75
Primer Write-Up Primer™ Microbiomic Superfuel™ Bacteria need to eat, too. Pamper them. A well fed microbiome is a happy and healthy microbiome. Give them the gourmet shit. Primer is a generously portioned blend of 11 prebiotic dishes and desserts meticulously chosen to entice the good bacterial denizens and citizens of your gut to feed and breed, prodigiously, while starving and poisoning unwelcome pathogenic bacterial inhabitants. Grow your own, right at home. Currently, probiotics are mostly thought of and used in relation to a healthy digestive system (reducing upset stomach, gas and bloating, diarrhea, and IBS type symptoms) and the immune system (coughs, colds, and general sinus and respiratory health). While they certainly are indeed useful for such applications, the ramifications of an unhealthy gut and microbiota go far, far beyond that. The gut and its microbiome are essentially a massive endocrine organ, controlling and influencing basically your entire body and brain. And, given that all of the trillions of bacteria that call it home originally came from outside your body – and entered without your permission – it is by far the most important organ in which we can take steps to manipulate and take back control. We will first look at some basic science and data on how this all works. Then, we will look at studies that have shown alterations in the microbiotic make-up of the gut, and the correlations they display in health and disease, suboptimal and optimal fitness, and just general things that everyone would consider part of good or bad life outcomes. It is a massive subject, far too much to discuss in complete depth, here, so we’ll do our best to keep it as short and sweet as possible while still giving you enough background in this field to understand the shocking reality, scope, and importance of this microscopic invasion.

neobium.orgneobium.org/wp-content/uploads/Primer Full Write-Up.docx · Web viewsuch as Lipopolysaccharide, TNF-alpha, interleukins, and prostaglandins, which subsequently escape the

  • Upload
    lymien

  • View
    215

  • Download
    2

Embed Size (px)

Citation preview

Primer Write-Up

Primer™Microbiomic Superfuel™Bacteria need to eat, too. Pamper them. A well fed microbiome is a happy and healthy microbiome. Give them the gourmet shit. Primer is a generously portioned blend of 11 prebiotic dishes and desserts meticulously chosen to entice the good bacterial denizens and citizens of your gut to feed and breed, prodigiously, while starving and poisoning unwelcome pathogenic bacterial inhabitants. Grow your own, right at home.

Currently, probiotics are mostly thought of and used in relation to a healthy digestive system (reducing upset stomach, gas and bloating, diarrhea, and IBS type symptoms) and the immune system (coughs, colds, and general sinus and respiratory health). While they certainly are indeed useful for such applications, the ramifications of an unhealthy gut and microbiota go far, far beyond that.

The gut and its microbiome are essentially a massive endocrine organ, controlling and influencing basically your entire body and brain. And, given that all of the trillions of bacteria that call it home originally came from outside your body – and entered without your permission – it is by far the most important organ in which we can take steps to manipulate and take back control.

We will first look at some basic science and data on how this all works. Then, we will look at studies that have shown alterations in the microbiotic make-up of the gut, and the correlations they display in health and disease, suboptimal and optimal fitness, and just general things that everyone would consider part of good or bad life outcomes.

It is a massive subject, far too much to discuss in complete depth, here, so we’ll do our best to keep it as short and sweet as possible while still giving you enough background in this field to understand the shocking reality, scope, and importance of this microscopic invasion.

Subsequently, we will get down to business and specifically get into the science of Shock Treatment™, the first step in the process of making yourself king or queen of your own castle, again. We’ll show you how it can immediately ameliorate symptoms, while preparing the gut for a permanent fix, with special emphasis on a lean, healthy body.

Deus Vult!

The BasicsIt basically works like this. The Western lifestyle, including diet, lack of exercise, and alcohol use (and, in all likelihood, genetics, though the data just isn’t there, yet) leads to an imbalance of the bacterial composition of the gut (1,2). This results in the excess production and release of inflammatory signals,

such as Lipopolysaccharide, TNF-alpha, interleukins, and prostaglandins, which subsequently escape the gut and enter the rest of your body (3).

Though, they all contribute to the pathologies we will cover in various ways, it is Lipopolysaccharide (LPS) that we will focus on the most. Within the gut, this leads to the general digestive issues and inflammatory bowel syndromes like IBS and colitis that you have commonly known probiotics as being used to alleviate (4).

While fixing digestive disorders will come along for the ride, our primary focus is going to be on body composition and metabolic health. In other words, we want to make you leaner, protect against diabetes, and help keep you from having a heart attack or stroke. However, there really is so much more to it than that, as a few quotes from the literature aptly demonstrate:

“Changes in the composition of the gut microbiota (dysbiosis) may be associated with several clinical conditions, including obesity and metabolic diseases, autoimmune diseases and allergy, acute and chronic intestinal inflammation, irritable bowel syndrome (IBS)…” (5)

“In this milieu… disturbance of the gut microbiota balance and the intestinal barrier permeability is a potential triggering factor for systemic inflammation in the onset and progression of obesity, type 2 diabetes and metabolic syndrome.” (6)

“Through these varied mechanisms, gut microbes shape the architecture of sleep and stress reactivity of the hypothalamic-pituitary-adrenal axis. They influence memory, mood, and cognition and are clinically and therapeutically relevant to a range of disorders, including alcoholism, chronic fatigue syndrome, fibromyalgia, and restless legs syndrome… Nutritional tools for altering the gut microbiome therapeutically include changes in diet, probiotics, and prebiotics.” (7)

As you can see, alterations in the microbiota can affect basically everything, but that there is also hope for change.

Getting back to the gut and body composition, the aforementioned Lipopolysaccharide (LPS) leads to overactivation of cannabinoid receptor 1 (CB1) within the gut, which causes an increase in intestinal motility (speed of food going through) in the proximal parts of the intestine. This leads to less absorption of nutrient feedback signals that tell the brain you are well fed, and that it is time to stop eating (8). Concurrent with this is an increase in transit time in the colon, which results in a greater total harvest of caloric energy from your food (9, 10).

In other words, the signal your brain is getting is that you are not getting enough food, while you are actually extracting more calories from what you eat. This not only directly leads to more fat accumulation from harvesting more calories, it lends itself to over-eating. This aggravates the cycle further, as overeating and increased adiposity are themselves inflammatory. So, what you have is more inflammation, more dysfunction, greater food intake, greater extraction of food, more fat accumulation, then REPEAT!

The carnage does not even end here. Along with this inflammatory state is a disruption in the intestinal barrier. Intestinal permeability is increased and these inflammatory agents spill out systemically. This is often called a “leaky gut”. This results in a low-level inflammatory state in the entire body. The biggest

culprit here is, once again, LPS (11).

LPS activates CB1 receptors in the body and brain, just as in the intestine. In the fat tissue, this leads to activation of PPAR-gamma, and an upregulation of triglyceride synthesis, fat cell formation, and fat storage (12). In the brain, activation of CB1 increases orexegenic pathways, thus increasing appetite, hunger, and ultimately, food intake (13). This should not much as much of a surprise considering “the munchies” that accompany intake of famous cannabinoid receptor agonist, marijuana.

And, LPS is not done yet, not at all. It also activates Toll-like Receptor 4 which, along with other inflammatory signals (TNF-alpha, interleukins), promotes both insulin and leptin insensitivity, peripherally and centrally (14, 15). At this point, your adipostat (the thermostat for your body fat level) is wrecked. Your ability to control food intake is gone, and you are a fat storing machine. Obviously, this is not what you want your body doing to itself. It is not what you want it doing to you. It is not what you want it doing to your life.

Oh, and to top it off, atherosclerosis, heart disease, and stroke are promoted by these same inflammatory pathways. Combined with the increased body fat and insulin resistance, you officially have all of the perfect ingredients for the dreaded Metabolic Syndrome (16, 17).

And, it is just a bunch of microscopic bacteria that call your gut “home” causing all of this devastation.

General DataThe most well-known genera of bacteria in commercial probiotics are Lactobacillus and Bifidobacterium. They are also among the most common in the body, along with several other ones which are not commercially available, but which we can manipulate with supplementation. We will talk about these in length in the SupraBiotic™ and Primer™ write-ups.

Unfortunately, Lactobacillus belong to the Firmicutes phylum which has been found to be associated with weight gain and obesity (18-20). Just a 20% increase in Firmicutes (which Lactobacillus is usually the primary genus) with an equal decrease in Bacteroides results in an increased energy harvest of 150 calories per day in humans (21). That is equal to 15lbs of fat per year! The Western style diet promotes these negative changes in microbial proportions (22). Thus, one can plainly see why it can be so difficult to get lean, as well as how easily obesity has become an epidemic.

Interestingly, smoking cessation produces the same negative changes in bacterial composition, while gastric bypass surgery improves it (23-24). The well-known effects on weight with both of these further highlights the negative body compositional effects of this intestinal dysbiosis.

In addition, probiotic treatment with several Lactobacillus species that are in a great number of commercial formulations, including Lactobacillus acidophilus, Lactobacillus fermentum, and Lactobacillus ingluviei , have been directly associated with weight gain and obesity (25). Type-2 diabetics had significantly more Lactobacillus, with L. acidophilus being particularly bad in this

regard (26). Further, L. reuteria and L. sakei have been found to be positively associated with obesity and body mass index (27-29). They probably don’t tell you that on the label.

More powerful evidence of the profound effect of the microbiota on body weight and metabolism come from studies on “fecal transfer”. And, yes, that is exactly what it sounds like – transferring poop from one subject’s intestine to another’s.

In twins, transfer of an obese microbiota to lean mice was accompanied by an increase in bodyweight, fat mass, and a dysbiotic alteration of the Firmicutes:Bacteroides ratio to reflect that of the obese model (30). A similar transfer replicated the obese phenotype with increased weight gain, lipogenesis, adipogenesis, overeating, and lower satiety, as well as inflammation and hyperglycemia in formerly lean, healthy subjects (31, 32).

On the other side of the coin, transferring the intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome, as well as reversing obesity and gastrointestinal issues (33). It also reduced markers of metabolic syndrome, inflammation, and oxidative stress in animals challenged with high-fructose diets (34).

Obviously, while it highlights the science, doing a fecal transfer is not terribly practical, appetizing, or readily available -- unless maybe you are in California.

Fortunately, there is good news. While several species and strains of lactobacillus have been found to promote weight gain, several have also been found to protect against it. And, of course, we only used the good ones. Furthermore, Bifidobacterium have shown only positive effects to a remarkable extent.

Bifidobacterium are anti-obesity and lipid lowering, decreasing fat weight, blood glucose, cholesterol, and triglyceride levels (35). They are higher in lean subjects, as well as being lower in obese (36, 37). They are significantly lower in type-II diabetics and have been shown to improve glucose tolerance as well to decrease inflammatory signaling (38-40). In addition, they increase levels of EPA, DHA, and CLA in fat tissue and the brain (40). They have also been found to decrease with aging (41).

We can also readily manipulate levels of the good bacteria that are not commercially available such as Bacteroides species, Akkermansia Muciniphilia, Faecalbacterium Prausnitzii, and Roseburia via supplementation of ingredients that ARE available.

So, let’s get to it.

You may have noticed that almost no probiotic formulas contain just a single species of bacteria, nowadays. And, if you did not, I will just say that it is for a good reason. They work better in combination. This applies to the prebiotics that feed them as well. You need a variety of prebiotics to grow a variety of probiotic bacterial species.

First of all, microbial diversity seems to be good, in and of itself. Essentially, a diverse gut is a healthy gut (42). Obesity has been associated with a lack of microbial diversity and, as you might expect, lean subjects have greater microbial diversity in the gut (43-45). Insulin sensitivity is also improved along with

diversity increases (46). Finally, in the interesting but not terribly shocking category, exercise increases microbial diversity (47, 48).

Increased diversity also works to specifically create an environment where probiotic bacteria can thrive, thus enhancing their ultimate performance (49). Compared to individual strains alone, this diversity increases adhesion to intestinal mucus, which is necessary for most survival, growth, and activity (50, 51). Conversely, bacteria inhibit adhesion of pathogenic bacteria better when in combination (52, 53).

However, you do not want to just try to have every single species and strain in existence growing inside of you. It needs to be done rationally. If not, they can interfere with each other’s actions and compete for space and resources (54-56).

But, maybe the most interesting benefit of supporting a combination of bacteria is through cross-feeding. This is when one bacterial species produces metabolic substrates the other species and strains use for fuel (57, 58). Bifidobacterium adolescentis is the most interesting and important species in this regard as it functions as THE archetypal cross-feeder for several of the most important and impressive strains of bacteria. And, those strains are not commercially available. B. adolescentis produces acetate and oligosacharrides which are then consumed by these acetate utilizing, butyrate and propionate producing bacteria (59).

The Best Bacterial Species That Money Can’t Buy.Unfortunately, several species of bacteria with some of the very best data are not available commercially, due to regulatory issues and well as practical challenges such as stability and viability of the bacteria themselves. We are working on these, as are several other groups, but it will happen later rather than sooner, at best.

Fortunately, there are a myriad of ways to specifically target and increase these strains using methods that ARE available. And, that is exactly what we have done. So, let’s take a look at these novel wonder-bacteria, and then we will get to the data on B. adolescentis as the ultimate cross-feeding probiotic.

