] [PubMed]
  65. Goseki-Sone, M.; Maruyama, R.; Sogabe, N.; Hosoi, T. Effects of dietary lactose on long-term high-fat-diet-induced obesity in rats. Obes. 2007, 15, 2605–2613. [Google Scholar] [CrossRef] [PubMed]
  66. El-Sherbeny, R.; El-Gharieb, M. A study on the effects of dietary lactose on ovarian function and body weight in normal and obese female albino rats. Bull. Egypt. Soc. Physiol. Sci. 2009, 29, 2009. [Google Scholar] [CrossRef]
  67. van de Heijning, B.J.; Kegler, D.; Schipper, L.; Voogd, E.; Oosting, A.; van der Beek, E.M. Acute and Chronic Effects of Dietary Lactose in Adult Rats Are not Explained by Residual Intestinal Lactase Activity. Nutrients 2015, 7, 5542–5555. [Google Scholar] [CrossRef]
  68. Bouwman, L.M.S.; Fernandez-Calleja, J.M.S.; van der Stelt, I.; Oosting, A.; Keijer, J.; van Schothorst, E.M. Replacing part of glucose with galactose in the postweaning diet protects female but not male mice from high-fat diet-induced adiposity in later life. J. Nutr. 2019, 149, 1140–1148. [Google Scholar] [CrossRef]
  69. Stahel, P.; Kim, J.J.; Xiao, C.; Cant, J.P. Of the milk sugars, galactose, but not prebiotic galacto-oligosaccharide, improves insulin sensitivity in male Sprague-Dawley rats. PLoS ONE 2017, 12, e0172260. [Google Scholar] [CrossRef]
  70. Salkovic-Petrisic, M.; Osmanovic-Barilar, J.; Knezovic, A.; Hoyer, S.; Mosetter, K.; Reutter, W. Long-term oral galactose treatment prevents cognitive deficits in male Wistar rats treated intracerebroventricularly with streptozotocin. Neuropharmacology 2014, 77, 68–80. [Google Scholar] [CrossRef]
  71. Hamilton, M.K.; Ronveaux, C.C.; Rust, B.M.; Newman, J.W.; Hawley, M.; Barile, D.; Mills, D.A.; Raybould, H.E. Prebiotic milk oligosaccharides prevent development of obese phenotype, impairment of gut permeability, and microbial dysbiosis in high fat-fed mice. Am. J. Physiol. Gastrointest. Liver Physiol. 2017, 312, G474–G487. [Google Scholar] [CrossRef]
  72. Boudry, G.; Hamilton, M.K.; Chichlowski, M.; Wickramasinghe, S.; Barile, D.; Kalanetra, K.M.; Mills, D.A.; Raybould, H.E. Bovine milk oligosaccharides decrease gut permeability and improve inflammation and microbial dysbiosis in diet-induced obese mice. J. Dairy Sci. 2017, 100, 2471–2481. [Google Scholar] [CrossRef] [PubMed]
  73. Yida, Z.; Imam, M.U.; Ismail, M.; Ismail, N.; Azmi, N.H.; Wong, W.; Altine Adamu, H.; Md Zamri, N.D.; Ideris, A.; Abdullah, M.A. N-Acetylneuraminic acid supplementation prevents high fat diet-induced insulin resistance in rats through transcriptional and nontranscriptional mechanisms. BioMed Res. Int. 2015, 2015, 602313. [Google Scholar] [CrossRef]
  74. Castañeda-Gutiérrez, E.; Moser, M.; García-Ródenas, C.; Raymond, F.; Mansourian, R.; Rubio-Aliaga, I.; Viguet-Carrin, S.; Metairon, S.; Ammon-Zufferey, C.; Avanti-Nigro, O.; et al. Effect of a mixture of bovine milk oligosaccharides, Lactobacillus rhamnosus NCC4007 and long-chain polyunsaturated fatty acids on catch-up growth of intra-uterine growth-restricted rats. Acta Physiol 2014, 210, 161–173. [Google Scholar] [CrossRef] [PubMed]
  75. Charbonneau, M.R.; O’Donnell, D.; Blanton, L.V.; Totten, S.M.; Davis, J.C.; Barratt, M.J.; Cheng, J.; Guruge, J.; Talcott, M.; Bain, J.R.; et al. Sialylated Milk Oligosaccharides Promote Microbiota-Dependent Growth in Models of Infant Undernutrition. Cell 2016, 164, 859–871. [Google Scholar] [CrossRef] [PubMed]
  76. Li, T.; Du, M.; Wang, H.; Mao, X. Milk fat globule membrane and its component phosphatidylcholine induce adipose browning both in vivo and in vitro. J. Nutr. Biochem. 2020, 81, 108372. [Google Scholar] [CrossRef] [PubMed]
  77. Li, T.; Yuan, Q.; Gong, H.; Du, M.; Mao, X. Gut microbiota mediates the alleviative effect of polar lipids-enriched milk fat globule membrane on obesity-induced glucose metabolism disorders in peripheral tissues in rat dams. Int. J. Obes. 2022, 46, 793–801. [Google Scholar] [CrossRef]
  78. Han, L.; Du, M.; Ren, F.; Mao, X. Milk Polar Lipids Supplementation to Obese Rats During Pregnancy and Lactation Benefited Skeletal Outcomes of Male Offspring. Mol. Nutr. Food Res. 2021, 65, 2001208. [Google Scholar] [CrossRef]
  79. Zhang, L.; Tian, R.; Yao, X.; Zhang, X.J.; Zhang, P.; Huang, Y.; She, Z.G.; Li, H.; Ji, Y.-X.; Cai, J. Milk fat globule-epidermal growth factor-factor 8 improves hepatic steatosis and inflammation. Hepatology 2021, 73, 586–605. [Google Scholar] [CrossRef]
  80. Yuan, Q.C.; Zhan, B.Y.; Du, M.; Chang, R.; Li, T.G.; Mao, X.Y. Dietary milk fat globule membrane regulates JNK and PI3K/Akt pathway and ameliorates type 2 diabetes in mice induced by a high-fat diet and streptozotocin. J. Funct. Foods 2019, 60, 103435. [Google Scholar] [CrossRef]
  81. Ye, L.; Zhang, Q.; Xin, F.; Cao, B.; Qian, L.; Dong, Y. Neonatal Milk Fat Globule Membrane Supplementation During Breastfeeding Ameliorates the Deleterious Effects of Maternal High-Fat Diet on Metabolism and Modulates Gut Microbiota in Adult Mice Offspring in a Sex-Specific Way. Front. Cell Infect. Microbiol. 2021, 11, 621957. [Google Scholar] [CrossRef]
  82. Milard, M.; Laugerette, F.; Durand, A.; Buisson, C.; Meugnier, E.; Loizon, E.; Louche-Pelissier, C.; Sauvinet, V.; Garnier, L.; Viel, S.; et al. Milk Polar Lipids in a High-Fat Diet Can Prevent Body Weight Gain: Modulated Abundance of Gut Bacteria in Relation with Fecal Loss of Specific Fatty Acids. Mol. Nutr. Food Res. 2019, 63, e1801078. [Google Scholar] [CrossRef]
  83. Yuan, Q.C.; Gong, H.; Du, M.; Mao, X.Y. Supplementation of milk polar lipids to obese dams improves neurodevelopment and cognitive function in male offspring. FASEB J. 2021, 35, e21454. [Google Scholar] [CrossRef]
  84. Teller, I.C.; Hoyer-Kuhn, H.; Brönneke, H.; Nosthoff-Horstmann, P.; Oosting, A.; Lippach, G.; Wohlfarth, M.; Rauh, M.; van der Beek, E.M.; Dötsch, J.; et al. Complex lipid globules in early-life nutrition improve long-term metabolic phenotype in intra-uterine growth-restricted rats. Br. J. Nutr. 2018, 120, 763–776. [Google Scholar] [CrossRef] [PubMed]
  85. 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.; et al. Buttermilk and Krill Oil Phospholipids Improve Hippocampal Insulin Resistance and Synaptic Signaling in Aged Rats. Mol. Neurobiol. 2018, 55, 7285–7296. [Google Scholar] [CrossRef] [PubMed]
  86. Norris, G.H.; Porter, C.M.; Jiang, C.; Millar, C.L.; Blesso, C.N. Dietary sphingomyelin attenuates hepatic steatosis and adipose tissue inflammation in high-fat-diet-induced obese mice. J. Nutr. Biochem. 2017, 40, 36–43. [Google Scholar] [CrossRef]
  87. Yamauchi, I.; Uemura, M.; Hoskawa, M.; Iwashima-Suzuki, A.; Shiota, M.; Miyashita, K. The dietary effect of milk sphingomyelin on the lipid metabolism of obese/diabetic KK-Aymice and wild-type C57BL/6J mice. Food Funct. 2016, 7, 3854–3867. [Google Scholar] [CrossRef]
  88. Yea, K.; Kim, J.; Yoon, J.H.; Kwon, T.; Kim, J.H.; Lee, B.D.; Lee, H.J.; Lee, S.J.; Kim, J.I.; Lee, T.G.; et al. Lysophosphatidylcholine activates adipocyte glucose uptake and lowers blood glucose levels in murine models of diabetes. J. Biol. Chem. 2009, 284, 33833–33840. [Google Scholar] [CrossRef] [PubMed]
  89. Saraswathi, V.; Kumar, N.; Gopal, T.; Bhatt, S.; Ai, W.; Ma, C.; Talmon, G.A.; Desouza, C. Lauric acid versus palmitic acid: Effects on adipose tissue inflammation, insulin resistance, and non-alcoholic fatty liver disease in obesity. Biology 2020, 9, 346. [Google Scholar] [CrossRef]
  90. Venn-Watson, S.; Lumpkin, R.; Dennis, E.A. Efficacy of dietary odd-chain saturated fatty acid pentadecanoic acid parallels broad associated health benefits in humans: Could it be essential? Sci. Rep. 2020, 10, 8161. [Google Scholar] [CrossRef]
