Effect of Consumption of Animal Products on the Gut Microbiome Composition and Gut Health (2024)

As a library, NLM provides access to scientific literature. Inclusion in an NLM database does not imply endorsem*nt of, or agreement with, the contents by NLM or the National Institutes of Health.
Learn more: PMC Disclaimer | PMC Copyright Notice

Effect of Consumption of Animal Products on the Gut MicrobiomeComposition and Gut Health (1)

Food Science of Animal Resources

Food Sci Anim Resour. 2023 Sep; 43(5): 723–750.

Published online 2023 Sep 1. doi:10.5851/kosfa.2023.e44

PMCID: PMC10493557

PMID: 37701742

Author information Article notes Copyright and License information PMC Disclaimer

Abstract

The gut microbiome is critical in human health, and various dietary factorsinfluence its composition and function. Among these factors, animal products,such as meat, dairy, and eggs, represent crucial sources of essential nutrientsfor the gut microbiome. However, the correlation and characteristics oflivestock consumption with the gut microbiome remain poorly understood. Thisreview aimed to delineate the distinct effects of meat, dairy, and egg productson gut microbiome composition and function. Based on the previous reports, theimpact of red meat, white meat, and processed meat consumption on the gutmicrobiome differs from that of milk, yogurt, cheese, or egg products. Inparticular, we have focused on animal-originated proteins, a significantnutrient in each livestock product, and revealed that the major proteins in eachfood elicit diverse effects on the gut microbiome. Collectively, this reviewhighlights the need for further insights into the interactions and mechanismsunderlying the impact of animal products on the gut microbiome. A deeperunderstanding of these interactions would be beneficial in elucidating thedevelopment of dietary interventions to prevent and treat diseases linked to thegut microbiome.

Keywords: animal products, gut microbiome, meat, dairy products, egg products

Introduction

The gut microbiome is a complex ecosystem comprising trillions of microorganisms thatplay a crucial role in human and animal health (Bäckhed et al., 2012; Lee et al.,2022; Oh et al., 2021). Diet is amodulator of the gut microbiome, and animal products cause changes in gut microbiomecomposition and function.

Animal products, such as meat, dairy, and eggs, are excellent sources of protein,vitamins, and minerals, all of which are essential or beneficial for human health(Hess et al., 2016; Puglisi and Fernandez, 2022; Udenigwe and Aluko, 2012; Wyness,2016). Animal products contain higher essential amino acid levels thanplant proteins (Day et al., 2022). Theseessential amino acids are derived from animal products and can be metabolized by gutbacteria to produce branched-chain amino acids that are important for the risk oftype 2 diabetes (Gojda and Cahova, 2021;Madsen et al., 2017). Menaquinone (MK) isknown to help stop and reverse bone loss and is contained mainly in the form of MK-4in meat, eggs, and dairy products (Walther et al.,2013).

Meat is a prominent source of high-quality protein and essential nutrients such asiron, zinc, and vitamin B12 (Ahmad et al.,2018). Some studies have demonstrated that moderate meat consumption andits proteins compensate for iron deficiency and positively affect the gutmicrobiome, such as a high abundance of Lactobacillus (Krebs et al., 2013; Zhu et al., 2015).

Dairy products refer to foods made from the milk of mammals, such as cows, goats, andsheep, and can include milk, cheese, yogurt, butter, and cream. Dairy products havelong been recognized as essential for a healthy and balanced diet because they arehigh in casein, calcium, and vitamins (Ortega etal., 2019). Nowadays, research has been conducted on the addition ofother substances, such as feeding probiotic culture fluids to cows or addingflaxseed to their feed, to improve milk yield or quality (Ababakri et al., 2021; Lim etal., 2021). Consumption of dairy products reduces cardiometabolic riskfactors in diet-induced obese mice (Perazza et al.,2020). These dairy products, such as milk, cheese, yogurt, and kefir,contain probiotics that can improve gut health by increasing the abundance ofbeneficial bacteria such as Bifidobacterium pseudolongum andLactococcus lactis (Aslam etal., 2020; Farag et al., 2020;Zhao et al., 2019a).

Eggs and egg products are nutrient-dense food containing various proteins, essentialamino acids, vitamins, and minerals, such as vitamin D, vitamin B12, choline, andselenium. These nutrients play an important role in supporting homeostasis in thebody, such as brain and nervous system function, metabolism, and immune function(Eckert et al., 2013). In addition, eggsand egg products contain nutrients and bioactive compounds, such as vitamin D andphospholipids, which improve gut health by reducing inflammation (Puglisi and Fernandez, 2022).

Moreover, eggs and egg products have been shown to increase gut microbial metabolitesthat help maintain a healthy gut lining, such as butyrate and propionic acid. Thehigh protein content in eggs provides a source of essential amino acids for thegrowth and maintenance of beneficial bacteria in the gut (Ge et al., 2021; Liu et al.,2022a).

Notably, the effects of animal products on the gut microbiome are complex and maydepend on the type of animal product and the individual’s gut microbiomecomposition. However, studies summarizing the effects of animal products andproteins on the gut microbiome are limited. In this review, we discussed the effectsof meat, dairy, and eggs, which are major animal products, and their processedproducts on gut microbiome composition and function. Furthermore, the effect ofproteins, a major nutritional component of animal products, on the gut microbiomewas summarized. Human studies were mainly summarized in this review, and the partslacking in human studies were reviewed based on animal studies.

Effects of Meat Products and Meat Protein on the Gut Microbiota

Meat products

The gut microbiome is significantly influenced by diet. In particular, meatproduct consumption affected gut microbiota composition (Table 1). Animal-based diets affect gut microbiotadifferently than plant-based diets (Muegge etal., 2011; Walker et al.,2011; Wu et al., 2011). Dependingon the type of diet, the gut microbiota can change even in a short time, such as3 days. There was no change in alpha-diversity for either animal- or plant-baseddiet, but the animal-based diet significantly improved beta-diversity (David et al., 2014). Another study found nosignificant differences in either the alpha-or beta-diversity of the gutmicrobiota between the animal- and plant-based diets, but did affect thecomposition of the gut microbiota (Kohnert etal., 2021). The meat-rich diet increased Roseburia,Faecalibacterium, and Blautia more thanthe strict vegan diet (Kohnert et al.,2021). High animal protein intake and low carbohydrates also affectedthe reduction of Bacteroides spp. while reducing the abundanceof the butyrate producer Roseburia/Eubacteriumrectale (Russell et al.,2011).

Table 1.

Human and animal studies assessing the effect of meat productconsumption on the composition of gut microbiota

CitationStudy designInterventionTrial durationParticipantsEffect on the gut microbiota
Kohnert et al. (2021)Randomized, controlled trialMeat-rich diet; >150 g ofmeat/d Strict vegan diet4 wk53 Healthy adults 18–60yrIn meat-rich group
Nochange in alpha- or beta-diversity CoprococcusRoseburiaFaecalibacteriumBlautia
Russell et al. (2011)Randomized, crossover trialMaintenance diet (M); 13%protein and 50% carbohydrates
High-proteinand moderate-carbohydrate diet (HPMC); 28% protein and35% carbohydrate
High-protein andlow-carbohydrate (HPLC); 29% protein and 5%carbohydrate
4 wk17 Healthy males 21–74 yrBody mass index (BMI) 27.88–48.48In HPLCgroup
Roseburia/EubacteriumrectaleBacteroides spp.↓
Foerster et al. (2014)Randomized, crossover trialLow-meat high-fiber period; wholegrain products, 40 g of fiber/d
High-meatlow-fiber period; 200 g of red meat/d
3 wk20 Healthy adults 20–60yrIn high-meat low-fiber periodClostridium sp. ↓
Hentges et al. (1977)Crossover trialHigh-beef diet; 380 g ofbeef/d
Meatless diet; 360 g milk; 150 g egg; 145 gcheese; 100 g peas
4 wk10 Healthy malesIn high-beefdiet
Genus Bacteroides
Species BacteroidesfragilisBifidobacteriumadolescentis
Zhaoet al. (2019b)Crossover, controlled trialBeef-based diet Chicken-baseddiet2 wk45 Healthy males 18–27yrIn beef-baseddiet
LachnospiraLachnospiraceae NK4A136 group ↓Ruminococcus 2 ↓
Dhakalet al. (2022)Randomized, crossover, controlledtrialPork group; 156 g pork/d Chickengroup; 156 g chicken/d10 d50 Healthy older adults66.44±7.44 yr BMI 29.8±5.59In both groups
Nochange in alpha- or beta-diversity
PhylumBacteriodetes
FamilyBacteriodaceaeChristencellaceae
Inpork group
Ruminiclostridium 5↑
In chickengroup
Roseburia
Shi etal. (2021)Cross-sectional studyNot applicable (NA)NA40 Healthy males 18–30 yr;chicken-eaters (n=20) and pork-eaters (n=20)In pork-eatergroup
Chao and Shannon index↓
Phylum FirmicutesBacteroidetes
Genus Clostridiales, Bacteroides,Firmicutes, Lachnospiraceae, Faecalibacterium, Roseburia,Ruminococcus 2, and Blautia
In chicken-eatergroup
Phylum FirmicutesBacteroidetes
GenusPrevotellaceae, Prevotella 9,Bacteroidales, Dialister, Prevotella 2,Ruminococcaceae UCG 002,Lactobacillus, andOlsenella
Sinhaet al. (2021)Dietary intervention (participantswere blinded to which group they were assigned)First phase (2 wk): conventionalprocessed meats (Diet A)
Second phase (2 wk):poultry (i.e., chicken and turkey) (Diet B)
Thirdphase (2 wk): Group 1) conventional processed meat supplementedwith natural phytochemical compounds (DietC)
Group 2) low-nitrite processed meatsupplemented with phytochemical compounds (DietD)
Final phase (1 wk): nitrate-enriched water withdiet A, B, C, or D
7 wk63 Healthy volunteers (participantsof each sex were randomly assigned to one of the twoexperimental groups)
(ages 18–70, in goodhealth, with a BMI between 18 and 25 kg/m2)
NA
Gao etal. (2021)Randomized controlled-feedingtrialControl group: restricted from friedmeat intake (n=58)
Fried meat group: friedmeat four times per week (n=59)
4 wk117 Participants, (18–35 yrold, BMI>24 kg/m2, and consumption of friedfood more than one time per wk)FamilyLachnospiraceae
Genus FlavonifractorDialisterDoreaVeillonella
Thøgersen et al. (2018)Chow,
Conventionalfrankfurter sausage
Inulin-enriched frankfurtersausage4 wk30 Healthy Sprague-Dawley ratsPhylumActinobacteria
Family unclassifiedLachnospiraceaeErysipelotrichaceae
Genus Bifidobacterium
Species Bacteroidesuniformis
Thøgersen et al. (2020)Control sausageSausage+inulin and calciumSausage+inulin Sausage+calcium4 wk48 Healthy Sprague-Dawley ratsPhylum Firmicutes↑ (Sausage+calcium, Sausage+inulin andcalcium) Bacteroidetes ↓(Sausage+calcium, Sausage+inulin and calcium)Proteobacteria,Cyanobacteria ↓(Sausage+inulin and calcium)Deferribacteres ↑(Sausage+calcium) Actinobacteria↑ (Sausage+inulin and calcium)
GenusMuribaculaceae, uncultivatedRuminococcaceae ↑(Sausage+Inulin) Clostridium 6,Staphylococcus, uncultivatedRuminococcaceae (PAC000661) ↑(Sausage+calcium) uncultivated genus (PAC002482),Parabacteroides,Alistipes, Clostridium,unclassified Peptostreptococcaceae ↓(Sausage+calcium)
Bifidobacterium,Staphylococcus, Blautia,uncultivated Ruminococcaceae (PAC000661),uncultivated Erysipelotrichaceae (CCMM),Faecalibaculum ↑(Sausage+inulin andcalcium)
Bacteroides,Parabacteroides,Alistipes, Clostridium,Dorea, uncultivatedBacteroidetes (PAC002482),Muribaculaceae (PAC001472),Lachnospiraceae (KE159600), unclassifiedFirmicutes ↓ (Sausage+inulinand calcium)
Lactobacillus↑ (q=0.63; Sausage+calcium,Sausage+inulin and calcium)
Fernandez et al. (2020)Control diet (universal feed)Acorn-fed Iberian commercialham2 wk20 Male Fischer 344 rats (5 wk old)(ulcerative colitis was induced with dextran sulfate sodium indrinking water ad libitum for 1 wk)Phylum Bacteroidetes,Actinobacteria, ProteobacteriaFirmicutes, Synergistetes, Deferribacteres
Family Coriobacteriaceae,Bacteroidaceae, Porphyromonadaceae, Rikenellaceae,Desulfovibrionaceae, Sutterellaceae, Staphylococcaceae,Enterococcaceae, Clostridiaceae FamilyXIII, Eubacteriaceae, Acidaminococcaceae,Erysipelotrichaceae, Enterobacteriaceae
Marinifilaceae,Prevotellaceae, Sphingobacteriaceae,Ruminococcaceae, Lachnospiraceae,Clostridiaceae,Veillonellaceae, Lactobacillaceae,Cohaesibacteriaceae
GenusBacteroides, Butyricimonas, Parabacteroides,Alistipes, Staphylococcus, Enterococcus, Blautia, Dorea,Absiella, Phascolarctobacterium, Parasutterella,Bilophila
Prevotella, Mucispirillum,Lactobacillus, Clostridium, Lachnoanaerobaculum,Ruminococcus, Oscillibacter, Desulfovibrio

Meat is a major component of an animal-based diet. Meat is classified as follows:red meat (beef and pork), white meat (chicken and fish), and processed meat.Recent epidemiological studies have reported higher mortality risks and certainchronic diseases in groups that consume more red and processed meat (Al-Shaar et al., 2020; Petermann-Rocha et al., 2021). Moreover, the WHO classifiedred meat as group 2A, a probable carcinogen (Bouvard et al., 2015). In a review of more than 800 epidemiologicalstudies, >10 studies found a 17% increased risk of colorectalcancer (CRC) for every 100 g increase in red meat intake. Nevertheless, researchon the effect of meat product consumption on the gut microbiome remains unclear,and evidence on the effect on health is insufficient.