Genus Bacteroides

Bacteroides are butyrate and propionate producing. Levels were 6-fold higher in lean vs. obese subjects, as well as being reduced in obese patients, in general, compared to control populations (60-63). The Firmicutes:Bacteroides ratio was also significantly worse in obese patients, even in comparison with the merely overweight (65, 66). It has a negative correlation with fat mass and waist circumference (66, 67). It was also 60% lower in obese pigs – yeah, apparently that is a thing (68).

Bacteroides levels in Type-2 diabetes were only half that of those with normal glucose tolerance (69). Lower Bacteroides was correlated with increased energy intake (70). Additionally, it was decreased after smoking cessation similar to differences in obese compared to lean subjects suggesting a link between Bacteroides and the weight gain of smoking cessation (71).

Among various species in the Bacteroides genus, B. uniformis reduced bodyweight gain, triglycerides, and adipocyte volume while improving insulin and leptin sensitivity. It also lowered LPS and other inflammatory signals (72). Bacteroides acidifaciens decreased bodyweight and fat gain, while increasing fatty acid oxidation via PPAR-alpha (73). In addition to an elevated Firmicutes:Bacteroides ratio, B. vulgatus levels were found to be lower in the obese (74).

B. fragilis releases a symbiotic immunomodulatory anti-inflammatory factor called Polysacharride A (75). This activates TLR-2, which releases anti-inflammatory interleukins. PSA is basically the opposite of LPS, and TLR-2 the opposite of TLR-4 (76). This has been shown not just to prevent but to cure experimental colitis, an extreme version of a leaky, inflammatory gut (77). It has also been shown to prevent demyelination of neurons in the central nervous system, indicative of protection against inflammation well outside of the gut (78).

A few of the Bacteroides species bind to mucins for colonization and consume these mucin polysaccharides (79, 79b). Bacteroides species also have greater glycan degrading capability than Firmicutes, thus they are preferentially increased by polyphenols (80). Primer™ contains both mucin and polyphenols.

Faecalibacterium prausnitzii

Faecalibacterium prausnitzii is butyrate producing and is considered a physiological sensor and marker of human health (81). It does not get much more important than that. It is lower in the obese and type-2 diabetics (82-84). Conversely, it is higher in normal glucose tolerance vs. prediabetic subjects (85).

Faecalibacterium prausnitzii is also negatively correlated with inflammatory markers and sharply decreased in inflammatory bowel diseases (84, 86). It is greatly reduced in ulcerative colitis and less abundant in Crohn’s disease (87, 88). As would be expected from the above, it improves intestinal barrier function (89).

Akkermansia muciniphilia

Akkermansia muciniphilia is mucin degrading, meaning it feeds on mucins (90). Levels are higher in lean subjects than the general population (91). It is also decreased in obesity and type-2 diabetes. Its administration reduced fat mass, adipose tissue inflammation, and enhanced insulin sensitivity. Along with this, improved gut barrier function and increased intestinal endocannabinoid levels were seen (92).

This species is also inversely related to fasting glucose, waist-to-hip ratio, subcutaneous adipocyte diameter, plasma triglyceride levels, visceral adipose tissue mass, and insulin resistance (93). Along with enhanced glucose tolerance, it reduced adipose tissue inflammation (94). Akkermansia levels are higher in normal glucose tolerance vs. pre-diabetic subjects (95). It decreased inflammatory cytokine production and protected intestinal barrier function in experimental colitis (96). Finally, its levels are reduced in ulcerative colitis (97).

Roseburia Species

Roseburia species are butyrate producing (98). An increase in this species is associated with decreased body weight, fat mass, insulin sensitivity, and triglycerides -- independent of calorie intake (99). Increased Roseburia correlated with reduced body weight, improved profile of lipid and obesity related gene expression, along with a normalized inflammatory status (100). It is also lower in type-2 diabetes (101). Levels are increased by a Mediterranean diet, as is insulin sensitivity (102). Roseburia is enriched in healthy populations vs. those with atherosclerosis (103). And, its levels display an inverse correlation with disease activity in ulcerative colitis (104).

High protein/low carbohydrate diets, which are so effective and popular, reduce Roseburia and SCFA levels (105, 106). This does not mean don’t use them, it just means make sure you make a point to get fiber/prebiotics to feed your good bacteria that produce SCFAs. Butyrate is especially important amongst the SCFAs, as it the preferred energy source, along with Glutamine, for epithelial cells in the colon (107). Butyrate is basically the fat to Glutamine’s protein and carbohydrate as far as feeding these cells. We will talk more on Glutamine in the Primer™ write-up.

Bifidobacterium adolescentis as Cross-Feeder

As mentioned, B. adolescentis is hugely important in helping to feed other bacteria, specifically the really good ones that we just talked about, which we cannot get commercially.

B. adolescentis is superior to other potential cross-feeding Bifidobacterium in that it provides a slow, steady degradation of oligosaccharides for a long, continuous release of substrate for these various bacteria to feed on. It is essentially time-released, allowing acetate feeding, butyrate producing bacteria to grow and thrive throughout the entire length of the gut (108).

Faecalibacterium prausnitzii is almost fully dependent on acetate, which B. adolescentis supplies. F. prausnitzii converts it to butyrate with 85% efficiency, and its growth is enhanced by co-culture with B. adolescentis (109, 110).

Roseburia is also an acetate user (111). It is, in fact, generally required for growth (112). In addition to acetate production, B. adolescentis increases Roseburia via partial breakdown of oligosaccharides, which it can then utilize (113).

Cross-feeding with Bifidobacterium modulates the prebiotic effect of inulin and arabinoxylan-oligosaccharides on Roseburia and F. prausnitzii by making acetate available (114). Roseburia was able to grow in pure complex carbohydrate cultures, which it cannot metabolize on its own, owing to cross-feeders (115).

Short Chain Fatty Acids (SCFAs)

One of the primary ways that probiotic bacteria work their magic is by fermenting prebiotics and producing SCFAs (primarily acetate, butyrate, and propionate), so we are going to talk about those, and how they work.

They primarily work through two mechanisms: 1) activation of free fatty acid receptors, FFA2 and FFA3. 2) Decreasing inflammation and permeability in the gut.

SCFAs protect against obesity and insulin resistance. Butyrate and propionate induce anorectic gut hormones, while acetate does so without reducing food intake (Supplementary 1). FFAR2 deficiency results in obesity on a normal diet, whereas with overexpression, subjects remain lean, even on an obesity promoting high-fat diet. Activation of FFAR2 suppresses insulin signaling in adipocytes, which inhibits fat accumulation in adipose tissue and promotes the metabolism of lipids and glucose in other tissues such as muscle (S2).

Propionate and butyrate activate intestinal gluconeogenesis. Butyrate does so through AMPK, while propionate works through a gut-brain neural circuit involving FFAR3 (S3). Propionate is sensed in the portal vein walls via FFAR3, initiating intestinal gluconeogenesis. This glucose then triggers a signal to the brain to modulate hunger sensations and normalize whole body glucose homeostasis (S4). In a fasting state, as much as 62% of infused propionate is converted to glucose, accounting for 69% of total glucose production (S5). This is quite applicable to lower carb diets. Basically, it makes your brain think you are plenty fed with carbs/glucose, so it signals not to eat more, as well as not to produce or pump out more glucose into the blood.

SCFAs also stimulate the release of anorectic and satiey inducing peptides like GLP-1 and PYY via FFAR2/3 (S6, S7). Activation of FFAR3 by SCFAs inhibits insulin secretion and increases sympathetic outflow. This raises energy expenditure and help to protect against obesity (S8, S9). Acetate has been found to increase brown adipose tissue, UCP1, and mitochondrial biogenesis via FFAR2 (S10).

Short-chain fatty acids also improve intestinal barrier function via activation of AMPK (S11). Sodium butyrate has been specifically found to be an AMPK agonist (S12). And, butyrate increase tight junction assembly, thus improving barrier function, specifically through AMPK (S13, S14).

This seems like as good of a place as any to add a bit more about AMPK, as it is one of the major targets in all of this.

AMPK

AMPK is a primary signaler in the maintenance of tight junction integrity and intestinal barrier function. It is one of the most important pathways in preventing the “leaky gut” we have spoken of earlier in regard to LPS and other inflammatory and infectious molecules escaping into the body to wreak havoc (S15, S16). Modern food processing and the Western diet is a particularly egregious malefactor in this (S17).

In addition to its involvement in barrier function, AMPK activation is extremely positive for the great bacteria that we can’t get commercially.

Metformin increased Akkermansia 18-fold through AMPK activation. Also, against a high-fat diet, it restored Bacteroides levels and the Firmicutes:Bacteroides ratio to that of lean subjects (S18-S20). It inhibited LPS induced inflammation and gut permeability increases, while improving glucose uptake and insulin sensitivity (S19). Akkermansia increases are likely at least partially due to greatly elevated production of its favorite food, mucin, which is stimulated by AMPK. It also reduces insulin resistance and adipose tissue inflammation in a high-fat diet (S20).

Primer™ IngredientsAs Primer™ is primarily a prebiotic, let’s start with those. Prebiotics have come a long way since oat bran and psyllium husks. Beginning with inulin, a huge array of oligosaccharide and glycan type compounds have been found to be fermented and fed on by intestinal bacteria. These newer prebiotics tend to be basically tasteless and dissolve effortlessly, which is quite handy.

With the importance of microbial diversity for optimal gut and body health, we want a number of different prebiotics for them to feed on. Likewise, we want to choose the ones that best increase the bacteria we want to increase, rather than just randomly feeding all of them.

Galacto-oligosaccharides (GOS)GOS reduced fat mass, food intake by 14%, and elevated expression of pro-satiety peptides. Combining them with Calcium increased propionate formation (116). In addition to reductions in food intake, appetite, bodyweight, and inflammation are also decreased (117). GOS increase beneficial bacteria, particularly Bifidobacterium, with 5-10 fold increases in some subjects being noted (118-121). They also raise Bacteroides levels (121). They are long-acting, providing prebiotic effect throughout the entire length of the colon, while strongly inhibiting pathogenic bacteria (122). This owes to high resistance to conditions early in gut digestion (123).

GOS provide direct enhancement of intestinal barrier function through interaction with goblet cells, separate from SCFA or anti-inflammatory mediated mechanisms. They also showed a 2-4 fold mucin elevation, which would create a positive environment for mucin feeders such as Akkermansia and Bacteroides (124). They inhibited inflammatory responses, augmented protein junction assembly by 85%, and prevented loss of barrier function (125). GOS displayed a microbiota independent increase in tight junction assembly and improved barrier function (126). Finally, they mitigate LPS induced inflammation and protect against stress induced LPS activity (127, 128).

Arabinoxylan-oligosaccharides (AXOS)AXOS are strongly Bifidogenic. They increase satiety inducing peptides, while decreasing weight gain, fat mass, and insulin resistance (129). They raise butyrate levels along with Bifidobacterium suggestive of subsequent cross-feeding to butyrate producing bacteria (130). They also reduce protein fermentation in the gut (130, 131). This spares amino acids for more useful purposes as well as preventing toxic breakdown products.

AXOS are long-acting, with bacterial fermentation occurring throughout length of colon. They significantly promote Bacteroides as well as Lactobacillus (132). They are even better than inulin at providing fermentation products to the distal portions of the colon (133). AXOS elevated Roseburia and butyrate levels, with total SCFA increases as high as 2-3 fold (134, 135). Finally, they increase tight junction proteins, improve barrier function, and inhibit inflammation in adipose tissue (129).

Xylo-oligosaccharides (XOS)XOS increased Bifidobacterium along with acetate and butyrate. Combining with inulin further augmented butyrate formation, as well as increasing propionate, suggesting cross-feeding to butyrate and propionate producing bacteria like Roseburia and Bacteroides (136, 137). And, XOS have indeed been found to promote both Roseburia and Bacteroides, as well as improving the Firmicutes:Bacteroides ratio (138, 139). Bacteroides possess special xylan degrading enzymes, making them a preferred fermenter of XOS (140). Elevated Bacteroides and butyrate from XOS protected against genotoxicity in a colonic simulator (141). They also decreased LPS and increased epithelial cell proliferation (137, 142).

Lactulose Lactulose inhibits adipogenesis and fat accumulation, down-regulates adipogenic genes, and reduces caloric extraction efficiency, while increasing energy expenditure and lipolysis (143). It also improves post-prandial blood glucose and insulin levels (144, 145). Lactulose raises Bifidobacterium counts, particularly of the ideal cross-feeder B. adolescentis, as well as Akkermansia (146-148). Finally, it decreases intestinal permeability and proteolysis of amino acids in the gut (147, 149).

Inulin Inulin improved glucose uptake in insulin resistant cells, and activated AMPK (150). It increases Bifidobacterium and butyrate, while reducing protein fermentation (151, 152). It has a prolonged Bifidogenic effect, with more distal fermentation and SCFA production vs. fructo-oligosaccharides, particularly of butyrate and propionate – again, suggestive of cross-feeding (153, 154). In fact, it was found to increase B. adolescentis more than 4-fold and F. Prausnitzii by 50% (155). It also increased

Roseburia, while augmenting mucin production 6-fold, leading to large elevations in Akkermansia and propionate, distally (156).