  91. Jenkins, B.J.; Seyssel, K.; Chiu, S.; Pan, P.-H.; Lin, S.-Y.; Stanley, E.; Ament, Z.; West, J.A.; Summerhill, K.; Griffin, J.L.; et al. Odd Chain Fatty Acids; New Insights of the Relationship Between the Gut Microbiota, Dietary Intake, Biosynthesis and Glucose Intolerance. Sci. Rep. 2017, 7, 44845. [Google Scholar] [CrossRef]
  92. Bishop, C.A.; Machate, T.; Henkel, J.; Schulze, M.B.; Klaus, S.; Piepelow, K. Heptadecanoic acid is not a key mediator in the prevention of diet-induced hepatic steatosis and insulin resistance in mice. Nutrients 2023, 15, 2052. [Google Scholar] [CrossRef]
  93. Jacome-Sosa, M.M.; Borthwick, F.; Mangat, R.; Uwiera, R.; Reaney, M.J.; Shen, J.; Quiroga, A.D.; Jacobs, R.L.; Lehner, R.; Proctor, S.D. Diets enriched in trans-11 vaccenic acid alleviate ectopic lipid accumulation in a rat model of NAFLD and metabolic syndrome. J. Nutr. Biochem. 2014, 25, 692–701. [Google Scholar] [CrossRef]
  94. Wang, X.; England, A.; Sinclair, C.; Merkosky, F.; Chan, C.B. Trans-11 vaccenic acid improves glucose homeostasis in a model of type 2 diabetes by promoting insulin secretion via GPR40. J. Funct. Foods 2019, 60, 103410. [Google Scholar] [CrossRef]
  95. Wang, X.; Gupta, J.; Kerslake, M.; Rayat, G.; Proctor, S.D.; Chan, C.B. Trans-11 vaccenic acid improves insulin secretion in models of type 2 diabetes in vivo and in vitro. Mol. Nutr. Food Res. 2016, 60, 846–857. [Google Scholar] [CrossRef] [PubMed]
  96. Mohankumar, S.K.; Hanke, D.; Siemens, L.; Cattini, A.; Enns, J.; Shen, J.; Reaney, M.; Zahradka, P.; Taylor, C.G. Dietary supplementation of trans-11-vaccenic acid reduces adipocyte size but neither aggravates nor attenuates obesity-mediated metabolic abnormalities in fa/fa Zucker rats. Br. J. Nutr. 2013, 109, 1628–1636. [Google Scholar] [CrossRef] [PubMed]
  97. Halade, G.V.; Rahman, M.M.; Fernades, G. Differential effects of conjugated linoleic acid isomers in insulin-resistant female C57Bl/6J mice. J. Nutr. Biochem. 2010, 21, 332–337. [Google Scholar] [CrossRef]
  98. Pang, K.; Zhu, Z.; Zhu, S.; Han, L. A high dose of conjugated linoleic acid increases fatty liver and insulin resistance in lactating mice. PLoS ONE 2019, 14, e0214903. [Google Scholar] [CrossRef]
  99. Sain, J.; Scanarotti, I.G.; Gerstner, C.D.; Fariña, A.C.; Lavandera, J.V.; Bernal, C.A. Enriched functional milk fat ameliorates glucose intolerance and triacylglycerol accumulation in skeletal muscle of rats fed high-fat diets. Eur. J. Nutr. 2023, 62, 1535–1550. [Google Scholar] [CrossRef]
  100. Moloney, F.; Toomey, S.; Noone, E.; Nugent, A.; Allan, B.; Loscher, C.; Roche, H. Antidiabetic effects of cis-9, trans-11-conjugated linoleic acid may be mediated via anti-inflammatory effects in white adipose tissue. Diabetes 2007, 56, 574–582. [Google Scholar] [CrossRef]
  101. de Almeida, M.M.; de Souza, Y.O.; Dutra Luquetti, S.C.; Sabarense, C.M.; do Amaral Corrêa, J.O.; da Conceição, E.P.; Lisboa, P.C.; de Moura, E.G.; Andrade Soares, S.M.; Moura Gualberto, A.C.; et al. Cis-9, trans-11 and trans-10, cis-12 CLA mixture does not change body composition, induces insulin resistance and increases serum HDL cholesterol level in rats. J. Oleo Sci. 2015, 64, 539–551. [Google Scholar] [CrossRef]
  102. Kraft, J.; Jetton, T.; Satish, B.; Gupta, D. Dairy-derived bioactive fatty acids improve pancreatic ß-cell function. FASEB J. 2015, 29, 608–625. [Google Scholar] [CrossRef]
  103. Morato, P.N.; Lollo, P.C.; Moura, C.S.; Batista, T.M.; Camargo, R.L.; Carneiro, E.M.; Amaya-Farfan, J. Whey protein hydrolysate increases translocation of GLUT-4 to the plasma membrane independent of insulin in wistar rats. PLoS ONE 2013, 8, e71134. [Google Scholar] [CrossRef]
  104. Gregersen, S.; Bystrup, S.; Overgaard, A.; Jeppesen, P.B.; Sonderstgaard Thorup, A.C.; Jensen, E.; Hermansen, K. Effects of whey proteins on glucose metabolism in normal Wistar rats and Zucker diabetic fatty (ZDF) rats. Rev. Diabet. Stud. 2013, 10, 252–269. [Google Scholar] [CrossRef]
  105. D’Souza, K.; Acquah, C.; Mercer, A.; Paudel, Y.; Pulinilkunnil, T.; Udenigwe, C.C.; Kienesberger, P.C. Whey peptides exacerbate body weight gain and perturb systemic glucose and tissue lipid metabolism in male high-fat fed mice. Food Funct. 2021, 12, 3552–3561. [Google Scholar] [CrossRef] [PubMed]
  106. Gaudel, C.; Nongonierma, A.B.; Maher, S.; Flynn, S.; Krause, M.; Murray, B.A.; Kelly, P.M.; Baird, A.W.; FitzGerald, R.J.; Newsholme, P. A whey protein hydrolysate promotes insulinotropic activity in a clonal pancreatic beta-cell line and enhances glycemic function in ob/ob mice. J. Nutr. 2013, 143, 1109–1114. [Google Scholar] [CrossRef]
  107. Yuan, Q.; Zhan, B.; Chang, R.; Du, M.; Mao, X. Antidiabetic effect of casein glycomacropeptide hydrolysates on high-fat diet and STZ-induced diabetic mice via regulating insulin signaling in skeletal muscle and modulating gut microbiota. Nutrients 2020, 12, 220. [Google Scholar] [CrossRef]
  108. Lillefosse, H.H.; Tastesen, H.S.; Du, Z.Y.; Ditlev, D.B.; Thorsen, F.A.; Madsen, L.; Kristiansen, K.; Liaset, B. Hydrolyzed casein reduces diet-induced obesity in male C57BL/6J mice. J. Nutr. 2013, 143, 1367–1375. [Google Scholar] [CrossRef] [PubMed]
  109. Ogiwara, M.; Ota, W.; Mizushige, T.; Kanamoto, R.; Ohinata, K. Enzymatic digest of whey protein and wheylin-1, a dipeptide released in the digest, increase insulin sensitivity in an Akt phosphorylation-dependent manner. Food Funct. 2018, 9, 4635–4641. [Google Scholar] [CrossRef] [PubMed]
  110. Healy, N.P.; Kirwan, A.M.; McArdle, M.A.; Holohan, K.; Nongonierma, A.B.; Keane, D.; Kelly, S.; Celkova, L.; Lyons, C.L.; McGillicuddy, F.C.; et al. A casein hydrolysate protects mice against high fat diet induced hyperglycemia by attenuating NLRP3 inflammasome-mediated inflammation and improving insulin signaling. Mol. Nutr. Food Res. 2016, 60, 2421–2432. [Google Scholar] [CrossRef]
  111. Liu, L.; Yu, S.; Bu, T.; He, G.; Li, S.; Wu, J. Casein Hydrolysate Alleviates Adipose Chronic Inflammation in High Fat-Diet Induced Obese C57BL/6J Mice through MAPK Pathway. Nutrients 2023, 15, 1813. [Google Scholar] [CrossRef]
  112. Wang, C.; Zheng, L.; Su, G.; Zeng, X.A.; Sun, B.; Zhao, M. Evaluation and Exploration of Potentially Bioactive Peptides in Casein Hydrolysates against Liver Oxidative Damage in STZ/HFD-Induced Diabetic Rats. J. Agric. Food Chem. 2020, 68, 2393–2405. [Google Scholar] [CrossRef]
  113. Mitkin, N.A.; Pavshintcev, V.V.; Sukhanova, I.A.; Doronin, I.I.; Babkin, G.A.; Sadagurski, M.; Malyshev, A.V. The novel peptide Chm-273s has therapeutic potential for metabolic disorders: Evidence from in vitro studies and high-sucrose diet and high-fat diet rodent models. Pharmaceutics 2022, 14, 2088. [Google Scholar] [CrossRef]
  114. Matsunaga, Y.; Tamura, Y.; Sakata, Y.; Nonaka, Y.; Saito, N.; Nakamura, H.; Shimizu, T.; Takeda, Y.; Terada, S.; Hatta, H. Comparison between pre-exercise casein peptide and intact casein supplementation on glucose tolerance in mice fed a high-fat diet. Appl. Physiol. Nutr. Metab. 2018, 43, 355–362. [Google Scholar] [CrossRef]
  115. Sauve, M.F.; Feldman, F.; Koudoufio, M.; Ould-Chikh, N.E.; Ahmarani, L.; Sane, A.; N’Timbane, T.; El-Jalbout, R.; Patey, N.; Spahis, S.; et al. Glycomacropeptide for management of insulin resistance and liver metabolic perturbations. Biomedicines 2021, 9, 1140. [Google Scholar] [CrossRef] [PubMed]
  116. Sawada, Y.; Sakamoto, Y.; Toh, M.; Ohara, N.; Hatanaka, Y.; Naka, A.; Kishimoto, Y.; Kondo, K.; Iida, K. Milk-derived peptide Val-Pro-Pro (VPP) inhibits obesity-induced adipose inflammation via an angiotensin-converting enzyme (ACE) dependent cascade. Mol. Nutr. Food Res. 2015, 59, 2502–2510. [Google Scholar] [CrossRef] [PubMed]