Consumption of red meat affects the gut microbiome. Consuming 200 g of red meatdaily significantly reduced Clostridium sp. (Foerster et al., 2014). However, findingson the health effect of Clostridium sp. are inconsistentbecause the changes that occur at the species level are very distinct. Inaddition, consuming 380 g of beef for 4 weeks affected the reduction ofdiarrheal disease-related Bacteroidesfragilis and the increase of intestinal movement regulatorBifidobacterium adolescentis (Hentges et al., 1977). Meanwhile, there are studiescomparing the effects of red and white meat consumption on the gut microbiome. Abeef-based diet increased the relative abundance ofLachnospira, the Lachnospiraceae NK4A136group, and Ruminococcus 2, whereas the chicken-based diet didnot show significant changes (Zhao et al.,2022). After consuming a beef-based diet, Akkermansiamuciniphila was reduced and blood cell counts were elevated,altering markers associated with inflammation. Replacing the beef-based dietwith a chicken-based diet reduced inflammation-related monocytes and basophils.This appears to be related to the chicken-based diet’s suppression ofBacteroides ovatus, a factor in generating immunoglobulinA. In an intervention study in which pork or chicken was consumed, both groupsaltered the gut microbiota in a similar pattern, with reductions in theBacteroidetes phylum, Bacteroidaceae, andChristencellaceae families (Dhakal et al., 2022). However, in a cross-sectional study observingpork and chicken eaters, high Bacteroidetes levels were foundin the chicken eaters, and differences were observed at the species level (Shi et al., 2021).

It is well-recognized that meat consumption is harmful to persons with diseases.Red meat may increase the gut microbiota’s production of uremic toxinssuch as trimethylamine (TMA) n-oxide (TMAO), indoxyl sulfate, and p-cresylsulfate. These uremic toxins are linked to a higher risk of cardiovascular death(Mafra et al., 2018). In addition,red meat, unlike white meat, is associated with CRC (Sasso and Latella, 2018). This is mainly because of thered-colored heme iron found in large quantities in the muscle myoglobin of redmeat. Heme iron causes direct harm, such as causing cytotoxic damage to colonicepithelial cells, and indirect harm by inducing alterations of the gutmicrobiota (Ijssennagger et al., 2012). Ared meat diet rich in heme iron increases Streptococcusbovis, Fusobacterium,Clostridium, and Helicobacterpylori, which are related to colorectal carcinogenesis (Sasso and Latella, 2018). However, it isunclear whether the increase or decrease in the gut microbiome caused by certainsubstances, such as toxins and heme iron, contributes to diseases.

Meanwhile, in inflammatory bowel disease (IBD)-induced mouse experiments,high-dose red meat induced intestinal microbial imbalance and reduced therelative abundances ofLachnospiraceae_NK4A136_group,Faecalibaculum, Blautia, andDubosiella (Li et al.,2021). This results from a study that contradicts the previoushealthy human study in which beef intake increasedLachnospiraceae_NK4A136_group.Lachnospiraceae_NK4A136_group is abutyrate-producing bacteria that protects the intestinal mucosal and reducesinflammation (Li et al., 2021; Zhao et al., 2022).

Taken together, red, white, and processed meats are all observed to havedifferent effects on the gut microbiome. However, there are currentlyinsufficient studies that compile adequate information to determine consistentchanges in the gut microbiome. In addition, like other food consumption,excessive meat consumption has also been linked in studies to the emergence ofdisease, albeit the exact mechanisms related to alterations in the gutmicrobiome are unknown.

Processed meat products

The WHO classified processed meats as group 1 carcinogens (Bouvard et al., 2015). The consumption of processed meat isknown to increase the incidence of CRC. N-nitroso compounds, such as nitratesused as preservatives in processed meat, are considered the leading cause ofCRC. Moreover, certain microorganisms in the gut can reduce nitrate to nitritethrough metabolic processes (González-Soltero et al., 2020). However, a diet containingprocessed meat and an intake of nitrate-rich water did not show significantchanges in the fecal microbiome (Sinha et al.,2021).

One human study confirmed the correlation between fried meat intake and gutmicrobiota. A randomized controlled trial was conducted for 4 weeks, and the gutmicrobiota of 59 participants in the group treated with fried meat and 58participants in the control group with restricted fried meat intake werecompared. In the gut microbiome of the fried meat intake group,Lachnospiraceae and Flavonifractordecreased, and Dialister, Dorea, andVeillonella increased (Gaoet al., 2021).

There are two studies in rats on the correlation between consuming sausage andthe modulation of gut microbiota. In one study, rats were fed for 4 weeks on oneof three diets: inulin-fortified pork sausage, control pork sausage, or standardchow diet. Rats in the inulin-fortified pork sausage group had increasedabundances of Actinobacteria, unclassifiedLachnospiraceae, Erysipelotrichaceae, Bifidobacterium, andBacteroides uniformis. In addition, unlike conventionalsausages, the sausage fortified with inulin showed similar effects to thegeneral dietary fiber intake, such as increasing short-chain fatty acid (SCFA)and Bifidobacteria (Thøgersen et al., 2018). In a subsequent study, rats were fedfor 4 weeks on one of four diets: control sausage, sausage with added inulin andcalcium, sausage with added inulin, and sausage with added calcium. In the gutmicrobiota of the two groups of rats fed the calcium-rich sausage andcalcium-added inulin-fortified sausage, Firmicutes increased,and Bacteroidetes decreased at the phylum level. At the genuslevel, Ruminococcaceae and Staphylococcus wereincreased in both groups, and in particular, Bifidobacteriawere increased in the intestinal microflora of rats in the group fed sausagescalcium-rich and fortified with inulin (Thøgersen et al., 2020).

Acorn-fed Iberian ham is a traditional cured meat product. A study was conductedon changes in gut microbiota caused by an acorn-fed ham diet using rats as anexperimental model. The acorn-fed ham diet had a lower carbohydrate content andhigher protein content than that of the control diet, resulting in increasedproteolytic metabolism-related Bacteroidetes andProteobacteria and decreased saccharolyticmetabolism-related Firmicutes at the phylum level. In addition,Dorea, Phascolarctobacterium, orButyricimonas, which are highly related to ulcerativecolitis disease, were decreased at the genus level (Fernández et al., 2020).

In conclusion, consumption of different types of processed meats can causechanges in the gut microbiome. However, processed meat consumption was notassociated with adverse changes in the gut microbiome, such as an increase inpathogenic bacteria or dysbiosis. These results show that there is no clearcausal link between processed meat consumption and adverse health effects ofchanges in the gut microbiome.

Meat protein

Proteins can show different changes in the composition of the intestinalmicrobiota depending on the type, such as plant and animal origin. Animalfood-originated protein is a great source of nutrients and can affect thecomposition of the gut microbiome (Lang et al.,2018). However, no human study has directly tested the effect of meatprotein intake on the modulation of the gut microbiome; however, there have beenanimal studies.

In one study using a rat model, the effect of various dietary proteins, such asplant (soy), dairy (casein), red meat (beef and pork), and white meat (chickenand fish) proteins, on the composition of gut microbiota was assessed (Zhu et al., 2015). At the phylum level,the group fed white meat protein had a higher abundance ofFirmicutes but lower Bacteroidetes thanthe other groups. Conversely, the chicken protein-fed group had a higherabundance of Actinobacteria, and the rats fed beef protein hada higher abundance of Proteobacteria. At the genus level,Ruminococcaceae and Lactobacillaceae wereabundant in the group fed red meat protein, andLactobacillaceae were more abundant in the group fed whitemeat protein. Consumption of meat protein reduced serumlipopolysaccharide-binding protein (LBP) compared to soy protein. Typically, thepresence of LBP in the blood is regarded as a biomarker of an inflammatoryreaction. This may be related to the composition of the gut microbiome.Bacteroidetes are major lipopolysaccharide (LPS)-producingbacteria, which are reduced by consuming meat proteins. Therefore, casein andmeat protein intake may help maintain gut microbiome balance and reduce antigenload and inflammatory response. In addition, when comparing each group, meatproteins, mainly white meat proteins, contained moreLactobacillus than non-meat proteins (casein, soy).Lactobacillus is well known as representative probioticbacteria. In other words, the recommended level of meat protein intake couldhelp the growth of beneficial intestinal bacteria such asLactobacillus (Zhu et al.,2015).

White meat-originated protein consumption has been linked to alterations in thegut microbiome in two rat model studies. Zhu et al. compared the feces of4-week-old and 64-week-old rats fed a chicken protein diet. The results showedthat the gut microbiome composition differed significantly between young andmiddle-aged mice. At the phylum level, Firmicutes decreased formiddle-aged rats but increased for young rats. However, the level ofBacteroidetes increased for middle-aged rats and decreasedfor young rats. At the genus level, the relative abundance of the beneficialbacterium Lactobacillus increased by chicken protein in theyoung group. In contrast, it had the opposite effect in the middle-aged group(Zhu et al., 2016a). In anotherstudy, the relative abundance of Lactobacillus was higher inthe chicken protein group than in the casein control group; it also showed thehighest levels of organic acids, including lactate, which can promote the growthof Lactobacillus (Zhu et al.,2017).

A. muciniphila is considered a next-generation probioticbacteria (Ross, 2022). It playssignificant roles in lipid metabolism and enhances intestinal immune function toprevent obesity, IBD, and diabetes (Rodrigues etal., 2022). In a study that compared the effects of soy and chickenprotein-based diet intake on the composition of intestinal microbiota using agerm-free mice model, compared to a soy protein-based diet, a chickenprotein-based diet helped the growth of A. muciniphila andmaintained mucus barrier function and intestinal homeostasis (Zhao et al., 2019b).

Overall, red meat proteins can modulate the alteration of the gut microbiome in adirection that reduces LPS-producing bacteria. White meat protein may helpmaintain gut homeostasis by increasing Lactobacillus orA. muciniphila levels. However, numerous perspectives onthe gut microbiota of meat protein consumption have been reported in priorresearch. Therefore, more research is required to provide clear scientificevidence.

Effects of Dairy Products and Dairy Protein on the Gut Microbiota

Dairy products

Investigations have examined how dairy products, including milk, yogurt, andcheese, affect the gut microbiome (Table2). Two studies reported the effects of the quantity of dairyproducts on the gut microbiota. Swarte et al.(2020) divided 46 healthy overweight adults into high-dairy diet(HDD) and a low-dairy diet (LDD) groups for 6 weeks. HDD showed relativelyhigher Streptococcus, Leuconostoc, andLactococcus abundances and lowerFaecalibacterium and Bilophila abundances.At the species level, the abundances of Streptococcusthermophilus and Leuconostoc mesenteroides wereincreased; however, Faecalibacterium prausnitzii andClostridium aldenense were decreased. Predicted metabolicpathways were also studied; however, there were no significant changes by HDD(Swarte et al., 2020). Alternatively,a randomized controlled trial was performed in one human study for 24 weeks;however, there was no significant change in gut microbiota composition ordiversity according to the difference in dairy intake (Bendtsen et al., 2018).