Resistant Starch 3 (RS3)Resistant Starch 3 is formed when starchy foods such as potatoes and rice are cooked and then cooled. This turns formerly digestible starches into resistant starches via a process called retrogradation. RS3 is particularly, and somewhat uniquely, highly prebiotic for Ruminococcus bromii, with increases up to 4-fold (157-159). R. bromii has superior ability to degrade this resistant starch, which is the most prevalent fermentable carbohydrate in the average diet, making it a “keystone species” by acting as a cross-feeder for other species (160, 161).

It was also found to be readily consumed by Bacteroides, elevating faecal propionate, rather than butyrate as is often observed following resistant starch feeding of other types. This propionate formation reflects a gut community dominated by the Bacteroides, and it actually became the primary lineage in this study (162).

Amylopectin Amylopectin was found to be superior to several other prebiotics for increasing butyrate, as well as butyrate producers F. prausnitzii and Roseburia (163). It also raises Bacteroides, with increases in Roseburia and Bacteroides being found to be proportional to the amylopectin content of barley and oats (164, 165).

Mucin Mucin is the glycoprotein constituent of the mucus which lines the wall of the intestines and protects it. Several species of bacteria, including some of the really good ones, feed off of it. Akkermansia is the most well characterized mucin consumer (166, 167). Verrucomicrobia, of which Akkermansia is the primary genus, was increased from .03% to 5.25% by mucin, and in combination with inulin, Bacteroides was raised as well (168).

Bacteroides thetaiotamicron is a known mucin degrading specialist (169-171). Bacteroides fragillis consumes mucins as well (172, 173). Roseburia intestinalis also colonizes the mucosal layer and feeds on mucins (174).

With these bacteria colonizing the mucus and being close to the epithelium, particularly with the butyrate producers, bioavailability for epithelial cell regeneration and barrier function is enhanced.

RhamnoseRhamnose is a preferred sugar for the propanediol pathway of propionate production by Roseburia inulinivorans (175, 176). It is quite selectively metabolized to propionate (177, 178). It is much more selective for propionate formation than lactulose or glucose, which utilize different, less selective pathways, resulting in 4 times more propionate than with lactulose or glucose (179). Rhamnose was also found to decrease triglyceride synthesis and serum triglyceride levels, likely due to propionates effects on the SCFA receptors FFAR 2/3 (180).

Glutamine Glutamine is the primary substrate of rapidly diving cells, a category to which the epithelial cells of the digestive tract belong. It increases tight junction protein production (181). It does so by activating the mammalian target of rapamycin (mTOR) cell signaling in enterocytes. It enhances intestinal growth, enterocyte proliferation and survival, and regulates intestinal barrier function in injury, infection, stress, inflammation, and other catabolic conditions (182). It is basically both the leucine (protein) and the glucose (carbohydrate), to go along with butyrate as the fat, for the fueling of survival, growth, and reproduction of the enterocyte. This makes it quite possibly the most important nutrient for intestinal barrier health and function.

Glutamine also reduces utilization of other amino acids (asparagine, aspartate, serine, lysine, leucine, valine, ornithine, and arginine) in the gut, preserving them for more useful things while reducing toxic metabolites (183, 184). It decreases intestinal permeability and enhances intestinal mucosa and barrier function (185). Glutamine improves intestinal barrier impairment and quells the LPS mediated inflammatory cascade (186). It also prevents mucosal injury and promotes recovery from LPS induced inflammatory damage, as well as downregulating TLR-4 expression (187, 188).

Inflammatory conditions increase the requirements for Glutamine to maintain the intestinal barrier (189). It has specifically been shown to protect the intestinal barrier against processed, Western diet style foods (190). AMPK mediates its enhancement of tight junction integrity and barrier preservation (191).

Conversion to glutamate and subsequent cellular uptake is a pivotal step in its protective effects (192). Monosodium Glutamate has been found to promote the colonization of F. prausnitzii and Roseburia (193). And, finally, L-glutamate enhances barrier function (194).

Calcium Phosphate Increasing dietary Calcium produced a reduction in weight gain and fat pad mass of 26-39% with a 51% inhibition of adipocyte fatty acid synthase expression and activity, while stimulating lipolysis by 3 to 5-fold (195). In another study, an almost 50% increase in weight loss was found (196). A high-Calcium diet decreased fat gain by 55%, stimulated adipose tissue uncoupling protein (UCP2) and skeletal muscle UCP3 expression, increased thermogenesis and lipolysis, while lowering fatty acid synthase expression

and activity (197, 198). Calcium also elevated peptides GLP-1 and GLP-2, which increase satiety and decrease food intake (199, 200).

Calcium improves intestinal permeability, strengthens the mucosal barrier, reduces inflammation, and alleviates colitis (201). Prebiotics have actually been found to have negative effects on intestinal permeability and inflammation without Calcium Phosphate rather than the positive effects produced when it is present (202, 203). This protection is dependent on Phosphate, thus Calcium likely pulls it into the colon, improving luminal buffering capability (204). This is because SCFAs produced by prebiotic fermentation could lower pH too much in its absence.

Finally, Calcium is necessary for the Calcium/Calmodulin-dependent Protein Kinase Kinase 2 (CaMKK2) mediated AMPK signaling and barrier maintenance produced by Glutamine (205).

Multi-Berry Powder

Multi-berry powder has the power of berries!! A day’s worth of Primer™ is equal to ¼ cup of mixed berries. It has the polyphenols and fiber and such that berries have, but it is mostly in here because it gives it a nice, subtle berry flavor.

ConclusionPrimer™ takes the concept of prebiotic far beyond where anyone has previously taken it before. It starts by carefully and selectively feeding the most beneficial bacterial species, including novel probiotic species that you cannot attain, anywhere. It does so in a way that no other product comes even close to doing. It protects against dysfunction of the gut and microbiota to promote better health, better appetite control, better metabolism, and better fat loss. Finally, its supporting ingredients go to work on making your inflamed and leaky gut as good as new, leaving your body functioning in the optimal way it is intended to.

Primer™ is a gourmet meal for your microbiota and a happy-ending massage for your gut. It is a one of a kind product that fits in perfectly with and enhances any diet and exercise program, any supplement regimine, any lifestyle.

References1. Ann Nutr Metab. 2013;63 Suppl 2:28-40. doi: 10.1159/000354902. Epub 2013 Nov 8.

Clinical consequences of diet-induced dysbiosis.

Chan YK(1), Estaki M, Gibson DL.

2. Gut. 2014 Jan;63(1):5-6. doi: 10.1136/gutjnl-2013-304969. Epub 2013 Jun 5.

Western lifestyle: a 'master' manipulator of the intestinal microbiota?

Hold GL.

3. Clin Sci (Lond). 2015 Dec;129(12):1083-96. doi: 10.1042/CS20150431.

Immunometabolism of obesity and diabetes: microbiota link compartmentalizedimmunity in the gut to metabolic tissue inflammation.

McPhee JB(1), Schertzer JD(2).

4. World J Gastroenterol. 2014 Mar 14;20(10):2456-69. doi: 10.3748/wjg.v20.i10.2456.

Irritable bowel syndrome: emerging paradigm in pathophysiology.

Lee YJ(1), Park KS(1).

5. Nutr Rev. 2015 Aug;73 Suppl 1:32-40. doi: 10.1093/nutrit/nuv039.

Potential role of the intestinal microbiota in programming health and disease.

Goulet O(1).

6. Benef Microbes. 2016;7(2):181-94. doi: 10.3920/bm2015.0062. Epub 2015 Dec 8.

Gut microbiota in health and disease: an overview focused on metabolicinflammation.

Nagpal R(1), Kumar M(2), Yadav AK(2), Hemalatha R(2), Yadav H(3), Marotta F(4),Yamashiro Y(1).

7. J Med Food. 2014 Dec;17(12):1261-72. doi: 10.1089/jmf.2014.7000.

The gut microbiome and the brain.

Galland L(1).

8. Ann Transl Med. 2013 Jul 1;1(2):14.

Effect of high fat-diet and obesity on gastrointestinal motility.

Mushref MA(1), Srinivasan S(1).

8b. Neurosci Lett. 2010 May 3;474(3):148-53. doi: 10.1016/j.neulet.2010.03.026. Epub 2010 Mar 15.

Oleamide suppresses lipopolysaccharide-induced expression of iNOS and COX-2through inhibition of NF-kappaB activation in BV2 murine microglial cells.

Oh YT(1), Lee JY, Lee J, Lee JH, Kim JE, Ha J, Kang I.

9. Obesity (Silver Spring). 2012 May;20(5):954-62. doi: 10.1038/oby.2011.357. Epub2011 Dec 15.

Effects of chronic oral rimonabant administration on energy budgets ofdiet-induced obese C57BL/6 mice.

Zhang LN(1), Gamo Y, Sinclair R, Mitchell SE, Morgan DG, Clapham JC, Speakman JR.

10. Am J Clin Nutr. 2011 Jul;94(1):58-65. doi: 10.3945/ajcn.110.010132. Epub 2011 May4.

Energy-balance studies reveal associations between gut microbes, caloric load,and nutrient absorption in humans.

Jumpertz R(1), Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, Krakoff J.

11. Brain Behav Immun. 2012 Jul;26(5):691-8. doi: 10.1016/j.bbi.2012.01.004. Epub2012 Jan 17.

Cannabinoid signalling regulates inflammation and energy balance: the importance of the brain-gut axis.

Cluny NL(1), Reimer RA, Sharkey KA.

12: Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NM, Cani PD. The endocannabinoid system links gut microbiota to adipogenesis. Mol Syst

Biol. 2010 Jul;6:392. doi: 10.1038/msb.2010.46. PubMed PMID: 20664638; PubMedCentral PMCID: PMC2925525.

13. Endocrinology. 2012 Sep;153(9):4136-43. doi: 10.1210/en.2012-1405. Epub 2012 Jul 9.

Hypothalamic CB1 cannabinoid receptors regulate energy balance in mice.

Cardinal P(1), Bellocchio L, Clark S, Cannich A, Klugmann M, Lutz B, Marsicano G,Cota D.

14. Diabetes. 2012 Jun;61(6):1455-62. doi: 10.2337/db11-0390. Epub 2012 Apr 20.

Inhibition of hypothalamic inflammation reverses diet-induced insulin resistance in the liver.

Milanski M(1), Arruda AP, Coope A, Ignacio-Souza LM, Nunez CE, Roman EA,Romanatto T, Pascoal LB, Caricilli AM, Torsoni MA, Prada PO, Saad MJ, Velloso LA.

15. Mol Endocrinol. 2013 Jun;27(6):990-1003. doi: 10.1210/me.2012-1338. Epub 2013 Apr11.

Cellular insulin resistance disrupts leptin-mediated control of neuronalsignaling and transcription.

Nazarians-Armavil A(1), Menchella JA, Belsham DD.

16: Rana JS, Nieuwdorp M, Jukema JW, Kastelein JJ. Cardiovascular metabolicsyndrome - an interplay of, obesity, inflammation, diabetes and coronary heartdisease. Diabetes Obes Metab. 2007 May;9(3):218-32. Review. PubMed PMID:17391148.

17. Nat Commun. 2012;3:1245. doi: 10.1038/ncomms2266.

Symptomatic atherosclerosis is associated with an altered gut metagenome.

Karlsson FH(1), Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, BäckhedF, Nielsen J.

18. Obesity (Silver Spring). 2013 Dec;21(12):E607-15. doi: 10.1002/oby.20466. Epub2013 Jun 22.

Human intestinal microbiota composition is associated with local and systemicinflammation in obesity.

Verdam FJ(1), Fuentes S, de Jonge C, Zoetendal EG, Erbil R, Greve JW, Buurman WA,de Vos WM, Rensen SS.

19. P R Health Sci J. 2015 Jun;34(2):60-4.

Association of the Intestinal Microbiota and Obesity.

López-Cepero AA(1), Palacios C(1).

20. J Clin Gastroenterol. 2012 Jan;46(1):16-24. doi: 10.1097/MCG.0b013e31823711fd.

The intestinal microbiota and obesity.

Kallus SJ(1), Brandt LJ.

21. Am J Clin Nutr. 2011 Jul;94(1):58-65. doi: 10.3945/ajcn.110.010132. Epub 2011 May4.

Energy-balance studies reveal associations between gut microbes, caloric load,and nutrient absorption in humans.

Jumpertz R(1), Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, Krakoff J.

22. Obes Rev. 2012 Sep;13(9):799-809. doi: 10.1111/j.1467-789X.2012.01009.x. Epub2012 Jun 11.

Gut microbial adaptation to dietary consumption of fructose, artificialsweeteners and sugar alcohols: implications for host-microbe interactionscontributing to obesity.

Payne AN(1), Chassard C, Lacroix C.

23. PLoS One. 2013;8(3):e59260. doi: 10.1371/journal.pone.0059260. Epub 2013 Mar 14.

Smoking cessation induces profound changes in the composition of the intestinalmicrobiota in humans.

Biedermann L(1), Zeitz J, Mwinyi J, Sutter-Minder E, Rehman A, Ott SJ,

Steurer-Stey C, Frei A, Frei P, Scharl M, Loessner MJ, Vavricka SR, Fried M,Schreiber S, Schuppler M, Rogler G.

24. Proc Natl Acad Sci U S A. 2009 Feb 17;106(7):2365-70. doi:10.1073/pnas.0812600106. Epub 2009 Jan 21.

Human gut microbiota in obesity and after gastric bypass.