  117. Zheng, L.; Xu, Q.; Lin, L.; Zeng, X.A.; Sun, B.; Zhao, M. In vitro metabolic stability of a casein-derived dipeptidyl peptidase-IV (DPP-IV) inhibitory peptide VPYPQ and its controlled release from casein by enzymatic hydrolysis. J. Agric. Food Chem. 2019, 67, 10604–10613. [Google Scholar] [CrossRef]
  118. Deng, Y.; Misselwitz, B.; Dai, N.; Fox, M. Lactose intolerance in adults: Biological mechanism and dietary management. Nutrients 2015, 7, 8020–8035. [Google Scholar] [CrossRef]
  119. Swallow, D.M. Genetics of lactase persistence and lactose intolerance. Annu. Rev. Genet. 2003, 37, 197–219. [Google Scholar] [CrossRef] [PubMed]
  120. Anguita-Ruiz, A.; Aguilera, C.M.; Gil, Á. Genetics of Lactose Intolerance: An Updated Review and Online Interactive World Maps of Phenotype and Genotype Frequencies. Nutrients 2020, 12, 2689. [Google Scholar] [CrossRef] [PubMed]
  121. Weiss, K.M. The unkindest cup. Lancet 2004, 363, 1489–1490. [Google Scholar] [CrossRef]
  122. Itan, Y.; Powell, A.; Beaumont, M.A.; Burger, J.; Thomas, M.G. The origins of lactase persistence in Europe. PLoS Comput. Biol. 2009, 5, e1000491. [Google Scholar] [CrossRef]
  123. Ahmadi-Abhari, S.; Luben, R.N.; Powell, N.; Bhaniani, A.; Chowdhury, R.; Wareham, N.J.; Forouhi, N.G.; Khaw, K.T. Dietary intake of carbohydrates and risk of type 2 diabetes: The European Prospective Investigation into Cancer-Norfolk study. Br. J. Nutr. 2014, 111, 342–352. [Google Scholar] [CrossRef]
  124. Jaffiol, C. [Milk and dairy products in the prevention and therapy of obesity, type 2 diabetes and metabolic syndrome]. Bull. Acad. Natl. Med. 2008, 192, 749–758. [Google Scholar] [PubMed]
  125. Steudle, J.; Schon, C.; Wargenau, M.; Pauly, L.; Schwejda-Guttes, S.; Gaigg, B.; Kuchinka-Koch, A.; Stover, J.F. Blood glucose response after oral intake of lactulose in healthy volunteers: A randomized, controlled, cross-over study. World J. Gastrointest. Pharmacol. Ther. 2018, 9, 22–30. [Google Scholar] [CrossRef] [PubMed]
  126. Aro, A.; Pelkonen, R.; Leino, U. Glucose and insulin responses to meals containing milk, lactose, glucose or fructose in subjects with non-insulin-dependent diabetes. Diabete Metab. 1987, 13, 603–606. [Google Scholar] [PubMed]
  127. Rosendo-Silva, D.; Viana, S.; Carvalho, E.; Reis, F.; Matafome, P. Are gut dysbiosis, barrier disruption, and endotoxemia related to adipose tissue dysfunction in metabolic disorders? Overview of the mechanisms involved. Intern. Emerg. Med. 2023, 18, 1287–1302. [Google Scholar] [CrossRef]
  128. Cani, P.D.; Neyrinck, A.M.; Fava, F.; Knauf, C.; Burcelin, R.G.; Tuohy, K.M.; Gibson, G.R.; Delzenne, N.M. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia 2007, 50, 2374–2383. [Google Scholar] [CrossRef]
  129. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481. [Google Scholar] [CrossRef]
  130. Teixeira, T.F.; Collado, M.C.; Ferreira, C.L.; Bressan, J.; Peluzio Mdo, C. Potential mechanisms for the emerging link between obesity and increased intestinal permeability. Nutr. Res. 2012, 32, 637–647. [Google Scholar] [CrossRef]
  131. Shen, J.; Obin, M.S.; Zhao, L. The gut microbiota, obesity and insulin resistance. Mol. Asp. Med. 2013, 34, 39–58. [Google Scholar] [CrossRef]
  132. Szilagyi, A. Review article: Lactose--a potential prebiotic. Aliment. Pharmacol. Ther. 2002, 16, 1591–1602. [Google Scholar] [CrossRef]
  133. Salminen, S.; Bouley, C.; Boutron-Ruault, M.C.; Cummings, J.H.; Franck, A.; Gibson, G.R.; Isolauri, E.; Moreau, M.C.; Roberfroid, M.; Rowland, I. Functional food science and gastrointestinal physiology and function. Br. J. Nutr. 1998, 80 (Suppl. 1), S147–S171. [Google Scholar] [CrossRef]
  134. Nath, A.; Haktanirlar, G.; Varga, A.; Molnar, M.A.; Albert, K.; Galambos, I.; Koris, A.; Vatai, G. Biological activities of lactose-derived prebiotics and symbiotic with probiotics on gastrointestinal system. Medicina 2018, 54, 18. [Google Scholar] [CrossRef] [PubMed]
  135. Schaafsma, G. Lactose and lactose derivatives as bioactive ingredients in human nutrition. Int. Dairy J. 2008, 18, 458–465. [Google Scholar] [CrossRef]
  136. Sieber, R.; Stransky, M.; de Vrese, M. Lactose intolerance and consumption of milk and milk products. Z. Ernahrungswiss 1997, 36, 375–393. [Google Scholar] [CrossRef] [PubMed]
  137. Savaiano, D.A. Lactose digestion from yogurt: Mechanism and relevance. Am. J. Clin. Nutr. 2014, 99, 1251S–1255S. [Google Scholar] [CrossRef]
  138. Daly, K.; Darby, A.C.; Hall, N.; Nau, A.; Bravo, D.; Shirazi-Beechey, S.P. Dietary supplementation with lactose or artificial sweetener enhances swine gut Lactobacillus population abundance. Br. J. Nutr. 2014, 111 (Suppl. 1), S30–S35. [Google Scholar] [CrossRef]
  139. Anachad, O.; Taouil, A.; Taha, W.; Bennis, F.; Chegdani, F. The Implication of Short-Chain Fatty Acids in Obesity and Diabetes. Microbiol. Insights 2023, 16, 11786361231162720. [Google Scholar] [CrossRef] [PubMed]
  140. Ruan, Y.; Sun, J.; He, J.; Chen, F.; Chen, R.; Chen, H. Effect of Probiotics on Glycemic Control: A Systematic Review and Meta-Analysis of Randomized, Controlled Trials. PLoS ONE 2015, 10, e0132121. [Google Scholar] [CrossRef]
  141. Meng, H.; Lee, Y.; Ba, Z.; Peng, J.; Lin, J.; Boyer, A.S.; Fleming, J.A.; Furumoto, E.J.; Roberts, R.F.; Kris-Etherton, P.M.; et al. Consumption of Bifidobacterium animalis subsp. lactis BB-12 impacts upper respiratory tract infection and the function of NK and T cells in healthy adults. Mol. Nutr. Food Res. 2016, 60, 1161–1171. [Google Scholar] [CrossRef]
  142. Venema, K. Intestinal fermentation of lactose and prebiotic lactose derivatives, including human milk oligosaccharides. Int. Dairy J. 2012, 22, 123–140. [Google Scholar] [CrossRef]
  143. Ohlsson, J.A.; Johansson, M.; Hansson, H.; Abrahamson, A.; Byberg, L.; Smedman, A.; Lindmark-Månsson, H.; Lundh, Å. Lactose, glucose and galactose content in milk, fermented milk and lactose-free milk products. Int. Dairy J. 2017, 73, 151–154. [Google Scholar] [CrossRef]
  144. Rostami Dovom, M.; Moslehi, N.; Mirmiran, P.; Azizi, F.; Ramezani Tehrani, F. Habitual dietary lactose and galactose intakes in association with age at menopause in non-galactosemic women. PLoS ONE 2019, 14, e0214067. [Google Scholar] [CrossRef]
  145. Yamakawa, M.; Wada, K.; Nakashima, Y.; Nagata, C. Dietary lactose and galactose intakes are associated with a later onset of natural menopause among women in a Japanese community. Br. J. Nutr. 2023, 129, 1607–1614. [Google Scholar] [CrossRef] [PubMed]
  146. Knezovic, A.; Osmanovic Barilar, J.; Babic, A.; Bagaric, R.; Farkas, V.; Riederer, P.; Salkovic-Petrisic, M. Glucagon-like peptide-1 mediates effects of oral galactose in streptozotocin-induced rat model of sporadic Alzheimer’s disease. Neuropharmacology 2018, 135, 48–62. [Google Scholar] [CrossRef] [PubMed]
  147. Adam, T.C.; Westerterp-Plantenga, M.S. Glucagon-like peptide-1 release and satiety after a nutrient challenge in normal-weight and obese subjects. Br. J. Nutr. 2005, 93, 845–851. [Google Scholar] [CrossRef] [PubMed]
  148. Tygstrup, N. Effect of sites of blood sampling in determination of the galactose elimination capacity. Scand. J. Clin. Lab. Invest. 1977, 37, 333–338. [Google Scholar] [CrossRef] [PubMed]
  149. Coelho, A.I.; Berry, G.T.; Rubio-Gozalbo, M.E. Galactose metabolism and health. Curr. Opin. Clin. Nutr. Metab. Care 2015, 18, 422–427. [Google Scholar] [CrossRef]
  150. Holst, J.J. The incretin system in healthy humans: The role of GIP and GLP-1. Metabolism 2019, 96, 46–55. [Google Scholar] [CrossRef] [PubMed]