Table 2.

Human and animal studies assessing the effect of dairy productconsumption on the composition of gut microbiota

CitationStudy designInterventionTrial durationParticipantsEffect on the gut microbiotaOther health or physiologicalobservations
Swarteet al. (2020)Randomized, cross-over trialQuantity of dairy High-dairy diet(HDD); 5–6 dairy portion/d Low-dairy diet (LDD);≤1 dairy portion/d6 wk46 Healthy overweight participants45–65 yr
Body mass index (BMI)25–30
In HDD Genus Streptococcus,Leuconostoc, and LactococcusFaecalibacterium andBilophila
SpeciesStreptococcus thermophilusErysipelatoclostridium ramosumLeuconostoc mesenteroidesFaecalibacterium prausnitziiClostridium aldenenseAcetivibrio ethanolgignensBilophila wadsworthiaLactococcus lactis
Predicted metabolic pathways werenot significantly altered due to a HDD
Bendtsen et al. (2018)Randomized, controlled, paralleltrialQuantity of dairy High dairy (HD);1,500 mg calcium/d Low dairy (LD); 600 mg calcium/d24 wk80 Overweight or obese participants18–60 yr BMI 28–36; 40 consumed HD and 40 consumedLDNo significant taxonomic changes inphylum and genus level
No significant changes inalpha- or beta-diversity Veillonella ↓in LD (vs baseline)
In both groups Body weight ↓(vs baseline) Fat mass ↓ (vsbaseline)
Respiratory quotient (RQ) ↓ in HDRQ ↑ in LD
Fernandez-Raudales et al. (2012)Randomized, double blind trialBovine milk, 500 mL/d3 mon64 Male participants 20–45 yrBMI 25–44; 24 consumed bovine milkPhylumProteobacteria
GenusLactobacillus ↑ (vsbaseline)
Alpha-diversity ACE ↓ Chao1↓
NA
Alvaroet al. (2007)Cross-sectional studyYogurt 200–400 g/dNot applicable (NA)51 Healthy participants 35–60yr; 30 consumed yogurtEnterobacteriaceae↓ No significant difference in short-chain fatty acid(SCFA) concentrationβ-galactosidase ↑
González et al. (2019)Cross-sectional studyNANA130 Healthy participants mean age of58.18Natural yogurt consumersAkkermansia
Sweetened yogurt consumersBacteroides
Cheeseconsumers SCFA (acetate, propionate and butyrate) ↑
Serum levels of C-reactive protein(CRP) were also significantly reduced in yogurt consumers
Le Royet al. (2022)Cross-sectional studyYogurt 125 g at least once a wkNA4,117 Adult participants mean age of67.6 yrBifidobacteriumanimalis subsp. lactis
Streptococcusthermophilus
Visceral fat mass ↓
Tillisch et al. (2013)Randomized, controlled, paralleltrialFermented milk 2×125 g/d4 wk36 Healthy female participants18–55 yr; 12 consumed fermented milkNo significant changes in the gutmicrobiota composition after the interventionActivity of brain regions thatcontrol central processing of emotion and sensation was affectedby intervention
Liskoet al. (2017)Parallel trialYogurt 250 g/d6 wk6 Healthy participants 18–54yrNo significant changes in the gutmicrobiota composition and diversityNA
Link-Amster et al. (1994)Randomized, controlled trialFermented milk 3×125 g/d3 wk30 Healthy adult participantsF=14, M=16 19–59 yr; 16 consumed fermentedmilkGenus LactobacillusBifidobacterium
Species Lactobacillusacidophilus
Serum IgA and IgG ↑
Volokhet al. (2019)Before and after trialYogurt 2×125 g/d30 d150 Healthy adult participants18–40 yr BMI 18–28No significant change inalpha-diversity after intervention
GenusBifidobacteriumLachnoclostridium/unclassified↓
Roseburia
Species B. bifidum, B.adolescentis,B. animalis, B. bifidum,B. longumAdlercreutziaequolifaciensSlackiaisoflavoniconvertensCollinsellaaerofaciensCatenibacterium.mitsuokaiStreptococcus.thermophilus/vestibularis
NA
Burtonet al. (2017)Randomized, double-blind, cross-overtrialYogurt 400 g/d2 wk14 Healthy male participants22–27 yr; 7 consumed yogurtLactobacillusdelbrueckii spp. bulgaricus
Streptococcussalivarius spp. thermophilus
Tumor necrosis factor (TNF)α,interleukin (IL)-6 and C-C motif chemokine ligand 5 (CCL5)↓ (vs baseline)
Alvarezet al. (2020)Randomized, double-blind,controlled, parallel trialYogurt 100 g/d or 3×100g/d4 wk96 Healthy adult participants18–55 yr BMI 18.5–30.0; 25 consumed 100 g/d yogurtand 24 consumed 3×100 g/d yogurtLactobacillusparacaseiLactobacillusrhamnosus ↑ No significant difference ineither alpha- or beta-diversityNo clinically significant changes indefecation frequency, stool consistency scores, composite scoreand frequency of digestive symptoms (abdominal pain, bloating,flatulence and rumbling) or vital signs
García-Albiach et al. (2008)Randomized, double-blind, cross-overtrialYogurt 3×125 g/d2 wk79 Healthy young participants meanage of 23.6 yr; 32 consumed fresh yogurtLactic acid bacteria ↑Clostridium perfringensBacteroidesNA
Unnoet al. (2015)Before and after trialFermented milk 2×140mL/d3 wk6 Healthy female participants20–24 yrPhylum FirmicutesBacteroidetes ↓ Alpha-diversity(Shannon index) ↓NA
Yangand Sheu (2012)Parallel trialYogurt 200 mL/d4 wk38 Helicobacterpylori-infected and 38 healthy children 4–12yrE. coliBifidobacterium spp. ↑Bifidobacterium spp./E.coli ratio ↑In H. pyloriinfected children 13C-Urea breath test ↓ (vsbaseline) IgA ↑ IL-6 ↓
Veigaet al. (2014)Randomized, double blind,controlled, parallel trialFermented milk 2×125 g/d4 wk28 Female inflammatory bowel diseasepatients 20–69 yr; 13 consumed fermented milkBilophilawadsworthia ↓ Butyrate-producing bacteria↑ SCFA ↑NA
Yılmaz et al. (2019)Randomized, controlled, open-label,parallel trialKefir 400 mL/d4 wk45 Inflammatory bowel diseasepatients; 10 Crohn disease (CD) patients and 15 ulcerativecolitis patients consumed kefirLactobacillusIn CD erythrocyte sedimentation rate↓ CRP ↓ hemoglobin ↑ bloating ↓feeling good scores ↑
Bellikci-Koyu et al. (2019)Randomized, controlled, paralleltrialKefir 180 mL/d12 wk22 Metabolic syndrome patients18–65 yr; 12 consumed kefirActinobacteria↑ No significant difference in either alpha- orbeta-diversityFasting insulin, homeostatic modelassessment for insulin resistance
(HOMA-IR),TNF-α, interferron (IFN)-γ, and systolic anddiastolic blood pressure ↓ (vs baseline)
Hricet al. (2021)Randomized, controlled, paralleltrialBryndza cheeses 30 g/d4 wk22 Female participants 18–65yr BMI 20–40; 13 consumed Bryndza cheeseNo significant change inalpha-diversity within and between thegroups.
Order Lactobacillales↑ (vs baseline)
FamilyStreptococcaceae ↑ (vsbaseline)
Genus Lactococcus andStreptococcus ↑ (vs baseline)Phascolarctobacterium andButyricimonas ↑ (vs baseline)
NA
Milaniet al. (2019)Randomized, controlled, paralleltrialParmesan cheese 45 g/d7 d20 Healthy participantsBifidobacteriummongolienseNA
Firmesse et al. (2007)Before and after trialCamembert cheese 2×40g/d4 wk12 Healthy participants (no agespecified)Enterococcusfaecalis ↑ (vs baseline)NA
Firmesse et al. (2008)Before and after trialCamembert cheese 2×40g/d4 wk12 Healthy participants 19–40yrLactococcus lactisLeuconostoc mesenteroides ↑ Nosignificant changes in bacterial enzyme activities and SCFAconcentrationNA

Another human study analyzed the correlation between consuming whole milk and gutmicrobiota. In the 3-month randomized, double-blind study, 24 of 64 maleparticipants received 500 mL of bovine milk daily. Then, the relative abundanceof members in the phylum Proteobacteria significantlyincreased. At the genus level, Lactobacillus andRoseburia tended to increase, whereasPrevotella decreased (Fernandez-Raudales et al., 2012).

There was more research on yogurt intake and gut microbiota change than on otherdairy products. First, three cross-sectional studies were identified from eachcohort study performed in France, Spain, and the United Kingdom. Thirty healthyadults who consumed at least 200–400 g of yogurt daily had significantlylower Enterobacteriaceae levels than those who did not consumeyogurt daily. Although there was no difference in the intestinal SCFAconcentration between the groups, β-galactosidase activity wassignificantly increased in the yogurt intake group (Alvaro et al., 2007). In another study, annual dietaryfermented food intake was investigated by a food frequency questionnaire (FFQ)from 130 people. Natural yogurt consumers showed significantly higher fecallevels of Akkermansia, and sweetened yogurt consumers displayedsignificantly lower fecal levels of Bacteroides. Additionally,cheese consumers presented significantly higher levels of the major fecal SCFAs(acetate, propionate, and butyrate; González et al., 2019). According to another FFQ survey from4,117 participants, >73% consumed 125 g of yogurt at least onceweekly and had higher abundances of Bifidobacterium animalissubsp. Lactis and S. thermophilus (Le Roy et al., 2022).

In a study of 36 healthy adult women who consumed fermented milk for 4 weeks, nosignificant changes in gut microbiota were found (Tillisch et al., 2013). Similarly, six healthy adults whoconsumed yogurt for 6 weeks showed no significant changes in gut microbiotacomposition or diversity (Lisko et al.,2017). However, unlike these two studies, most studies found thatconsuming fermented dairy products affected gut microbiota, particularlyLactobacillus and Bifidobacterium.Fermented milk intake was associated with increased intestinalLactobacillus (particularly Lactobacillusacidophilus) and Bifidobacterium (Link-Amster et al., 1994). It was alsoconfirmed that the intestinal Bifidobacterium increased, whileRoseburia decreased after consuming fermented milk (Volokh et al., 2019). In addition, threerandomized, double-blind studies found that yogurt intake affected the increasein intestinal Lactobacillus spp. Specifically, consuming 400 gof yogurt daily for 2 weeks was associated with increased intestinalLactobacillus delbrueckii spp. bulgaricusand Streptococcus salivarius spp. thermophiles(Burton et al., 2017). One studyfound that yogurt consumption increased Lactobacillus paracaseiand Lactobacillus rhamnosus but did not affect alpha- orbeta-diversity (Alvarez et al., 2020). Theincreased L. rhamnosus in this study has been shown to improvethe immune response in another study (Kang etal., 2021a). García-Albiach etal. (2008) reported that yogurt consumption increased the intestinaldensity of lactic acid bacteria (LAB) and the pathogen Clostridiumperfringens (García-Albiachet al., 2008). Similarly, fermented milk intake increasedFirmicutes and decreased Bacteroidetes atthe phylum level (Unno et al., 2015).

Consumption of fermented dairy products also inhibits pathogens in pathologicalconditions. In 38 H. pylori-infected children, when 200 mL ofyogurt was consumed daily for 4 weeks, the representative pathogenEscherichia coli decreased, and the beneficial bacteriaBifidobacterium spp. increased (Yang and Sheu, 2012). Additionally, fermented milkconsumption decreased Bilophila wadsworthia levels in patientswith IBD, which is a strain known to cause IBD by inducing barrier collapse byproducing hydrogen sulfide. Meanwhile, the intake of fermented milk increasedintestinal SCFA concentration and significantly increased the level ofbutyrate-producing bacteria. Among SCFAs, butyrate is particularly helpful instrengthening the gut barrier (Veiga et al.,2014). Kefir intake significantly increased the abundance ofLactobacillus in the gut microbiota of patients with IBD(Yılmaz et al., 2019).Another study showed that ingestion of kefir increased the relative abundance ofActinobacteria but had no significant effect onBacteroidetes, Proteobacteria, orVerrucomicrobia (Bellikci-Koyu et al., 2019).