Zhang H(1), DiBaise JK, Zuccolo A, Kudrna D, Braidotti M, Yu Y, Parameswaran P,Crowell MD, Wing R, Rittmann BE, Krajmalnik-Brown R.

25. Microb Pathog.2012;53:100–108

Comparative meta-analysis of the effect of Lactobacillus species on weight gain in humans and animals.

Million M, Angelakis E, Paul M, Armougom F, Leibovici L, Raoult D.

26. Front Physiol. 2013 Jan 31;3:496. doi: 10.3389/fphys.2012.00496. eCollection2012.

Alterations in fecal Lactobacillus and Bifidobacterium species in type 2 diabeticpatients in Southern China population.

Lê KA(1), Li Y, Xu X, Yang W, Liu T, Zhao X, Tang YG, Cai D, Go VL, Pandol S, HuiH.

27. Int J Obes (Lond). 2012 Jun;36(6):817-25. doi: 10.1038/ijo.2011.153. Epub 2011Aug 9.

Obesity-associated gut microbiota is enriched in Lactobacillus reuteri anddepleted in Bifidobacterium animalis and Methanobrevibacter smithii.

Million M(1), Maraninchi M, Henry M, Armougom F, Richet H, Carrieri P, Valero R, Raccah D, Vialettes B, Raoult D.

28. Int J Obes (Lond). 2013 Nov;37(11):1460-6. doi: 10.1038/ijo.2013.20. Epub 2013Mar 5.

Correlation between body mass index and gut concentrations of Lactobacillusreuteri, Bifidobacterium animalis, Methanobrevibacter smithii and Escherichiacoli.

Million M(1), Angelakis E, Maraninchi M, Henry M, Giorgi R, Valero R, Vialettes B, Raoult D.

29. Br J Nutr. 2011 Apr;105(8):1235-44. doi: 10.1017/S0007114510004770. Epub 2011 Feb8.

Diversity and metabolic impact of intestinal Lactobacillus species in healthyadults and the elderly.

Štšepetova J(1), Sepp E, Kolk H, Lõivukene K, Songisepp E, Mikelsaar M.

30. Science. 2013 Sep 6;341(6150):1241214. doi: 10.1126/science.1241214.

Gut microbiota from twins discordant for obesity modulate metabolism in mice.

Ridaura VK(1), Faith JJ, Rey FE, Cheng J, Duncan AE, Kau AL, Griffin NW, Lombard V, Henrissat B, Bain JR, Muehlbauer MJ, Ilkayeva O, Semenkovich CF, Funai K,Hayashi DK, Lyle BJ, Martini MC, Ursell LK, Clemente JC, Van Treuren W, WaltersWA, Knight R, Newgard CB, Heath AC, Gordon JI.

31. Diabetes. 2014 May;63(5):1624-36. doi: 10.2337/db13-1526. Epub 2014 Jan 15.

Replication of obesity and associated signaling pathways through transfer ofmicrobiota from obese-prone rats.

Duca FA(1), Sakar Y, Lepage P, Devime F, Langelier B, Doré J, Covasa M.

32. Gut. 2013 Dec;62(12):1787-94. doi: 10.1136/gutjnl-2012-303816. Epub 2012 Nov 29.

Intestinal microbiota determines development of non-alcoholic fatty liver diseasein mice.

Le Roy T(1), Llopis M, Lepage P, Bruneau A, Rabot S, Bevilacqua C, Martin P,Philippe C, Walker F, Bado A, Perlemuter G, Cassard-Doulcier AM, Gérard P.

33. Gastroenterology. 2012 Oct;143(4):913-6.e7. doi: 10.1053/j.gastro.2012.06.031.Epub 2012 Jun 20.

Transfer of intestinal microbiota from lean donors increases insulin sensitivity in individuals with metabolic syndrome.

Vrieze A(1), Van Nood E, Holleman F, Salojärvi J, Kootte RS, Bartelsman JF,Dallinga-Thie GM, Ackermans MT, Serlie MJ, Oozeer R, Derrien M, Druesne A, Van

Hylckama Vlieg JE, Bloks VW, Groen AK, Heilig HG, Zoetendal EG, Stroes ES, de VosWM, Hoekstra JB, Nieuwdorp M.

34. PLoS One. 2015 Aug 5;10(8):e0134893. doi: 10.1371/journal.pone.0134893.eCollection 2015.

Rescue of Fructose-Induced Metabolic Syndrome by Antibiotics or FaecalTransplantation in a Rat Model of Obesity.

Di Luccia B(1), Crescenzo R(1), Mazzoli A(1), Cigliano L(1), Venditti P(1),Walser JC(2), Widmer A(3), Baccigalupi L(1), Ricca E(1), Iossa S(1).

35. Lipids Health Dis. 2011 Jul 12;10:116. doi: 10.1186/1476-511X-10-116.

Antiobesity and lipid-lowering effects of Bifidobacterium spp. in high fatdiet-induced obese rats.

36. Clin Microbiol Infect. 2016 Mar;22(3):258.e1-8. doi: 10.1016/j.cmi.2015.10.031.Epub 2015 Nov 10.

Correlation between body mass index and faecal microbiota from children.

Ignacio A(1), Fernandes MR(1), Rodrigues VA(1), Groppo FC(2), Cardoso AL(3),Avila-Campos MJ(1), Nakano V(4).

37. Gut Pathog. 2013 Apr 30;5(1):10. doi: 10.1186/1757-4749-5-10.

Differences in gut microbiota composition between obese and lean children: across-sectional study.

Bervoets L(1), Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C,Goossens H, Desager KN, Vankerckhoven V.

38. Curr Microbiol. 2010 Jul;61(1):69-78. doi: 10.1007/s00284-010-9582-9. Epub 2010Jan 20.

Molecular characterisation of the faecal microbiota in patients with type IIdiabetes.

Wu X(1), Ma C, Han L, Nawaz M, Gao F, Zhang X, Yu P, Zhao C, Li L, Zhou A, WangJ, Moore JE, Millar BC, Xu J.

39. Diabetologia. 2007 Nov;50(11):2374-83. Epub 2007 Sep 6.

Selective increases of bifidobacteria in gut microflora improvehigh-fat-diet-induced diabetes in mice through a mechanism associated withendotoxaemia.

Cani PD(1), Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, Gibson GR,Delzenne NM.

40. Am J Clin Nutr. 2009 May;89(5):1393-401. doi: 10.3945/ajcn.2008.27023. Epub 2009 Apr 8.

Metabolic activity of the enteric microbiota influences the fatty acidcomposition of murine and porcine liver and adipose tissues.

Wall R(1), Ross RP, Shanahan F, O'Mahony L, O'Mahony C, Coakley M, Hart O, LawlorP, Quigley EM, Kiely B, Fitzgerald GF, Stanton C.

41. Curr Issues Intest Microbiol. 2001 Sep;2(2):43-53.

The Lactobacillus and Bifidobacterium microflora of the human intestine:composition and succession.

Reuter G(1).

42. Microb Biotechnol. 2013 Jul;6(4):335-40. doi: 10.1111/1751-7915.12049. Epub 2013 Apr 18.

Prebiotics, faecal transplants and microbial network units to stimulatebiodiversity of the human gut microbiome.

Van den Abbeele P(1), Verstraete W, El Aidy S, Geirnaert A, Van de Wiele T.

43. Nutr Metab Cardiovasc Dis. 2013 Mar;23(3):160-8. doi:10.1016/j.numecd.2012.09.002. Epub 2012 Nov 10.

The role of gut microbiota in human obesity: recent findings and futureperspectives.

Tagliabue A(1), Elli M.

44. Nature. 2009 Jan 22;457(7228):480-4. doi: 10.1038/nature07540. Epub 2008 Nov 30.

A core gut microbiome in obese and lean twins.

Turnbaugh PJ(1), Hamady M, Yatsunenko T, Cantarel BL, Duncan A, Ley RE, Sogin ML,Jones WJ, Roe BA, Affourtit JP, Egholm M, Henrissat B, Heath AC, Knight R, GordonJI.

45. J Clin Biochem Nutr. 2016 Jul;59(1):65-70. doi: 10.3164/jcbn.15-152. Epub 2016Apr 29.

Comparison of the gut microbial community between obese and lean peoples using16S gene sequencing in a Japanese population.

46. Diabetes Care. 2015 Jan;38(1):159-65. doi: 10.2337/dc14-0769.

Insights into the role of the microbiome in obesity and type 2 diabetes.

Hartstra AV(1), Bouter KE(1), Bäckhed F(2), Nieuwdorp M(3).

47. Gut. 2014 Dec;63(12):1913-20. doi: 10.1136/gutjnl-2013-306541. Epub 2014 Jun 9.

Exercise and associated dietary extremes impact on gut microbial diversity.

Clarke SF(1), Murphy EF(2), O'Sullivan O(3), Lucey AJ(4), Humphreys M(5), HoganA(6), Hayes P(6), O'Reilly M(2), Jeffery IB(7), Wood-Martin R(8), Kerins DM(9),Quigley E(6), Ross RP(10), O'Toole PW(11), Molloy MG(12), Falvey E(13), Shanahan F(14), Cotter PD(10).

48. Exerc Sport Sci Rev. 2016 Oct 25. [Epub ahead of print]

Exercise Is a Novel Promoter of Intestinal Health and Microbial Diversity.

Campbell SC(1), Wisniewski PJ 2nd.

49. Int J Food Microbiol. 2004 Nov 15;96(3):219-33.

Monostrain, multistrain and multispecies probiotics--A comparison offunctionality and efficacy.

Timmerman HM(1), Koning CJ, Mulder L, Rombouts FM, Beynen AC.

50. Lett Appl Microbiol. 2000 Jan;30(1):10-3.

The mucus binding of Bifidobacterium lactis Bb12 is enhanced in the presence ofLactobacillus GG and Lact. delbrueckii subsp. bulgaricus.

Ouwehand AC(1), Isolauri E, Kirjavainen PV, Tölkko S, Salminen SJ.

51. J Dairy Sci. 2007 Jun;90(6):2710-6.

Development of new probiotics by strain combinations: is it possible to improvethe adhesion to intestinal mucus?

Collado MC(1), Meriluoto J, Salminen S.

52. Asia Pac J Clin Nutr. 2006;15(4):570-5.

Protection mechanism of probiotic combination against human pathogens: in vitroadhesion to human intestinal mucus.

Collado MC(1), Jalonen L, Meriluoto J, Salminen S.

53. Int J Food Microbiol. 2007 Feb 28;114(1):113-9. Epub 2007 Jan 17.

The effect of calcium ions on adhesion and competitive exclusion of Lactobacillusssp. and E. coli O138.

Larsen N(1), Nissen P, Willats WG.

54. Clin Vaccine Immunol. 2008 Sep;15(9):1472-82. doi: 10.1128/CVI.00080-08. Epub2008 Jun 25.

Effects of multispecies probiotic combination on helicobacter pylori infection invitro.

Myllyluoma E(1), Ahonen AM, Korpela R, Vapaatalo H, Kankuri E.

55. Curr Microbiol. 2010 May;60(5):327-35. doi: 10.1007/s00284-009-9545-1. Epub 2009 Dec 1.

An in vitro study on bacterial growth interactions and intestinal epithelial celladhesion characteristics of probiotic combinations.

Moussavi M(1), Adams MC.

56. Mol Biosyst. 2014 Dec;10(12):3044-65. doi: 10.1039/c3mb70602h.

Evolutionary game theory: cells as players.

Hummert S(1), Bohl K, Basanta D, Deutsch A, Werner S, Theissen G, Schroeter A,Schuster S.

57. J Mol Biol. 2016 Aug 14;428(16):3230-52. doi: 10.1016/j.jmb.2016.06.021. Epub2016 Jul 6.

Polysaccharide Degradation by the Intestinal Microbiota and Its Influence onHuman Health and Disease.

Cockburn DW(1), Koropatkin NM(2).

58. Appl Environ Microbiol. 2006 Dec;72(12):7835-41. Epub 2006 Oct 20.

Cross-feeding between Bifidobacterium longum BB536 and acetate-converting,butyrate-producing colon bacteria during growth on oligofructose.

Falony G(1), Vlachou A, Verbrugghe K, De Vuyst L.

59. Appl Environ Microbiol. 2006 May;72(5):3593-9.

Two routes of metabolic cross-feeding between Bifidobacterium adolescentis andbutyrate-producing anaerobes from the human gut.

Belenguer A(1), Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ.

60. Environ Microbiol. 2013 Jan;15(1):211-26. doi: 10.1111/j.1462-2920.2012.02845.x. Epub 2012 Aug 14.

Microbiota from the distal guts of lean and obese adolescents exhibit partialfunctional redundancy besides clear differences in community structure.

Ferrer M(1), Ruiz A, Lanza F, Haange SB, Oberbach A, Till H, Bargiela R, CampoyC, Segura MT, Richter M, von Bergen M, Seifert J, Suarez A.

61. PLoS One. 2009 Sep 23;4(9):e7125. doi: 10.1371/journal.pone.0007125.

Monitoring bacterial community of human gut microbiota reveals an increase inLactobacillus in obese patients and Methanogens in anorexic patients.

Armougom F(1), Henry M, Vialettes B, Raccah D, Raoult D.

62. BMC Microbiol. 2012 Nov 28;12:283. doi: 10.1186/1471-2180-12-283.

Correlation of intestinal microbiota with overweight and obesity in Kazakh schoolchildren.