  151. Unger, J.R.; Parkin, C.G. Glucagon-like peptide-1 (GLP-1) receptor agonists: Differentiating the new medications. Diabetes Ther. 2011, 2, 29–39. [Google Scholar] [CrossRef]
  152. Nauck, M.A.; Quast, D.R.; Wefers, J.; Meier, J.J. GLP-1 receptor agonists in the treatment of type 2 diabetes-state-of-the-art. Mol. Metab. 2021, 46, 101102. [Google Scholar] [CrossRef]
  153. Tao, N.; DePeters, E.J.; German, J.B.; Grimm, R.; Lebrilla, C.B. Variations in bovine milk oligosaccharides during early and middle lactation stages analyzed by high-performance liquid chromatography-chip/mass spectrometry. J. Dairy Sci. 2009, 92, 2991–3001. [Google Scholar] [CrossRef]
  154. Nwosu, C.C.; Aldredge, D.L.; Lee, H.; Lerno, L.A.; Zivkovic, A.M.; German, J.B.; Lebrilla, C.B. Comparison of the human and bovine milk N-glycome via high-performance microfluidic chip liquid chromatography and tandem mass spectrometry. J. Proteome Res. 2012, 11, 2912–2924. [Google Scholar] [CrossRef]
  155. Quinn, E.M.; O’Callaghan, T.F.; Tobin, J.T.; Murphy, J.P.; Sugrue, K.; Slattery, H.; O’Donovan, M.; Hickey, R.M. Changes to the Oligosaccharide Profile of Bovine Milk at the Onset of Lactation. Dairy 2020, 1, 284–296. [Google Scholar] [CrossRef]
  156. Wong, J.M.; de Souza, R.; Kendall, C.W.; Emam, A.; Jenkins, D.J. Colonic health: Fermentation and short chain fatty acids. J. Clin. Gastroenterol. 2006, 40, 235–243. [Google Scholar] [CrossRef]
  157. Nath, A.; Mondal, S.; Csighy, A.; Molnar, M.A.; Pasztorne-Huszar, K.; Kovacs, Z.; Koris, A.; Vatai, G. Biochemical activities of lactose-derived prebiotics-a review. Acta Aliment. 2017, 46, 449–456. [Google Scholar] [CrossRef]
  158. Oliveira, D.L.; Wilbey, R.A.; Grandison, A.S.; Roseiro, L.B. Milk oligosaccharides: A review. Int. J. Dairy Technol. 2015, 68, 305–321. [Google Scholar] [CrossRef]
  159. Westreich, S.T.; Salcedo, J.; Durbin-Johnson, B.; Smilowitz, J.T.; Korf, I.; Mills, D.A.; Barile, D.; Lemay, D.G. Fecal metatranscriptomics and glycomics suggest that bovine milk oligosaccharides are fully utilized by healthy adults. J. Nutr. Biochem. 2020, 79, 108340. [Google Scholar] [CrossRef]
  160. Jensen, R.G. The composition of bovine milk lipids: January 1995 to December 2000. J. Dairy Sci. 2002, 85, 295–350. [Google Scholar] [CrossRef]
  161. Månsson, H.L. Fatty acids in bovine milk fat. Food Nutr. Res. 2008, 52, 1821. [Google Scholar] [CrossRef]
  162. German, J.B.; Dillard, C.J. Composition, structure and absorption of milk lipids: A source of energy, fat-soluble nutrients and bioactive molecules. Crit. Rev. Food Sci. Nutr. 2006, 46, 57–92. [Google Scholar] [CrossRef]
  163. Hirahatake, K.M.; Bruno, R.S.; Bolling, B.W.; Blesso, C.; Alexander, L.M.; Adams, S.H. Dairy Foods and Dairy Fats: New Perspectives on Pathways Implicated in Cardiometabolic Health. Adv. Nutr. 2020, 11, 266–279. [Google Scholar] [CrossRef]
  164. Dewettinck, K.; Rombaut, R.; Thienpont, N.; Le, T.T.; Messens, K.; Van Camp, J. Nutritional and technological aspects of milk fat globule membrane material. Int. Dairy J. 2008, 18, 436–457. [Google Scholar] [CrossRef]
  165. Douellou, T.; Montel, M.C.; Thevenot Sergentet, D. Invited review: Anti-adhesive properties of bovine oligosaccharides and bovine milk fat globule membrane-associated glycoconjugates against bacterial food enteropathogens. J. Dairy Sci. 2017, 100, 3348–3359. [Google Scholar] [CrossRef]
  166. Fontecha, J.; Brink, L.; Wu, S.; Pouliot, Y.; Visioli, F.; Jimenez-Flores, R. Sources, production, and clinical treatments of milk fat globule membrane for infant nutrition and well-being. Nutrients 2020, 12, 1607. [Google Scholar] [CrossRef]
  167. Nie, C.; Zhao, Y.; Wang, X.; Li, Y.; Fang, B.; Wang, R.; Wang, X.; Liao, H.; Li, G.; Wang, P.; et al. Structure, Biological Functions, Separation, Properties, and Potential Applications of Milk Fat Globule Membrane (MFGM): A Review. Nutrients 2024, 16, 587. [Google Scholar] [CrossRef]
  168. Martinez-Sanchez, V.; Calvo, M.V.; Viera, I.; Giron-Calle, J.; Fontecha, J.; Perez-Galvez, A. Mechanisms for the interaction of the milk fat globule membrane with the plasma membrane of gut epithelial cells. Food Res. Int. 2023, 173, 113330. [Google Scholar] [CrossRef]
  169. Singh, H. Symposium review: Fat globules in milk and their structural modifications during gastrointestinal digestion. J. Dairy Sci. 2019, 102, 2749–2759. [Google Scholar] [CrossRef]
  170. Yao, D.; Ranadheera, C.S.; Shen, C.; Wei, W.; Cheong, L.Z. Milk fat globule membrane: Composition, production and its potential as encapsulant for bioactives and probiotics. Crit. Rev. Food Sci. Nutr. 2023, 1–16. [Google Scholar] [CrossRef]
  171. Quarles, W.R.; Pokala, A.; Shaw, E.L.; Ortega-Anaya, J.; Hillmann, L.; Jimenez-Flores, R.; Bruno, R.S. Alleviation of Metabolic Endotoxemia by Milk Fat Globule Membrane: Rationale, Design, and Methods of a Double-Blind, Randomized, Controlled, Crossover Dietary Intervention in Adults with Metabolic Syndrome. Curr. Dev. Nutr. 2020, 4, nzaa130. [Google Scholar] [CrossRef]
  172. Sabio, G.; Kennedy, N.J.; Cavanagh-Kyros, J.; Jung, D.Y.; Ko, H.J.; Ong, H.; Barrett, T.; Kim, J.K.; Davis, R.J. Role of muscle c-Jun NH2-terminal kinase 1 in obesity-induced insulin resistance. Mol. Cell Biol. 2010, 30, 106–115. [Google Scholar] [CrossRef] [PubMed]
  173. Henstridge, D.C.; Bruce, C.R.; Pang, C.P.; Lancaster, G.I.; Allen, T.L.; Estevez, E.; Gardner, T.; Weir, J.M.; Meikle, P.J.; Lam, K.S.L.; et al. Skeletal muscle-specific overproduction of constitutively activated c-Jun N-terminal kinase (JNK) induces insulin resistance in mice. Diabetologia 2012, 55, 2769–2778. [Google Scholar] [CrossRef]
  174. Ning, C.; Wang, X.; Gao, S.; Mu, J.; Wang, Y.; Liu, S.; Zhu, J.; Meng, X. Chicory inulin ameliorates type 2 diabetes mellitus and suppresses JNK and MAPK pathways in vivo and in vitro. Mol. Nutr. Food Res. 2017, 61, 1600673. [Google Scholar] [CrossRef]
  175. Zhao, L.; Du, M.; Gao, J.; Zhan, B.; Mao, X. Label-free quantitative proteomic analysis of milk fat globule membrane proteins of yak and cow and identification of proteins associated with glucose and lipid metabolism. Food Chem. 2019, 275, 59–68. [Google Scholar] [CrossRef]
  176. Venkat, M.; Chia, L.W.; Lambers, T.T. Milk polar lipids composition and functionality: A systematic review. Crit. Rev. Food Sci. Nutr. 2024, 64, 31–75. [Google Scholar] [CrossRef]
  177. Li, J.; Lin, S.; Vanhoutte, P.M.; Woo, C.W.; Xu, A. Akkermansia Muciniphila Protects Against Atherosclerosis by Preventing Metabolic Endotoxemia-Induced Inflammation in Apoe-/- Mice. Circulation 2016, 133, 2434–2446. [Google Scholar] [CrossRef]
  178. Schneeberger, M.; Everard, A.; Gómez-Valadés, A.G.; Matamoros, S.; Ramírez, S.; Delzenne, N.M.; Gomis, R.; Claret, M.; Cani, P.D. Akkermansia muciniphila inversely correlates with the onset of inflammation, altered adipose tissue metabolism and metabolic disorders during obesity in mice. Sci. Rep. 2015, 5, 16643. [Google Scholar] [CrossRef]
  179. Traber, M.G.; Buettner, G.R.; Bruno, R.S. The relationship between vitamin C status, the gut-liver axis, and metabolic syndrome. Redox Biol. 2019, 21, 101091. [Google Scholar] [CrossRef]
  180. Lumeng, C.N.; Deyoung, S.M.; Bodzin, J.L.; Saltiel, A.R. Increased inflammatory properties of adipose tissue macrophages recruited during diet-induced obesity. Diabetes 2007, 56, 16–23. [Google Scholar] [CrossRef]
  181. Yang, Y.; Zhang, T.; Zhou, G.; Jiang, X.; Tao, M.; Zhang, J.; Zeng, X.; Wu, Z.; Pan, D.; Guo, Y. Prevention of Necrotizing Enterocolitis through Milk Polar Lipids Reducing Intestinal Epithelial Apoptosis. J. Agric. Food Chem. 2020, 68, 7014–7023. [Google Scholar] [CrossRef] [PubMed]