Cheese is a high-protein-containing dairy product, a densely nutrient-rich solidfood, unlike raw milk or yogurt. Four studies reported the effect of cheeseconsumption on gut microbiota. When 13 female adults consumed 30 g of Bryndzacheese daily for 4 weeks, there was no change in alpha-diversity; however, therelative abundance of LAB (Lactobacillales,Streptococcaceae, Lactococcus, andStreptococcus) significantly increased. Additionally, SCFAproducers such as Phascolarctobacterium andButyricimonas increased significantly in Bryndza cheeseconsumers (Hric et al., 2021). A humanpilot study of Parmesan cheese consumption for 7 days showed thatBifidobacterium mongoliense strains from cheese couldtransiently colonize the human gut (Milani etal., 2019). In two studies of Camembert cheese intake affecting gutmicrobiota, higher Enterococcus faecalis, L.lactis, and L. mesenteroides levels were found infecal samples (Firmesse et al., 2007;Firmesse et al., 2008).

In conclusion, intake and types of dairy products are important gutmicrobiome-changing factors. A higher dairy product intake increases theintestinal Lactobacillus and Bifidobacteriumlevels. In H. pylori infection or IBD condition, consumingfermented dairy products may help hosts suppress the pathogen proliferation.Most reported studies have confirmed that consuming dairy products can improvehost health by increasing the number of beneficial bacteria in the gut.

Dairy protein

Numerous studies are underway to determine why consuming dairy products altersthe gut microbiome. Dairy nutrients have been the subject of several studies(Ha et al., 2021; Lim et al., 2020). It is also used toenhance the quality of other foods (Kang et al.,2021b). Several studies have examined how dairy proteins affect thegut microbiome. The gut microbiome of rats changed according to the intake ofmilk protein. When 14% or 53% of whole milk protein was fed torats, there were changes in microbiota composition, such as a decrease in genecopy numbers in Clostridium coccoides and Clostridiumleptum groups in the high protein group compared to that in thenormal protein group (Liu et al., 2014).However, studies on casein or whey proteins have been increasing. Casein andwhey proteins are known as the main proteins in milk. In particular, caseinconstitutes approximately 80% of the total protein in milk, while wheyprotein accounts for approximately 20% (Davoodi et al., 2016).

One study fed casein to mice for 2 weeks. The abundance ofBacteroidetes at the phylum level significantly increasedin the casein-treated group, while Lachnospiraceae andRuminococcaceae decreased at the family level, andParabacteroides and Bacteroides increasedat the genus level (Kim et al., 2016).Other studies have also shown that the gut microbiome changes when high caseinconcentrations are ingested. When the rats were fed diets containing either19.4% or 52% casein for 24 weeks, the gut microbiota of the groupsfed high casein concentrations were altered. At the phylum level, there was arelative expansion of Actinobacteria and a relative contractionof Saccaribacteria. At the genus level, there was an expansionof Bifidobacterium, Bacteroides,Parabacteroides, and Oscillospira (Snelson et al., 2021). Alternatively,another study reported that changes in the gut microbiota caused by a highcasein intake could be detrimental to intestinal health. This is because therelative abundance of pathogens in the colon increased in the 54%high-concentration casein intake (HCD) group compared to that in the normalintake (20% casein; NCD) group.Escherichia/Shigella,Enterococcus, Streptococcus, andsulfate-reducing bacteria increased >2–5 times in the HCD groupcompared to the NCD group. In contrast, Ruminococcus,Akkermansia, and F.prausnitzii, which are generally regarded as beneficialbacteria in the large intestine, were reduced (Mu et al., 2016). In another study, the number of E.coli increased in the HCD group, whereas bacteria that protectintestinal epithelium (A.muciniphila, Bifidobacterium),propionate-producing bacteria (Prevotella), butyrate-producingbacteria (Roseburia/Eubacteriumrectale), and acetate producing bacteria(Ruminococcusbromii), were reduced (Mu etal., 2017). However, studies remain insufficient to suggest thatchanges in the gut microbiota caused by a high casein intake may be detrimentalto gut health.

Conversely, casein is a non-meat protein that changes gut microbiota close to soyprotein, and there was no difference in alpha- and beta-diversity between humangroups consuming casein or soy protein (Beaumontet al., 2017). In animal experiments, casein intake was similar tothe gut bacteria composition of soy protein-treated groups rather than chicken,beef, and fish protein-treated groups. One study showed that casein consumptionwas related to the relative abundance of Lachnospiraceae (Zhu et al., 2016b). Similar to this study,rats fed with soy protein and casein had similar gut bacterial profiles at thefamily level that was characteristic of Lachnospiraceae (Zhu et al., 2015). Members of theLachnospiraceae family are known to protect the gut againsthuman colon cancer by producing butyrate. Additionally, the lower relativeabundance of Lactobacillus is associated with casein intake(Rist et al., 2014; Zhu et al., 2015; Zhu et al., 2016b).

Whey protein affects gut microbiota composition differently compared to casein.When high-fat diet mice were treated with casein or whey protein, differences inbeta-diversity were observed between groups (Boscaini et al., 2019). Another study showed a significantly higherproportion of Streptococcaceae at the family level in the groupfed a diet containing casein as the primary protein source (Boscaini et al., 2019; Boscaini et al., 2020; Nilaweera et al., 2017). In addition, highproportions of Lactobacillus andBifidobacterium were mainly observed in the group thatconsumed whey protein (Boscaini et al.,2019; Boscaini et al., 2020;Boudry et al., 2013; McAllan et al., 2014; Schaafsma et al., 2021; Sprong et al., 2010). The combination ofLactobacillus and Bifidobacterium has beenmentioned in one study as a potential candidate strain to be used for immuneenhancement, thus an increase in the proportion ofLactobacillus and Bifidobacterium in thegut microbiome could potentially associated with immune enhancing effects (Yu et al., 2022). In two studies byBoscaini et al. comparing casein and whey protein, at the family level, therelative abundance of Streptococcaceae was higher in the caseingroup, and the relative abundance of Lactobacillaceae washigher in the whey protein group. At the genus level, the relative abundance ofLactococcus was higher in the casein group, and therelative abundances of Parabacteroides,Lactobacillus, and Bifidobacterium werehigher in the whey protein group (Boscaini etal., 2019; Boscaini et al.,2020). In a study by Nilaweera et al.(2017) casein intake increased the proportion ofEnterobacteriaceae and Streptococcaceaecompared to the whey protein intake group (Nilaweera et al., 2017). Compared to casein, cheese whey proteinincreased fecal Lactobacilli andBifidobacteria counts in a colitis-induced rat model (Sprong et al., 2010), and a lactoperoxidaseand lactoferrin-enriched whey protein isolate increasedLactobacillaceae/Lactobacillus anddecreased Clostridiaceae/Clostridium inhigh-fat diet-fed mice (McAllan et al.,2014). In a human study to examine sleep quality and stress, those ona whey protein-based diet had significantly increased relative abundances ofBifidobacterium compared to the casein intake group (Schaafsma et al., 2021). Whey peptideextracts with a molecular weight of <1 kDa increasedLactobacillus spp. and Bifidobacteriumspp. (Boudry et al., 2013).

In conclusion, a long-term high-protein diet causes gut microbiota imbalance andincreases intestinal permeability. However, normal levels of casein intakeconstitute a gut microbiome that regulates the Lachnospiraceaefamily similarly to soy protein. Changes in the gut microbiome by whey proteinintake increased beneficial bacteria such as Lactobacillus andBifidobacterium, similar to the changes in the intake ofdairy products.

Effects of Egg Products and Egg Protein on the Gut Microbiota

Egg products

We summarized previous studies on the impact of egg products and their proteinson the composition and function of the gut microbiome (Table 3). Eggs have different effects on the gut microbiomedepending on the species and processing method. There are studies on hen eggwhite (HEW), duck egg white (DEW), and preserved duck egg white (PEW) using ratsas experimental models. Akkermansia andPeptostreptococcaceae were relatively high in abundance inthe HEW and DEW groups, respectively. In the PEW group, the intestinal microberichness was significantly lower than that of other groups, and in particular,compared to DEW, Proteobacteria abundance was relatively low.Egg consumption effects on gut microbes may differ depending on the egg type andprocessing method (Yu et al., 2020).

Table 3.

Human and animal studies assessing the effect of eggs consumption onthe composition of the gut microbiota

CitationStudy designInterventionTrial durationParticipantsEffect on the gut microbiotaOther health or physiologicalobservations
Thomaset al. (2022)Randomized, controlledcross-over3 eggs/d (4 wk) choline bitartrate(CB) supplement/d (4 wk)13 wk23 Men and women classified withmetabolic syndrome (MetS) (35–70 yr old)Phylum Firmicutes/Bacteroidetes -
Shannon -
Not applicable (NA)
Liu etal. (2022a)Prospective nonrandomized2 eggs/d (90–100 g,460–500 mg cholesterol)2 wk9 Healthy males [29±1 yr old,body mass index (BMI) 22±1 kg/m2]Phylum Firmicutes/Bacteroidetes -
Shannon - Simpson- Chao1 -
NA
Zhuet al. (2020)Randomized, cross-over2 whole eggs (100 g)/d (4 wk)yolk-free eggs (100 g)/d (4 wk)12 wk20 Overweight/obese postmenopausalwomen [57.7 (±5.64) yr old, average BMI of 28.34(±2.96)]Genus Prevotella -Anaeroplasma - Clostridium- Peptostreptococcaceae -NA
CitationControl treatmentExperimental treatmentFeeding durationBody weightAnimal modelEffect on the gut microbiota
Avirineni et al. (2022)Control diet (CON; 4.63 kcal/g)Egg albumen+cellulose (EC;4.38 kcal/g) Egg albumen+inulin (EI; 4.63 kcal/g) Wheyprotein isolate+cellulose (WC; 4.38 kcal/g) Whey proteinisolate+inulin (WI; 4.63 kcal/g)9 wk442.2±28 g40 Male obesity-prone Sprague-Dawleyrats (3 wk old)NA
Ge etal. (2021)Dextran sodium sulfate (DSS)DSS+egg white peptide (50 mg)DSS+egg white peptide (100 mg) DSS+egg whitepeptide (200 mg)2 wk20.00±2 gMale BALB/c mice (SPF level)PhylumFirmicutes/Bacteroidetes↑
Genus Lactobacillus↑ norank_f_Ruminococcaceae ↓Ruminiclostridium ↓Candidatus_Saccharimonas ↑
Zhanget al. (2020)Distilled waterEgg ovotransferrin (OVT)(≈400 mg)3 wk control, 3 wk OVT, 8 wkcontrol, and 8 wk OVTNAMale C57BL/6J mice (young: 3 wk old)(adult: 8 wk old)PhylumActinobacteria ↓ (Young)Actinobacteria ↑ (Adult)Proteobacteria ↑ (Young) TM7↓ (Adult)
GenusAkkermansia ↑ (Young,Adult)
Shannon - (Young, Adult)
Requena et al. (2017)Tap waterEgg white hydrolyzed withpepsin12 wk250–275 g [Zucker fatty(fa/fa) rats]
150–175 g [Zucker lean(+/+) rats]
20 Male Zucker fatty (fa/fa) rats (8wk old)
10 Male Zucker lean (+/+)rats (8 wk old)
Phylum Bacteroides
Genus Lactobacillus /EnterococcusBifidobacterium
Species Clostridiumleptum
Yu etal. (2020)CaseinHen egg white (‘HEW’group); duck egg white (‘DEW’ group); preservedegg white (‘PEW’ group)8 wk(117 g±10 g)40 Male Sprague–DawleyratsPhylumFirmicutes/Bacteroidetes -Proteobacteria (DEW ↑)Verrucomicrobia (HEW↑)
FamilyPeptostreptococcaceae,Moraxellase (DEW ↑)Lactobacillaceae,Lachnospiraceae (DEW↓)
Genus Akkermansia (HEW↑)
Shannon (HEW ↑) Simpson (HEW↑) Ace (PEW ↓) Chao 1 (PEW ↓)
Menget al. (2020)Fresh duck eggsPreserved duck eggs8 wk110–130 g24 Male Sprague–DawleyratsPhylum Firmicutes -Bacteroidetes -Firmicutes/BacteroidetesProteobacteria -
GenusRuminococcaceae UCG-005 ↓AllobaculumChristensenellaceae R-7 group ↓unclassified ClostridialesEubacteriumruminantium group ↑Eubacteriumxylanophilum group ↑Ruminococcaceae UCG-009↑
EubacteriumventriosumTyzzerella
Shannon ↑
Simpson↓
f*ckunaga et al. (2020)Casein-beef tallow-based dietCasein-egg yolk-based diet14 d35.0±0.4 g 35.1±0.6g16 Male Kwl:ddY mice 5 wk oldPhylum FirmicutesEpsilonbacteraeota
Family LachnospiraceaeRuminococcaceaeErysipelotrichaceae
Genus
Parasutterella
coprostanoligenes
Operational taxanomic unit (OTU) ↓Shannon ↓ Simpson -

In a study comparing the effects of duck eggs and preserved duck egg intake onchanges in gut microbial composition using rats as experimental models, theingestion of preserved duck eggs increased the α-diversity of gutmicrobes compared to the duck egg group. There was no significant difference atthe phylum level; however,Firmicutes/Bacteroidetes were decreased.At the genus level, Veillonella,Phascolarctobacterium, Alpinimonas,Coprococcus 3, Coprococcus 2,Gelria, and unclassified Methylocystaceae,which were not found in the duck egg group, were found (Meng et al., 2020).