Xu P(1), Li M, Zhang J, Zhang T.

63. BMC Gastroenterol. 2015 Aug 11;15:100. doi: 10.1186/s12876-015-0330-2.

Comparison of the gut microbiota composition between obese and non-obeseindividuals in a Japanese population, as analyzed by terminal restrictionfragment length polymorphism and next-generation sequencing.

Kasai C(1), Sugimoto K(2,)(3), Moritani I(4), Tanaka J(5), Oya Y(6), Inoue H(7), Tameda M(8,)(9), Shiraki K(10), Ito M(11), Takei Y(12), Takase K(13).

64. Nutr Res. 2015 Aug;35(8):655-63. doi: 10.1016/j.nutres.2015.05.001. Epub 2015 May14.

Schisandra chinensis fruit modulates the gut microbiota composition inassociation with metabolic markers in obese women: a randomized, double-blindplacebo-controlled study.

Song MY(1), Wang JH(2), Eom T(3), Kim H(4).

65. Br J Nutr. 2010 Apr;103(7):1070-8. doi: 10.1017/S0007114509992807. Epub 2009 Nov 24.

Effect of overweight on gastrointestinal microbiology and immunology: correlationwith blood biomarkers.

Tiihonen K(1), Ouwehand AC, Rautonen N.

66. Lett Appl Microbiol. 2008 Nov;47(5):367-73. doi:10.1111/j.1472-765X.2008.02408.x.

Development of a real-time PCR method for Firmicutes and Bacteroidetes in faeces and its application to quantify intestinal population of obese and lean pigs.

Guo X(1), Xia X, Tang R, Zhou J, Zhao H, Wang K.

67. PLoS One. 2013 Aug 27;8(8):e71108. doi: 10.1371/journal.pone.0071108. eCollection2013.

Human gut microbiota changes reveal the progression of glucose intolerance.

Zhang X(1), Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji L.

68. J Nutr. 2013 Apr;143(4):417-23. doi: 10.3945/jn.112.166322. Epub 2013 Jan 23.

Habitual dietary intake is associated with stool microbiota composition inmonozygotic twins.

Simões CD(1), Maukonen J, Kaprio J, Rissanen A, Pietiläinen KH, Saarela M.

69. PLoS One. 2013 Aug 27;8(8):e71108. doi: 10.1371/journal.pone.0071108. eCollection2013.

Human gut microbiota changes reveal the progression of glucose intolerance.

Zhang X(1), Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji L.

70. J Nutr. 2013 Apr;143(4):417-23. doi: 10.3945/jn.112.166322. Epub 2013 Jan 23.

Habitual dietary intake is associated with stool microbiota composition inmonozygotic twins.

Simões CD(1), Maukonen J, Kaprio J, Rissanen A, Pietiläinen KH, Saarela M.

71. PLoS One. 2013;8(3):e59260. doi: 10.1371/journal.pone.0059260. Epub 2013 Mar 14.

Smoking cessation induces profound changes in the composition of the intestinalmicrobiota in humans.

Biedermann L(1), Zeitz J, Mwinyi J, Sutter-Minder E, Rehman A, Ott SJ,Steurer-Stey C, Frei A, Frei P, Scharl M, Loessner MJ, Vavricka SR, Fried M,

Schreiber S, Schuppler M, Rogler G.

72. PLoS One. 2012;7(7):e41079. doi: 10.1371/journal.pone.0041079. Epub 2012 Jul 26.

Bacteroides uniformis CECT 7771 ameliorates metabolic and immunologicaldysfunction in mice with high-fat-diet induced obesity.

Gauffin Cano P(1), Santacruz A, Moya Á, Sanz Y.

73. Mucosal Immunol. 2016 Apr 27. doi: 10.1038/mi.2016.42. [Epub ahead of print]

Gut commensal Bacteroides acidifaciens prevents obesity and improves insulinsensitivity in mice.

Yang JY(1,)(2), Lee YS(1), Kim Y(1), Lee SH(1), Ryu S(2), Fukuda S(3), Hase K(4),Yang CS(5), Lim HS(6), Kim MS(6), Kim HM(7), Ahn SH(8), Kwon BE(9), Ko HJ(9),Kweon MN(1).

74. Gut Pathog. 2013 Apr 30;5(1):10. doi: 10.1186/1757-4749-5-10.

Differences in gut microbiota composition between obese and lean children: across-sectional study.

Bervoets L(1), Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C,Goossens H, Desager KN, Vankerckhoven V.

75. PLoS One. 2012;7(11):e49653. doi: 10.1371/journal.pone.0049653. Epub 2012 Nov 15.

Evidence of Bacteroides fragilis protection from Bartonella henselae-induceddamage.

Sommese L(1), Pagliuca C, Avallone B, Ippolito R, Casamassimi A, Costa V,Colicchio R, Cerciello R, D'Armiento M, Scarpato M, Giovane A, Pastore G, InfanteT, Ciccodicola A, Fiorito C, D'Armiento FP, Salvatore P, Napoli C.

76. Science. 2011 May 20;332(6032):974-7. doi: 10.1126/science.1206095. Epub 2011 Apr21.

The Toll-like receptor 2 pathway establishes colonization by a commensal of the

human microbiota.

Round JL(1), Lee SM, Li J, Tran G, Jabri B, Chatila TA, Mazmanian SK.

77. Proc Natl Acad Sci U S A. 2010 Jul 6;107(27):12204-9. doi:10.1073/pnas.0909122107. Epub 2010 Jun 21.

Inducible Foxp3+ regulatory T-cell development by a commensal bacterium of theintestinal microbiota.

Round JL(1), Mazmanian SK.

78. J Immunol. 2010 Oct 1;185(7):4101-8. doi: 10.4049/jimmunol.1001443. Epub 2010 Sep3.

Central nervous system demyelinating disease protection by the human commensalBacteroides fragilis depends on polysaccharide A expression.

Ochoa-Repáraz J(1), Mielcarz DW, Ditrio LE, Burroughs AR, Begum-Haque S, DasguptaS, Kasper DL, Kasper LH.

79. Anaerobe. 2011 Aug;17(4):137-41. doi: 10.1016/j.anaerobe.2011.05.017. Epub 2011Jun 2.

The human commensal Bacteroides fragilis binds intestinal mucin.

Huang JY(1), Lee SM, Mazmanian SK.

80. Chem Biol Interact. 2011 Jan 15;189(1-2):1-8. doi: 10.1016/j.cbi.2010.10.002.Epub 2010 Oct 15.

High polyphenol, low probiotic diet for weight loss because of intestinalmicrobiota interaction.

Rastmanesh R(1).

81. BMC Biol. 2013 May 21;11:61. doi: 10.1186/1741-7007-11-61.

Bacteroides thetaiotaomicron and Faecalibacterium prausnitzii influence theproduction of mucus glycans and the development of goblet cells in the colonicepithelium of a gnotobiotic model rodent.

Wrzosek L(1), Miquel S, Noordine ML, Bouet S, Joncquel Chevalier-Curt M, Robert

V, Philippe C, Bridonneau C, Cherbuy C, Robbe-Masselot C, Langella P, Thomas M.

82. Gene. 2014 Mar 1;537(1):85-92. doi: 10.1016/j.gene.2013.11.081. Epub 2013 Dec 8.

Effects of short chain fatty acid producing bacteria on epigenetic regulation of FFAR3 in type 2 diabetes and obesity.

Remely M(1), Aumueller E(1), Merold C(1), Dworzak S(1), Hippe B(1), Zanner J(1), Pointner A(1), Brath H(2), Haslberger AG(3).

83. Benef Microbes. 2016 Sep;7(4):511-7. doi: 10.3920/BM2015.0075. Epub 2016 Apr 6.

Faecalibacterium prausnitzii phylotypes in type two diabetic, obese, and leancontrol subjects.

Hippe B(1), Remely M(1), Aumueller E(1), Pointner A(1), Magnet U(1), HaslbergerAG(1).

84. Diabetes. 2010 Dec;59(12):3049-57. doi: 10.2337/db10-0253. Epub 2010 Sep 28.

Differential adaptation of human gut microbiota to bariatric surgery-inducedweight loss: links with metabolic and low-grade inflammation markers.

Furet JP(1), Kong LC, Tap J, Poitou C, Basdevant A, Bouillot JL, Mariat D,Corthier G, Doré J, Henegar C, Rizkalla S, Clément K.

85. PLoS One. 2013 Aug 27;8(8):e71108. doi: 10.1371/journal.pone.0071108. eCollection2013.

Human gut microbiota changes reveal the progression of glucose intolerance.

Zhang X(1), Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji L.

86. Epub 2013 Nov 13.

Increased proportions of Bifidobacterium and the Lactobacillus group and loss of butyrate-producing bacteria in inflammatory bowel disease.

Wang W(1), Chen L, Zhou R, Wang X, Song L, Huang S, Wang G, Xia B.

87. Gut. 2014 Aug;63(8):1275-83. doi: 10.1136/gutjnl-2013-304833. Epub 2013 Sep 10.

A decrease of the butyrate-producing species Roseburia hominis andFaecalibacterium prausnitzii defines dysbiosis in patients with ulcerativecolitis.

Machiels K(1), Joossens M(2), Sabino J(1), De Preter V(3), Arijs I(1), EeckhautV(4), Ballet V(1), Claes K(1), Van Immerseel F(4), Verbeke K(3), Ferrante M(1),Verhaegen J(5), Rutgeerts P(1), Vermeire S(1).

88. BMC Syst Biol. 2014 Apr 3;8:41. doi: 10.1186/1752-0509-8-41.

Genome-scale metabolic reconstructions of Bifidobacterium adolescentis L2-32 and Faecalibacterium prausnitzii A2-165 and their interaction.

El-Semman IE, Karlsson FH, Shoaie S, Nookaew I, Soliman TH, Nielsen J(1).

89. Scand J Gastroenterol. 2013 Oct;48(10):1136-44. doi:10.3109/00365521.2013.828773. Epub 2013 Aug 26.

Faecalibacterium prausnitzii supernatant improves intestinal barrier function in mice DSS colitis.

Carlsson AH(1), Yakymenko O, Olivier I, Håkansson F, Postma E, Keita AV,Söderholm JD.

90. Appl Environ Microbiol. 2015 Jun;81(11):3655-62. doi: 10.1128/AEM.04050-14. Epub 2015 Mar 20.

Akkermansia muciniphila Adheres to Enterocytes and Strengthens the Integrity ofthe Epithelial Cell Layer.

Reunanen J(1), Kainulainen V(1), Huuskonen L(1), Ottman N(2), Belzer C(2),Huhtinen H(3), de Vos WM(4), Satokari R(5).

91. Clin Nutr. 2013 Dec;32(6):1017-22. doi: 10.1016/j.clnu.2013.02.008. Epub 2013 Feb21.

Faecal levels of Bifidobacterium and Clostridium coccoides but not plasmalipopolysaccharide are inversely related to insulin and HOMA index in women.

F S Teixeira T(1), Grześkowiak LM, Salminen S, Laitinen K, Bressan J, GouveiaPeluzio Mdo C.

92. Proc Natl Acad Sci U S A. 2013 May 28;110(22):9066-71. doi:10.1073/pnas.1219451110. Epub 2013 May 13.

Cross-talk between Akkermansia muciniphila and intestinal epithelium controlsdiet-induced obesity.

Everard A(1), Belzer C, Geurts L, Ouwerkerk JP, Druart C, Bindels LB, Guiot Y,Derrien M, Muccioli GG, Delzenne NM, de Vos WM, Cani PD.

93. Gut. 2016 Mar;65(3):426-36. doi: 10.1136/gutjnl-2014-308778. Epub 2015 Jun 22.

Akkermansia muciniphila and improved metabolic health during a dietaryintervention in obesity: relationship with gut microbiome richness and ecology.

Dao MC(1), Everard A(2), Aron-Wisnewsky J(1), Sokolovska N(1), Prifti E(3),Verger EO(1), Kayser BD(3), Levenez F(4), Chilloux J(5), Hoyles L(5); MICRO-Obes Consortium, Dumas ME(5), Rizkalla SW(3), Doré J(4), Cani PD(2), Clément K(1).

94. Gut. 2014 May;63(5):727-35. doi: 10.1136/gutjnl-2012-303839. Epub 2013 Jun 26.

An increase in the Akkermansia spp. population induced by metformin treatmentimproves glucose homeostasis in diet-induced obese mice.

Shin NR(1), Lee JC, Lee HY, Kim MS, Whon TW, Lee MS, Bae JW.

95. PLoS One. 2013 Aug 27;8(8):e71108. doi: 10.1371/journal.pone.0071108. eCollection2013.

Human gut microbiota changes reveal the progression of glucose intolerance.

Zhang X(1), Shen D, Fang Z, Jie Z, Qiu X, Zhang C, Chen Y, Ji L.

96. PLoS One. 2013 Oct 24;8(10):e76520. doi: 10.1371/journal.pone.0076520.eCollection 2013.

Extracellular vesicles derived from gut microbiota, especially Akkermansiamuciniphila, protect the progression of dextran sulfate sodium-induced colitis.

Kang CS(1), Ban M, Choi EJ, Moon HG, Jeon JS, Kim DK, Park SK, Jeon SG, Roh TY,Myung SJ, Gho YS, Kim JG, Kim YK.