  182. Snow, D.R.; Ward, R.E.; Olsen, A.; Jimenez-Flores, R.; Hintze, K.J. Membrane-rich milk fat diet provides protection against gastrointestinal leakiness in mice treated with lipopolysaccharide. J. Dairy Sci. 2011, 94, 2201–2212. [Google Scholar] [CrossRef] [PubMed]
  183. Palmano, K.P.; MacGibbon, A.K.H.; Gunn, C.A.; Schollum, L.M. In Vitro and In Vivo Anti-inflammatory Activity of Bovine Milkfat Globule (MFGM)-derived Complex Lipid Fractions. Nutrients 2020, 12, 2089. [Google Scholar] [CrossRef] [PubMed]
  184. Nagasawa, T.; Nakamichi, H.; Hama, Y.; Higashiyama, S.; Igarashi, Y.; Mitsutake, S. Phytosphingosine is a novel activator of GPR120. J. Biochem. 2018, 164, 27–32. [Google Scholar] [CrossRef]
  185. Ramstedt, B.; Leppimäki, P.; Axberg, M.; Slotte, J.P. Analysis of natural and synthetic sphingomyelins using high-performance thin-layer chromatography. Eur. J. Biochem. 1999, 266, 997–1002. [Google Scholar] [CrossRef] [PubMed]
  186. Djordjevic, J.; Ledina, T.; Baltic, M.Z.; Trbovic, D.; Babic, M.; Bulajic, S. Fatty acid profile of milk. IOP Conf. Ser. Earth Environ. Sci. 2019, 333, 012057. [Google Scholar] [CrossRef]
  187. de Wit, N.; Derrien, M.; Bosch-Vermeulen, H.; Oosterink, E.; Keshtkar, S.; Duval, C.; de Vogel-van den Bosch, J.; Kleerebezem, M.; Müller, M.; van der Meer, R. Saturated fat stimulates obesity and hepatic steatosis and affects gut microbiota composition by an enhanced overflow of dietary fat to the distal intestine. Am. J. Physiol. Gastrointest. Liver Physiol. 2012, 303, G589–G599. [Google Scholar] [CrossRef]
  188. Žáček, P.; Bukowski, M.; Mehus, A.; Johnson, L.; Zeng, H.; Raatz, S.; Idso, J.P.; Picklo, M. Dietary saturated fatty acid type impacts obesity-induced metabolic dysfunction and plasma lipidomic signatures in mice. J. Nutr. Biochem. 2019, 64, 32–44. [Google Scholar] [CrossRef] [PubMed]
  189. Liu, X.; Zeng, X.; Chen, X.; Luo, R.; Li, L.; Wang, C.; Liu, J.; Cheng, J.; Lu, Y.; Chen, Y. Oleic acid protects insulin-secreting INS-1E cells against palmitic acid-induced lipotoxicity along with an amelioration of ER stress. Endocrine 2019, 64, 512–524. [Google Scholar] [CrossRef] [PubMed]
  190. Bhavadharini, B.; Dehghan, M.; Mente, A.; Rangarajan, S.; Sheridan, P.; Mohan, V.; Iqbal, R.; Gupta, R.; Lear, S.; Wentzel-Viljoen, E.; et al. Association of dairy consumption with metabolic syndrome, hypertension and diabetes in 147 812 individuals from 21 countries. BMJ Open Diabetes Res. Care 2020, 8, e000826. [Google Scholar] [CrossRef]
  191. Mozaffarian, D. Dairy foods, dairy fat, diabetes, and death: What can be learned from 3 large new investigations? Am. J. Clin. Nutr. 2019, 110, 1053–1054. [Google Scholar] [CrossRef]
  192. Abdoul-Aziz, S.K.A.; Zhang, Y.; Wang, J. Milk Odd and Branched Chain Fatty Acids in Dairy Cows: A Review on Dietary Factors and Its Consequences on Human Health. Animals 2021, 11, 3210. [Google Scholar] [CrossRef] [PubMed]
  193. Dąbrowski, G.; Konopka, I. Update on food sources and biological activity of odd-chain, branched and cyclic fatty acids –– A review. Trends Food Sci. Technol. 2022, 119, 514–529. [Google Scholar] [CrossRef]
  194. Sun, Q.; Ma, J.; Campos, H.; Hu, F.B. Plasma and erythrocyte biomarkers of dairy fat intake and risk of ischemic heart disease. Am. J. Clin. Nutr. 2007, 86, 929–937. [Google Scholar] [CrossRef]
  195. Krachler, B.; Norberg, M.; Eriksson, J.W.; Hallmans, G.; Johansson, I.; Vessby, B.; Weinehall, L.; Lindahl, B. Fatty acid profile of the erythrocyte membrane preceding development of Type 2 diabetes mellitus. Nutr. Metab. Cardiovasc. Dis. 2008, 18, 503–510. [Google Scholar] [CrossRef]
  196. Jenkins, B.; West, J.A.; Koulman, A.J.M. A review of odd-chain fatty acid metabolism and the role of pentadecanoic acid (C15: 0) and heptadecanoic acid (C17: 0) in health and disease. Molecules 2015, 20, 2425–2444. [Google Scholar] [CrossRef]
  197. Imamura, F.; Fretts, A.; Marklund, M.; Ardisson Korat, A.V.; Yang, W.S.; Lankinen, M.; Qureshi, W.; Helmer, C.; Chen, T.A.; Wong, K.; et al. Fatty acid biomarkers of dairy fat consumption and incidence of type 2 diabetes: A pooled analysis of prospective cohort studies. PLoS Med. 2018, 15, e1002670. [Google Scholar] [CrossRef] [PubMed]
  198. Santaren, I.D.; Watkins, S.M.; Liese, A.D.; Wagenknecht, L.E.; Rewers, M.J.; Haffner, S.M.; Lorenzo, C.; Hanley, A.J. Serum pentadecanoic acid (15:0), a short-term marker of dairy food intake, is inversely associated with incident type 2 diabetes and its underlying disorders. Am. J. Clin. Nutr. 2014, 100, 1532–1540. [Google Scholar] [CrossRef] [PubMed]
  199. Luo, C.; Liu, H.; Wang, X.; Xia, L.; Huang, H.; Peng, X.; Xia, C.; Liu, L. The associations between individual plasma SFAs, serine palmitoyl-transferase long-chain base subunit 3 gene rs680379 polymorphism, and type 2 diabetes among Chinese adults. Am. J. Clin. Nutr. 2021, 114, 704–712. [Google Scholar] [CrossRef]
  200. Fu, W.C.; Li, H.Y.; Li, T.T.; Yang, K.; Chen, J.X.; Wang, S.J.; Liu, C.H.; Zhang, W. Pentadecanoic acid promotes basal and insulin-stimulated glucose uptake in C2C12 myotubes. Food Nutr. Res. 2021, 65. [Google Scholar] [CrossRef] [PubMed]
  201. McCall, A.L. Glucose Transport. In Stress: Physiology, Biochemistry, and Pathology; Fink, G., Ed.; Academic Press: Cambridge, MA, USA, 2019; pp. 293–307. [Google Scholar]
  202. Venn-Watson, S.; Schork, N.J. Pentadecanoic Acid (C15:0), an Essential Fatty Acid, Shares Clinically Relevant Cell-Based Activities with Leading Longevity-Enhancing Compounds. Nutrients 2023, 15, 4607. [Google Scholar] [CrossRef]
  203. Pipoyan, D.; Stepanyan, S.; Stepanyan, S.; Beglaryan, M.; Costantini, L.; Molinari, R.; Merendino, N. The Effect of Trans Fatty Acids on Human Health: Regulation and Consumption Patterns. Foods 2021, 10, 2452. [Google Scholar] [CrossRef]
  204. Wang, Y.; Jacome-Sosa, M.M.; Ruth, M.R.; Lu, Y.; Shen, J.; Reaney, M.J.; Scott, S.L.; Dugan, M.E.; Anderson, H.D.; Field, C.J.; et al. The intestinal bioavailability of vaccenic acid and activation of peroxisome proliferator-activated receptor-α and -γ in a rodent model of dyslipidemia and the metabolic syndrome. Mol. Nutr. Food Res. 2012, 56, 1234–1246. [Google Scholar] [CrossRef]
  205. Tholstrup, T. Dairy products and cardiovascular disease. Curr. Opin. Lipidol. 2006, 17, 1–10. [Google Scholar] [CrossRef] [PubMed]
  206. Tardy, A.L.; Lambert-Porcheron, S.; Malpuech-Brugère, C.; Giraudet, C.; Rigaudière, J.P.; Laillet, B.; Leruyet, P.; Peyraud, J.L.; Boirie, Y.; Laville, M.; et al. Dairy and industrial sources of trans fat do not impair peripheral insulin sensitivity in overweight women. Am. J. Clin. Nutr. 2009, 90, 88–94. [Google Scholar] [CrossRef] [PubMed]
  207. Badawy, S.; Liu, Y.; Guo, M.; Liu, Z.; Xie, C.; Marawan, M.A.; Ares, I.; Lopez-Torres, B.; Martínez, M.; Maximiliano, J.E.; et al. Conjugated linoleic acid (CLA) as a functional food: Is it beneficial or not? Food Res. Int. 2023, 172, 113158. [Google Scholar] [CrossRef] [PubMed]
  208. Sun, X.; Wang, Y.; Ma, X.; Li, S.; Wang, W. Producing natural functional and low-carbon milk by regulating the diet of the cattle-The fatty acid associated rumen fermentation, biohydrogenation, and microorganism response. Front. Nutr. 2022, 9, 955846. [Google Scholar] [CrossRef] [PubMed]
  209. Chinnadurai, K.; Tyagi, A. Conjugated Linoleic Acid: A Milk Fatty Acid with Unique Health Benefit Properties. In Soybean and Health; Hany, E.-S., Ed.; IntechOpen: Rijeka, Croatia, 2011; Chapter 6. [Google Scholar]