Egg yolk is a food ingredient commonly used in various processed foods. A studyusing the mouse as an experimental model found that Firmicutesand Epsilonbacteraeota were relatively decreased at the phylumlevel when egg yolk was fed. In addition, Lachnospiraceae andRuminococcaceae were significantly reduced at the familylevel, and Erysipelotrichaceae was significantly increased. Atthe genus level, Parasutterella andCoprostanoligenes showed a relatively high abundance (f*ckunaga et al., 2020).Parasutterella has been defined as a core component of thehuman and mouse gut microbiota and has been correlated with bile acidmaintenance and cholesterol metabolism (Ju etal., 2019). Coprostanoligenes is a bacteria that caninfluence host cholesterol levels. Therefore, these results indicate that eggyolk consumption can induce changes in gut microbiota related to cholesterolmetabolism (Kenny et al., 2020).

In a study on the change in gut microbes by comparing the gut microbes of obeseand lean rats and treating the obese rats with egg white hydrolyzed with pepsin(EWH), the gut microbe composition of obese and lean rats showed significantdifferences at the phylum and genus levels. However, in the gut microbiomecomposition of obese rats fed EWH,Lactobacillus/Enterococcus andC.leptum were similar to those of lean rats. Conversely, thechange in the microbial composition observed after ingestion of EWH did not showthe final weight loss effect in obese rats. Thus, this study suggests that,although consumption of EWH can induce changes in gut microbiota composition, itmay have difficulty elucidating the direct link between changes in gutmicrobiota composition and its effects on metabolism and health (Requena et al., 2017).

Unlike consuming only eggs compared to consuming eggs with prebiotic fibertogether, the results of cecal microbiota were changed in 111 species’abundance. The abundances of Actinobacteria,Deinococcus-Thermus, Deferribacteres, andVerrucomicrobia were relatively higher than that of thecontrol, and the abundance of Firmicutes species was relativelyless than that of the control group. These changes in bacterial composition arecorrelated with the production of plasma metabolites, such as plasma butyricacid, propionic acid, and other metabolites derived from carbohydrate, protein,and fat metabolism, in an obese male rat model (Avirineni et al., 2022).

Choline contained in eggs can produce TMA by enzyme reaction of intestinalmicrobes, and TMA can be converted into harmful substances which may triggermetabolic diseases such as TMAO (Salzano et al.,2022). In patients with metabolic syndrome, intestinal microbiota andtheir correlation to metabolites were the subject of a study on the impact ofegg consumption. It was found that there was no significant effect on microbialdiversity or abundance of taxa (Thomas et al.,2022). Phosphatidylcholine and choline bitartrate are two differentforms of choline. Phosphatidylcholine is a type of choline derived from soybeanand egg yolk. Choline bitartrate is a type of choline produced through chemicalsynthesis (Smolders et al., 2019). Therewas no significant difference between the baseline group, the group withphosphatidylcholine provided by eggs, and the group with choline bitartratesupplement. There was no significant correlation in the correlation analysisbetween intestinal microflora and TMAO. These results suggest that cholinesupplied from eggs may not be a major influencer on TMAO production (Thomas et al., 2022).

Whole egg consumption increased plasma choline and betaine in overweightpostmenopausal women with mild hypercholesterolemia; however, it did notincrease plasma TMAO or alter gut microbiota composition, such asPrevotella, Anaeroplasma,Clostridium, and Peptostreptococcaceae,which are associated with TMAO concentrations (Zhu et al., 2020). In addition, whole egg consumption is believed tocause an increase in cholesterol and thereby induce cardiovascular disease.However, consuming two eggs daily for 2 weeks without changing their usual dietin people at low risk of developing metabolic diseases did not cause any changesin the gut microbiota. However, it rather positively modulated the functions ofthe gut microbiota, improving vascular health and intestinal function (Liu et al., 2022a).

Egg protein

Egg protein refers to the protein found in eggs, which is a complete proteincontaining all essential amino acids needed by the human body (Puglisi and Fernandez, 2022). One of theegg protein-derived peptides, Isoleucine-Arginine-Tryptophan (lle-Arg-Trp; IRW)and Isoleucine-Glutamine-Tryptophan (lle-Gln-Trp; IQW), reduce tumor necrosisfactor (TNF)-induced inflammatory responses and oxidative stress in endothelialcells (Majumder et al., 2013). In onestudy, consumption of the egg protein transferrin-derived peptides IRW and IQWincreased the ACE and Shannon index but decreased the Simpson index in obesemice induced by a high-fat diet. In addition, administration of IRW and IQWreduced the relative abundances of Firmicutes andParabacteroides, and IRW increased the abundance ofBacteroides, known as the major microorganisms that exhibitanti-obesity effects in the intestine. This study showed that ingesting eggprotein-derived peptides alleviates high-fat diet-induced obesity byreprogramming the gut microbiome (Liu et al.,2022b).

Egg yolks and egg whites have different nutritional compositions. Compared to eggyolk, egg white has a relatively low percentage of fat and proteins. In adextran sulfate sodium (DSS)-induced colitis mouse model study, 200 mg/kg bw ofegg white peptide ingestion decreased Ruminiclostridium andsignificantly increased Lactobacillus andCandidatus_Saccharimonas in the gutmicrobiome compared to the DSS group. In the correlation analysis,Lactobacillus andCandidatus_Saccharimonassignificantly reduced pro-inflammatory cytokines such as interleukin(IL)-1β and TNF-α. This indicates that the ingestion of egg whitepeptides can alleviate colonic inflammation by increasing the relative abundanceof beneficial bacteria and reducing pro-inflammatory cytokines (Ge et al., 2021). Ovotransferrin (OVT) isan egg white protein well known to have a wide range of biological activities,such as anti-inflammatory, antioxidant, and immunomodulatory functions (Lee et al., 2021). In a clinical study, adiet with OVT positively affected gut health by increasing the proportion ofAkkermansia, which promotes host immune regulation andintestinal epithelial cell integrity at the genus level of the gut microbiota(Zhang et al., 2020).

Conversely, in a study comparing the cecum microbiota of rats ingesting soy,milk, meat, fish, and egg proteins, along with oligosaccharides,Erysipelotrichaceae, Ruminococcaceae, andLachnospiraceae were the most abundant families in eggprotein and cellulose-fed rats. Erysipelotrichaceae,Bifidobacteriaceae, and Lachnospiraceaewere the most abundant families in egg protein and raffinose-fed rats,respectively (Sivixay et al., 2021).Erysipelotrichaceae, which was most abundantly increasedthrough the consumption of egg protein and prebiotics, is a characteristicbacterium that is decreased in patients with atopic dermatitis and increased inpatients with remission of the disease. For example,Erysipelatrichaceae_ UCG-003 is a potentialprobiotic used in a probiotic formula along with other beneficial bacteria andprebiotics (Wang et al., 2022). Theseresults indicate that the growth of Erysipelotrichaceae, whichcan have a positive effect on atopic dermatitis, can be promoted by egg protein(Sivixay et al., 2021).

Conclusion

Previous studies have indicated that the consumption of animal products can affectthe gut microbiome, with protein being a key nutritional characteristic of theseproducts. The ingestion of red or processed meat is associated with alterations inthe abundance of intestinal Lachnospiraceae, although the mechanismof the changes is uncertain. In contrast, white meat protein, such as chicken, isassociated with an increased level of Lactobacillus in the gut.Consumption of dairy products results in an increase in bothLactobacillus and Bifidobacterium abundances,and moderate intake of casein and whey protein is associated with elevated levels ofLAB. Egg yolk or egg protein can also impact the growth ofErysipelotrichaceae. Although the available scientific evidenceis insufficient to confirm a correlation between animal products or their proteinand the gut microbiome, current accumulating reports and results point to arelatively consistent direction of future research. Taken together, the presentreview investigates the effects of these animal products and their protein on thecomposition of the gut microbiome to enhance our understanding of the metabolism andfunction of intestinal microorganisms.

Acknowledgements

This research was supported by National Research Foundation of Korea Grant funded bythe Korean government (MEST) (NRF-2021R1A2C3011051) and by High Value-Added FoodTechnology Development Program of the Korean Institute of Planning and Evaluationfor Technology in Food, Agriculture, Forestry, and Fisheries (IPET), the Ministryfor Food, Agriculture, Forestry, and Fisheries of the Korea (321037-05-3-HD040) andby the Main Research Program (E0211100-03) of the Korea Food Research Institute(KFRI) funded by the Ministry of Science and ICT (Korea).

Conflicts of Interest

The authors declare no potential conflicts of interest.

Author Contributions

Contributed by

Conceptualization: Lee C, Lee J, Eor JY, Kwak MJ, Huh CS, Kim Y. Investigation:Lee C, Lee J, Eor JY, Kwak MJ, Huh CS, Kim Y. Writing - original draft: Lee C,Lee J, Eor JY, Huh CS, Kim Y. Writing - review & editing: Lee C, Lee J,Eor JY, Kwak MJ, Huh CS, Kim Y.

Ethics Approval

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

References

  • Ababakri R, Dayani O, Khezri A, Naserian AA. Influence of flaxseed with rumen undegradable protein level onmilk yield, milk fatty acids and blood metabolites in transitionewes. J Anim Sci Technol. 2021;63:475–490. doi:10.5187/jast.2021.e50. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ahmad RS, Imran A, Hussain MB. Nutritional composition of meat. Meat Sci Nutr. 2018;61:61–75. doi:10.5772/intechopen.77045. [CrossRef] [Google Scholar]
  • Al-Shaar L, Satija A, Wang DD, Rimm EB, Smith-Warner SA, Stampfer MJ, Hu FB, Willett WC. Red meat intake and risk of coronary heart disease among US men:Prospective cohort study. BMJ. 2020;371:m4141. doi:10.1136/bmj.m4141. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Alvarez AS, Tap J, Chambaud I, Cools-Portier S, Quinquis L, Bourlioux P, Marteau P, Guillemard E, Schrezenmeir J, Derrien M. Safety and functional enrichment of gut microbiome in healthysubjects consuming a multi-strain fermented milk product: A randomisedcontrolled trial. Sci Rep. 2020;10:15974. doi:10.1038/s41598-020-72161-w. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Alvaro E, Andrieux C, Rochet V, Rigottier-Gois L, Lepercq P, Sutren M, Galan P, Duval Y, Juste C, Doré J. Composition and metabolism of the intestinal microbiota inconsumers and non-consumers of yogurt. Br J Nutr. 2007;97:126–133. doi:10.1017/S0007114507243065. [PubMed] [CrossRef] [Google Scholar]
  • Aslam H, Marx W, Rocks T, Loughman A, Chandrasekaran V, Ruusunen A, Dawson SL, West M, Mullarkey E, Pasco JA, Jacka FN. The effects of dairy and dairy derivatives on the gut microbiota:A systematic literature review. Gut Microbes. 2020;12:1799533. doi:10.1080/19490976.2020.1799533. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Avirineni BS, Singh A, Zapata RC, Stevens RD, Phillips CD, Chelikani PK. Diets containing egg or whey protein and inulin fiber improveenergy balance and modulate gut microbiota in exercising obeserats. Mol Nutr Food Res. 2022;66:2100653. doi:10.1002/mnfr.202100653. [PubMed] [CrossRef] [Google Scholar]
  • Bäckhed F, Fraser CM, Ringel Y, Sanders ME, Sartor RB, Sherman PM, Versalovic J, Young V, Finlay BB. Defining a healthy human gut microbiome: Current concepts, futuredirections, and clinical applications. Cell Host Microbe. 2012;12:611–622. doi:10.1016/j.chom.2012.10.012. [PubMed] [CrossRef] [Google Scholar]
  • Beaumont M, Portune KJ, Steuer N, Lan A, Cerrudo V, Audebert M, Dumont F, Mancano G, Khodorova N, Andriamihaja M, Airinei G, Tomé D, Benamouzig R, Davila AM, Claus SP, Sanz Y, Blachier F. Quantity and source of dietary protein influence metaboliteproduction by gut microbiota and rectal mucosa gene expression: Arandomized, parallel, double-blind trial in overweighthumans. Am J Clin Nutr. 2017;106:1005–1019. doi:10.3945/ajcn.117.158816. [PubMed] [CrossRef] [Google Scholar]
  • Bellikci-Koyu E, Sarer-Yurekli BP, Akyon Y, Aydin-Kose F, Karagozlu C, Ozgen AG, Brinkmann A, Nitsche A, Ergunay K, Yilmaz E, Buyuktuncer Z. Effects of regular kefir consumption on gut microbiota inpatients with metabolic syndrome: A parallel-group, randomized, controlledstudy. Nutrients. 2019;11:2089. doi:10.3390/nu11092089. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Bendtsen LQ, Blædel T, Holm JB, Lorenzen JK, Mark AB, Kiilerich P, Kristiansen K, Astrup A, Larsen LH. High intake of dairy during energy restriction does not affectenergy balance or the intestinal microflora compared with low dairy intakein overweight individuals in a randomized controlled trial. Appl Physiol Nutr Metab. 2018;43:1–10. doi:10.1139/apnm-2017-0234. [PubMed] [CrossRef] [Google Scholar]
  • Boscaini S, Cabrera-Rubio R, Nychyk O, Speakman JR, Cryan JF, Cotter PD, Nilaweera KN. Age- and duration-dependent effects of whey protein on high-fatdiet-induced changes in body weight, lipid metabolism, and gut microbiota inmice. Physiol Rep. 2020;8:e14523. doi:10.14814/phy2.14523. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Boscaini S, Cabrera-Rubio R, Speakman JR, Cotter PD, Cryan JF, Nilaweera KN. Dietary α-lactalbumin alters energybalance, gut microbiota composition and intestinal nutrient transporterexpression in high-fat diet-fed mice. Br J Nutr. 2019;121:1097–1107. doi:10.1017/S0007114519000461. [PubMed] [CrossRef] [Google Scholar]
  • Boudry G, Jamin A, Chatelais L, Gras-Le Guen C, Michel C, Le Huёrou-Luron I. Dietary protein excess during neonatal life alters colonicmicrobiota and mucosal response to inflammatory mediators later in life infemale pigs. J Nutr. 2013;143:1225–1232. doi:10.3945/jn.113.175828. [PubMed] [CrossRef] [Google Scholar]
  • Bouvard V, Loomis D, Guyton KZ, Grosse Y, El Ghissassi F, Benbrahim-Tallaa L, Guha N, Mattock H, Straif K. Carcinogenicity of consumption of red and processedmeat. Lancet Oncol. 2015;16:1599–1600. doi:10.1016/S1470-2045(15)00444-1. [PubMed] [CrossRef] [Google Scholar]
  • Burton KJ, Rosikiewicz M, Pimentel G, Bütikofer U, von Ah U, Voirol MJ, Croxatto A, Aeby S, Drai J, McTernan PG, Greub G, Pralong FP, Vergères G, Vionnet N. Probiotic yogurt and acidified milk similarly reduce postprandialinflammation and both alter the gut microbiota of healthy, youngmen. Br J Nutr. 2017;117:1312–1322. doi:10.1017/S0007114517000885. [PubMed] [CrossRef] [Google Scholar]
  • David LA, Maurice CF, Carmody RN, Gootenberg DB, Button JE, Wolfe BE, Ling AV, Devlin AS, Varma Y, Fischbach MA, Biddinger SB, Dutton RJ, Tumbaugh PJ. Diet rapidly and reproducibly alters the human gutmicrobiome. Nature. 2014;505:559–563. doi:10.1038/nature12820. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Davoodi SH, 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. [PMC free article] [PubMed] [Google Scholar]
  • Day L, Cakebread JA, Loveday SM. Food proteins from animals and plants: Differences in thenutritional and functional properties. Trends Food Sci Technol. 2022;119:428–442. doi:10.1016/j.tifs.2021.12.020. [CrossRef] [Google Scholar]
  • Dhakal S, Moazzami Z, Perry C, Dey M. Effects of lean pork on microbiota and microbial-metabolitetrimethylamine-n-oxide: A randomized controlled non-inferiority feedingtrial based on the dietary guidelines for Americans. Mol Nutr Food Res. 2022;66:2101136. doi:10.1002/mnfr.202101136. [PubMed] [CrossRef] [Google Scholar]
  • Eckert E, Zambrowicz A, Pokora M, Polanowski A, Chrzanowska J, Szoltysik M, Dąbrowska A, Różański H, Trziszka T. Biologically active peptides derived from eggproteins. Worlds Poult Sci J. 2013;69:375–386. doi:10.1017/S0043933913000366. [CrossRef] [Google Scholar]
  • Farag MA, Jomaa SA, Abd El-Wahed A, El-Seedi HR. The many faces of kefir fermented dairy products: Qualitycharacteristics, flavour chemistry, nutritional value, health benefits, andsafety. Nutrients. 2020;12:346. doi:10.3390/nu12020346. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fernández J, de la Fuente VG, García MTF, Sánchez JG, Redondo BI, Villar CJ, Lombó F. A diet based on cured acorn-fed ham with oleic acid contentpromotes anti-inflammatory gut microbiota and prevents ulcerative colitis inan animal model. Lipids Health Dis. 2020;19:28. doi:10.1186/s12944-020-01205-x. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Fernandez-Raudales D, Hoeflinger JL, Bringe NA, Cox SB, Dowd SE, Miller MJ, de Mejia EG. Consumption of different soymilk formulations differentiallyaffects the gut microbiomes of overweight and obese men. Gut Microbes. 2012;3:490–500. doi:10.4161/gmic.21578. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Firmesse O, Alvaro E, Mogenet A, Bresson JL, Lemée R, Le Ruyet P, Bonhomme C, Lambert D, Andrieux C, Doré J, Corthier G, Furet JP, Rigottier-Gois L. Fate and effects of camembert cheese micro-organisms in the humancolonic microbiota of healthy volunteers after regular camembertconsumption. Int J Food Microbiol. 2008;125:176–181. doi:10.1016/j.ijfoodmicro.2008.03.044. [PubMed] [CrossRef] [Google Scholar]
  • Firmesse O, Rabot S, Bermúdez-Humarán LG, Corthier G, Furet JP. Consumption of camembert cheese stimulates commensal enterococciin healthy human intestinal microbiota. FEMS Microbiol Lett. 2007;276:189–192. doi:10.1111/j.1574-6968.2007.00933.x. [PubMed] [CrossRef] [Google Scholar]
  • Foerster J, Maskarinec G, Reichardt N, Tett A, Narbad A, Blaut M, Boeing H. The influence of whole grain products and red meat on intestinalmicrobiota composition in normal weight adults: A randomized crossoverintervention trial. PLOS ONE. 2014;9:e109606. doi:10.1371/journal.pone.0109606. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • f*ckunaga M, Kuda T, Xia Y, Nakamura S, Takahashi H, Kimura B. Detection and isolation of the typical gut indigenous bacteriafrom ddY mice fed a casein-beef tallow-based or egg yolk-baseddiet. J Food Biochem. 2020;44:e13246. doi:10.1111/jfbc.13246. [PubMed] [CrossRef] [Google Scholar]
  • Gao J, Guo X, Wei W, Li R, Hu K, Liu X, Jiang W, Liu S, Wang W, Sun H, Wu H, Zhang Y, Gu W, Li Y, Sun C, Han T. The association of fried meat consumption with the gut microbiotaand fecal metabolites and its impact on glucose hom*oeostasis, intestinalendotoxin levels, and systemic inflammation: A randomized controlled-feedingtrial. Diabetes Care. 2021;44:1970–1979. doi:10.2337/dc21-0099. [PubMed] [CrossRef] [Google Scholar]
  • García-Albiach R, José M, de Felipe P, Angulo S, Morosini MI, Bravo D, Baquero F, del Campo R. Molecular analysis of yogurt containing Lactobacillusdelbrueckii subsp. bulgaricus andStreptococcus thermophilus in human intestinalmicrobiota. Am J Clin Nutr. 2008;87:91–96. doi:10.1093/ajcn/87.1.91. [PubMed] [CrossRef] [Google Scholar]
  • Ge H, Cai Z, Chai J, Liu J, Liu B, Yu Y, Liu J, Zhang T. Egg white peptides ameliorate dextran sulfate sodium-inducedacute colitis symptoms by inhibiting the production of pro-inflammatorycytokines and modulation of gut microbiota composition. Food Chem. 2021;360:129981. doi:10.1016/j.foodchem.2021.129981. [PubMed] [CrossRef] [Google Scholar]
  • Gojda J, Cahova M. Gut microbiota as the link between elevated BCAA serum levels andinsulin resistance. Biomolecules. 2021;11:1414. doi:10.3390/biom11101414. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • González S, Fernández-Navarro T, Arboleya S, de los Reyes-Gavilán CG, Salazar N, Gueimonde M. Fermented dairy foods: Impact on intestinal microbiota andhealth-linked biomarkers. Front Microbiol. 2019;10:1046. doi:10.3389/fmicb.2019.01046. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • González-Soltero R, Bailén M, de Lucas B, Ramírez-Goercke MI, Pareja-Galeano H, Larrosa M. Role of oral and gut microbiota in dietary nitrate metabolism andits impact on sports performance. Nutrients. 2020;12:3611. doi:10.3390/nu12123611. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ha HK, Hong JY, Ayu IL, Lee MR, Lee WJ. Development and evaluation of probiotic delivery systems usingthe rennet-induced gelation of milk proteins. J Anim Sci Technol. 2021;63:1182–1193. doi:10.5187/jast.2021.e92. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Hentges DJ, Maier BR, Burton GC, Flynn MA, Tsutakawa RK. Effect of a high-beef diet on the fecal bacterial flora ofhumans. Cancer Res. 1977;37:568–571. [PubMed] [Google Scholar]
  • Hess JM, Jonnalagadda SS, Slavin JL. Dairy foods: Current evidence of their effects on bone,cardiometabolic, cognitive, and digestive health. Compr Rev Food Sci Food Saf. 2016;15:251–268. doi:10.1111/1541-4337.12183. [PubMed] [CrossRef] [Google Scholar]
  • Hric I, Ugrayová S, Penesová A, Rádiková Ž, Kubáňová L, Šardzíková S, Baranovičová E, Klučár L’, Beke G, Grendar M, Kolisek M, Šoltys K, Bielik V. The efficacy of short-term weight loss programs and consumptionof natural probiotic bryndza cheese on gut microbiota composition inwomen. Nutrients. 2021;13:1753. doi:10.3390/nu13061753. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ijssennagger N, Derrien M, van Doorn GM, Rijnierse A, van den Bogert B, Müller M, Dekker J, Kleerebezem M, van der Meer R. Dietary heme alters microbiota and mucosa of mouse colon withoutfunctional changes in host-microbe cross-talk. PLOS ONE. 2012;7:e49868. doi:10.1371/journal.pone.0049868. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ju T, Kong JY, Stothard P, Willing BP. Defining the role of parasutterella, a previously uncharacterizedmember of the core gut microbiota. ISME J. 2019;13:1520–1534. doi:10.1038/s41396-019-0364-5. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kang J, Lee JJ, Cho JH, Choe J, Kyoung H, Kim SH, Kim HB, Song M. Effects of dietary inactivated probiotics on growth performanceand immune responses of weaned pigs. J Anim Sci Technol. 2021a;63:520–530. doi:10.5187/jast.2021.e44. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kang KM, Lee SH, Kim HY. Quality properties of whole milk powder on chicken breastemulsion-type sausage. J Anim Sci Technol. 2021b;63:405–416. doi:10.5187/jast.2021.e30. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kenny DJ, Plichta DR, Shungin D, Koppel N, Hall AB, Fu B, Vasan RS, Shaw SY, Vlamakis H, Balskus EP, Xavier RJ. Cholesterol metabolism by uncultured human gut bacteriainfluences host cholesterol level. Cell Host Microbe. 2020;28:245–257. doi:10.1016/j.chom.2020.05.013. E6. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kim E, Kim DB, Park JY. Changes of mouse gut microbiota diversity and composition bymodulating dietary protein and carbohydrate contents: A pilotstudy. Prev Nutr Food Sci. 2016;21:57–61. doi:10.3746/pnf.2016.21.1.57. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Kohnert E, Kreutz C, Binder N, Hannibal L, Gorkiewicz G, Müller A, Storz MA, Huber R, Lederer AK. Changes in gut microbiota after a four-week intervention withvegan vs. meat-rich diets in healthy participants: A randomized controlledtrial. Microorganisms. 2021;9:727. doi:10.3390/microorganisms9040727. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Krebs NF, Sherlock LG, Westcott J, Culbertson D, Hambidge KM, Feazel LM, Robertson CE, Frank DN. Effects of different complementary feeding regimens on ironstatus and enteric microbiota in breastfed infants. J Pediatr. 2013;163:416–423. doi:10.1016/j.jpeds.2013.01.024. E4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lang JM, Pan C, Cantor RM, Tang WHW, Garcia-Garcia JC, Kurtz I, Hazen SL, Bergeron N, Krauss RM, Lusis AJ. Impact of individual traits, saturated fat, and protein source onthe gut microbiome. mBio. 2018;9:e01604-18. doi:10.1128/mBio.01604-18. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Le Roy CI, Kurilshikov A, Leeming ER, Visconti A, Bowyer RCE, Menni C, Falchi M, Koutnikova H, Veiga P, Zhernakova A, Derrien M, Spector TD. Yoghurt consumption is associated with changes in the compositionof the human gut microbiome and metabolome. BMC Microbiol. 2022;22:39. doi:10.1186/s12866-021-02364-2. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lee D, Goh TW, Kang MG, Choi HJ, Yeo SY, Yang J, Huh CS, Kim YY, Kim Y. Perspectives and advances in probiotics and the gut microbiome incompanion animals. J Anim Sci Technol. 2022;64:197–217. doi:10.5187/jast.2022.e8. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lee JH, Kim HJ, Ahn DU, Paik HD. Improved immune-enhancing activity of egg white proteinovotransferrin after enzyme hydrolysis. J Anim Sci Technol. 2021;63:1159–1168. doi:10.5187/jast.2021.e82. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Li DP, Cui M, Tan F, Liu XY, Yao P. High red meat intake exacerbates dextran sulfate-induced colitisby altering gut microbiota in mice. Front Nutr. 2021;8:646819. doi:10.3389/fnut.2021.646819. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lim DH, Han MH, Ki KS, Kim TI, Park SM, Kim DH, Kim Y. Changes in milk production and blood metabolism of lactatingdairy cows fed Saccharomycescerevisiae culture fluid under heat stress. J Anim Sci Technol. 2021;63:1433–1442. doi:10.5187/jast.2021.e114. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Lim DH, Mayakrishnan V, Lee HJ, Ki KS, Kim TI, Kim Y. A comparative study on milk composition of Jersey and Holsteindairy cows during the early lactation. J Anim Sci Technol. 2020;62:565–576. doi:10.5187/jast.2020.62.4.565. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Link-Amster H, Rochat F, Saudan KY, Mignot O, Aeschlimann JM. Modulation of a specific humoral immune response and changes inintestinal flora mediated through fermented milk intake. FEMS Immunol Med Microbiol. 1994;10:55–63. doi:10.1111/j.1574-695X.1994.tb00011.x. [PubMed] [CrossRef] [Google Scholar]
  • Lisko DJ, Johnston GP, Johnston CG. Effects of dietary yogurt on the healthy human gastrointestinal(GI) microbiome. Microorganisms. 2017;5:6. doi:10.3390/microorganisms5010006. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu X, Blouin JM, Santacruz A, Lan A, Andriamihaja M, Wilkanowicz S, Benetti PH, Tomé D, Sanz Y, Blachier F, Davila AM. High-protein diet modifies colonic microbiota and luminalenvironment but not colonocyte metabolism in the rat model: The increasedluminal bulk connection. Am J Physiol Gastrointest Liver Physiol. 2014;307:G459–G470. doi:10.1152/ajpgi.00400.2013. [PubMed] [CrossRef] [Google Scholar]
  • Liu X, Shao Y, Sun J, Tu J, Wang Z, Tao J, Chen J. Egg consumption improves vascular and gut microbiota functionwithout increasing inflammatory, metabolic, and oxidative stressmarkers. Food Sci Nutr. 2022a;10:295–304. doi:10.1002/fsn3.2671. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Liu Z, Ding S, Jiang H, Fang J. Egg protein transferrin-derived peptides Irw (Lle-Arg-Trp) andIqw (Lle-Gln-Trp) prevent obesity mouse model induced by a high-fat diet viareducing lipid deposition and reprogramming gut microbiota. Int J Mol Sci. 2022b;23:11227. doi:10.3390/ijms231911227. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Madsen L, Myrmel LS, Fjære E, Liaset B, Kristiansen K. Links between dietary protein sources, the gut microbiota, andobesity. Front Physiol. 2017;8:1047. doi:10.3389/fphys.2017.01047. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mafra D, Borges NA, Cardozo LFMF, Anjos JS, Black AP, Moraes C, Bergman P, Lindholm B, Stenvinkel P. Red meat intake in chronic kidney disease patients: Two sides ofthe coin. Nutrition. 2018;46:26–32. doi:10.1016/j.nut.2017.08.015. [PubMed] [CrossRef] [Google Scholar]
  • Majumder K, Chakrabarti S, Davidge ST, Wu J. Structure and activity study of egg protein ovotransferrinderived peptides (IRW and IQW) on endothelial inflammatory response andoxidative stress. J Agric Food Chem. 2013;61:2120–2129. doi:10.1021/jf3046076. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • McAllan L, Skuse P, Cotter PD, O’Connor P, Cryan JF, Ross RP, Fitzgerald G, Roche HM, Nilaweera KN. Protein quality and the protein to carbohydrate ratio within ahigh fat diet influences energy balance and the gut microbiota in C57BL/6Jmice. PLOS ONE. 2014;9:e88904. doi:10.1371/journal.pone.0088904. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Meng Y, Chen C, Qiu N, Keast R. Modulation of gut microbiota in rats fed whole egg diets byprocessing duck egg to preserved egg. J Biosci Bioeng. 2020;130:54–62. doi:10.1016/j.jbiosc.2020.02.015. [PubMed] [CrossRef] [Google Scholar]
  • Milani C, Duranti S, Napoli S, Alessandri G, Mancabelli L, Anzalone R, Longhi G, Viappiani A, Mangifesta M, Lugli GA, Bernasconi S, Ossiprandi MC, van Sinderen D, Ventura M, Turroni F. Colonization of the human gut by bovine bacteria present inParmesan cheese. Nat Commun. 2019;10:1286. doi:10.1038/s41467-019-09303-w. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Mu C, Yang Y, Luo Z, Guan L, Zhu W. The colonic microbiome and epithelial transcriptome are alteredin rats fed a high-protein diet compared with a normal-proteindiet. J Nutr. 2016;146:474–483. doi:10.3945/jn.115.223990. [PubMed] [CrossRef] [Google Scholar]
  • Mu C, Yang Y, Luo Z, Zhu W. Temporal microbiota changes of high-protein diet intake in a ratmodel. Anaerobe. 2017;47:218–225. doi:10.1016/j.anaerobe.2017.06.003. [PubMed] [CrossRef] [Google Scholar]
  • Muegge BD, Kuczynski J, Knights D, Clemente JC, González A, Fontana L, Henrissat B, Knight R, Gordon JI. Diet drives convergence in gut microbiome functions acrossmammalian phylogeny and within humans. Science. 2011;332:970–974. doi:10.1126/science.1198719. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Nilaweera KN, Cabrera-Rubio R, Speakman JR, O’Connor PM, McAuliffe A, Guinane CM, Lawton EM, Crispie F, Aguilera M, Stanley M, Boscaini S, Joyce S, Melgar S, Cryan JF, Cotter PD. Whey protein effects on energy balance link the intestinalmechanisms of energy absorption with adiposity and hypothalamic neuropeptidegene expression. Am J Physiol Endocrinol Metab. 2017;313:E1–E11. doi:10.1152/ajpendo.00356.2016. [PubMed] [CrossRef] [Google Scholar]
  • Oh JK, Vasquez R, Kim SH, Hwang IC, Song JH, Park JH, Kim IH, Kang DK. Multispecies probiotics alter fecal short-chain fatty acids andlactate levels in weaned pigs by modulating gut microbiota. J Anim Sci Technol. 2021;63:1142–1158. doi:10.5187/jast.2021.e94. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ortega RM, Ortega AIJ, Sánchez JMP, Soto EC, Vizuete AA, López-Sobaler AM. Nutritional value of dairy products and recommended dailyconsumption. Nutr Hosp. 2019;36:25–29. doi:10.20960/nh.02803. [PubMed] [CrossRef] [Google Scholar]
  • Perazza LR, Daniel N, Dubois MJ, Pilon G, Varin TV, Blais M, Martinez Gonzales JL, Bouchard M, Asselin C, Lessard M, Pouliot Y, Roy D, Marette A. Distinct effects of milk-derived and fermented dairy protein ongut microbiota and cardiometabolic markers in diet-induced obesemice. J Nutr. 2020;150:2673–2686. doi:10.1093/jn/nxaa217. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Petermann-Rocha F, Parra-Soto S, Gray S, Anderson J, Welsh P, Gill J, Sattar N, Ho FK, Celis-Morales C, Pell JP. Vegetarians, fish, poultry, and meat-eaters: Who has higher riskof cardiovascular disease incidence and mortality? A prospective study fromUK Biobank. Eur Heart J. 2021;42:1136–1143. doi:10.1093/eurheartj/ehaa939. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Puglisi MJ, Fernandez ML. The health benefits of egg protein. Nutrients. 2022;14:2904. doi:10.3390/nu14142904. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Requena T, Miguel M, Garcés-Rimón M, Martínez-Cuesta MC, López-Fandiño R, Peláez C. Pepsin egg white hydrolysate modulates gut microbiota in Zuckerobese rats. Food Funct. 2017;8:437–443. doi:10.1039/C6FO01571A. [PubMed] [CrossRef] [Google Scholar]
  • Rist VTS, Weiss E, Sauer N, Mosenthin R, Eklund M. Effect of dietary protein supply originating from soybean meal orcasein on the intestinal microbiota of piglets. Anaerobe. 2014;25:72–79. doi:10.1016/j.anaerobe.2013.10.003. [PubMed] [CrossRef] [Google Scholar]
  • Rodrigues VF, Elias-Oliveira J, Pereira ÍS, Pereira JA, Barbosa SC, Machado MSG, Carlos D. Akkermansia muciniphila and gut immune system: Agood friendship that attenuates inflammatory bowel disease, obesity, anddiabetes. Front Immunol. 2022;13:934695. doi:10.3389/fimmu.2022.934695. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Ross SM. Akkermansia muciniphila: A next-generationbeneficial gut microbe and its role in obesity and diabetes. Holist Nurs Pract. 2022;36:372–374. doi:10.1097/HNP.0000000000000556. [PubMed] [CrossRef] [Google Scholar]
  • Russell WR, Gratz SW, Duncan SH, Holtrop G, Ince J, Scobbie L, Duncan G, Johnstone AM, Lobley GE, Wallace RJ, Duthie GG, Flint HJ. High-protein, reduced-carbohydrate weight-loss diets promotemetabolite profiles likely to be detrimental to colonichealth. Am J Clin Nutr. 2011;93:1062–1072. doi:10.3945/ajcn.110.002188. [PubMed] [CrossRef] [Google Scholar]
  • Salzano A, Cassambai S, Yazaki Y, Israr MZ, Bernieh D, Wong M, Suzuki T. The gut axis involvement in heart failure: Focus ontrimethylamine N-oxide. Cardiol Clin. 2022;40:161–169. doi:10.1016/j.ccl.2021.12.004. [PubMed] [CrossRef] [Google Scholar]
  • Sasso A, Latella G. Role of heme iron in the association between red meat consumptionand colorectal cancer. Nutr Cancer. 2018;70:1173–1183. doi:10.1080/01635581.2018.1521441. [PubMed] [CrossRef] [Google Scholar]
  • Schaafsma A, Mallee L, van den Belt M, Floris E, Kortman G, Veldman J, van den Ende D, Kardinaal A. The effect of a whey-protein and galacto-oligosaccharides basedproduct on parameters of sleep quality, stress, and gut microbiota inapparently healthy adults with moderate sleep disturbances: A randomizedcontrolled cross-over study. Nutrients. 2021;13:2204. doi:10.3390/nu13072204. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Shi J, Zhao D, Zhao F, Wang C, Zamaratskaia G, Li C. Chicken-eaters and pork-eaters have different gut microbiota andtryptophan metabolites. Sci Rep. 2021;11:11934. doi:10.1038/s41598-021-91429-3. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sinha R, Zhao N, Goedert JJ, Byrd DA, Wan Y, Hua X, Hullings AG, Knight R, van Breda S, Mathijs K, de Kok TM, Ward MH. Effects of processed meat and drinking water nitrate on oral andfecal microbial populations in a controlled feeding study. Environ Res. 2021;197:111084. doi:10.1016/j.envres.2021.111084. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Sivixay S, Bai G, Tsuruta T, Nishino N. Cecum microbiota in rats fed soy, milk, meat, fish, and eggproteins with prebiotic oligosaccharides. AIMS Microbiol. 2021;7:1–12. doi:10.3934/microbiol.2021001. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Smolders L, de Wit NJW, Balvers MGJ, Obeid R, Vissers MMM, Esser D. Natural choline from egg yolk phospholipids is more efficientlyabsorbed compared with choline bitartrate; outcomes of a randomized trial inhealthy adults. Nutrients. 2019;11:2758. doi:10.3390/nu11112758. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Snelson M, Clarke RE, Nguyen TV, Penfold SA, Forbes JM, Tan SM, Coughlan MT. Long term high protein diet feeding alters the microbiome andincreases intestinal permeability, systemic inflammation and kidney injuryin mice. Mol Nutr Food Res. 2021;65:2000851. doi:10.1002/mnfr.202000851. [PubMed] [CrossRef] [Google Scholar]
  • Sprong RC, Schonewille AJ, van der Meer R. Dietary cheese whey protein protects rats against mild dextransulfate sodium–induced colitis: Role of mucin andmicrobiota. J Dairy Sci. 2010;93:1364–1371. doi:10.3168/jds.2009-2397. [PubMed] [CrossRef] [Google Scholar]
  • Swarte JC, Eelderink C, Douwes RM, Said MY, Hu S, Post A, Westerhuis R, Bakker SJL, Harmsen HJM. Effect of high versus low dairy consumption on the gutmicrobiome: Results of a randomized, cross-over study. Nutrients. 2020;12:2129. doi:10.3390/nu12072129. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Thøgersen R, Castro-Mejía JL, Sundekilde UK, Hansen LH, Gray N, Kuhnle G, Rye Jørgensen N, Kornerup Hansen A, Sandris Nielsen D, Bertram HC. Inulin and milk mineral fortification of a pork sausage exhibitsdistinct effects on the microbiome and biochemical activity in the gut ofhealthy rats. Food Chem. 2020;331:127291. doi:10.1016/j.foodchem.2020.127291. [PubMed] [CrossRef] [Google Scholar]
  • Thøgersen R, Castro-Mejía JL, Sundekilde UK, Hansen LH, Hansen AK, Nielsen DS, Bertram HC. Ingestion of an inulin-enriched pork sausage product positivelymodulates the gut microbiome and metabolome of healthy rats. Mol Nutr Food Res. 2018;62:1800608. doi:10.1002/mnfr.201800608. [PubMed] [CrossRef] [Google Scholar]
  • Thomas MS, DiBella M, Blesso CN, Malysheva O, Caudill M, Sholola M, Cooperstone JL, Fernandez ML. Comparison between egg intake versus choline supplementation ongut microbiota and plasma carotenoids in subjects with metabolicsyndrome. Nutrients. 2022;14:1179. doi:10.3390/nu14061179. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Tillisch K, Labus J, Kilpatrick L, Jiang Z, Stains J, Ebrat B, Guyonnet D, Legrain–Raspaud S, Trotin B, Naliboff B, Mayer EA. Consumption of fermented milk product with probiotic modulatesbrain activity. Gastroenterology. 2013;144:1394–1401. doi:10.1053/j.gastro.2013.02.043. E4. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Udenigwe CC, Aluko RE. Food protein-derived bioactive peptides: Production, processing,and potential health benefits. J Food Sci. 2012;77:R11–R24. doi:10.1111/j.1750-3841.2011.02455.x. [PubMed] [CrossRef] [Google Scholar]
  • Unno T, Choi JH, Hur HG, Sadowsky MJ, Ahn YT, Huh CS, Kim GB, Cha CJ. Changes in human gut microbiota influenced by probiotic fermentedmilk ingestion. J Dairy Sci. 2015;98:3568–3576. doi:10.3168/jds.2014-8943. [PubMed] [CrossRef] [Google Scholar]
  • Veiga P, Pons N, Agrawal A, Oozeer R, Guyonnet D, Brazeilles R, Faurie JM, van Hylckama Vlieg JET, Houghton LA, Whorwell PJ, Ehrlich SD, Kennedy SP. Changes of the human gut microbiome induced by a fermented milkproduct. Sci Rep. 2014;4:6328. doi:10.1038/srep06328. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Volokh O, Klimenko N, Berezhnaya Y, Tyakht A, Nesterova P, Popenko A, Alexeev D. Human gut microbiome response induced by fermented dairy productintake in healthy volunteers. Nutrients. 2019;11:547. doi:10.3390/nu11030547. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Walker AW, Ince J, Duncan SH, Webster LM, Holtrop G, Ze X, Brown D, Stares MD, Scott P, Bergerat A, Louis P, McIntosh F, Johnstone AM, Lobley GE, Parkhill J, Flint HJ. Dominant and diet-responsive groups of bacteria within the humancolonic microbiota. ISME J. 2011;5:220–230. doi:10.1038/ismej.2010.118. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Walther B, Karl JP, Booth SL, Boyaval P. Menaquinones, bacteria, and the food supply: The relevance ofdairy and fermented food products to vitamin K requirements. Adv Nutr. 2013;4:463–473. doi:10.3945/an.113.003855. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wang Y, Choy CT, Lin Y, Wang L, Hou J, Tsui JCC, Zhou J, Wong CH, Yim TK, Tsui WK, Chan UK, Siu PLK, Loo SKF, Tsui SKW. Effect of a novel E3 probiotics formula on the gut microbiome inatopic dermatitis patients: A pilot study. Biomedicines. 2022;10:2904. doi:10.3390/biomedicines10112904. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wu GD, Chen J, Hoffmann C, Bittinger K, Chen YY, Keilbaugh SA, Bewtra M, Knights D, Walters WA, Knight R, Sinha R, Gilroy E, Gupta K, Baldassano R, Nessel L, Li H, Bushman FD, Lewis JD. Linking long-term dietary patterns with gut microbialenterotypes. Science. 2011;334:105–108. doi:10.1126/science.1208344. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Wyness L. The role of red meat in the diet: Nutrition and healthbenefits. Proc Nutr Soc. 2016;75:227–232. doi:10.1017/S0029665115004267. [PubMed] [CrossRef] [Google Scholar]
  • Yang YJ, Sheu BS. Probiotics-containing yogurts suppress Helicobacter pylori loadand modify immune response and intestinal microbiota in the Helicobacterpylori-infected children. Helicobacter. 2012;17:297–304. doi:10.1111/j.1523-5378.2012.00941.x. [PubMed] [CrossRef] [Google Scholar]
  • Yılmaz İ, Dolar ME, Özpınar H. Effect of administering kefir on the changes in fecal microbiotaand symptoms of inflammatory bowel disease: A randomized controlledtrial. Turk J Gastroenterol. 2019;30:242–253. doi:10.5152/tjg.2018.18227. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yu DY, Oh SH, Kim IS, Kim GI, Kim JA, Moon YS, Jang JC, Lee SS, Jung JH, Park J, Cho KK. Intestinal microbial composition changes induced byLactobacillusplantarum GBL 16, 17 fermented feed and intestinal immunehomeostasis regulation in pigs. J Anim Sci Technol. 2022;64:1184–1198. doi:10.5187/jast.2022.e89. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Yu H, Qiu N, Meng Y, Keast R. A comparative study of the modulation of the gut microbiota inrats by dietary intervention with different sources of egg-whiteproteins. J Sci Food Agric. 2020;100:3622–3629. doi:10.1002/jsfa.10387. [PubMed] [CrossRef] [Google Scholar]
  • Zhang Q, Tan W, Yang L, Lu M, Dong S, Liu X, Duan X. Multi-omics analysis of the effects of egg ovotransferrin on thegut environment in mice: Mucosal gene expression, microbiota composition,and intestinal structural homeostasis. Mol Nutr Food Res. 2020;64:1901024. doi:10.1002/mnfr.201901024. [PubMed] [CrossRef] [Google Scholar]
  • Zhao D, Shan K, Xie Y, Zhang G, An Q, Yu X, Zhou G, Li C. Body weight index indicates the responses of the fecalmicrobiota, metabolome and proteome to beef/chicken-based diet alterationsin Chinese volunteers. NPJ Biofilms Microbiomes. 2022;8:56. doi:10.1038/s41522-022-00319-7. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhao F, Song S, Ma Y, Xu X, Zhou G, Li C. A short-term feeding of dietary casein increases abundance ofLactococcus lactis and upregulates gene expressioninvolving obesity prevention in cecum of young rats compared with dietarychicken protein. Front Microbiol. 2019a;10:2411. doi:10.3389/fmicb.2019.02411. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhao F, Zhou G, Liu X, Song S, Xu X, Hooiveld G, Müller M, Liu L, Kristiansen K, Li C. Dietary protein sources differentially affect the growth ofAkkermansia muciniphila and maintenance of the gutmucus barrier in mice. Mol Nutr Food Res. 2019b;63:1900589. doi:10.1002/mnfr.201900589. [PubMed] [CrossRef] [Google Scholar]
  • Zhu C, Sawrey-Kubicek L, Bardagjy AS, Houts H, Tang X, Sacchi R, Randolph JM, Steinberg FM, Zivkovic AM. Whole egg consumption increases plasma choline and betainewithout affecting TMAO levels or gut microbiome in overweight postmenopausalwomen. Nutr Res. 2020;78:36–41. doi:10.1016/j.nutres.2020.04.002. [PubMed] [CrossRef] [Google Scholar]
  • Zhu Y, Li H, Xu X, Li C, Zhou G. The gut microbiota in young and middle-aged rats showed differentresponses to chicken protein in their diet. BMC Microbiol. 2016a;16:281. doi:10.1186/s12866-016-0895-0. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhu Y, Lin X, Li H, Li Y, Shi X, Zhao F, Xu X, Li C, Zhou G. Intake of meat proteins substantially increased the relativeabundance of genus Lactobacillus in ratfeces. PLOS ONE. 2016b;11:e0152678. doi:10.1371/journal.pone.0152678. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhu Y, Lin X, Zhao F, Shi X, Li H, Li Y, Zhu W, Xu X, Li C, Zhou G. Meat, dairy and plant proteins alter bacterial composition of ratgut bacteria. Sci Rep. 2015;5:15220. doi:10.1038/srep15220. [PMC free article] [PubMed] [CrossRef] [Google Scholar]
  • Zhu Y, Shi X, Lin X, Ye K, Xu X, Li C, Zhou G. Beef, chicken, and soy proteins in diets induce different gutmicrobiota and metabolites in rats. Front Microbiol. 2017;8:1395. doi:10.3389/fmicb.2017.01395. [PMC free article] [PubMed] [CrossRef] [Google Scholar]

Articles from Food Science of Animal Resources are provided here courtesy of The Korean Society for Food Science of Animal Resources

Effect of Consumption of Animal Products on the Gut Microbiome
Composition and Gut Health (2024)
Top Articles
Latest Posts
Article information

Author: Tish Haag

Last Updated:

Views: 5893

Rating: 4.7 / 5 (67 voted)

Reviews: 90% of readers found this page helpful

Author information

Name: Tish Haag

Birthday: 1999-11-18

Address: 30256 Tara Expressway, Kutchburgh, VT 92892-0078

Phone: +4215847628708

Job: Internal Consulting Engineer

Hobby: Roller skating, Roller skating, Kayaking, Flying, Graffiti, Ghost hunting, scrapbook

Introduction: My name is Tish Haag, I am a excited, delightful, curious, beautiful, agreeable, enchanting, fancy person who loves writing and wants to share my knowledge and understanding with you.