97. Benef Microbes. 2012 Dec 1;3(4):287-97. doi: 10.3920/BM2012.0018.

Gram-negative bacteria account for main differences between faecal microbiotafrom patients with ulcerative colitis and healthy controls.

Vigsnæs LK(1), Brynskov J, Steenholdt C, Wilcks A, Licht TR.

98. FEMS Microbiol Ecol. 2014 Jan;87(1):30-40. doi: 10.1111/1574-6941.12186. Epub2013 Aug 28.

Prebiotic stimulation of human colonic butyrate-producing bacteria andbifidobacteria, in vitro.

Scott KP(1), Martin JC, Duncan SH, Flint HJ.

99. J Nutr Biochem. 2012 Jan;23(1):51-9. doi: 10.1016/j.jnutbio.2010.10.008. Epub2011 Mar 15.

Dietary modulation of clostridial cluster XIVa gut bacteria (Roseburia spp.) bychitin-glucan fiber improves host metabolic alterations induced by high-fat diet in mice.

Neyrinck AM(1), Possemiers S, Verstraete W, De Backer F, Cani PD, Delzenne NM.

100. Br J Nutr. 2014 Nov 14;112(9):1447-58. doi: 10.1017/S0007114514002396. Epub 2014 Sep 19.

Finger millet bran supplementation alleviates obesity-induced oxidative stress,inflammation and gut microbial derangements in high-fat diet-fed mice.

Murtaza N(1), Baboota RK(1), Jagtap S(2), Singh DP(1), Khare P(1), Sarma SM(1),Podili K(3), Alagesan S(4), Chandra TS(5), Bhutani KK(2), Boparai RK(6), Bishnoi M(1), Kondepudi KK(1).

101. Gut. 2014 Sep;63(9):1513-21. doi: 10.1136/gutjnl-2014-306928. Epub 2014 May 15.

Microbiota and diabetes: an evolving relationship.

Tilg H(1), Moschen AR(1).

102. J Clin Endocrinol Metab. 2016 Jan;101(1):233-42. doi: 10.1210/jc.2015-3351. Epub 2015 Oct 27.

Two Healthy Diets Modulate Gut Microbial Community Improving Insulin Sensitivity in a Human Obese Population.

Haro C(1), Montes-Borrego M(1), Rangel-Zúñiga OA(1), Alcalá-Díaz JF(1),Gómez-Delgado F(1), Pérez-Martínez P(1), Delgado-Lista J(1), Quintana-NavarroGM(1), Tinahones FJ(1), Landa BB(1), López-Miranda J(1), Camargo A(1),Pérez-Jiménez F(1).

103. Nat Commun. 2012;3:1245. doi: 10.1038/ncomms2266.

Symptomatic atherosclerosis is associated with an altered gut metagenome.

Karlsson FH(1), Fåk F, Nookaew I, Tremaroli V, Fagerberg B, Petranovic D, BäckhedF, Nielsen J.

104. Gut. 2014 Aug;63(8):1275-83. doi: 10.1136/gutjnl-2013-304833. Epub 2013 Sep 10.

A decrease of the butyrate-producing species Roseburia hominis andFaecalibacterium prausnitzii defines dysbiosis in patients with ulcerativecolitis.

Machiels K(1), Joossens M(2), Sabino J(1), De Preter V(3), Arijs I(1), EeckhautV(4), Ballet V(1), Claes K(1), Van Immerseel F(4), Verbeke K(3), Ferrante M(1),Verhaegen J(5), Rutgeerts P(1), Vermeire S(1).

105. Am J Clin Nutr. 2011 May;93(5):1062-72. doi: 10.3945/ajcn.110.002188. Epub 2011Mar 9.

High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health.

Russell WR(1), Gratz SW, Duncan SH, Holtrop G, Ince J, Scobbie L, Duncan G,Johnstone AM, Lobley GE, Wallace RJ, Duthie GG, Flint HJ.

106. Appl Environ Microbiol. 2007 Feb;73(4):1073-8. Epub 2006 Dec 22.

Reduced dietary intake of carbohydrates by obese subjects results in decreased

concentrations of butyrate and butyrate-producing bacteria in feces.

Duncan SH(1), Belenguer A, Holtrop G, Johnstone AM, Flint HJ, Lobley GE.

107. Front Microbiol. 2016 Jun 28;7:979. doi: 10.3389/fmicb.2016.00979. eCollection2016.

Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategiesfor Their Stimulation in the Human Gut.

Rivière A(1), Selak M(1), Lantin D(1), Leroy F(1), De Vuyst L(1).

108. Int J Food Microbiol. 2016 Aug 16;231:76-85. doi:10.1016/j.ijfoodmicro.2016.05.015. Epub 2016 May 12.

Bifidobacterial inulin-type fructan degradation capacity determines cross-feedinginteractions between bifidobacteria and Faecalibacterium prausnitzii.

Moens F(1), Weckx S(1), De Vuyst L(2).

109. BMC Syst Biol. 2014 Apr 3;8:41. doi: 10.1186/1752-0509-8-41.

Genome-scale metabolic reconstructions of Bifidobacterium adolescentis L2-32 and Faecalibacterium prausnitzii A2-165 and their interaction.

El-Semman IE, Karlsson FH, Shoaie S, Nookaew I, Soliman TH, Nielsen J(1).

110. FEMS Microbiol Lett. 2015 Nov;362(21). pii: fnv176. doi: 10.1093/femsle/fnv176.Epub 2015 Sep 28.

Enhanced butyrate formation by cross-feeding between Faecalibacterium prausnitziiand Bifidobacterium adolescentis

Rios-Covian D(1), Gueimonde M(2), Duncan SH(3), Flint HJ(3), de los Reyes-GavilanCG(4).

111. Int J Syst Evol Microbiol. 2006 Oct;56(Pt 10):2437-41.

Proposal of Roseburia faecis sp. nov., Roseburia hominis sp. nov. and Roseburiainulinivorans sp. nov., based on isolates from human faeces.

Duncan SH(1), Aminov RI, Scott KP, Louis P, Stanton TB, Flint HJ.

112. Appl Environ Microbiol. 2006 Dec;72(12):7835-41. Epub 2006 Oct 20.

Cross-feeding between Bifidobacterium longum BB536 and acetate-converting,butyrate-producing colon bacteria during growth on oligofructose.

Falony G(1), Vlachou A, Verbrugghe K, De Vuyst L.

113. Appl Environ Microbiol. 2006 May;72(5):3593-9.

Two routes of metabolic cross-feeding between Bifidobacterium adolescentis andbutyrate-producing anaerobes from the human gut.

Belenguer A(1), Duncan SH, Calder AG, Holtrop G, Louis P, Lobley GE, Flint HJ.

114. Front Microbiol. 2016 Jun 28;7:979. doi: 10.3389/fmicb.2016.00979. eCollection2016.

Bifidobacteria and Butyrate-Producing Colon Bacteria: Importance and Strategiesfor Their Stimulation in the Human Gut.

Rivière A(1), Selak M(1), Lantin D(1), Leroy F(1), De Vuyst L(1).

115. Appl Environ Microbiol. 2003 Feb;69(2):1136-42.

Effects of alternative dietary substrates on competition between human colonicbacteria in an anaerobic fermentor system.

Duncan SH(1), Scott KP, Ramsay AG, Harmsen HJ, Welling GW, Stewart CS, Flint HJ.

116. Br J Nutr. 2013 Apr 14;109(7):1338-48. doi: 10.1017/S0007114512003066. Epub 2012 Jul 31.

Dietary galacto-oligosaccharides and calcium: effects on energy intake, fat-padweight and satiety-related, gastrointestinal hormones in rats.

Overduin J(1), Schoterman MH, Calame W, Schonewille AJ, Ten Bruggencate SJ.

117. J Nutr. 2015 Sep;145(9):2052-9. doi: 10.3945/jn.114.204909. Epub 2015 Jul 15.

α-Galacto-oligosaccharides Dose-Dependently Reduce Appetite and Decrease

Inflammation in Overweight Adults.

Morel FB(1), Dai Q(2), Ni J(3), Thomas D(4), Parnet P(5), Fança-Berthon P(6).

118. Am J Clin Nutr. 2008 Nov;88(5):1438-46.

Modulation of the fecal microflora profile and immune function by a noveltrans-galactooligosaccharide mixture (B-GOS) in healthy elderly volunteers.

Vulevic J(1), Drakoularakou A, Yaqoob P, Tzortzis G, Gibson GR.

119. PLoS One. 2011;6(9):e25200. doi: 10.1371/journal.pone.0025200. Epub 2011 Sep 26.

Barcoded pyrosequencing reveals that consumption of galactooligosaccharidesresults in a highly specific bifidogenic response in humans.

Davis LM(1), Martínez I, Walter J, Goin C, Hutkins RW.

120. Am J Clin Nutr. 2008 Mar;87(3):785-91.

Prebiotic evaluation of a novel galactooligosaccharide mixture produced by the enzymatic activity of Bifidobacterium bifidum NCIMB 41171, in healthy humans: a randomized, double-blind, crossover, placebo-controlled intervention study.

Depeint F(1), Tzortzis G, Vulevic J, I'anson K, Gibson GR.

121. Br J Nutr. 2015 Aug 28;114(4):586-95. doi: 10.1017/S0007114515001889. Epub 2015Jul 28.

Influence of galacto-oligosaccharide mixture (B-GOS) on gut microbiota, immune parameters and metabonomics in elderly persons.

Vulevic J(1), Juric A(2), Walton GE(2), Claus SP(2), Tzortzis G(1), Toward RE(2),Gibson GR(2).

122. J Nutr. 2005 Jul;135(7):1726-31.

A novel galactooligosaccharide mixture increases the bifidobacterial population numbers in a continuous in vitro fermentation system and in the proximal colonic contents of pigs in vivo.

Tzortzis G(1), Goulas AK, Gee JM, Gibson GR.

123. J Nutr. 2012 Jul;142(7):1232-9. doi: 10.3945/jn.111.155762. Epub 2012 May 30.

Monomer and linkage type of galacto-oligosaccharides affect their resistance toileal digestion and prebiotic properties in rats.

Hernández-Hernández O(1), Marín-Manzano MC, Rubio LA, Moreno FJ, Sanz ML,Clemente A.

124. Mol Nutr Food Res. 2015 Mar;59(3):566-73. doi: 10.1002/mnfr.201400639. Epub 2014 Dec 28.

Galacto-oligosaccharides may directly enhance intestinal barrier function throughthe modulation of goblet cells.

Bhatia S(1), Prabhu PN, Benefiel AC, Miller MJ, Chow J, Davis SR, Gaskins HR.

125. J Nutr. 2015 Jul;145(7):1604-13. doi: 10.3945/jn.114.209486. Epub 2015 May 27.

Galacto-oligosaccharides Protect the Intestinal Barrier by Maintaining the Tight Junction Network and Modulating the Inflammatory Responses after a Challenge withthe Mycotoxin Deoxynivalenol in Human Caco-2 Cell Monolayers and B6C3F1 Mice.

Akbari P(1), Braber S(2), Alizadeh A(1), Verheijden KA(3), Schoterman MH(4),Kraneveld AD(3), Garssen J(5), Fink-Gremmels J(6).

126. Eur J Nutr. 2016 Jun 13. [Epub ahead of print]

Characterizing microbiota-independent effects of oligosaccharides on intestinalepithelial cells: insight into the role of structure and size :Structure-activity relationships of non-digestible oligosaccharides.

Akbari P(1,)(2), Fink-Gremmels J(1), Willems RH(3), Difilippo E(3), Schols HA(3),Schoterman MH(4), Garssen J(2,)(5), Braber S(6).

127. BMC Vet Res. 2013 Jul 22;9:147. doi: 10.1186/1746-6148-9-147.

In vitro evaluation of defined oligosaccharide fractions in an equine model ofinflammation.

Vendrig JC(1), Coffeng LE, Fink-Gremmels J.

128. Brain Behav Immun. 2016 Feb;52:120-31. doi: 10.1016/j.bbi.2015.10.007.

Prebiotic administration normalizes lipopolysaccharide (LPS)-induced anxiety and cortical 5-HT2A receptor and IL1-β levels in male mice.

Savignac HM(1), Couch Y(2), Stratford M(3), Bannerman DM(4), Tzortzis G(1),Anthony DC(2), Burnet PW(5).

129. Nutr Diabetes. 2012 Jan 23;2:e28. doi: 10.1038/nutd.2011.24.

Wheat-derived arabinoxylan oligosaccharides with prebiotic effect increasesatietogenic gut peptides and reduce metabolic endotoxemia in diet-induced obese mice.

Neyrinck AM(1), Van Hée VF, Piront N, De Backer F, Toussaint O, Cani PD, DelzenneNM.

130. Nutr J. 2012 Jun 1;11:36. doi: 10.1186/1475-2891-11-36.

A randomised, double-blind, placebo controlled cross-over study to determine the gastrointestinal effects of consumption of arabinoxylan-oligosaccharides enrichedbread in healthy volunteers.

Walton GE(1), Lu C, Trogh I, Arnaut F, Gibson GR.

131. Br J Nutr. 2010 Mar;103(5):703-13. doi: 10.1017/S0007114509992248. Epub 2009 Dec 10.

Tolerance of arabinoxylan-oligosaccharides and their prebiotic activity inhealthy subjects: a randomised, placebo-controlled cross-over study.

Cloetens L(1), Broekaert WF, Delaedt Y, Ollevier F, Courtin CM, Delcour JA,Rutgeerts P, Verbeke K.

132. Microb Biotechnol. 2009 Jan;2(1):101-13. doi: 10.1111/j.1751-7915.2008.00064.x.Epub 2008 Oct 13.

Arabinoxylan-oligosaccharides (AXOS) affect the protein/carbohydrate fermentationbalance and microbial population dynamics of the Simulator of Human IntestinalMicrobial Ecosystem.

Sanchez JI(1), Marzorati M, Grootaert C, Baran M, Van Craeyveld V, Courtin CM,Broekaert WF, Delcour JA, Verstraete W, Van de Wiele T.

133. FEMS Microbiol Ecol. 2009 Aug;69(2):231-42. doi:10.1111/j.1574-6941.2009.00712.x. Epub 2009 Jun 9.

Comparison of prebiotic effects of arabinoxylan oligosaccharides and inulin in a simulator of the human intestinal microbial ecosystem.

Grootaert C(1), Van den Abbeele P, Marzorati M, Broekaert WF, Courtin CM, DelcourJA, Verstraete W, Van de Wiele T.

134. Mol Nutr Food Res. 2011 Dec;55(12):1862-74. doi: 10.1002/mnfr.201100377. Epub2011 Nov 7.

Prebiotic effects and intestinal fermentation of cereal arabinoxylans andarabinoxylan oligosaccharides in rats depend strongly on their structuralproperties and joint presence.

Damen B(1), Verspreet J, Pollet A, Broekaert WF, Delcour JA, Courtin CM.

135. J Agric Food Chem. 2012 Feb 1;60(4):946-54. doi: 10.1021/jf203820j. Epub 2012 Jan19.

In vitro fermentation of arabinoxylan oligosaccharides and low molecular massarabinoxylans with different structural properties from wheat (Triticum aestivum L.) bran and psyllium (Plantago ovata Forsk) seed husk.

Pollet A(1), Van Craeyveld V, Van de Wiele T, Verstraete W, Delcour JA, CourtinCM.

136. Benef Microbes. 2010 Mar;1(1):81-91. doi: 10.3920/BM2009.0025.

Xylo-oligosaccharides enhance the growth of bifidobacteria and Bifidobacteriumlactis in a simulated colon model.

Mäkeläinen H(1), Forssten S, Saarinen M, Stowell J, Rautonen N, Ouwehand AC.

137. Br J Nutr. 2012 Nov 28;108(10):1847-58. doi: 10.1017/S0007114511007252. Epub 2012Jan 23.

Xylo-oligosaccharide (XOS) in combination with inulin modulates both theintestinal environment and immune status in healthy subjects, while XOS aloneonly shows prebiotic properties.

Lecerf JM(1), Dépeint F, Clerc E, Dugenet Y, Niamba CN, Rhazi L, Cayzeele A,Abdelnour G, Jaruga A, Younes H, Jacobs H, Lambrey G, Abdelnour AM, Pouillart PR.

138. FEMS Microbiol Ecol. 2016 Nov 2. pii: fiw226. [Epub ahead of print]

Mannan- and Xylo-oligosaccharides modulate cecal microbiota and expression ofinflammatory related cytokines and reduce cecal Salmonella Enteritidiscolonization in young chickens.

Pourabedin M(1), Chen Q(2), Yang M(2), Zhao X(3).

139. Benef Microbes. 2010 Sep;1(3):271-81. doi: 10.3920/BM2010.0016.

Analysis of the intestinal microbiota of oligosaccharide fed mice exhibitingreduced resistance to Salmonella infection.

Petersen A(1), Bergström A, Andersen JB, Hansen M, Lahtinen SJ, Wilcks A, LichtTR.

140. Sci Rep. 2016 Sep 29;6:34360. doi: 10.1038/srep34360.

Bacteroides intestinalis DSM 17393, a member of the human colonic microbiome,upregulates multiple endoxylanases during growth on xylan.

Wang K(1,)(2), Pereira GV(1,)(2,)(3), Cavalcante JJ(1,)(2), Zhang M(1,)(2,)(3),Mackie R(1,)(2,)(3), Cann I(1,)(2,)(3,)(4).

141. Nutrients. 2013 Sep 23;5(9):3740-56. doi: 10.3390/nu5093740.

Xylo-oligosaccharides and inulin affect genotoxicity and bacterial populationsdifferently in a human colonic simulator challenged with soy protein.

Christophersen CT(1), Petersen A, Licht TR, Conlon MA.

142. J Nutr. 1995 Oct;125(10):2604-9.

Dietary fructooligosaccharide, xylooligosaccharide and gum arabic have variableeffects on cecal and colonic microbiota and epithelial cell proliferation in miceand rats.

Howard MD(1), Gordon DT, Garleb KA, Kerley MS.

143. Life Sci. 2016 Mar 1;148:112-7. doi: 10.1016/j.lfs.2016.02.018. Epub 2016 Feb 13.

Pharmacological inhibition of galectin-1 by lactulose alleviates weight gain indiet-induced obese rats.

Mukherjee R(1), Yun JW(2).

144. Ital J Gastroenterol. 1994 May;26(4):174-8.

Lowering effects of a preparation containing fibres and lactulose on glucose and insulin levels in obesity.

Bianchi GP(1), De Mitri MS, Bugianesi E, Abbiati R, Fabbri A, Marchesini G.

145. Scand J Gastroenterol Suppl. 1997;222:62-4.

Effect of lactulose on carbohydrate metabolism and diabetes mellitus.

Bianchi G(1), Ronchi M, Marchesini G.

146. Aliment Pharmacol Ther. 2006 Apr 1;23(7):963-74.

Effect of lactulose and Saccharomyces boulardii administration on the colonicurea-nitrogen metabolism and the bifidobacteria concentration in healthy humansubjects.

De Preter V(1), Vanhoutte T, Huys G, Swings J, Rutgeerts P, Verbeke K.

147. Appl Environ Microbiol. 2006 Sep;72(9):5990-7.

Molecular monitoring of the fecal microbiota of healthy human subjects duringadministration of lactulose and Saccharomyces boulardii.

Vanhoutte T(1), De Preter V, De Brandt E, Verbeke K, Swings J, Huys G.

148. J Agric Food Chem. 2016 Aug 10;64(31):6240-7. doi: 10.1021/acs.jafc.6b02305. Epub2016 Aug 2.

Lactulose Differently Modulates the Composition of Luminal and Mucosal Microbiotain C57BL/6J Mice.

Mao B, Li D, Ai C(1), Zhao J, Zhang H, Chen W(2).

149. J Am Coll Nutr. 2006 Dec;25(6):541-9.

Influence of long-term administration of lactulose and Saccharomyces boulardii onthe colonic generation of phenolic compounds in healthy human subjects.

De Preter V(1), Coopmans T, Rutgeerts P, Verbeke K.

150. J Med Food. 2009 Oct;12(5):1023-8. doi: 10.1089/jmf.2009.0128.

Inulin increases glucose transport in C2C12 myotubes and HepG2 cells viaactivation of AMP-activated protein kinase and phosphatidylinositol 3-kinasepathways.

Yun H(1), Lee JH, Park CE, Kim MJ, Min BI, Bae H, Choe W, Kang I, Kim SS, Ha J.

151. Am J Physiol Gastrointest Liver Physiol. 2007 Jan;292(1):G358-68. Epub 2006 Sep21.

Effects of Lactobacillus casei Shirota, Bifidobacterium breve, andoligofructose-enriched inulin on colonic nitrogen-protein metabolism in healthyhumans.

De Preter V(1), Vanhoutte T, Huys G, Swings J, De Vuyst L, Rutgeerts P, VerbekeK.

152. Br J Nutr. 2005 May;93(5):677-84.

Brussels sprouts, inulin and fermented milk alter the faecal microbiota of human microbiota-associated rats as shown by PCR-temporal temperature gradient gelelectrophoresis using universal, Lactobacillus and Bifidobacterium 16S rRNA gene primers.

Humblot C(1), Bruneau A, Sutren M, Lhoste EF, Doré J, Andrieux C, Rabot S.

153. Anaerobe. 2008 Nov;14(5):280-6. doi: 10.1016/j.anaerobe.2008.07.002. Epub 2008Aug 5.

In vitro comparison of the prebiotic effects of two inulin-type fructans.

Pompei A(1), Cordisco L, Raimondi S, Amaretti A, Pagnoni UM, Matteuzzi D, RossiM.

154 J Appl Microbiol. 2007 Feb;102(2):452-60.

Inulin-type fructans of longer degree of polymerization exert more pronounced in vitro prebiotic effects.

van de Wiele T(1), Boon N, Possemiers S, Jacobs H, Verstraete W.

155. Br J Nutr. 2009 Feb;101(4):541-50. doi: 10.1017/S0007114508019880. Epub 2008 Jul 1.

Effect of inulin on the human gut microbiota: stimulation of Bifidobacteriumadolescentis and Faecalibacterium prausnitzii.

Ramirez-Farias C(1), Slezak K, Fuller Z, Duncan A, Holtrop G, Louis P.

156. Environ Microbiol. 2011 Oct;13(10):2667-80. doi:10.1111/j.1462-2920.2011.02533.x. Epub 2011 Aug 30.

Arabinoxylans and inulin differentially modulate the mucosal and luminal gutmicrobiota and mucin-degradation in humanized rats.

Van den Abbeele P(1), Gérard P, Rabot S, Bruneau A, El Aidy S, Derrien M,Kleerebezem M, Zoetendal EG, Smidt H, Verstraete W, Van de Wiele T, Possemiers S.

157. Microbiome. 2015 Apr 16;3:16. doi: 10.1186/s40168-015-0078-5. eCollection 2015.

Resistant starch diet induces change in the swine microbiome and a predominanceof beneficial bacterial populations.

Umu ÖC(1), Frank JA(1), Fangel JU(2), Oostindjer M(1), da Silva CS(3), BolhuisEJ(4), Bosch G(5), Willats WG(2), Pope PB(1), Diep DB(1).

158. ISME J. 2011 Feb;5(2):220-30. doi: 10.1038/ismej.2010.118. Epub 2010 Aug 5.

Dominant and diet-responsive groups of bacteria within the human colonicmicrobiota.

Walker AW(1), 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.

159. FEMS Microbiol Ecol. 2008 Dec;66(3):505-15. doi:10.1111/j.1574-6941.2008.00527.x. Epub 2008 Jun 24.

Phylotypes related to Ruminococcus bromii are abundant in the large bowel ofhumans and increase in response to a diet high in resistant starch.

Abell GC(1), Cooke CM, Bennett CN, Conlon MA, McOrist AL.

160. Gut Microbes. 2013 May-Jun;4(3):236-40. doi: 10.4161/gmic.23998. Epub 2013 Apr 2.

Some are more equal than others: the role of "keystone" species in thedegradation of recalcitrant substrates.

Ze X(1), Le Mougen F, Duncan SH, Louis P, Flint HJ.

161. MBio. 2015 Sep 29;6(5):e01058-15. doi: 10.1128/mBio.01058-15.

Unique Organization of Extracellular Amylases into Amylosomes in the ResistantStarch-Utilizing Human Colonic Firmicutes Bacterium Ruminococcus bromii.

Ze X(1), Ben David Y(2), Laverde-Gomez JA(1), Dassa B(2), Sheridan PO(1), Duncan SH(1), Louis P(1), Henrissat B(3), Juge N(4), Koropatkin NM(5), Bayer EA(2),Flint HJ(6).

162. J Appl Microbiol. 2013 May;114(5):1516-28. doi: 10.1111/jam.12157. Epub 2013 Feb 21.

Temporal change in the gut community of rats fed high amylose cornstarch isdriven by endogenous urea rather than strictly on carbohydrate availability.

Kalmokoff M(1), Zwicker B, O'Hara M, Matias F, Green J, Shastri P, Green-Johnson J, Brooks SP.

163. Br J Nutr. 2004 Jun;91(6):915-23.

Contribution of acetate to butyrate formation by human faecal bacteria.

Duncan SH(1), Holtrop G, Lobley GE, Calder AG, Stewart CS, Flint HJ.

164. Appl Microbiol Biotechnol. 2015 Dec;99(24):10627-38. doi:

10.1007/s00253-015-6917-2. Epub 2015 Aug 30.

Responses in ileal and cecal bacteria to low and high amylose/amylopectin ratiodiets in growing pigs.

Luo YH(1), Yang C(1), Wright AD(2), He J(1), Chen DW(3).

165. Appl Environ Microbiol. 2009 Nov;75(22):7006-16. doi: 10.1128/AEM.01343-09. Epub 2009 Sep 25.

Effect of carbohydrate composition in barley and oat cultivars on microbialecophysiology and proliferation of Salmonella enterica in an in vitro model ofthe porcine gastrointestinal tract.

Pieper R(1), Bindelle J, Rossnagel B, Van Kessel A, Leterme P.

166. Environ Microbiol. 2011 Oct;13(10):2667-80. doi:

10.1111/j.1462-2920.2011.02533.x. Epub 2011 Aug 30.

Arabinoxylans and inulin differentially modulate the mucosal and luminal gutmicrobiota and mucin-degradation in humanized rats.

Van den Abbeele P(1), Gérard P, Rabot S, Bruneau A, El Aidy S, Derrien M,Kleerebezem M, Zoetendal EG, Smidt H, Verstraete W, Van de Wiele T, Possemiers S.

167. Int J Syst Evol Microbiol 2004 Sep;54(Pt 5):1469–76.

Akkermansia muciniphila gen. nov., sp. nov., a human intestinal mucindegrading bacterium.

Derrien M, Vaughan EE, Plugge CM, de Vos WM

168. FEMS Microbiol Ecol. 2016 Feb;92(2). pii: fiv165. doi: 10.1093/femsec/fiv165.Epub 2015 Dec 20.

Adding mucins to an in vitro batch fermentation model of the large intestineinduces changes in microbial population isolated from porcine feces depending on the substrate.

Tran TH(1), Boudry C(2), Everaert N(2), Théwis A(2), Portetelle D(3), Daube G(4),Nezer C(5), Taminiau B(6), Bindelle J(7).

169. Best Pract Res Clin Gastroenterol. 2013 Feb;27(1):25-38. doi:10.1016/j.bpg.2013.03.001.

Glycobiome: bacteria and mucus at the epithelial interface.

Ouwerkerk JP(1), de Vos WM, Belzer C.

170. Science 2003 Mar 28;299(5615):2074–6.

A genomic view of the human-Bacteroides thetaiotaomicron symbiosis.

Xu J, Bjursell MK, Himrod J, Deng S, Carmichael LK, Chiang HC, et al.

171. J Biol Chem. 2014 Aug 29;289(35):24289-303. doi: 10.1074/jbc.M114.573303. Epub2014 Jul 7.

Characterization of glycosaminoglycan (GAG) sulfatases from the human gutsymbiont Bacteroides thetaiotaomicron reveals the first GAG-specific bacterialendosulfatase.

Ulmer JE(1), Vilén EM(2), Namburi RB(3), Benjdia A(1), Beneteau J(1), MalleronA(4), Bonnaffé D(4), Driguez PA(5), Descroix K(5), Lassalle G(5), Le Narvor C(4),Sandström C(2), Spillmann D(6), Berteau O(7).

172. Anaerobe. 2011 Aug;17(4):137-41. doi: 10.1016/j.anaerobe.2011.05.017. Epub 2011Jun 2.

The human commensal Bacteroides fragilis binds intestinal mucin.

Huang JY(1), Lee SM, Mazmanian SK.

B. fragilis binds to murine colonic mucus. We further demonstrate that B. fragilisspecifically and quantitatively binds to highly purified mucins (the majorconstituent in intestinal mucus)

173. FEMS Microbiol Lett 1991 Jan 15;61(2–3):289–93

Formation of glycoprotein degrading enzymes by Bacteroides fragilis.

Macfarlane GT, Gibson GR.

174. ISME J. 2013 May;7(5):949-61. doi: 10.1038/ismej.2012.158. Epub 2012 Dec 13.

Butyrate-producing Clostridium cluster XIVa species specifically colonize mucins

in an in vitro gut model.

Van den Abbeele P(1), Belzer C, Goossens M, Kleerebezem M, De Vos WM, Thas O, De Weirdt R, Kerckhof FM, Van de Wiele T.

175. ISME J. 2014 Jun;8(6):1323-35. doi: 10.1038/ismej.2014.14. Epub 2014 Feb 20.

Phylogenetic distribution of three pathways for propionate production within the human gut microbiota.

Reichardt N(1), Duncan SH(2), Young P(2), Belenguer A(2), McWilliam Leitch C(2), Scott KP(2), Flint HJ(2), Louis P(2).

176. Am J Clin Nutr. 2004 Jul;80(1):89-94.

L-Rhamnose increases serum propionate in humans.

Vogt JA(1), Pencharz PB, Wolever TM.

177. Nutr Rev. 2011 May;69(5):245-58. doi: 10.1111/j.1753-4887.2011.00388.x.

Propionate as a health-promoting microbial metabolite in the human gut.

Hosseini E(1), Grootaert C, Verstraete W, Van de Wiele T.

178. Am J Clin Nutr. 2004 Nov;80(5):1254-61.

L-Rhamnose increases serum propionate after long-term supplementation, butlactulose does not raise serum acetate.

Vogt JA(1), Ishii-Schrade KB, Pencharz PB, Wolever TM.

179. Fernandes J, Rao AV, Wolever TMS. Different substrates and methane producing status affect short-chain fatty acid pro- files produced by in vitro fermentation of human feces. J Nutr. 2000;130:1932–1936

180. J Nutr. 2006 Aug;136(8):2160-6.

L-rhamnose and lactulose decrease serum triacylglycerols and their rates ofsynthesis, but do not affect serum cholesterol concentrations in men.

Vogt JA(1), Ishii-Schrade KB, Pencharz PB, Jones PJ, Wolever TM.

181. JPEN J Parenter Enteral Nutr. 2015 May 13. pii: 0148607115587330. [Epub ahead of print]

Glutamine Restores Tight Junction Protein Claudin-1 Expression in Colonic Mucosa of Patients With Diarrhea-Predominant Irritable Bowel Syndrome.

Bertrand J(1), Ghouzali I(1), Guérin C(1), Bôle-Feysot C(1), Gouteux M(1),Déchelotte P(2), Ducrotté P(3), Coëffier M(4).

182. Amino Acids. 2015 Oct;47(10):2143-54. doi: 10.1007/s00726-014-1773-4. Epub 2014Jun 26.

Glutamine and intestinal barrier function.

Wang B(1), Wu G(1,)(2), Zhou Z(3), Dai Z(1), Sun Y(1), Ji Y(1), Li W(1), WangW(1), Liu C(1), Han F(1), Wu Z(4).

183. Amino Acids. 2013 Sep;45(3):501-12. doi: 10.1007/s00726-012-1264-4. Epub 2012 Mar24.

L-Glutamine regulates amino acid utilization by intestinal bacteria.

Dai ZL(1), Li XL, Xi PB, Zhang J, Wu G, Zhu WY.

184. Aliment Pharmacol Ther. 2016 Jan;43(2):181-96. doi: 10.1111/apt.13456. Epub 2015 Nov 2.

Review article: insights into colonic protein fermentation, its modulation andpotential health implications.

Yao CK(1), Muir JG(1), Gibson PR(1).

185. JPEN J Parenter Enteral Nutr. 2010 Jul-Aug;34(4):408-13. doi:10.1177/0148607110362530.

Glutamine supplementation decreases intestinal permeability and preserves gutmucosa integrity in an experimental mouse model.

dos Santos Rd(1), Viana ML, Generoso SV, Arantes RE, Davisson Correia MI, CardosoVN.

186. Amino Acids. 2012 Aug;43(2):813-21. doi: 10.1007/s00726-011-1137-2. Epub 2011 Nov9.

Preventive oral supplementation with glutamine and arginine has beneficialeffects on the intestinal mucosa and inflammatory cytokines in endotoxemic rats.

Zhou X(1), Wu X, Yin Y, Zhang C, He L.

187. J Surg Res. 2007 Dec;143(2):379-84. Epub 2007 Jun 14.

Oral glutamine prevents gut mucosal injury and improves mucosal recoveryfollowing lipopolysaccharide endotoxemia in a rat.

Sukhotnik I(1), Agam M, Shamir R, Shehadeh N, Lurie M, Coran AG, Shiloni E,Mogilner J.

188. Clin Exp Immunol. 2008 Feb;151(2):341-7. Epub 2007 Dec 6.

Treatment with glutamine is associated with down-regulation of Toll-likereceptor-4 and myeloid differentiation factor 88 expression and decrease inintestinal mucosal injury caused by lipopolysaccharide endotoxaemia in a rat.

Kessel A(1), Toubi E, Pavlotzky E, Mogilner J, Coran AG, Lurie M, Karry R,Sukhotnik I.

189. Clin Nutr. 2004 Oct;23(5):1209-16.

Inflammation rather than nutritional depletion determines glutamineconcentrations and intestinal permeability.

Hulsewé KW(1), van der Hulst RW, van Acker BA, von Meyenfeldt MF, Soeters PB.

190. Clinics (Sao Paulo). 2010 Jun;65(6):635-43. doi: 10.1590/S1807-59322010000600012.

Possible links between intestinal permeability and food processing: A potentialtherapeutic niche for glutamine.

Rapin JR(1), Wiernsperger N.

191. J Nutr. 2016 Mar;146(3):501-8. doi: 10.3945/jn.115.224857. Epub 2016 Feb 10.

L-Glutamine Enhances Tight Junction Integrity by Activating CaMK Kinase2-AMP-Activated Protein Kinase Signaling in Intestinal Porcine Epithelial Cells.

Wang B(1), Wu Z(2), Ji Y(1), Sun K(1), Dai Z(1), Wu G(3).

192. World J Gastroenterol. 2011 Mar 28;17(12):1569-73. doi: 10.3748/wjg.v17.i12.1569.

Glutamate reduces experimental intestinal hyperpermeability and facilitatesglutamine support of gut integrity.

Vermeulen MA(1), de Jong J, Vaessen MJ, van Leeuwen PA, Houdijk AP.

193. Obes Facts. 2015;8(2):87-100. doi: 10.1159/000380889. Epub 2015 Mar 10.

Monosodium L-Glutamate and Dietary Fat Differently Modify the Composition of the Intestinal Microbiota in Growing Pigs.

Feng ZM(1), Li TJ, Wu L, Xiao DF, Blachier F, Yin YL.

194. J Nutr. 2015 Oct;145(10):2258-64. doi: 10.3945/jn.115.217661. Epub 2015 Sep 2.

L-Glutamate Enhances Barrier and Antioxidative Functions in Intestinal PorcineEpithelial Cells.

Jiao N(1), Wu Z(2), Ji Y(1), Wang B(1), Dai Z(1), Wu G(3).

195. FASEB J. 2000 Jun;14(9):1132-8.

Regulation of adiposity by dietary calcium.

Zemel MB(1), Shi H, Greer B, Dirienzo D, Zemel PC.

196. Nutr Metab Cardiovasc Dis. 2011 Jul;21(7):499-503. doi:10.1016/j.numecd.2009.11.013. Epub 2010 Mar 12.

Comparison of the effects of cows' milk, fortified soy milk, and calciumsupplement on weight and fat loss in premenopausal overweight and obese women.

Faghih Sh(1), Abadi AR, Hedayati M, Kimiagar SM.

197. J Nutr. 2004 Nov;134(11):3054-60.

Calcium and dairy products inhibit weight and fat regain during ad libitumconsumption following energy restriction in Ap2-agouti transgenic mice.

Sun X(1), Zemel MB.

198. J Am Coll Nutr. 2002 Apr;21(2):146S-151S.

Regulation of adiposity and obesity risk by dietary calcium: mechanisms andimplications.

Zemel MB(1).

199. Br J Nutr. 2014 Jan 28;111(2):287-93. doi: 10.1017/S0007114513002341. Epub 2013Jul 22.

Effect of supplementary calcium phosphate on plasma gastrointestinal hormones in a double-blind, placebo-controlled, cross-over human study.

Trautvetter U(1), Jahreis G(1).

200. J Endocrinol. 2012 Jun;213(3):277-84. doi: 10.1530/JOE-12-0092. Epub 2012 Mar 28.

Food intake in lean and obese mice after peripheral administration ofglucagon-like peptide 2.

Baldassano S(1), Bellanca AL, Serio R, Mulè F.

201. J Nutr. 2009 Aug;139(8):1525-33. doi: 10.3945/jn.109.105205. Epub 2009 Jun 17.

Supplemental calcium attenuates the colitis-related increase in diarrhea,intestinal permeability, and extracellular matrix breakdown in HLA-B27 transgenicrats.

Schepens MA(1), Schonewille AJ, Vink C, van Schothorst EM, Kramer E, Hendriks T, Brummer RJ, Keijer J, van der Meer R, Bovee-Oudenhoven IM.

202. J Nutr. 2007 Nov;137(11 Suppl):2568S-2571S.

Studies with inulin-type fructans on intestinal infections, permeability, andinflammation.

Guarner F(1).

203. Br J Nutr. 2010 Dec;104(12):1780-6. doi: 10.1017/S0007114510002990. Epub 2010 Aug9.

Dietary calcium decreases but short-chain fructo-oligosaccharides increasecolonic permeability in rats.

Schepens MA(1), Rijnierse A, Schonewille AJ, Vink C, Brummer RJ, Willemsen LE,van der Meer R, Bovee-Oudenhoven IM.

204. Br J Nutr. 2012 Apr;107(7):950-6. doi: 10.1017/S0007114511003977. Epub 2011 Aug19.

The protective effect of supplemental calcium on colonic permeability depends on a calcium phosphate-induced increase in luminal buffering capacity.

Schepens MA(1), ten Bruggencate SJ, Schonewille AJ, Brummer RJ, van der Meer R,Bovee-Oudenhoven IM.

205. J Nutr. 2016 Mar;146(3):501-8. doi: 10.3945/jn.115.224857. Epub 2016 Feb 10.

L-Glutamine Enhances Tight Junction Integrity by Activating CaMK Kinase2-AMP-Activated Protein Kinase Signaling in Intestinal Porcine Epithelial Cells.

Wang B(1), Wu Z(2), Ji Y(1), Sun K(1), Dai Z(1), Wu G(3).