  210. Rastgoo, S.; Shimi, G.; Shiraseb, F.; Karbasi, A.; Ashtary-Larky, D.; Yousefi, M.; Golalipour, E.; Asbaghi, O.; Zamani, M. The effects of conjugated linoleic acid supplementation on inflammatory cytokines and adipokines in adults: A GRADE-assessed systematic review and dose-response meta-analysis. Front. Immunol. 2023, 14, 1092077. [Google Scholar] [CrossRef]
  211. Suksatan, W.; Putera, H.D.; Abdulkadhim, A.H.; Hammid, A.T.; Ismailov, J.A.; Jannat, B.; Parvizi, R.; Izadi, F. The effect of conjugated linoleic acid supplementation on oxidative stress markers: A systematic review and meta-analysis of randomized controlled trials. Clin. Nutr. ESPEN 2022, 49, 121–128. [Google Scholar] [CrossRef]
  212. Kim, B.; Lim, H.R.; Lee, H.; Lee, H.; Kang, W.; Kim, E. The effects of conjugated linoleic acid (CLA) on metabolic syndrome patients: A systematic review and meta-analysis. J. Funct. Foods 2016, 25, 588–598. [Google Scholar] [CrossRef]
  213. Riserus, U.; Vessby, B.; Arner, P.; Zethelius, B. Supplementation with trans10cis12-conjugated linoleic acid induces hyperproinsulinaemia in obese men: Close association with impaired insulin sensitivity. Diabetologia 2004, 47, 1016–1019. [Google Scholar] [CrossRef]
  214. Mohankumar, S.K.; Taylor, C.G.; Siemens, L.; Zahradka, P. Acute exposure of L6 myotubes to cis-9, trans-11 and trans-10, cis-12 conjugated linoleic acid isomers stimulates glucose uptake by modulating Ca2+/calmodulin-dependent protein kinase II. Int. J. Biochem. Cell Biol. 2012, 44, 1321–1330. [Google Scholar] [CrossRef]
  215. Chai, B.K.; Al-Shagga, M.; Pan, Y.; Then, S.M.; Ting, K.N.; Loh, H.S.; Mohankumar, S.K. Cis-9, Trans-11 Conjugated Linoleic Acid Reduces Phosphoenolpyruvate Carboxykinase Expression and Hepatic Glucose Production in HepG2 Cells. Lipids 2019, 54, 369–379. [Google Scholar] [CrossRef]
  216. Taormina, V.M.; Unger, A.L.; Schiksnis, M.R.; Torres-Gonzalez, M.; Kraft, J. Branched-Chain Fatty Acids—An Underexplored Class of Dairy-Derived Fatty Acids. Nutrients 2020, 12, 2875. [Google Scholar] [CrossRef]
  217. Shingfield, K.J.; Chilliard, Y.; Toivonen, V.; Kairenius, P.; Givens, D.I. Trans fatty acids and bioactive lipids in ruminant milk. Adv. Exp. Med. Biol. 2008, 606, 3–65. [Google Scholar] [CrossRef]
  218. Vetter, W.; Schröder, M. Concentrations of phytanic acid and pristanic acid are higher in organic than in conventional dairy products from the German market. Food Chem. 2010, 119, 746–752. [Google Scholar] [CrossRef]
  219. Schröder, M.; Lutz, N.L.; Tangwan, E.C.; Hajazimi, E.; Vetter, W. Phytanic acid concentrations and diastereomer ratios in milk fat during changes in the cow’s feed from concentrate to hay and back. Eur. Food Res. Technol. 2012, 234, 955–962. [Google Scholar] [CrossRef]
  220. Mika, A.; Stepnowski, P.; Kaska, L.; Proczko, M.; Wisniewski, P.; Sledzinski, M.; Sledzinski, T. A comprehensive study of serum odd- and branched-chain fatty acids in patients with excess weight. Obesity 2016, 24, 1669–1676. [Google Scholar] [CrossRef]
  221. Pakiet, A.; Wilczynski, M.; Rostkowska, O.; Korczynska, J.; Jabłonska, P.; Kaska, L.; Proczko-Stepaniak, M.; Sobczak, E.; Stepnowski, P.; Magkos, F.; et al. The Effect of One Anastomosis Gastric Bypass on Branched-Chain Fatty Acid and Branched-Chain Amino Acid Metabolism in Subjects with Morbid Obesity. Obes. Surg. 2020, 30, 304–312. [Google Scholar] [CrossRef] [PubMed]
  222. Su, X.; Magkos, F.; Zhou, D.; Eagon, J.C.; Fabbrini, E.; Okunade, A.L.; Klein, S. Adipose tissue monomethyl branched-chain fatty acids and insulin sensitivity: Effects of obesity and weight loss. Obesity 2015, 23, 329–334. [Google Scholar] [CrossRef]
  223. Utzschneider, K.M.; Prigeon, R.L.; Faulenbach, M.V.; Tong, J.; Carr, D.B.; Boyko, E.J.; Leonetti, D.L.; McNeely, M.J.; Fujimoto, W.Y.; Kahn, S.E. Oral disposition index predicts the development of future diabetes above and beyond fasting and 2-h glucose levels. Diabetes Care 2009, 32, 335–341. [Google Scholar] [CrossRef]
  224. Garcia Caraballo, S.C.; Comhair, T.M.; Houten, S.M.; Dejong, C.H.; Lamers, W.H.; Koehler, S.E. High-protein diets prevent steatosis and induce hepatic accumulation of monomethyl branched-chain fatty acids. J. Nutr. Biochem. 2014, 25, 1263–1274. [Google Scholar] [CrossRef]
  225. Liu, L.; Xiao, D.; Lei, H.; Peng, T.; Li, J.; Cheng, T.; He, J. Branched-chain fatty acids lower triglyceride levels in a fatty liver model in vitro. FASEB J. 2017, 31, 969.4. [Google Scholar] [CrossRef]
  226. Ran-Ressler, R.R.; Khailova, L.; Arganbright, K.M.; Adkins-Rieck, C.K.; Jouni, Z.E.; Koren, O.; Ley, R.E.; Brenna, J.T.; Dvorak, B. Branched chain fatty acids reduce the incidence of necrotizing enterocolitis and alter gastrointestinal microbial ecology in a neonatal rat model. PLoS ONE 2011, 6, e29032. [Google Scholar] [CrossRef] [PubMed]
  227. Yan, Y.; Wang, Z.; Greenwald, J.; Kothapalli, K.S.; Park, H.G.; Liu, R.; Mendralla, E.; Lawrence, P.; Wang, X.; Brenna, J.T. BCFA suppresses LPS induced IL-8 mRNA expression in human intestinal epithelial cells. Prostaglandins Leukot. Essent. Fat. Acids 2017, 116, 27–31. [Google Scholar] [CrossRef] [PubMed]
  228. Davoodi, S.H.; Shahbazi, R.; Esmaeili, S.; Sohrabvandi, S.; Mortazavian, A.; Jazayeri, S.; Taslimi, A. Health-Related Aspects of Milk Proteins. Iran. J. Pharm. Res. 2016, 15, 573–591. [Google Scholar] [PubMed]
  229. Auestad, N.; Layman, D.K. Dairy bioactive proteins and peptides: A narrative review. Nutr. Rev. 2021, 79, 36–47. [Google Scholar] [CrossRef]
  230. Mohammadi, S.; Asbaghi, O.; Dolatshahi, S.; Omran, H.S.; Amirani, N.; Koozehkanani, F.J.; Garmjani, H.B.; Goudarzi, K.; Ashtary-Larky, D. Effects of supplementation with milk protein on glycemic parameters: A GRADE-assessed systematic review and dose-response meta-analysis. Nutr. J. 2023, 22, 49. [Google Scholar] [CrossRef] [PubMed]
  231. Amirani, E.; Milajerdi, A.; Reiner, Ž.; Mirzaei, H.; Mansournia, M.A.; Asemi, Z. Effects of whey protein on glycemic control and serum lipoproteins in patients with metabolic syndrome and related conditions: A systematic review and meta-analysis of randomized controlled clinical trials. Lipids Health Dis. 2020, 19, 209. [Google Scholar] [CrossRef] [PubMed]
  232. Nouri, M.; Pourghassem Gargari, B.; Tajfar, P.; Tarighat-Esfanjani, A. A systematic review of whey protein supplementation effects on human glycemic control: A mechanistic insight. Diabetes Metab. Syndr. 2022, 16, 102540. [Google Scholar] [CrossRef] [PubMed]
  233. Chiang, S.W.; Liu, H.W.; Loh, E.W.; Tam, K.W.; Wang, J.Y.; Huang, W.L.; Kuan, Y.C. Whey protein supplementation improves postprandial glycemia in persons with type 2 diabetes mellitus: A systematic review and meta-analysis of randomized controlled trials. Nutr. Res. 2022, 104, 44–54. [Google Scholar] [CrossRef] [PubMed]
  234. Gobbetti, M.; Stepaniak, L.; De Angelis, M.; Corsetti, A.; Di Cagno, R. Latent bioactive peptides in milk proteins: Proteolytic activation and significance in dairy processing. Crit. Rev. Food Sci. Nutr. 2002, 42, 223–239. [Google Scholar] [CrossRef]
  235. Holt, C.; Carver, J.A.; Ecroyd, H.; Thorn, D.C. Invited review: Caseins and the casein micelle: Their biological functions, structures, and behavior in foods. J. Dairy Sci. 2013, 96, 6127–6146. [Google Scholar] [CrossRef]
  236. Kitts, D.D.; Weiler, K. Bioactive proteins and peptides from food sources. Applications of bioprocesses used in isolation and recovery. Curr. Pharm. Des. 2003, 9, 1309–1323. [Google Scholar] [CrossRef] [PubMed]
  237. Lahov, E.; Regelson, W. Antibacterial and immunostimulating casein-derived substances from milk: Casecidin, isracidin peptides. Food Chem. Toxicol. 1996, 34, 131–145. [Google Scholar] [CrossRef]
  238. Meisel, H. Biochemical properties of regulatory peptides derived from milk proteins. Biopolymers 1997, 43, 119–128. [Google Scholar] [CrossRef]
  239. Dziuba, J.; Minkiewicz, P.; Nałecz, D.; Iwaniak, A. Database of biologically active peptide sequences. Nahrung 1999, 43, 190–195. [Google Scholar] [CrossRef]
  240. Meisel, H. Multifunctional peptides encrypted in milk proteins. Biofactors 2004, 21, 55–61. [Google Scholar] [CrossRef]
  241. Marcone, S.; Belton, O.; Fitzgerald, D.J. Milk-derived bioactive peptides and their health promoting effects: A potential role in atherosclerosis. Br. J. Clin. Pharmacol. 2017, 83, 152–162. [Google Scholar] [CrossRef] [PubMed]
  242. Nakamura, Y.; Yamamoto, N.; Sakai, K.; Okubo, A.; Yamazaki, S.; Takano, T. Purification and characterization of angiotensin I-converting enzyme inhibitors from sour milk. J. Dairy Sci. 1995, 78, 777–783. [Google Scholar] [CrossRef]
  243. Boelsma, E.; Kloek, J. IPP-rich milk protein hydrolysate lowers blood pressure in subjects with stage 1 hypertension, a randomized controlled trial. Nutr. J. 2010, 9, 52. [Google Scholar] [CrossRef]
  244. Cadee, J.A.; Chang, C.Y.; Chen, C.W.; Huang, C.N.; Chen, S.L.; Wang, C.K. Bovine casein hydrolysate (c12 Peptide) reduces blood pressure in prehypertensive subjects. Am. J. Hypertens. 2007, 20, 1–5. [Google Scholar] [CrossRef]
  245. Cicero, A.F.; Gerocarni, B.; Laghi, L.; Borghi, C. Blood pressure lowering effect of lactotripeptides assumed as functional foods: A meta-analysis of current available clinical trials. J. Hum. Hypertens. 2011, 25, 425–436. [Google Scholar] [CrossRef]
  246. de Leeuw, P.W.; van der Zander, K.; Kroon, A.A.; Rennenberg, R.M.; Koning, M.M. Dose-dependent lowering of blood pressure by dairy peptides in mildly hypertensive subjects. Blood Press. 2009, 18, 44–50. [Google Scholar] [CrossRef] [PubMed]
  247. Chen, Y.C.; Smith, H.A.; Hengist, A.; Chrzanowski-Smith, O.J.; Mikkelsen, U.R.; Carroll, H.A.; Betts, J.A.; Thompson, D.; Saunders, J.; Gonzalez, J.T. Co-ingestion of whey protein hydrolysate with milk minerals rich in calcium potently stimulates glucagon-like peptide-1 secretion: An RCT in healthy adults. Eur. J. Nutr. 2020, 59, 2449–2462. [Google Scholar] [CrossRef]
  248. Geerts, B.F.; van Dongen, M.G.; Flameling, B.; Moerland, M.M.; de Kam, M.L.; Cohen, A.F.; Romijn, J.A.; Gerhardt, C.C.; Kloek, J.; Burggraaf, J. Hydrolyzed casein decreases postprandial glucose concentrations in T2DM patients irrespective of leucine content. J. Diet. Suppl. 2011, 8, 280–292. [Google Scholar] [CrossRef]
  249. Zhou, S.; Xu, T.; Zhang, X.; Luo, J.; An, P.; Luo, Y. Effect of Casein Hydrolysate on Cardiovascular Risk Factors: A Systematic Review and Meta-Analysis of Randomized Controlled Trials. Nutrients 2022, 14, 4207. [Google Scholar] [CrossRef]
  250. Elbira, A.; Hafiz, M.; Hernández-Álvarez, A.J.; Zulyniak, M.A.; Boesch, C. Protein Hydrolysates and Bioactive Peptides as Mediators of Blood Glucose-A Systematic Review and Meta-Analysis of Acute and Long-Term Studies. Nutrients 2024, 16, 323. [Google Scholar] [CrossRef] [PubMed]
  251. Isolauri, E.; Sütas, Y.; Mäkinen-Kiljunen, S.; Oja, S.S.; Isosomppi, R.; Turjanmaa, K. Efficacy and safety of hydrolyzed cow milk and amino acid-derived formulas in infants with cow milk allergy. J. Pediatr. 1995, 127, 550–557. [Google Scholar] [CrossRef] [PubMed]
  252. Maryniak, N.Z.; Sancho, A.I.; Hansen, E.B.; Bøgh, K.L. Alternatives to Cow’s Milk-Based Infant Formulas in the Prevention and Management of Cow’s Milk Allergy. Foods 2022, 11, 926. [Google Scholar] [CrossRef]
  253. Tenenbaum, M.; Dugardin, C.; Moro, J.; Auger, J.; Baniel, A.; Boulier, A.; Ravallec, R.; Cudennec, B. In vitro comparison of whey protein isolate and hydrolysate for their effect on glucose homeostasis markers. Food Funct. 2023, 14, 4173–4182. [Google Scholar] [CrossRef]
  254. Manders, R.J.; Wagenmakers, A.J.; Koopman, R.; Zorenc, A.H.; Menheere, P.P.; Schaper, N.C.; Saris, W.H.; van Loon, L.J. Co-ingestion of a protein hydrolysate and amino acid mixture with carbohydrate improves plasma glucose disposal in patients with type 2 diabetes. Am. J. Clin. Nutr. 2005, 82, 76–83. [Google Scholar] [CrossRef]
  255. Wang, C.; Luo, D.; Zheng, L.; Zhao, M. Anti-diabetic mechanism and potential bioactive peptides of casein hydrolysates in STZ/HFD-induced diabetic rats. J. Sci. Food Agric. 2024, 104, 2947–2958. [Google Scholar] [CrossRef]
  256. Iwasa, M.; Takezoe, S.; Kitaura, N.; Sutani, T.; Miyazaki, H.; Aoi, W. A milk casein hydrolysate-derived peptide enhances glucose uptake through the AMP-activated protein kinase signalling pathway in skeletal muscle cells. Exp. Physiol. 2021, 106, 496–505. [Google Scholar] [CrossRef]
  257. Ramzan, I.; Ardavani, A.; Vanweert, F.; Mellett, A.; Atherton, P.J.; Idris, I. The Association between Circulating Branched Chain Amino Acids and the Temporal Risk of Developing Type 2 Diabetes Mellitus: A Systematic Review & Meta-Analysis. Nutrients 2022, 14, 4411. [Google Scholar] [CrossRef] [PubMed]
  258. Siddik, M.A.B.; Shin, A.C. Recent Progress on Branched-Chain Amino Acids in Obesity, Diabetes, and Beyond. Endocrinol. Metab. 2019, 34, 234–246. [Google Scholar] [CrossRef] [PubMed]
  259. Tremblay, F.; Lavigne, C.; Jacques, H.; Marette, A. Role of dietary proteins and amino acids in the pathogenesis of insulin resistance. Annu. Rev. Nutr. 2007, 27, 293–310. [Google Scholar] [CrossRef]
  260. Gannon, N.P.; Schnuck, J.K.; Vaughan, R.A. BCAA Metabolism and Insulin Sensitivity-Dysregulated by Metabolic Status? Mol. Nutr. Food Res. 2018, 62, e1700756. [Google Scholar] [CrossRef] [PubMed]
  261. Rietman, A.; Schwarz, J.; Tomé, D.; Kok, F.J.; Mensink, M. High dietary protein intake, reducing or eliciting insulin resistance? Eur. J. Clin. Nutr. 2014, 68, 973–979. [Google Scholar] [CrossRef]
  262. Abdualkader, A.M.; Karwi, Q.G.; Lopaschuk, G.D.; Al Batran, R. The role of branched-chain amino acids and their downstream metabolites in mediating insulin resistance. J. Pharm. Pharm. Sci. 2024, 27, 13040. [Google Scholar] [CrossRef]
  263. Bloomgarden, Z. Diabetes and branched-chain amino acids: What is the link? J. Diabetes 2018, 10, 350–352. [Google Scholar] [CrossRef]
  264. White, P.J.; McGarrah, R.W.; Herman, M.A.; Bain, J.R.; Shah, S.H.; Newgard, C.B. Insulin action, type 2 diabetes, and branched-chain amino acids: A two-way street. Mol. Metab. 2021, 52, 101261. [Google Scholar] [CrossRef]
  265. Carniero de Campos Zani, S.; Son, M.; Bhullar, K.; Chan, C.; Wu, J. IRW improves glucose tolerance in high fat diet fed C57BL/6 mice via activation of insulin signaling and AMPK pathways in skeletal muscle. Biomedicines 2022, 10, 1235. [Google Scholar] [CrossRef]
  266. Jahandideh, F.; de Campos Zani, S.C.; Davidge, S.T.; Proctor, S.D.; Chan, C.B.; Wu, J. Egg white hydrolysate enhances insulin sensitivity in high fat diet induced insulin resistant rats via AKT activation. Br. J. Nutr. 2019, 122, 14–24. [Google Scholar] [CrossRef] [PubMed]
  267. Gerosa, S.; Skoet, J. Milk Availability: Trends in Production and Demand and Medium-Term Outlook; FAO: Rome, Italy, 2012. [Google Scholar]
  268. Potočnik, K.; Gantner, V.; Kuterovac, K.; Cividini, A. Mare’s milk: Composition and protein fraction in comparison with different milk species. Mljekarstvo 2011, 61, 107–113. [Google Scholar]
  269. Guha, S.; Sharma, H.; Deshwal, G.K.; Rao, P.S. A comprehensive review on bioactive peptides derived from milk and milk products of minor dairy species. Food Prod. Process. Nutr. 2021, 3, 2. [Google Scholar] [CrossRef]
  270. Anusha Siddiqui, S.; Mahmood Salman, S.H.; Ali Redha, A.; Zannou, O.; Chabi, I.B.; Oussou, K.F.; Bhowmik, S.; Nirmal, N.P.; Maqsood, S. Physicochemical and nutritional properties of different non-bovine milk and dairy products: A review. Int. Dairy J. 2024, 148, 105790. [Google Scholar] [CrossRef]
  271. Flis, Z.; Molik, E. Importance of Bioactive Substances in Sheep’s Milk in Human Health. Int. J. Mol. Sci. 2021, 22, 4364. [Google Scholar] [CrossRef]
  272. dos Santos, W.M.; Guimarães Gomes, A.C.; de Caldas Nobre, M.S.; de Souza Pereira, Á.M.; dos Santos Pereira, E.V.; dos Santos, K.M.O.; Florentino, E.R.; Alonso Buriti, F.C. Goat milk as a natural source of bioactive compounds and strategies to enhance the amount of these beneficial components. Int. Dairy J. 2023, 137, 105515. [Google Scholar] [CrossRef]
  273. Mirmiran, P.; Ejtahed, H.S.; Angoorani, P.; Eslami, F.; Azizi, F. Camel Milk Has Beneficial Effects on Diabetes Mellitus: A Systematic Review. Int. J. Endocrinol. Metab. 2017, 15, e42150. [Google Scholar] [CrossRef]
  274. Han, B.; Zhang, L.; Hou, Y.; Zhong, J.; Hettinga, K.; Zhou, P. Phosphoproteomics reveals that camel and goat milk improve glucose homeostasis in HDF/STZ-induced diabetic rats through activation of hepatic AMPK and GSK3-GYS axis. Food Res. Int. 2022, 157, 111254. [Google Scholar] [CrossRef]
  275. Almohmadi, W. Camel Milk as an Antidiabetic Agent: A Review of the Impact of In Vitro Digestion and Pasteurization on Glucose Regulatory Hormones. Ph.D. Dissertation, Graduate Faculty of North Carolina State University, Raleigh, NC, USA, 2020. [Google Scholar]
  276. Arain, M.A.; Khaskheli, G.B.; Shah, A.H.; Marghazani, I.B.; Barham, G.S.; Shah, Q.A.; Khand, F.M.; Buzdar, J.A.; Soomro, F.; Fazlani, S.A. Nutritional significance and promising therapeutic/medicinal application of camel milk as a functional food in human and animals: A comprehensive review. Anim. Biotechnol. 2023, 34, 1988–2005. [Google Scholar] [CrossRef]
  277. Verruck, S.; Dantas, A.; Prudencio, E.S. Functionality of the components from goat’s milk, recent advances for functional dairy products development and its implications on human health. J. Funct. Foods 2019, 52, 243–257. [Google Scholar] [CrossRef]
Figure 1. A summary of the evidence that bioactive compounds from milk can regulate glucose homeostasis. Abbreviations: BW, body weight; GTT, glucose tolerance test; IR, insulin resistance; AKT, protein kinase B; FBG, fasting blood (plasma) glucose; FM, fat mass; GLUT4, glucose transporter-4. Created with BioRender.com (accessed on 30 August 2024).
Figure 1. A summary of the evidence that bioactive compounds from milk can regulate glucose homeostasis. Abbreviations: BW, body weight; GTT, glucose tolerance test; IR, insulin resistance; AKT, protein kinase B; FBG, fasting blood (plasma) glucose; FM, fat mass; GLUT4, glucose transporter-4. Created with BioRender.com (accessed on 30 August 2024).
Foods 13 02837 g001
Table 1. Results of experimental studies in humans investigating the use of cow’s milk bioactive molecules on glucoregulation-related outcomes.
Table 1. Results of experimental studies in humans investigating the use of cow’s milk bioactive molecules on glucoregulation-related outcomes.
Obesity PhenotypeBlood Glucose Parameters
ImproveNeutral/WorseImproveNeutral/Worse
Carbohydrate bioactives
Lactose--BMI [38]GTT [39,40]FBG, PPG [38]
Galactose----G-Ra [41,42]FPG, HOMA-IR [41]
PPG [41,43]
GTT [44,45]
FPG [42,46]
Oligosaccharides—No human studies
Lipid bioactives
Milk fat globule membrane--BMI [47]GTT [48]FBG [47,48,49,50]
GTT [50]
HOMA-IR [47]
Polar lipidsWC [51]BW [52]--FBG [51,52]
HOMA-IR [52]
Even-chain fatty acids—No human studies
Odd-chain fatty acidsBW, FM [53]--FBG, HOMA-IR [53]--
Trans fatty acidsCLA: BW or BMI, FM (MA [54,55])----CLA: FBG, HOMA-IR (MA [56])
RA: FBG [57,58]
RA: Clamp [58]
Branched-chain fatty acids—No human studies
Protein bioactives
Whey hydrolysateBW [59]--FBG [60,61]HOMA-IR [62]
GTT [61,63]
HbA1c [63]
CGMS [60]
Casein hydrolysate----FBG [60,61]
GTT [61]
CGMS [60]
Peptides—No human studies
Abbreviations: G-Ra, glucose rate of appearance; BW, body weight; BMI, body mass index; FM, fat mass; FBG, fasting blood glucose; PPG, postprandial glucose; GTT, glucose tolerance test; HOMA-IR, Homeostatic Model Assessment for Insulin Resistance; MA, meta-analysis; CGMS, continuous glucose monitoring system, -- no data. This table provides a summary of relevant literature but does not represent a comprehensive systematic review.
Table 2. Results of experimental studies in animal models investigating the use of cow’s milk bioactive molecules on glucoregulation-related outcomes.
Table 2. Results of experimental studies in animal models investigating the use of cow’s milk bioactive molecules on glucoregulation-related outcomes.
Obesity PhenotypeBlood GlucoseInsulin Signaling
ImproveNeutral/WorseImproveNeutral/WorseImproveNeutral/Worse
Carbohydrate bioactives
LactoseBW [64,65,66]--FPG [64,65]------
FM [65] GTT [67]
GalactoseBW, FM [68]BW, FM [69]FPG [70]FPG [68]Irs2 [68]--
BW, FM [68]Clamp [69]FPG, HOMA-IR [68]
GTT, HOMA-IR [68]
OligosaccharidesFM [71]BW [72]GTT [73]FBG [72]Pi3k, Irs2 [73]--
BW [69,71,73,74] FPG [75]GTT, HOMA-IR [74]
Clamp [69]
Lipid bioactives
Milk fat globule membraneBW [76,77,78,79]BW [80,81]FBG [76,77,79,80]--Pi3k, Akt [80]--
FM [77] GTT [76,77,80,81] PI3K, p-AKT) [77]
IRS, AKT [78]
AMPK, AKT [79]
IP [78]
Milk polar lipidsBW [82,83,84]BW [52,85,86,87]FBG [82,84,88]FBG [52,86](IRS, AKT [85]--
GTT [88]HOMA-IR [52,84,86]
HOMA-IR [85]
Even-chain fatty acids--BW [89]--FBG, GTT [89] --
Odd-chain fatty acidsBW [90,91]BW [92]FBG [90]FBG [92]----
GTT [91,92]
Trans fatty acidsVA: BW, FM [93]VA: BW [94,95,96]VA: FBG, Clamp [94,95]VA: FBG, HOMA-IR, GTT [96] VA: IR [95]--
VA: FBG, HOMA-IR [93]
RA: BW, FM [97]RA: BW [98,99,100] RA: FBG, HOMA-IR, ITT [98]RA: Ir, Irs [100]
RA: FBG, GTT [99]
RA: FBG, HOMA-IR, QUICKI, GTT [97]
RA: FBG, HOMA-IR) [100]
CLA: BW, FM [101] CLA: FBG, HOMA-IR, R-QUICKI [101]
Branched-chain fatty acids--------PDX1, PPAR-γ [102]--
Protein bioactives
Whey hydrolysate--BW [103,104]HbA1c [104]HOMA-IR [105,106]GLUT-4 [103]--
GTT [104,106]IP [106]
Casein hydrolysateBW [107,108,109]BW [110,111]GTT [107,109,110,112]GTT [108]p-AKT [109,110]--
FBG [107,108,111,112] GLUT-4, AKT, IRS-1 [107]
ITT [109]ITT [110]
HbA1c [112]
HOMA-IR [111]
Bioactive peptidesBW [113,114]BW [115,116]GTT [113,116,117]GTT [115]p-AKT [115]--
HOMA-IR [113,114]HOMA-IR [115]
FBG [113,114] ITT [116]FBG [116]
BW, body weight; FM, fat mass; FBG, fasting blood (or plasma) glucose; GTT, glucose tolerance test; ITT, insulin tolerance test; HOMA-IR, Homeostatic Model Assessment for Insulin Resistance; IP, insulin production; IR and Ir, insulin receptor; IRS and Irs, insulin receptor substrate protein and gene; --, no data. This table provides a summary of relevant literature but does not represent a comprehensive systematic review.
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yuzbashian, E.; Berg, E.; de Campos Zani, S.C.; Chan, C.B. Cow’s Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies. Foods 2024, 13, 2837. https://doi.org/10.3390/foods13172837

AMA Style

Yuzbashian E, Berg E, de Campos Zani SC, Chan CB. Cow’s Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies. Foods. 2024; 13(17):2837. https://doi.org/10.3390/foods13172837

Chicago/Turabian Style

Yuzbashian, Emad, Emily Berg, Stepheny C. de Campos Zani, and Catherine B. Chan. 2024. "Cow’s Milk Bioactive Molecules in the Regulation of Glucose Homeostasis in Human and Animal Studies" Foods 13, no. 17: 2837. https://doi.org/10.3390/foods13172837

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop