neobium.orgneobium.org/wp-content/uploads/suprabiotic full write-up.docx · web viewneobium.org

108
SupraBiotic™ The Slimming Probiotic™ Beyond the cutting edge of probiotic research and development, SupraBiotic™ combines 6 probiotic strains, 100 billion colony forming units, with an unrivalled auxillary formula, optimized to turn the bacterial milieu of your gut into a mean, lean body machine. It does not stop at fat loss, though, fighting and fixing inflammatory pathways that wreak havoc on insulin sensitivity, cardiovascular and digestive health, the immune system, and even your skin. 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.

Upload: hathuy

Post on 08-Apr-2019

220 views

Category:

Documents


0 download

TRANSCRIPT

Page 1: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

SupraBiotic™The Slimming Probiotic™Beyond the cutting edge of probiotic research and development, SupraBiotic™ combines 6 probiotic strains, 100 billion colony forming units, with an unrivalled auxillary formula, optimized to turn the bacterial milieu of your gut into a mean, lean body machine. It does not stop at fat loss, though, fighting and fixing inflammatory pathways that wreak havoc on insulin sensitivity, cardiovascular and digestive health, the immune system, and even your skin.

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).

Page 2: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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).

Page 3: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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 a bit, as well as in the Primer™ write-up.

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.

Page 4: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

SupraBiotic™ IngredientsBifidobacterium 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 fish oils EPA and DHA, as well as conjugated linoleic acid (CLA), in fat tissue and the brain (40). They have also been found to be reduced with aging (41).

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

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.

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

Page 5: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

microbial diversity (47, 48).

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

However, you do not want to just throw every single commercially available species and strain into a product as so many companies do. They need to be rationally combined. If not, they can interfere with each other’s actions and compete for space and resources (54-56).

But, the most interesting benefit of probiotic combinations is through cross-feeding. This is when one bacterial strain produces metabolites the others use for fuel (57).

We will get into this in detail, in a bit. Right now, let’s get to the SupraBiotic™ probiotic combination.

Bifidobacterium breveB. breve supplementation significantly suppressed the accumulation of body weight and fat mass, while improving serum levels of total cholesterol, fasting glucose, and insulin (58). The expression of genes related to fat metabolism and insulin sensitivity in both the gut and fat tissue was upregulated by its administration (59). It also improved lipid levels and insulin resistance while lowering bodyweight (60).

In addition, B. breve combats the cycle of LPS inflammation, leaky gut, and insulin/leptin resistance. It reduced LPS activity 60% and quelled general colonic inflammation, particularly TNF-alpha, a downstream signal of LPS (61-63). It also upregulated anti-inflammatory pathways such as interleukin-8 and Toll-like Receptor-2 (64, 65). The latter having the opposite effect as TLR-4. Ultimately, it is reinforcing on intestinal epithelial cells and mucosa, improving the physical barrier of the intestine (66, 67).

Bifidobacterium animalis subsp. lactisB. animalis subsp. lactis ferments a wide range of oligosaccharides quite extensively, so it is very versatile, being viable under numerous different conditions (68). It increased short chain fatty acid (SCFA) production to distal parts of the colon, meaning it has a long acting mechanism of action (69). We will discuss SCFAs a good bit more below, but they provide fuel for intestinal barrier repair, as well as having other metabolic benefits – and, their production is one of the main ways probiotics exert their positive effects.

This species is negatively associated with body mass index in humans, and increased levels are associated with resistance to obesity (70-71). It prevents weight gain, reduces fat mass accumulation and LPS levels, while preserving glucose tolerance in the face of a high-fat diet (72, 73). Administration shifts the microbiota toward that of a lean phenotype while reducing inflammatory activity (73). Interestingly, it also improved the efficacy of diabetic drug and AMPK agonist metformin, suggesting

Page 6: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

potentiation with that pathway (74). Other direct metabolic improvements were enhanced energy and lipid metabolism, as well as an increase in markers of satiety (75).

Switching to complementary mechanisms, B. animalis subsp. lactis limits increases of pro-inflammatory signals, supporting mucosal recovery to stress (76). It increases tight junction proteins, restoring normal intestinal permeability and preserving gut barrier function in the face of inflammation (77). Finally, it prevents translocation of pathogenic bacteria from intestine to body tissue, reversing inflammation induced insulin resistance (77).

Lactobacillis PlantarumL. plantarum has a great deal of good data. Levels are higher in lean subjects than in the overfat (78). It lowered plasma glucose, insulin, triglycerides, and oxidative stress levels (78b). Further, it reduced lipogenesis and increased fatty acid oxidation via up-regulation of PPARalpha (79). It inhibits the formation of fat cells while decreasing adipose size as well as white adipose tissue mass (80, 81). L. plantarum also reduced weight gain and fat accumulation, upregulated fatty acid oxidation, while improving insulin and leptin sensitivity against an obesity promoting diet (82, 83).

In addition to reducing weight gain and fat mass, it also lowered blood triglyceride levels, while improving leptin sensitivity and intestinal permeability (83). Remarkably, it led to a significant increase in leptin levels, concurrent with weight loss (84). Weight loss typically results in augmented leptin sensitivity, but decreased leptin levels, which is one of the primary causes of hitting the wall on fat loss with prolonged dieting. L. plantarum also improved glucose levels and insulin sensitivity – and, continuing with its remarkable effects, it increased weight with same body fat (85). This means it seemingly helped direct calories toward muscle formation instead of fat.

It was more potent in combination with other probiotics as well as with polyphenols in reducing fat accumulation and improving metabolic alterations (86, 87). Another nice perk, in a comprehensive formula like SupraBiotic™, is that it increased levels of the genus Bacteroides while reducing the Firmicutes:Bacteroides ratio that is associated with obesity (88, 89).

L. plantarum also displays potent anti-inflammatory actions, attenuating signaling of LPS and TLR-4, as well as COX-2, TNF-alpha, and inflammatory interleukins (90, 91). The reduction in inflammatory responses downstream of the LPS signaling pathway was consistently found in several studies (92, 93). Improvements of inflammatory colitis were also seen with L. plantarum (94, 95). Finally, it increased tight junction protein formation and improved intestinal barrier function (96-98).

Lactobacillis gasseriL. gasseri is consistently associated with weight loss in both animals and humans in the literature (99). It mitigates bodyweight and fat mass increases in obesity promoting diets (100). It decreases body fat in both in visceral and subcutaneous adipose, while also increasing insulin and leptin sensitizing peptide

Page 7: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

adiponectin (100-101). This loss of visceral adipose tissue was associated with attenuation in inflammatory gene expression (102).

This species elevated total energy expenditure, while diminishing body weight gain and improving glucose tolerance (103). It reduced bodyweight, triglycerides, and lipogenic genes, while augmenting insulin and leptin sensitivity (104, 105).

Finally, L. gasseri increases tight-junction protein expression and improves intestinal barrier function (106). It elevated levels of the short chain fatty acid, butyrate, relieving inflammatory signaling (103). And, in kind, it inhibits intestinal permeability, LPS production, and adipose tissue inflammation (107, 108).

Lactobacillis RhamnosumL. rhamnosum reduces fat mass, fat synthesis, and improves the obesity associated Firmicutes:Bacteroides ratio (109). It prevented weight gain in diet induced obesity (110). In women, it decreased fat mass, increased weight loss, and improved leptin sensitivity (111). Perhaps most notably, it reduced bodyweight and increased insulin and leptin sensitizing peptide adiponectin while also increasing leptin levels (112). As previously mentioned, leptin normally decreases along with metabolism and appetite control during weight loss, so this is a really nice effect.

Administration of L. rhamnosum resulted in decreased weight gain, with enhanced fatty acid oxidation, insulin sensitivity, and adiponectin via activation of AMPK in both adipose and skeletal muscle tissue (113). It has also been shown to reduce bodyweight and adipose tissue, with increased conjugated linoleic acid (CLA) formation and upregulation of thermogenic protein UCP2 and leptin sensitivity (114, 115).

On the inflammation and gut barrier side of things, L. rhamnosum decreased LPS and LPS induced systemic inflammatory markers including IL-6, COX-2, and TNF-alpha (116-118). Relatedly, it also reduces TLR-4 expression (119). Further, it increases tight junction proteins and restores intestinal barrier function, while inhibiting inflammation downstream of LPS (120-122). A novel action of this species is the production of soluble proteins, p40 and p75, which protect against tight junction and barrier function disruption (123). In addition, it raised Bacteroide levels, tight junction proteins, reduced inflammation, and protected barrier function against a high fructose diet (124).

While this very likely goes for our other probiotic bacteria to some extent, L. rhamnosum has a good bit of data in regard to alcohol consumption. It protects against ethanol induced microbiomal changes, inflammation, and pathology (125). It is also protective against ethanol stimulated inflammation and damage via AMPK (126). Finally, it prevented not just inflammation but also gut barrier disruption in response to ethanol (127). Somewhat related, given alcohol’s use as self-medication, it increased GABA activity and was anxiolytic in response to stress (128).

Page 8: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Bifidobacterium adolescentisB. adolescentis does not have a lot of direct data on body composition. It is higher in lean than obese populations and levels predict leanness, in general (129, 130). Administration resulted in reduction in bodyweight, visceral adipose tissue, and fat mass, while improving insulin resistance (131). It also is synergistic with polyphenols as an anti-inflammatory (132).

However, it is probably the most interesting and important species that is commercially available. That is because it functions as THE archetypal cross-feeder of several of the most important and impressive strains of bacteria. And, those strains are not commercially available.

As a reminder, cross-feeding is production of metabolic substrates by one bacterial species which other species and strains can utilize (133). B. adolescentis produces acetate and oligosacharrides which are then consumed by acetate utilizing, butyrate and propionate producing bacteria (134).

The Best Probiotics 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 (135-138). The Firmicutes:Bacteroides ratio was also significantly worse in obese patients, even in comparison with the merely overweight (137, 138). It has a negative correlation with fat mass and waist circumference (139, 140). It was also 60% lower in obese pigs – yeah, apparently that is a thing (141).

Bacteroides levels in Type-2 diabetes were only half that of the those with normal glucose tolerance (142). Lower Bacteroides was correlated with increased energy intake (143). 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 (144).

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 (145). Bacteroides acidifaciens decreased bodyweight and fat gain, while increasing

Page 9: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

fatty acid oxidation via PPAR-alpha (146). In addition to an elevated Firmicutes:Bacteroides ratio, B. vulgatus levels were found to be lower in the obese (147).

B. fragilis releases a symbiotic immunomodulatory anti-inflammatory factor called Polysacharride A (148). This activates TLR-2, which releases anti-inflammatory interleukins. PSA is basically the opposite of LPS, and TLR-2 the opposite of TLR-4 (149). This has been shown not just to prevent but to cure experimental colitis, an extreme version of a leaky, inflammatory gut (150). 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 (151).

Finally, a few tidbits that will make more sense after reading the Primer™ write-up. A few of the Bacteroides species bind to mucins for colonization and consume these mucin polysaccharides (152, 152b). Bacteroides species also have greater glycan degrading capability than Firmicutes, thus they are preferentially increased by polyphenols (153). Hint: Primer™ contains both mucin and polyphenols.

Faecalibacterium prausnitzii

Faecalibacterium prausnitzii is butyrate producing and is considered a physiological sensor and marker of human health (154). It does not get much more important than that. It is lower in the obese and type-2 diabetics (155-157). Conversely, it is higher in normal glucose tolerance vs. prediabetic subjects (158).

Faecalibacterium prausnitzii is also negatively correlated with inflammatory markers and sharply decreased in inflammatory bowel diseases (157, 159). It is greatly reduced in ulcerative colitis and less abundant in Crohn’s disease (160, 161). As would be expected from the above, it improves intestinal barrier function (162).

Akkermansia muciniphilia

Akkermansia muciniphilia is mucin degrading, meaning it feeds on mucins (163). Levels are higher in lean subjects than the general population (164). 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 (165).

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 (166). Along with enhanced glucose tolerance, it reduced adipose tissue inflammation (167). Akkermansia levels are higher in normal glucose tolerance vs. pre-diabetic subjects (168). It decreased inflammatory cytokine production and protected intestinal barrier function in experimental colitis (169). Finally, its levels are reduced in ulcerative colitis (170).

Page 10: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Roseburia Species

Roseburia species are butyrate producing (171). An increase in this species is associated with decreased body weight, fat mass, insulin sensitivity, and triglycerides -- independent of calorie intake (172). Increased Roseburia correlated with reduced body weight, improved profile of lipid and obesity related gene expression, along with a normalized inflammatory status (173). It is also lower in type-2 diabetes (174). Levels are increased by a Mediterranean diet, as is insulin sensitivity (175). Roseburia is enriched in healthy populations vs. those with atherosclerosis (176). And, its levels display an inverse correlation with disease activity in ulcerative colitis (177).

High protein/low carbohydrate diets, which are so effective and popular, reduce Roseburia and SCFA levels (178, 179). 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 (180). 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, the most important contribution of B. adolescentis is to feed other bacteria, specifically the really good ones that we just talked about and 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 (181).

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 (182, 183).

Roseburia is also an acetate user (184). It is, in fact, generally required for growth (185). In addition to acetate production, B. adolescentis increases Roseburia via partial breakdown of oligosaccharides, which it can then utilize (186).

Cross-feeding with Bifidobacterium modulates the prebiotic effect of inulin and arabinoxylan-oligosaccharides on Roseburia and F. prausnitzii by making acetate available (187). Roseburia was able to grow in pure complex carbohydrate cultures, which it cannot metabolize on its own, owing to cross-feeders (188).

Short Chain Fatty Acids (SCFAs)

Page 11: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

Page 12: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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).

Pomegranate ExtractPomegranate Extract is an extremely rich source of polyphenols. Polyphenols are generally prebiotic for good bacteria (Bifidobacterium, Akkermansia, Bacteroides, and Roseburia), and antibacterial for less favorable and pathogenic ones (189-191). Fruit/berry based polyphenols seem to be particularly favorable toward Bacteroides, the Firmicutes:Bacteroides ratio, and Akkermansia compared to other polyphenol sources. Lactobacillus (Firmicute) lack glycan degrading enzymes, thus do not grow on them particularly well compared to the others (192).

Strawberry polyphenols elevate Bifidobacterium and Bacteroides, butyrate and propionate, as well as decreasing Firmicutes (193). Red wine polyphenols raise Bifidobacterium and Bacteroides as well (194, 195). Polyphenols improve the Firmicutes:Bacteroides ratio, while also increasing Roseburia (196, 197).

Akkermansia REALLY loves polyphenols (198). Grape polyphenols gave a 10-fold increase in Akkermansia and decreased the Firmicutes:Bacteroides ratio, while also reducing weight gain, triglyceride storage, insulin resistance, LPS, and inflammation (199). Cranberry polyphenols produced a 30-fold increase in Akkermansia and decreased weight gain, visceral adipose tissue, triglyceride synthesis, insulin resistance, LPS, and inflammation (200).

Finally, Pomegranate Extract, itself, produced a massive 33 to 47 fold increase in Akkermansia (201). Caffeic acid, a component of Pomegranate Extract, increased Akkermansia 15-fold vs control and several hundred fold vs. subjects with induced colitis! It also improved the Firmicutes:Bacteroides ratio (202).

Polyphenols activate AMPK, enhancing intestinal barrier function (203). They increase tight junction proteins, decrease tight junction pore formation, and ameliorate inflammatory bowel disease (204). Pomegranate Extract activates AMPK at 2 times the potency of metformin (205). It also displayed extremely potent alpha-glucosidase inhibitory activity (this is an ezyme that metabolizes carbohydrates to glucose), being 10 times as potent as acarbose, lowering blood glucose after sucrose intake, but not after glucose (206, 207). It consistently decreases glucose levels, as well as being anti-inflammatory (208, 209).

Fermented Herbs

Page 13: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Fermentation of herbs results in much higher concentrations of active compounds compared to unfermented (210). This same fermentation is done in the body, but it is highly dependent upon the microbial make-up of the individual’s gut, so it can vary widely from person to person (211, 212). As just one example, a fermented herb preparation inhibited LPS mediated inflammatory damage, while the unfermented was ineffective (213, 214)

Fermented KudzuKudzu is a group of polyphenol rich plants belonging to the pea family. Its administration reduced body weight, fat mass, and lipogenesis while stimulating lipolysis and thermogenesis (215). It also lowers body mass index and visceral fat (216). Kudzu increased fatty acid oxidation, and decreased weight gain, triglyceride levels, and visceral adipose tissue on a high-fat diet (217). In addition, it improves insulin sensitivity and lipid metabolism (218).

Kudzu is anti-inflammatory, with components inhibiting LPS, TNF-alpha, and ROS induced inflammation (219, 220). It is also a potent inhibitor of COX-2 (221). In addition, Kudzu reduced intestinal permeability and improved intestinal barrier function (222, 223). Finally, it reduces expression of the dreaded TLR-4 (224, 225).

Fermented GinsengThe anti-obesity effect of unfermented ginseng was shown to be dependent on bacterial make-up of the microbiota (226). It increased mucins (the Akkermansia and Bacteroides food) by 50% (227). It is metabolized by Bifidobacterium as well as Bacteroides. There is a dramatic difference in levels of those bacteria between metabolizers and non-metabolizers, suggesting strong prebiotic specificity toward them (228). And, it was indeed found to enhance growth of Bacteroides (229).

Absorption of one of its main active ingredients, Compound K, is increases by prebiotics (230). And, there are plenty of those in Primer™. Fermented Ginseng decreased bodyweight, fat mass, and food efficiency, while improving insulin and leptin sensitivity (231, 232). In addition to reduced body weight, decreases in fat mass, adipocyte size, and glucose uptake were also observed. And, all of these effects were superior with fermented vs. regular ginseng (233, 234). Finally, it decreases inflammatory cytokines and protects the intestinal barrier (235, 235b).

Mulberry Extract SupraBiotic™ contains an industry-best Mulberry Extract, standardized to over 5% 1-Deoxynojirimycin (1-DNJ) and containing relevant amounts of d-Fagomine, as well. Like all berries, it also has high polyphenol content, the benefits of which we have already talked about.

1-DNJ1-DNJ is a naturally occurring carbohydrate mimic. Its use significantly lowered body weight, blood glucose, and serum insulin levels, and it conversely improved glucose tolerance and insulin sensitivity (236). It increased the insulin and leptin sensitizing peptide adiponectin (which activates AMPK),

Page 14: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

reduced visceral adipose tissue, adipose mass, triglycerides, lipid accumulation, and increased fatty acid oxidation (237).

It is remarkably potent, elevating adiponectin, GLUT4, and AMPK at just .5uM (238). This is 1000 times as potent as metformin. Along with increasing AMPK, it improved mitochondrial function and lipid metabolism (239). Finally, it is a more potent alpha-glucosidase inhibitor than acarbose, which futher helps with glucose and insulin (240).

D-fagomine D-fagomine is also a naturally occurring sugar mimic. It reduced weight gain, plasma triglycerides, glucose, and enhanced leptin and insulin sensitivity (241). It attentuated fat gain on high-fat diet (242). D-fagomine was also found to inhibit intestinal sucrase, lowering post-prandial glucose levels (with either sucrose or starch), as well as modulating bacterial adhesion, inhibiting pathogenic ones without effecting Bifidobacterium or Lactobacillus (243).

Selenium Selenium increases microbial diversity, and it is synergistic with probiotics for this gut bacteria modulation (244, 245). It works by enhancing the fermentation activity of gut bacteria resulting in better bacteria growth as well as output of SCFAs (246).

Queen’s Bee Acid (10-hydroxy-2-decenoic acid) QBA is a medium chain fatty acid from Royal Jelly. It is a very potent AMPK agonist, being effective at just 20uM. This is 25 times as potent as research standard AICAR and the pharmaceutical metformin. (247)

We have mentioned it over and over, but it bears repeating more about the machinations that result in the vicious cycle of gut dysbiosis. Inflammation in the gut, followed by low-level but constant systemic inflammation, PRECEDES obesity and insulin/leptin resistance. It is what gets them started. If you are not either quite lean, or have been on a diet long enough that fat loss has basically stopped, insulin and leptin resistance are already at work, particularly in the brain/adipostat (248). This is bad news for appetite control, metabolism, and body weight regulation.

LPS levels of just 2 to 3-fold above normal, which occur during a Western-style/high-fat diet, initiate the low level inflammatory response that leads to reduced insulin and leptin senstivity, and ultimately type-2 diabetes and Metabolic Syndrome. And, again, this happens before the weight gain starts (249). It sets you up for it to begin in earnest.

AMPK is one of the primary brakes on this ride. It inhibits the LPS induced inflammatory response, as well as the leaky gut. Super potent AMPK agonist, like 1-DNJ and QBA, are basically airbrakes on a runaway train.

Page 15: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

QBA inhibits LPS induced cytokine production (250). It increases GLUT4, glucose uptake, and insulin signaling (247). It enhances intestinal barrier function and tight junction assembly in an AMPK dependent manner (251-253). AMPK activation also improves LPS induced blood-brain-barrier disruption much as it does with the intestinal barrier (254). This improves central insulin and leptin signaling, keeping your adipostat functioning properly.

Palmitoylethanolamide (PEA)PEA is a naturally occurring cannabinoid-like lipid, from a class called acylethanolamides. It is a competitive FAAH inhibitor (this is the enzyme that breaks down endocannabinoids). Decreased levels of endocannabinoids lead to upregulation of cannabinoid receptors, which leads to the increased activity at CB1 via LPS that gets the increased fat storage and over-eating part of the vicious cycle really going into overdrive.

Endocannabinoid activation of CB2, along with PEA/acylethanolamide activation of PPAR-alpha, normally keeps this inflammatory response balanced and in check, but when it gets out of whack, LPS CB1 reigns supreme.

Let’s take a closer look at all of this and what PEA does to fix it.

Intake of dietary fat stimulates production of PEA, and the other acylethanolamides (255, 256). They increase satiety, and reduce food intake and body weight (257, 258). It creates satiety via activation of PPAR-alpha in the intestine followed by direct vagal signal to brain -- i.e. immediate, no gene transcription needed (259, 260). This is one of the steps in which fast transit times in the proximal small intestine become problematic. You get less PEA, thus less satiety signaling to the brain.

In addition, PEA specifically decreases this intestinal transit rate, so it is double plus good on appetite and satiety signaling (261). It both directly makes you feel full and prolongs its own duration of activity in doing so. PEA does, in fact, ultimately reach the brain, where it is active in nM concentrations through gene transcription (258). But, you would have already finished eating too much at that sitting, and each of your other meals, before it did anything, centrally.

In addition to the appetite side of things, PEA decreases intestinal inflammation and permeability. It does so through CB2 and PPAR-alpha as this is blocked by antagonists of either one (262, 263). It is also anti-inflammatory in the intestine via selective targeting of TLR-4 (263).

All sounds great, right? Well, the problem is that with a Western-style or chronic high-fat diet, the PEA release trigger becomes desensitized, ultimately resulting in reduced levels (256, 259, 265). This disrupts normal functioning of the whole system, leading to LPS induction of CB1 becoming dominant. And, this is on top of losing its direct beneficial effects on satiety and food intake.

An increase in inflammation happens in parallel with decreases in PEA, endocannabinoids, and FAAH – and, an upregulation of the cannabinoid receptors (266). It all happens together, at the same time,

Page 16: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

because they are all connected. And, FAAH inhibitors restore levels of all of these and suppress this inflammation (266). FAAH inhibitors also decrease intestinal motility, as we would expect (267). They do so by restoring PEA and the endocannabinoids, thus normalizing the system.

The final bit of evidence on how this operates is that chronic administration of tetrahydrocannabinol (THC), the main active ingredient in marijuana, reduces weight gain, fat mass gain, and energy intake in obese but not lean mice, despite an initial increase in food consumption (268). This is because it is both a CB1 and CB2 agonist. So, you have anti-inflammatory CB2 activation by THC snuffing out the inflammatory LPS pathway, as well as it down-regulating and competing for CB1, such that LPS cannot act on it to wreak its havoc, unchallenged.

This THC administration even improved select gut microbiota profiles. It increased Akkermansia by 4-fold, and improved the Firmicutes:Bacteroides ratio by 6-fold (268). It did not have these same effects in lean subjects because these pathways were not messed up to begin with for them.

So, you can see that PEA helps correct the system at basically all levels that we have been talking about.

One more thing of note is that, as we mentioned with high protein diets negatively altering the bacterial make-up of the microbiota if one does not make a point to get plenty of fiber/prebiotics, the high-fat side of such diets will negatively affect PEA levels, long-term, thus the entire system will be functioning sub-optimally.

Again, I am not bashing these diets at all, they are quite effective. Consuming an excess of glucose and/or fructose is the worst thing you can do to your body as far as inflammation and insulin and everything we have been talking about, just to be clear. But, we want to be optimal, and we can be, quite easily with supplementation.

Ginger Extract (20% Gingerols)Ginger is well known as a digestive aid. The extract is an all-around nice ingredient, aiding through several pathways, and it is particularly effective as an anti-inflammatory and protector of barrier function. The primary component of the extract is 6-gingerol, though 8-, and 10-gingerol, as well as 6-, 8, and 10-shogaol are present in significant, pharmacologically relevant numbers. They all pretty much do the same thing, just at different potencies.

Mechanistically, it is primarily an anti-inflammatory and antioxidant. It decreases basically all inflammatory cytokines (269, 275, 276). It inhibits LPS induced inflammation as well as TNF-alpha (270). It reduced interleukins 3-fold at just 50uM, and its COX inhibition is comparable to aspirin (271-273). It also displays extremely potent anti-oxidant activity, being effective against various radicals at just 1-25uM (274).

Gingerols activate PPAR-alpha, as well as AMPK, with 5 times the potency of metformin and AICAR in suppression of inflammatory cytokines (275, 276). They increase tight junction proteins and integrity via protection against inflammatory assault (277, 278). They suppress colitis via anti-inflammatory and anti-oxidant activity (279). Gingerols enhanced the survival and proliferation of intestinal epithelial cells via reductions in pro-inflammatory cytokines (TNF-α, IL-6 and IL-1β), while also elevating anti-inflammatory

Page 17: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

cytokines (IL-10 and IL-22) in colitis models (280). They also reduce spasms of smooth muscle in the digestive tract (281).

In addition, gingerols displayed some modest inhibitory activity on α-glucosidase and α-amylase, being about 1/6 as potent as acarbose (282). Finally, it increased uptake of Calcium (100+%) and Glutamic acid (60%), both of which we will talk in detail about in the Primer™ write-up (283).

CONCLUSIONAs you can see, SupraBiotic™ takes the concept of probiotic far beyond where anyone has previously taken it before. It starts with bacterial species carefully and purposefully selected to protect against dysfunction of the gut and microbiota to promote better health, better appetite control, better metabolism, and better fat loss. On top of this, SupraBiotic™ addresses and supports novel probiotic bacterial species that you cannot attain, anywhere. And, it does so in a way that no other product is even close to doing. Finally, its supporting ingredients crush inflammation and repair your leaky gut, leaving your body functioning in the optimal way it is intended to.

SupraBiotic™ is a one of a kind product that fits in perfectly with and enhances any diet and exercise program, any supplement regimine, any lifestyle.

References

1. 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.

Page 18: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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).

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.

Page 19: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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: Mol Syst Biol. 2010 Jul;6:392. doi: 10.1038/msb.2010.46.

The endocannabinoid system links gut microbiota to adipogenesis.

Muccioli GG, Naslain D, Bäckhed F, Reigstad CS, Lambert DM, Delzenne NM, Cani PD.

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 Apr

Page 20: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

11.

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

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

16: Diabetes Obes Metab. 2007 May;9(3):218-32. Review.

Cardiovascular metabolic syndrome - an interplay of, obesity, inflammation, diabetes and coronary heartdisease.

Rana JS, Nieuwdorp M, Jukema JW, Kastelein JJ.

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.

Page 21: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

Page 22: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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, VialettesB, 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.

Page 23: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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, VanHylckama 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.

Page 24: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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:

Page 25: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

Page 26: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

Page 27: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

Page 28: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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

58. J Nutr Sci. 2015 May 4;4:e17. doi: 10.1017/jns.2015.5. eCollection 2015.

Oral administration of Bifidobacterium breve B-3 modifies metabolic functions in adults with obese tendencies in a randomised controlled trial.

Minami J(1), Kondo S(1), Yanagisawa N(2), Odamaki T(1), Xiao JZ(1), Abe F(1),Nakajima S(3), Hamamoto Y(3), Saitoh S(4), Shimoda T(4).

59. Benef Microbes. 2013 Sep;4(3):247-51. doi: 10.3920/BM2012.0019.

Bifidobacterium breve B-3 exerts metabolic syndrome-suppressing effects in theliver of diet-induced obese mice: a DNA microarray analysis.

Kondo S(1), Kamei A, Xiao JZ, Iwatsuki K, Abe K.

60. Zhongguo Dang Dai Er Ke Za Zhi. 2013 Dec;15(12):1123-7.

[Probiotics improve obesity-associated dyslipidemia and insulin resistance inhigh-fat diet-fed rats].

[Article in Chinese]

Yu RQ(1), Yuan JL, Ma LY, Qin QX, Wu XY.

61. Gut. 2004 Jun;53(6):821-8.

Lactic acid bacteria secrete metabolites retaining anti-inflammatory propertiesafter intestinal transport.

Ménard S(1), Candalh C, Bambou JC, Terpend K, Cerf-Bensussan N, Heyman M.

62. Dig Dis Sci. 2015 Nov;60(11):3386-92. doi: 10.1007/s10620-015-3769-7. Epub 2015Jul 2.

Page 29: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Administration of Bifidobacterium breve Decreases the Production of TNF-α inChildren with Celiac Disease.

Klemenak M(1), Dolinšek J(2), Langerholc T(3), Di Gioia D(4), Mičetić-Turk D(5).

63. Microbiology. 2012 Nov;158(Pt 11):2826-34. doi: 10.1099/mic.0.060830-0. Epub 2012Aug 17.

Pre-treatment with Bifidobacterium breve UCC2003 modulates Citrobacterrodentium-induced colonic inflammation and organ specificity.

Collins JW(1), Akin AR, Kosta A, Zhang N, Tangney M, Francis KP, Frankel G.

64. Can J Microbiol. 2013 Jan;59(1):9-17. doi: 10.1139/cjm-2012-0446. Epub 2012 Nov1.

Lactic acid bacteria and bifidobacteria attenuate the proinflammatory response inintestinal epithelial cells induced by Salmonella enterica serovar Typhimurium.

Carey CM(1), Kostrzynska M.

65. FEMS Immunol Med Microbiol. 2011 Oct;63(1):129-39. doi:10.1111/j.1574-695X.2011.00837.x. Epub 2011 Jul 29.

Toll-like receptor-2-activating bifidobacteria strains differentially regulateinflammatory cytokines in the porcine intestinal epithelial cell culture system: finding new anti-inflammatory immunobiotics.

Fujie H(1), Villena J, Tohno M, Morie K, Shimazu T, Aso H, Suda Y, Shimosato T,Iwabuchi N, Xiao JZ, Yaeshima T, Iwatsuki K, Saito T, Numasaki M, Kitazawa H.

66. Biomed Res Int. 2015;2015:479140. doi: 10.1155/2015/479140. Epub 2015 Feb 22.

The effects of Bifidobacterium breve on immune mediators and proteome of HT29cells monolayers.

Sánchez B(1), González-Rodríguez I(1), Arboleya S(1), López P(2), Suárez A(2),Ruas-Madiedo P(1), Margolles A(1), Gueimonde M(1).

Page 30: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

67. Inflamm Bowel Dis. 2012 Aug;18(8):1434-46. doi: 10.1002/ibd.22848. Epub 2011 Dec 11.

Commensal and probiotic bacteria influence intestinal barrier function andsusceptibility to colitis in Nod1-/-; Nod2-/- mice.

Natividad JM(1), Petit V, Huang X, de Palma G, Jury J, Sanz Y, Philpott D, GarciaRodenas CL, McCoy KD, Verdu EF.

68. BMC Genomics. 2013 May 10;14:312. doi: 10.1186/1471-2164-14-312.

Transcriptional analysis of oligosaccharide utilization by Bifidobacterium lactisBl-04.

Andersen JM(1), Barrangou R, Abou Hachem M, Lahtinen SJ, Goh YJ, Svensson B,Klaenhammer TR.

69. Int J Food Microbiol. 2011 Jan 5;144(3):342-51. doi:10.1016/j.ijfoodmicro.2010.10.016.

Safety and intestinal microbiota modulation by the exopolysaccharide-producingstrains Bifidobacterium animalis IPLA R1 and Bifidobacterium longum IPLA E44orally administered to Wistar rats.

Salazar N(1), Binetti A, Gueimonde M, Alonso A, Garrido P, González del Rey C,González C, Ruas-Madiedo P, de los Reyes-Gavilán CG.

70. 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, VialettesB, Raoult D.

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

Page 31: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

72. Benef Microbes. 2014 Dec;5(4):437-45. doi: 10.3920/BM2014.0014.

Potential probiotic Bifidobacterium animalis ssp. lactis 420 prevents weight gainand glucose intolerance in diet-induced obese mice.

Stenman LK(1), Waget A(2), Garret C(2), Klopp P(2), Burcelin R(2), Lahtinen S(1).

73. ISME J. 2015 Jan;9(1):1-15. doi: 10.1038/ismej.2014.99. Epub 2014 Jun 17.

Modulation of gut microbiota during probiotic-mediated attenuation of metabolicsyndrome in high fat diet-fed mice.

Wang J(1), Tang H(2), Zhang C(2), Zhao Y(3), Derrien M(4), Rocher E(4),van-Hylckama Vlieg JE(4), Strissel K(5), Zhao L(1), Obin M(5), Shen J(3).

74. Diabetol Metab Syndr. 2015 Sep 12;7:75. doi: 10.1186/s13098-015-0075-7.eCollection 2015.

Probiotic B420 and prebiotic polydextrose improve efficacy of antidiabetic drugs in mice.

Stenman LK(1), Waget A(2), Garret C(2), Briand F(3), Burcelin R(2), Sulpice T(3),Lahtinen S(1).

75. J Agric Food Chem. 2016 May 4;64(17):3462-72. doi: 10.1021/acs.jafc.5b05934. Epub2016 Apr 19.

Probiotic Strain Bifidobacterium animalis subsp. lactis CECT 8145 Reduces FatContent and Modulates Lipid Metabolism and Antioxidant Response in Caenorhabditiselegans.

Martorell P(1), Llopis S(1), González N(1), Chenoll E(1), López-Carreras N(2),Aleixandre A(2), Chen Y(3), Karoly ED(3), Ramón D(1), Genovés S(1).

Page 32: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

76. FEMS Microbiol Lett. 2014 Aug;357(2):157-63. doi: 10.1111/1574-6968.12515. Epub2014 Jul 14.

Bifidobacterium animalis ssp. lactis BI07 modulates the tumor necrosis factoralpha-dependent imbalances of the enterocyte-associated intestinal microbiotafraction.

Centanni M(1), Turroni S, Rampelli S, Biagi E, Quercia S, Consolandi C,Severgnini M, Brigidi P, Candela M.

77. Front Microbiol. 2016 May 6;7:608. doi: 10.3389/fmicb.2016.00608. eCollection2016.

Bifidobacterium animalis ssp. lactis CNCM-I2494 Restores Gut Barrier Permeabilityin Chronically Low-Grade Inflamed Mice.

Martín R(1), Laval L(2), Chain F(1), Miquel S(1), Natividad J(3), Cherbuy C(1),Sokol H(4), Verdu EF(3), van Hylckama Vlieg J(5), Bermudez-Humaran LG(1),Smokvina T(5), Langella P(1).

78. EMBO Mol Med. 2011 Sep;3(9):559-72. doi: 10.1002/emmm.201100159. Epub 2011 Aug 3.

Intestinal mucosal adherence and translocation of commensal bacteria at the earlyonset of type 2 diabetes: molecular mechanisms and probiotic treatment.

Amar J(1), Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán LG, Smirnova N,Bergé M, Sulpice T, Lahtinen S, Ouwehand A, Langella P, Rautonen N, SansonettiPJ, Burcelin R.

78b. 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.

79. World J Gastroenterol. 2013 Jan 14;19(2):274-83. doi: 10.3748/wjg.v19.i2.274.

Dual probiotic strains suppress high fructose-induced metabolic syndrome.

Page 33: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Park DY(1), Ahn YT, Huh CS, McGregor RA, Choi MS.

80. Korean J Food Sci Anim Resour. 2014;34(5):647-55. doi:10.5851/kosfa.2014.34.5.647. Epub 2014 Oct 31.

Physiological Characteristics and Anti-obesity Effect of Lactobacillus plantarum Q180 Isolated from Feces.

Park SY(1), Cho SA(2), Kim SH(3), Lim SD(2).

81. Exp Biol Med (Maywood). 2010 Jul;235(7):849-56. doi: 10.1258/ebm.2010.009377.

Lactobacillus plantarum strain No. 14 reduces adipocyte size in mice fed high-fatdiet.

Takemura N(1), Okubo T, Sonoyama K.

82. PLoS One. 2013;8(3):e59470. doi: 10.1371/journal.pone.0059470. Epub 2013 Mar 21.

Supplementation of Lactobacillus curvatus HY7601 and Lactobacillus plantarumKY1032 in diet-induced obese mice is associated with gut microbial changes andreduction in obesity.

Park DY(1), Ahn YT, Park SH, Huh CS, Yoo SR, Yu R, Sung MK, McGregor RA, Choi MS.

83. Evid Based Complement Alternat Med. 2015;2015:391767. doi: 10.1155/2015/391767.Epub 2015 Feb 23.

Effect of Lactobacillus plantarum Strain K21 on High-Fat Diet-Fed Obese Mice.

Wu CC(1), Weng WL(2), Lai WL(3), Tsai HP(2), Liu WH(2), Lee MH(4), Tsai YC(1).

84. Food Funct. 2015 Feb;6(2):558-65. doi: 10.1039/c4fo00843j.

Lactobacillus rhamnosus LA68 and Lactobacillus plantarum WCFS1 differentlyinfluence metabolic and immunological parameters in high fat diet-inducedhypercholesterolemia and hepatic steatosis.

Ivanovic N(1), Minic R, Dimitrijevic L, Radojevic Skodric S, Zivkovic I,Djordjevic B.

85. Benef Microbes. 2010 Jun;1(2):189-96. doi: 10.3920/BM2009.0036.

Page 34: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Probiotics lower plasma glucose in the high-fat fed C57BL/6J mouse.

Andersson U(1), Bränning C, Ahrné S, Molin G, Alenfall J, Onning G, Nyman M, HolmC.

86. Obesity (Silver Spring). 2013 Dec;21(12):2571-8. doi: 10.1002/oby.20428. Epub2013 Jun 11.

Probiotics L. plantarum and L. curvatus in combination alter hepatic lipidmetabolism and suppress diet-induced obesity.

Yoo SR(1), Kim YJ, Park DY, Jung UJ, Jeon SM, Ahn YT, Huh CS, McGregor R, ChoiMS.

87. Nutr Metab (Lond). 2012 Nov 26;9(1):105. doi: 10.1186/1743-7075-9-105.

Green tea powder and Lactobacillus plantarum affect gut microbiota, lipidmetabolism and inflammation in high-fat fed C57BL/6J mice.

Axling U(1), Olsson C, Xu J, Fernandez C, Larsson S, Ström K, Ahrné S, Holm C,Molin G, Berger K.

88. Lipids Health Dis. 2013 May 9;12:67. doi: 10.1186/1476-511X-12-67.

Effects of NS Lactobacillus strains on lipid metabolism of rats fed ahigh-cholesterol diet.

Hu X(1), Wang T, Li W, Jin F, Wang L.

89. Korean J Food Sci Anim Resour. 2015;35(2):171-8. doi:10.5851/kosfa.2015.35.2.171. Epub 2015 Apr 30.

Effect of Lactobacillus plantarum FH185 on the Reduction of Adipocyte Size andGut Microbial Changes in Mice with Diet-induced Obesity.

Park SY(1), Cho SA(1), Lee MK(1), Lim SD(1).

90. Mol Immunol. 2015 Mar;64(1):63-75. doi: 10.1016/j.molimm.2014.10.027. Epub 2014Nov 13.

The toll-like receptor family protein RP105/MD1 complex is involved in theimmunoregulatory effect of exopolysaccharides from Lactobacillus plantarum N14.

Page 35: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Murofushi Y(1), Villena J(2), Morie K(3), Kanmani P(4), Tohno M(5), Shimazu T(6),Aso H(7), Suda Y(8), Hashiguchi K(9), Saito T(10), Kitazawa H(11).

91. Eur J Nutr. 2015 Oct;54(7):1161-71. doi: 10.1007/s00394-014-0794-9. Epub 2014 Nov19.

Lactobacillus plantarum CECT 7315/7316 intake modulates the acute and chronicinnate inflammatory response.

Vilahur G(1), López-Bernal S(1), Camino S(1), Mendieta G(1), Padró T(1), Badimon L(2,)(3).

92. J Microbiol Biotechnol. 2013 Apr;23(4):518-26.

Effect of probiotics Lactobacillus and Bifidobacterium on gut-derivedlipopolysaccharides and inflammatory cytokines: an in vitro study using a humancolonic microbiota model.

Rodes L(1), Khan A, Paul A, Coussa-Charley M, Marinescu D, Tomaro-Duchesneau C,Shao W, Kahouli I, Prakash S.

93. Mol Cells. 2013 Feb;35(2):115-24. doi: 10.1007/s10059-013-2190-3. Epub 2013 Feb21.

Lipoteichoic acid isolated from Lactobacillus plantarum suppresses LPS-mediatedatherosclerotic plaque inflammation.

Kim JY(1), Kim H, Jung BJ, Kim NR, Park JE, Chung DK.

94. J Microbiol Biotechnol. 2009 Jun;19(6):616-21.

Glycosaminoglycan degradation-inhibitory lactic acid bacteria ameliorate2,4,6-trinitrobenzenesulfonic acid-induced colitis in mice.

Lee B(1), Lee JH, Lee HS, Bae EA, Huh CS, Ahn YT, Kim DH.

95. Int Immunopharmacol. 2014 Jul;21(1):186-92. doi: 10.1016/j.intimp.2014.04.021.Epub 2014 May 9.

Lactobacillus plantarum CLP-0611 ameliorates colitis in mice by polarizing M1 to M2-like macrophages.

Page 36: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Jang SE(1), Han MJ(2), Kim SY(3), Kim DH(4).

96. BMC Microbiol. 2010 Dec 9;10:316. doi: 10.1186/1471-2180-10-316.

Lactobacillus plantarum MB452 enhances the function of the intestinal barrier by increasing the expression levels of genes involved in tight junction formation.

Anderson RC(1), Cookson AL, McNabb WC, Park Z, McCann MJ, Kelly WJ, Roy NC.

97. FEMS Microbiol Lett. 2010 Aug 1;309(2):184-92. doi:10.1111/j.1574-6968.2010.02038.x. Epub 2010 Jun 16.

Lactobacillus plantarum DSM 2648 is a potential probiotic that enhancesintestinal barrier function.

Anderson RC(1), Cookson AL, McNabb WC, Kelly WJ, Roy NC.

98. Am J Physiol Gastrointest Liver Physiol. 2010 Jun;298(6):G851-9. doi:10.1152/ajpgi.00327.2009. Epub 2010 Mar 11.

Regulation of human epithelial tight junction proteins by Lactobacillus plantarumin vivo and protective effects on the epithelial barrier.

Karczewski J(1), Troost FJ, Konings I, Dekker J, Kleerebezem M, Brummer RJ, WellsJM.

99. Microb Pathog. 2012 Aug;53(2):100-8. doi: 10.1016/j.micpath.2012.05.007. Epub2012 May 24.

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

Million M(1), Angelakis E, Paul M, Armougom F, Leibovici L, Raoult D.

100. J Microbiol. 2010 Oct;48(5):712-4. doi: 10.1007/s12275-010-0363-8. Epub 2010 Nov 3.

Effects of Lactobacillus gasseri BNR17 on body weight and adipose tissue mass in diet-induced overweight rats.

Kang JH(1), Yun SI, Park HO.

101. Eur J Clin Nutr. 2010 Jun;64(6):636-43. doi: 10.1038/ejcn.2010.19. Epub 2010 Mar

Page 37: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

10.

Regulation of abdominal adiposity by probiotics (Lactobacillus gasseri SBT2055)in adults with obese tendencies in a randomized controlled trial.

Kadooka Y(1), Sato M, Imaizumi K, Ogawa A, Ikuyama K, Akai Y, Okano M, Kagoshima M, Tsuchida T.

102. Eur J Nutr. 2014;53(2):599-606. doi: 10.1007/s00394-013-0568-9. Epub 2013 Aug 6.

Anti-obesity effect of Lactobacillus gasseri SBT2055 accompanied by inhibition ofpro-inflammatory gene expression in the visceral adipose tissue in diet-inducedobese mice.

Miyoshi M(1), Ogawa A, Higurashi S, Kadooka Y.

103. Br J Nutr. 2016 Aug;116(3):451-8. doi: 10.1017/S0007114516002245. Epub 2016 Jun8.

Probiotic Lactobacillus gasseri SBT2055 improves glucose tolerance and reduces body weight gain in rats by stimulating energy expenditure.

Shirouchi B(1), Nagao K(2), Umegatani M(1), Shiraishi A(1), Morita Y(1), KaiS(2), Yanagita T(3), Ogawa A(4), Kadooka Y(4), Sato M(1).

104. PLoS One. 2013;8(1):e54617. doi: 10.1371/journal.pone.0054617. Epub 2013 Jan 30.

Anti-obesity effect of Lactobacillus gasseri BNR17 in high-sucrose diet-inducedobese mice.

Kang JH(1), Yun SI, Park MH, Park JH, Jeong SY, Park HO.

105. Biosci Microbiota Food Health. 2013;32(2):51-8. doi: 10.12938/bmfh.32.51. Epub2013 Apr 27.

Effect of Lactic Acid Bacteria on Lipid Metabolism and Fat Synthesis in Mice Fed a High-fat Diet.

Yonejima Y(1), Ushida K(2), Mori Y(2).

106. Appl Environ Microbiol. 2012 Feb;78(4):1140-7. doi: 10.1128/AEM.06983-11. Epub2011 Dec 16.

Page 38: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Differential targeting of the E-Cadherin/β-Catenin complex by gram-positiveprobiotic lactobacilli improves epithelial barrier function.

Hummel S(1), Veltman K, Cichon C, Sonnenborn U, Schmidt MA.

107. J Nutr Sci. 2016 May 30;5:e23. doi: 10.1017/jns.2016.12. eCollection 2016.

Lactobacillus gasseri SBT2055 inhibits adipose tissue inflammation and intestinalpermeability in mice fed a high-fat diet.

Kawano M(1), Miyoshi M(1), Ogawa A(1), Sakai F(1), Kadooka Y(1).

108. Br J Nutr. 2015 Oct 28;114(8):1180-7. doi: 10.1017/S0007114515002627. Epub 2015Aug 24.

Administration of Lactobacillus gasseri SBT2055 suppresses macrophageinfiltration into adipose tissue in diet-induced obese mice.

Ukibe K(1), Miyoshi M(1), Kadooka Y(1).

109. Benef Microbes. 2012 Mar 1;3(1):13-22. doi: 10.3920/BM2011.0046.

Modulation of the murine microbiome with a concomitant anti-obesity effect byLactobacillus rhamnosus GG and Lactobacillus sakei NR28.

Ji YS(1), Kim HN, Park HJ, Lee JE, Yeo SY, Yang JS, Park SY, Yoon HS, Cho GS,Franz CM, Bomba A, Shin HK, Holzapfel WH.

110. Pain Res Manag. 2016;2016:5080438. doi: 10.1155/2016/5080438. Epub 2016 Aug 25.

Potential Nociceptive Regulatory Effect of Probiotic Lactobacillus rhamnosus PB01(DSM 14870) on Mechanical Sensitivity in Diet-Induced Obesity Model.

Dardmeh F(1), Nielsen HI(2), Alipour H(2), Kjærgaard B(3), Brandsborg E(4),Gazerani P(1).

111. Br J Nutr. 2014 Apr 28;111(8):1507-19. doi: 10.1017/S0007114513003875. Epub 2013 Dec 3.

Effect of Lactobacillus rhamnosus CGMCC1.3724 supplementation on weight loss and maintenance in obese men and women.

Sanchez M(1), Darimont C(2), Drapeau V(3), Emady-Azar S(4), Lepage M(5),

Page 39: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Rezzonico E(2), Ngom-Bru C(5), Berger B(2), Philippe L(4), Ammon-Zuffrey C(2),Leone P(2), Chevrier G(6), St-Amand E(6), Marette A(6), Doré J(1), Tremblay A(1).

112. Food Funct. 2015 Feb;6(2):558-65. doi: 10.1039/c4fo00843j.

Lactobacillus rhamnosus LA68 and Lactobacillus plantarum WCFS1 differentlyinfluence metabolic and immunological parameters in high fat diet-inducedhypercholesterolemia and hepatic steatosis.

Ivanovic N(1), Minic R, Dimitrijevic L, Radojevic Skodric S, Zivkovic I,Djordjevic B.

113. Biochem Biophys Res Commun. 2013 Feb 8;431(2):258-63. doi:10.1016/j.bbrc.2012.12.121. Epub 2013 Jan 9.

Lactobacillus rhamnosus GG improves insulin sensitivity and reduces adiposity in high-fat diet-fed mice through enhancement of adiponectin production.

Kim SW(1), Park KY, Kim B, Kim E, Hyun CK.

114. Biochim Biophys Acta. 2006 Jul;1761(7):736-44. Epub 2006 May 20.

Human originated bacteria, Lactobacillus rhamnosus PL60, produce conjugatedlinoleic acid and show anti-obesity effects in diet-induced obese mice.

Lee HY(1), Park JH, Seok SH, Baek MW, Kim DJ, Lee KE, Paek KS, Lee Y, Park JH.

115. J Microbiol Biotechnol. 2009 Dec;19(12):1617-9.

Production of c9,t11- and t10,c12-conjugated linoleic acids in humans byLactobacillus rhamnosus PL60.

Lee K(1), Lee Y.

116. J Pediatr Gastroenterol Nutr. 2006 May;42(5):545-52.

Lactobacillus rhamnosus GG decreases lipopolysaccharide-induced systemicinflammation in a gastrostomy-fed infant rat model.

Zhang L(1), Li N, des Robert C, Fang M, Liboni K, McMahon R, Caicedo RA, Neu J.Decreased lipogenic genes, increase AMPK, PPAR-alpha, and fatty acid oxidation against ethanol

Page 40: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

117. PLoS One. 2014 May 15;9(5):e97861. doi: 10.1371/journal.pone.0097861. eCollection2014.

Lactobacillus rhamnosus GG and Bifidobacterium longum attenuate lung injury andinflammatory response in experimental sepsis.

Khailova L(1), Petrie B(1), Baird CH(1), Dominguez Rieg JA(1), Wischmeyer PE(1).

118. PLoS One. 2014 May 22;9(5):e98401. doi: 10.1371/journal.pone.0098401. eCollection2014.

Effects of Lactobacillus paracasei CNCM I-4034, Bifidobacterium breve CNCM I-4035and Lactobacillus rhamnosus CNCM I-4036 on hepatic steatosis in Zucker rats.

Plaza-Diaz J(1), Gomez-Llorente C(1), Abadia-Molina F(2), Saez-Lara MJ(3),Campaña-Martin L(4), Muñoz-Quezada S(1), Romero F(5), Gil A(1), Fontana L(1).

119. PLoS One. 2015 Feb 18;10(2):e0117451. doi: 10.1371/journal.pone.0117451.eCollection 2015.

Probiotics (Lactobacillus rhamnosus R0011 and acidophilus R0052) reduce theexpression of toll-like receptor 4 in mice with alcoholic liver disease.

Hong M(1), Kim SW(2), Han SH(3), Kim DJ(1), Suk KT(4), Kim YS(1), Kim MJ(5), Kim MY(6), Baik SK(6), Ham YL(7).

120. Gut Microbes. 2015;6(1):1-9. doi: 10.4161/19490976.2014.990784. Epub 2015 Jan 14.

Lactobacillus rhamnosus CNCM I-3690 and the commensal bacterium Faecalibacterium prausnitzii A2-165 exhibit similar protective effects to induced barrierhyper-permeability in mice.

Laval L(1), Martin R, Natividad JN, Chain F, Miquel S, Desclée de Maredsous C,Capronnier S, Sokol H, Verdu EF, van Hylckama Vlieg JE, Bermúdez-Humarán LG,Smokvina T, Langella P.

121. Microbiology. 2010 Nov;156(Pt 11):3288-97. doi: 10.1099/mic.0.040139-0. Epub 2010Jul 23.

Lactobacillus rhamnosus GG attenuates interferon-{gamma} and tumour necrosisfactor-alpha-induced barrier dysfunction and pro-inflammatory signalling.

Donato KA(1), Gareau MG, Wang YJ, Sherman PM.

Page 41: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

122. Neurogastroenterol Motil. 2013 Dec;25(12):984-e777. doi: 10.1111/nmo.12232. Epub 2013 Oct 1.

Lactobacillus rhamnosus protects human colonic muscle from pathogenlipopolysaccharide-induced damage.

Ammoscato F(1), Scirocco A, Altomare A, Matarrese P, Petitta C, Ascione B,Caronna R, Guarino M, Marignani M, Cicala M, Chirletti P, Malorni W, Severi C.

123. Am J Physiol Gastrointest Liver Physiol. 2008 Apr;294(4):G1060-9. doi:10.1152/ajpgi.00202.2007. Epub 2008 Feb 21.

Probiotics ameliorate the hydrogen peroxide-induced epithelial barrier disruptionby a PKC- and MAP kinase-dependent mechanism.

Seth A(1), Yan F, Polk DB, Rao RK.

124. PLoS One. 2014 Jan 27;9(1):e80169. doi: 10.1371/journal.pone.0080169. eCollection2014 (124)

Lactobacillus rhamnosus GG protects against non-alcoholic fatty liver disease in mice.

Ritze Y(1), Bárdos G(1), Claus A(1), Ehrmann V(1), Bergheim I(2), Schwiertz A(3),Bischoff SC(1).

125. PLoS One. 2013;8(1):e53028. doi: 10.1371/journal.pone.0053028. Epub 2013 Jan 9.

Metagenomic analyses of alcohol induced pathogenic alterations in the intestinal microbiome and the effect of Lactobacillus rhamnosus GG treatment.

Bull-Otterson L(1), Feng W, Kirpich I, Wang Y, Qin X, Liu Y, Gobejishvili L,Joshi-Barve S, Ayvaz T, Petrosino J, Kong M, Barker D, McClain C, Barve S.

126. J Nutr Biochem. 2015 Apr;26(4):337-44. doi: 10.1016/j.jnutbio.2014.10.016. Epub2014 Dec 5.

Enhanced AMPK phosphorylation contributes to the beneficial effects ofLactobacillus rhamnosus GG supernatant on chronic-alcohol-induced fatty liverdisease.

Zhang M(1), Wang C(2), Wang C(3), Zhao H(4), Zhao C(5), Chen Y(6), Wang Y(7),

Page 42: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

McClain C(8), Feng W(9).

127. PLoS One. 2013;8(1):e53028. doi: 10.1371/journal.pone.0053028. Epub 2013 Jan 9.

Metagenomic analyses of alcohol induced pathogenic alterations in the intestinal microbiome and the effect of Lactobacillus rhamnosus GG treatment.

Bull-Otterson L(1), Feng W, Kirpich I, Wang Y, Qin X, Liu Y, Gobejishvili L,Joshi-Barve S, Ayvaz T, Petrosino J, Kong M, Barker D, McClain C, Barve S.

128. Mod Trends Pharmacopsychiatri. 2013;28:90-9. doi: 10.1159/000343971. Epub 2013Feb 27.

The brain-gut axis: a target for treating stress-related disorders.

Scott LV(1), Clarke G, Dinan TG.

129. PLoS One. 2016 Jul 13;11(7):e0159236. doi: 10.1371/journal.pone.0159236.eCollection 2016.

The Gut Microbiota from Lean and Obese Subjects Contribute Differently to theFermentation of Arabinogalactan and Inulin.

Aguirre M(1,)(2,)(3), Bussolo de Souza C(3,)(4,)(5), Venema K(1,)(2,)(6).

130. 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).

131. Br J Nutr. 2012 May;107(10):1429-34. doi: 10.1017/S0007114511004491. Epub 2011Sep 14.

Bifidobacterium adolescentis supplementation ameliorates visceral fataccumulation and insulin sensitivity in an experimental model of the metabolicsyndrome.

Chen J(1), Wang R, Li XF, Wang RL.

132. Biofactors. 2013 Jul-Aug;39(4):422-9. doi: 10.1002/biof.1081. Epub 2013 Mar 29.

Page 43: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Flavonols enhanced production of anti-inflammatory substance(s) byBifidobacterium adolescentis: prebiotic actions of galangin, quercetin, andfisetin.

Kawabata K(1), Sugiyama Y, Sakano T, Ohigashi H.

133. 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.

134. 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.

135. 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.

136. 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.

Page 44: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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

137. 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.

138. 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).

139. 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).

140. 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.

141. 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.

Page 45: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

142. 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.

143. 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.

144. 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.

145. 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.

146. 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).

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

Page 46: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

148. 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.

149. 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 thehuman microbiota.

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

150. 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.

151. 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.

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

Page 47: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

The human commensal Bacteroides fragilis binds intestinal mucin.

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

152b. 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, RobertV, Philippe C, Bridonneau C, Cherbuy C, Robbe-Masselot C, Langella P, Thomas M.

153. 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).

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

155. 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).

156. 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.

Page 48: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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

157. 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.

158. 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.

159. J Clin Microbiol. 2014 Feb;52(2):398-406. doi: 10.1128/JCM.01500-13. Epub 2013Nov 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.

160. 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).

161. 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).

Page 49: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

162. 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.

163. 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).

164. 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.

165. 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.

166. 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

Page 50: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Consortium, Dumas ME(5), Rizkalla SW(3), Doré J(4), Cani PD(2), Clément K(1).

167. 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.

168. 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.

169. 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.

170. 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.

171. 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.

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

Page 51: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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.

173. 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).

174. 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).

175. 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).

176. 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.

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

Page 52: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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).

178. 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.

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

Reduced dietary intake of carbohydrates by obese subjects results in decreasedconcentrations of butyrate and butyrate-producing bacteria in feces.

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

180. 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).

181. 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).

182. 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.

Page 53: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

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

183. 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).

184. 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.

185. 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.

186. 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.

187. 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).

Page 54: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

188. 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.

189. Res Microbiol. 2006 Nov;157(9):876-84. Epub 2006 Aug 18.

Effect of tea phenolics and their aromatic fecal bacterial metabolites onintestinal microbiota.

Lee HC(1), Jenner AM, Low CS, Lee YK.

190. Int J Food Microbiol. 2008 Jun 10;124(3):295-8. doi:10.1016/j.ijfoodmicro.2008.03.017. Epub 2008 Apr 1.

The potential influence of fruit polyphenols on colonic microflora and human gut health.

Parkar SG(1), Stevenson DE, Skinner MA.

191. Oxid Med Cell Longev. 2016;2016:9346470. doi: 10.1155/2016/9346470. Epub 2016 Jul10.

Bioactivity of Polyphenols: Preventive and Adjuvant Strategies toward ReducingInflammatory Bowel Diseases-Promises, Perspectives, and Pitfalls.

Kaulmann A(1), Bohn T(2).

192. 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).

193. Eur J Nutr. 2016 Oct 15. [Epub ahead of print]

Bioactive compounds from regular diet and faecal microbial metabolites.

Page 55: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Fernández-Navarro T(1,)(2), Salazar N(2), Gutiérrez-Díaz I(1,)(2), Sánchez B(2), Rúas-Madiedo P(2), de Los Reyes-Gavilán CG(2), Margolles A(2), Gueimonde M(2),González S(3).

194. Am J Clin Nutr. 2012 Jun;95(6):1323-34. doi: 10.3945/ajcn.111.027847. Epub 2012May 2.

Influence of red wine polyphenols and ethanol on the gut microbiota ecology andbiochemical biomarkers.

Queipo-Ortuño MI(1), Boto-Ordóñez M, Murri M, Gomez-Zumaquero JM,Clemente-Postigo M, Estruch R, Cardona Diaz F, Andrés-Lacueva C, Tinahones FJ.

195. Food Funct. 2016 Jan;7(1):104-9. doi: 10.1039/c5fo00853k.

Phenolic compounds from red wine and coffee are associated with specificintestinal microorganisms in allergic subjects.

Cuervo A(1), Hevia A(2), López P(3), Suárez A(3), Diaz C(4), Sánchez B(2),Margolles A(2), González S(1).

196. FEMS Microbiol Ecol. 2015 Mar;91(3). pii: fiv008. doi: 10.1093/femsec/fiv008.Epub 2015 Jan 14.

Polyphenol-rich sorghum brans alter colon microbiota and impact species diversityand species richness after multiple bouts of dextran sodium sulfate-inducedcolitis.

Ritchie LE(1), Sturino JM(2), Carroll RJ(3), Rooney LW(4), Azcarate-Peril MA(5), Turner ND(6).

197. Food Funct. 2016 Apr 20;7(4):1775-87. doi: 10.1039/c5fo00886g.

Red wine polyphenols modulate fecal microbiota and reduce markers of themetabolic syndrome in obese patients.

Moreno-Indias I(1), Sánchez-Alcoholado L(2), Pérez-Martínez P(3), Andrés-Lacueva C(4), Cardona F(1), Tinahones F(1), Queipo-Ortuño MI(1).

198. Curr Obes Rep. 2015 Dec;4(4):389-400. doi: 10.1007/s13679-015-0172-9.

Gut Microbiota Dysbiosis in Obesity-Linked Metabolic Diseases and Prebiotic

Page 56: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Potential of Polyphenol-Rich Extracts.

Anhê FF(1,)(2,)(3), Varin TV(4), Le Barz M(5,)(6,)(7), Desjardins Y(8), LevyE(9,)(10,)(11), Roy D(12), Marette A(13,)(14,)(15).

199. Diabetes. 2015 Aug;64(8):2847-58. doi: 10.2337/db14-1916. Epub 2015 Apr 6.

Dietary Polyphenols Promote Growth of the Gut Bacterium Akkermansia muciniphilaand Attenuate High-Fat Diet-Induced Metabolic Syndrome.

Roopchand DE(1), Carmody RN(2), Kuhn P(3), Moskal K(4), Rojas-Silva P(3),Turnbaugh PJ(2), Raskin I(5).

200. Gut. 2015 Jun;64(6):872-83. doi: 10.1136/gutjnl-2014-307142. Epub 2014 Jul 30.

A polyphenol-rich cranberry extract protects from diet-induced obesity, insulinresistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice.

Anhê FF(1), Roy D(2), Pilon G(1), Dudonné S(2), Matamoros S(2), Varin TV(2),Garofalo C(3), Moine Q(3), Desjardins Y(2), Levy E(4), Marette A(1).

201. Food Funct. 2015 Aug;6(8):2487-95. doi: 10.1039/c5fo00669d. Epub 2015 Jul 20.

Pomegranate extract induces ellagitannin metabolite formation and changes stoolmicrobiota in healthy volunteers.

Li Z(1), Henning SM, Lee RP, Lu QY, Summanen PH, Thames G, Corbett K, Downes J,Tseng CH, Finegold SM, Heber D.

202. Oncotarget. 2016 May 11. doi: 10.18632/oncotarget.9306. [Epub ahead of print]

Caffeic acid ameliorates colitis in association with increased Akkermansiapopulation in the gut microbiota of mice.

Zhang Z(1), Wu X(1), Cao S(1), Wang L(1), Wang D(1), Yang H(1), Feng Y(1), WangS(2), Li L(1).

203. Nutrients. 2016 May 7;8(5). pii: E272. doi: 10.3390/nu8050272.

Polyphenol-Rich Propolis Extracts Strengthen Intestinal Barrier Function byActivating AMPK and ERK Signaling.

Page 57: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Wang K(1,)(2), Jin X(3), Chen Y(4), Song Z(5), Jiang X(6), Hu F(7), Conlon MA(8),Topping DL(9).

204. Mol Nutr Food Res. 2013 Dec;57(12):2253-7. doi: 10.1002/mnfr.201300146. Epub 2013Aug 21.

Dietary grape seed extract ameliorates symptoms of inflammatory bowel disease in IL10-deficient mice.

Wang H(1), Xue Y, Zhang H, Huang Y, Yang G, Du M, Zhu MJ.

205. Sci Rep. 2015 Sep 15;5:14014. doi: 10.1038/srep14014.

Punicalagin, an active component in pomegranate, ameliorates cardiacmitochondrial impairment in obese rats via AMPK activation.

Cao K(1,)(2), Xu J(1,)(2), Pu W(1,)(2), Dong Z(3), Sun L(4), Zang W(4), Gao F(5),Zhang Y(1,)(2,)(6), Feng Z(1,)(2), Liu J(1,)(2,)(6).

206. J Ethnopharmacol. 2005 Jun 3;99(2):239-44. Epub 2005 Apr 9.

Punica granatum flower extract, a potent alpha-glucosidase inhibitor, improvespostprandial hyperglycemia in Zucker diabetic fatty rats.

Li Y(1), Wen S, Kota BP, Peng G, Li GQ, Yamahara J, Roufogalis BD.

207. Int J Food Sci Nutr. 2015 Feb;66(1):85-92. doi: 10.3109/09637486.2014.953455.Epub 2014 Dec 18.

Pomegranate ellagitannins inhibit α-glucosidase activity in vitro and reducestarch digestibility under simulated gastro-intestinal conditions.

Bellesia A(1), Verzelloni E, Tagliazucchi D.

208. Nutr Res. 2013 May;33(5):341-8. doi: 10.1016/j.nutres.2013.03.003. Epub 2013 Apr 15.

Pomegranate and type 2 diabetes.

Banihani S(1), Swedan S, Alguraan Z.

209. Complement Ther Clin Pract. 2016 Feb;22:44-50. doi: 10.1016/j.ctcp.2015.12.003.

Page 58: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Epub 2015 Dec 12.

Effects of pomegranate extract supplementation on inflammation in overweight and obese individuals: A randomized controlled clinical trial.

Hosseini B(1), Saedisomeolia A(2), Wood LG(3), Yaseri M(4), Tavasoli S(5).

210. Appl Biochem Biotechnol. 2014 Apr;172(7):3583-92. doi: 10.1007/s12010-014-0774-9.Epub 2014 Feb 21.

Flavonoid-rich plants used as sole substrate to induce the solid-statefermentation of laccase.

Qiu W(1), Zhang W, Chen H.

211. J Ethnopharmacol. 2016 Feb 17;179:253-64. doi: 10.1016/j.jep.2015.12.031. Epub2015 Dec 23.

Could the gut microbiota reconcile the oral bioavailability conundrum oftraditional herbs?

Chen F(1), Wen Q(2), Jiang J(2), Li HL(2), Tan YF(2), Li YH(2), Zeng NK(2).

212. Evid Based Complement Alternat Med. 2013;2013:363076. doi: 10.1155/2013/363076.Epub 2013 Mar 14.

Evaluation of In Vitro Anti-Inflammatory Activities and Protective Effect ofFermented Preparations of Rhizoma Atractylodis Macrocephalae on IntestinalBarrier Function against Lipopolysaccharide Insult.

Bose S(1), Kim H.

213. Food Chem. 2012 Sep 15;134(2):758-65. doi: 10.1016/j.foodchem.2012.02.175. Epub2012 Mar 6.

In vitro and in vivo protective effects of fermented preparations of dietaryherbs against lipopolysaccharide insult.

Bose S(1), Song MY, Nam JK, Lee MJ, Kim H.

Page 59: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

214. Food Chem. 2012 Nov 15;135(2):452-9. doi: 10.1016/j.foodchem.2012.05.007. Epub2012 May 11.

Evaluation of the in vitro and in vivo protective effects of unfermented andfermented Rhizoma coptidis formulations against lipopolysaccharide insult.

Bose S(1), Jeon S, Eom T, Song MY, Kim H.

215. Evid Based Complement Alternat Med. 2012;2012:272710. doi: 10.1155/2012/272710.Epub 2012 May 27.

The crude extract from puerariae flower exerts antiobesity and antifatty livereffects in high-fat diet-induced obese mice.

Kamiya T(1), Sameshima-Kamiya M, Nagamine R, Tsubata M, Ikeguchi M, Takagaki K,Shimada T, Aburada M.

216. Biosci Biotechnol Biochem. 2012;76(8):1511-7. Epub 2012 Aug 7.

Consumption of Pueraria flower extract reduces body mass index via a decrease in the visceral fat area in obese humans.

Kamiya T(1), Takano A, Matsuzuka Y, Kusaba N, Ikeguchi M, Takagaki K, Kondo K.

217. Glob J Health Sci. 2012 Aug 12;4(5):147-55. doi: 10.5539/gjhs.v4n5p147.

The isoflavone-rich fraction of the crude extract of the Puerariae flowerincreases oxygen consumption and BAT UCP1 expression in high-fat diet-fed mice.

Kamiya T(1), Nagamine R, Sameshima-Kamiya M, Tsubata M, Ikeguchi M, Takagaki K.

218. Phytomedicine. 2012 Dec 15;20(1):17-23. doi: 10.1016/j.phymed.2012.09.017. Epub2012 Nov 2.

The Chinese Pueraria root extract (Pueraria lobata) ameliorates impaired glucose and lipid metabolism in obese mice.

Prasain JK(1), Peng N, Rajbhandari R, Wyss JM.

219. PLoS One. 2013 Jun 19;8(6):e66417. doi: 10.1371/journal.pone.0066417. Print 2013.

Page 60: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Tectorigenin Attenuates Palmitate-Induced Endothelial Insulin Resistance viaTargeting ROS-Associated Inflammation and IRS-1 Pathway.

Wang Q(1), Cheng XL, Zhang DY, Gao XJ, Zhou L, Qin XY, Xie GY, Liu K, Qin Y, Liu BL, Qin MJ.

220. Arch Pharm Res. 2012 May;35(5):823-37. doi: 10.1007/s12272-012-0508-x. Epub 2012 May 29.

Anti-inflammatory and antioxidant activities of constituents isolated fromPueraria lobata roots.

Jin SE(1), Son YK, Min BS, Jung HA, Choi JS.

221. Nat Prod Commun. 2015 Oct;10(10):1703-4.

Isoorientin, a Selective Inhibitor of Cyclooxygenase-2 (COX-2) from the Tubers ofPueraria tuberosa.

Sumalatha M, Munikishore R, Rammohan A, Gunasekar D, Kumar KA, Reddy KK, Azad R, Reddanna P, Bodo B.

222. J Ethnopharmacol. 2009 Nov 12;126(2):207-14. doi: 10.1016/j.jep.2009.08.044. Epub2009 Sep 6.

Effects of Puerariae radix extract on the increasing intestinal permeability inrat with alcohol-induced liver injury.

Zhang R(1), Hu Y, Yuan J, Wu D.

223. J Ethnopharmacol. 2009 Nov 12;126(2):207-14. doi: 10.1016/j.jep.2009.08.044. Epub2009 Sep 6.

Effects of Puerariae radix extract on the increasing intestinal permeability inrat with alcohol-induced liver injury.

Zhang R(1), Hu Y, Yuan J, Wu D.

224. Mol Med Rep. 2016 Aug;14(2):1365-70. doi: 10.3892/mmr.2016.5357. Epub 2016 Jun 2.

Puerarin attenuates inflammation and oxidation in mice with collagenantibody-induced arthritis via TLR4/NF-κB signaling.

Page 61: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Wang C(1), Wang W(1), Jin X(1), Shen J(1), Hu W(1), Jiang T(1).

225. Evid Based Complement Alternat Med. 2012;2012:234987. doi: 10.1155/2012/234987.Epub 2012 Oct 21.

Effects of Puerariae Radix Extract on Endotoxin Receptors and TNF-α ExpressionInduced by Gut-Derived Endotoxin in Chronic Alcoholic Liver Injury.

Peng JH(1), Cui T, Sun ZL, Huang F, Chen L, Xu L, Feng Q, Hu YY.

226. J Ginseng Res. 2014 Apr;38(2):106-15. doi: 10.1016/j.jgr.2013.12.004. Epub 2014Jan 9.

Influence of Panax ginseng on obesity and gut microbiota in obese middle-agedKorean women.

Song MY(1), Kim BS(2), Kim H(3).

227. Food Funct. 2014 Jul 25;5(7):1506-12. doi: 10.1039/c4fo00087k.

Korean ginseng modulates the ileal microbiota and mucin gene expression in thegrowing rat.

Han KS(1), Balan P, Hong HD, Choi WI, Cho CW, Lee YC, Moughan PJ, Singh H.

228. PLoS One. 2013 Apr 29;8(4):e62409. doi: 10.1371/journal.pone.0062409. Print 2013.

Comparative analysis of the gut microbiota in people with different levels ofginsenoside Rb1 degradation to compound K.

Kim KA(1), Jung IH, Park SH, Ahn YT, Huh CS, Kim DH.

229. Sci Rep. 2016 Mar 2;6:22474. doi: 10.1038/srep22474.

Gut microbiota-involved mechanisms in enhancing systemic exposure of ginsenosidesby coexisting polysaccharides in ginseng decoction.

Zhou SS(1,)(2), Xu J(3), Zhu H(1), Wu J(2), Xu JD(1), Yan R(4), Li XY(1,)(2), LiuHH(1,)(2), Duan SM(2), Wang Z(2), Chen HB(3), Shen H(2), Li SL(1,)(2).

230. J Ginseng Res. 2015 Apr;39(2):183-7. doi: 10.1016/j.jgr.2014.11.002. Epub 2014

Page 62: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Nov 24.

A prebiotic fiber increases the formation and subsequent absorption of compound Kfollowing oral administration of ginseng in rats.

Kim KA(1), Yoo HH(2), Gu W(3), Yu DH(3), Jin MJ(2), Choi HL(4), Yuan K(5),Guerin-Deremaux L(6), Kim DH(3).

231. Phytother Res. 2014 Apr;28(4):617-22. doi: 10.1002/ptr.5042. Epub 2013 Jul 19.

The anti-obesity effects of the dietary combination of fermented red ginseng withlevan in high fat diet mouse model.

Oh Js(1), Lee SR, Hwang KT, Ji GE.

232. J Ginseng Res. 2015 Oct;39(4):331-7. doi: 10.1016/j.jgr.2015.03.005. Epub 2015Mar 23.

Insulin sensitivity improvement of fermented Korean Red Ginseng (Panax ginseng)mediated by insulin resistance hallmarks in old-aged ob/ob mice.

Cheon JM(1), Kim DI(2), Kim KS(1).

233. Nutrients. 2016 Jun 16;8(6). pii: E369. doi: 10.3390/nu8060369.

Fermented Red Ginseng Potentiates Improvement of Metabolic Dysfunction inMetabolic Syndrome Rat Models.

Kho MC(1,)(2), Lee YJ(3,)(4), Park JH(5), Kim HY(6,)(7), Yoon JJ(8,)(9), AhnYM(10,)(11), Tan R(12,)(13), Park MC(14), Cha JD(15), Choi KM(16), KangDG(17,)(18), Lee HS(19,)(20).

234. Nutrients. 2013 Oct 30;5(11):4316-32. doi: 10.3390/nu5114316.

Red-koji fermented red ginseng ameliorates high fat diet-induced metabolicdisorders in mice.

Kim CM(1), Yi SJ, Cho IJ, Ku SK.

235. BMB Rep. 2015 Jul;48(7):419-25.

Oral administration of fermented wild ginseng ameliorates DSS-induced acutecolitis by inhibiting NF-κB signaling and protects intestinal epithelial barrier.

Page 63: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Seong MA(1), Woo JK(1), Kang JH(2), Jang YS(1), Choi S(3), Jang YS(1), Lee TH(1),Jung KH(4), Kang DK(5), Hurh BS(4), Kim DE(4), Kim SY(1), Oh SH(1).

235b. Cancer Prev Res (Phila). 2016 Jul 21. pii: canprevres.0372.2015. [Epub ahead ofprint]

American Ginseng Attenuates Colitis Associated Colon Carcinogenesis in Mice:Impact on Gut Microbiota and Metabolomics.

Wang CZ(1), Yu C(1), Wen XD(1), Chen L(1), Zhang CF(1), Calway T(1), Qiu Y(2),Wang Y(3), Zhang Z(1), Anderson S(1), Wang YT(4), Jia W(5), Yuan CS(6).

236. Molecules. 2015 Dec 4;20(12):21700-14. doi: 10.3390/molecules201219794.

1-Deoxynojirimycin Alleviates Insulin Resistance via Activation of InsulinSignaling PI3K/AKT Pathway in Skeletal Muscle of db/db Mice.

Liu Q(1), Li X(2), Li C(3,)(4), Zheng Y(5,)(6), Peng G(7,)(8).

237. Food Chem. 2013 Aug 15;139(1-4):16-23. doi: 10.1016/j.foodchem.2013.02.025. Epub 2013 Feb 20.

Intake of mulberry 1-deoxynojirimycin prevents diet-induced obesity throughincreases in adiponectin in mice.

Tsuduki T(1), Kikuchi I, Kimura T, Nakagawa K, Miyazawa T.

238. J Microbiol Biotechnol. 2013 May;23(5):637-43.

1-Deoxynojirimycin isolated from a Bacillus subtilis stimulates adiponectin and GLUT4 expressions in 3T3-L1 adipocytes.

Lee SM1, Do HJ, Shin MJ, Seong SI, Hwang KY, Lee JY, Kwon O, Jin T, Chung JH.

239. Food Chem Toxicol. 2015 Jan;75:1-7. doi: 10.1016/j.fct.2014.11.001. Epub 2014 Nov 10.

Page 64: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

1-deoxynojirimycin isolated from Bacillus subtilis improves hepatic lipid metabolism and mitochondrial function in high-fat-fed mice.

Do HJ1, Chung JH2, Hwang JW3, Kim OY4, Lee JY5, Shin MJ6.

240. J Agric Food Chem. 2008 Apr 23;56(8):2613-9. doi: 10.1021/jf073223i. Epub 2008Mar 26.

Antiobesity effects and improvement of insulin sensitivity by 1-deoxynojirimycin in animal models.

Kong WH(1), Oh SH, Ahn YR, Kim KW, Kim JH, Seo SW.

241. Food Funct. 2015 Aug;6(8):2614-9. doi: 10.1039/c5fo00591d. Epub 2015 Jul 1.

D-Fagomine attenuates metabolic alterations induced by a high-energy-dense dietin rats.

Molinar-Toribio E(1), Pérez-Jiménez J, Ramos-Romero S, Gómez L, Taltavull N,Nogués MR, Adeva A, Jáuregui O, Joglar J, Clapés P, Torres JL.

242. Obesity (Silver Spring). 2014 Apr;22(4):976-9. doi: 10.1002/oby.20640. Epub 2013 Dec 4.

Effect of (D)-fagomine on excreted Enterobacteria and weight gain in rats fed ahigh-fat high-sucrose diet.

Ramos-Romero S(1), Molinar-Toribio E, Gómez L, Pérez-Jiménez J, Casado M, Clapés P, Piña B, Torres JL.

243. Br J Nutr. 2012 Jun;107(12):1739-46. doi: 10.1017/S0007114511005009. Epub 2011Oct 3.

D-Fagomine lowers postprandial blood glucose and modulates bacterial adhesion.

Gómez L(1), Molinar-Toribio E, Calvo-Torras MÁ, Adelantado C, Juan ME, Planas JM,Cañas X, Lozano C, Pumarola S, Clapés P, Torres JL.

244 . FASEB J. 2011 Jul;25(7):2492-9. doi: 10.1096/fj.11-181990. Epub 2011 Apr 14.

Dietary selenium affects host selenoproteome expression by influencing the gutmicrobiota.

Page 65: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Kasaikina MV(1), Kravtsova MA, Lee BC, Seravalli J, Peterson DA, Walter J, Legge R, Benson AK, Hatfield DL, Gladyshev VN.

245. Front Immunol. 2013 Dec 12;4:445. doi: 10.3389/fimmu.2013.00445.

The role of probiotics and prebiotics in inducing gut immunity.

Vieira AT(1), Teixeira MM(1), Martins FS(2).

246. Biol Trace Elem Res. 1997 Feb;56(2):215-24.

Effects of selenium on colonic fermentation in the rat.

Kim J(1), Combs GF Jr.

247. Mol Nutr Food Res. 2013 Oct;57(10):1794-802. doi: 10.1002/mnfr.201300041. Epub2013 Jun 10.

10-Hydroxy-2-decenoic acid, a unique medium-chain fatty acid, activates5'-AMP-activated protein kinase in L6 myotubes and mice.

Takikawa M(1), Kumagai A, Hirata H, Soga M, Yamashita Y, Ueda M, Ashida H, Tsuda T.

248. PLoS One. 2010 Aug 16;5(8):e12191. doi: 10.1371/journal.pone.0012191.

High-fat diet: bacteria interactions promote intestinal inflammation whichprecedes and correlates with obesity and insulin resistance in mouse.

Ding S(1), Chi MM, Scull BP, Rigby R, Schwerbrock NM, Magness S, Jobin C, LundPK.

249. Diabetes. 2007 Jul;56(7):1761-72. Epub 2007 Apr 24.

Metabolic endotoxemia initiates obesity and insulin resistance.

Cani PD(1), Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, Neyrinck AM, FavaF, Tuohy KM, Chabo C, Waget A, Delmée E, Cousin B, Sulpice T, Chamontin B,Ferrières J, Tanti JF, Gibson GR, Casteilla L, Delzenne NM, Alessi MC, BurcelinR.

Page 66: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

250. Innate Immun. 2012 Jun;18(3):429-37. doi: 10.1177/1753425911416022. Epub 2011 Sep26.

Inhibitory effect of 10-hydroxy-trans-2-decenoic acid on LPS-induced IL-6production via reducing IκB-ζ expression.

Sugiyama T(1), Takahashi K, Tokoro S, Gotou T, Neri P, Mori H.

251. Nutrients. 2016 May 7;8(5). pii: E272. doi: 10.3390/nu8050272.

Polyphenol-Rich Propolis Extracts Strengthen Intestinal Barrier Function byActivating AMPK and ERK Signaling.

Wang K(1,)(2), Jin X(3), Chen Y(4), Song Z(5), Jiang X(6), Hu F(7), Conlon MA(8),Topping DL(9).

252. Biochem Pharmacol. 2015 Aug 1;96(3):225-36. doi: 10.1016/j.bcp.2015.05.016. Epub 2015 Jun 3.

Activation of AMPK by chitosan oligosaccharide in intestinal epithelial cells:Mechanism of action and potential applications in intestinal disorders.

Muanprasat C(1), Wongkrasant P(2), Satitsri S(2), Moonwiriyakit A(2),Pongkorpsakol P(2), Mattaveewong T(2), Pichyangkura R(3), Chatsudthipong V(2).

253. Proc Natl Acad Sci U S A. 2006 Nov 14;103(46):17272-7. Epub 2006 Nov 6.

AMP-activated protein kinase regulates the assembly of epithelial tightjunctions.

Zhang L(1), Li J, Young LH, Caplan MJ.

Activation of AMPK by5-aminoimidazole-4-carboxamide ribonucleoside facilitates tight junction assembly

254. Brain Inj. 2015;29(6):777-84. doi: 10.3109/02699052.2015.1004746. Epub 2015 Mar20.

Activation of AMPK improves lipopolysaccharide-induced dysfunction of theblood-brain barrier in mice.

Yu HY(1), Cai YB, Liu Z.

Page 67: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

255. J Neuroendocrinol. 2008 May;20 Suppl 1:94-9. doi:10.1111/j.1365-2826.2008.01687.x.

Endocannabinoids and nutrition.

Hansen HS(1), Artmann A.

256. Biochim Biophys Acta. 2008 Apr;1781(4):200-12. doi: 10.1016/j.bbalip.2008.01.006.Epub 2008 Feb 12.

Influence of dietary fatty acids on endocannabinoid and N-acylethanolamine levelsin rat brain, liver and small intestine.

Artmann A(1), Petersen G, Hellgren LI, Boberg J, Skonberg C, Nellemann C, Hansen SH, Hansen HS.

257. Best Pract Res Clin Endocrinol Metab. 2009 Feb;23(1):33-49. doi:10.1016/j.beem.2008.10.003.

Role of acylethanolamides in the gastrointestinal tract with special reference tofood intake and energy balance.

Borrelli F(1), Izzo AA.

258. Cell. 2008 Nov 28;135(5):813-24. doi: 10.1016/j.cell.2008.10.043.

N-acylphosphatidylethanolamine, a gut- derived circulating factor induced by fat ingestion, inhibits food intake.

Gillum MP(1), Zhang D, Zhang XM, Erion DM, Jamison RA, Choi C, Dong J,Shanabrough M, Duenas HR, Frederick DW, Hsiao JJ, Horvath TL, Lo CM, Tso P, ClineGW, Shulman GI.

259. Pharmacol Res. 2014 Aug;86:18-25. doi: 10.1016/j.phrs.2014.03.006. Epub 2014 Mar 28.

Role of anorectic N-acylethanolamines in intestinal physiology and satietycontrol with respect to dietary fat.

Hansen HS(1).

260. Biochem Pharmacol. 2009 Sep 15;78(6):553-60. doi: 10.1016/j.bcp.2009.04.024. Epub2009 May 4.

Page 68: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

N-acylethanolamines, anandamide and food intake.

Hansen HS(1), Diep TA.

261. Br J Pharmacol. 2001 Nov;134(5):945-50.

Inhibitory effect of palmitoylethanolamide on gastrointestinal motility in mice.

Capasso R(1), Izzo AA, Fezza F, Pinto A, Capasso F, Mascolo N, Di Marzo V.

262. Br J Pharmacol. 2015 Jan;172(1):142-58. doi: 10.1111/bph.12907. Epub 2014 Dec 1.

Palmitoylethanolamide, a naturally occurring lipid, is an orally effectiveintestinal anti-inflammatory agent.

Borrelli F(1), Romano B, Petrosino S, Pagano E, Capasso R, Coppola D, Battista G,Orlando P, Di Marzo V, Izzo AA.

263. Gut. 2014 Aug;63(8):1300-12. doi: 10.1136/gutjnl-2013-305005. Epub 2013 Sep 30.

Palmitoylethanolamide improves colon inflammation through an enteric glia/tolllike receptor 4-dependent PPAR-α activation.

Esposito G(1), Capoccia E(1), Turco F(2), Palumbo I(2), Lu J(3), Steardo A(4),Cuomo R(2), Sarnelli G(2), Steardo L(1).

264. 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.

265. Biofactors. 2014 Jul-Aug;40(4):363-72. doi: 10.1002/biof.1158. Epub 2014 Feb 14.

Classical endocannabinoid-like compounds and their regulation by nutrients.

Kleberg K(1), Hassing HA, Hansen HS.

266. Pharmacol Res. 2010 Apr;61(4):321-8. doi: 10.1016/j.phrs.2009.11.005. Epub 2009

Page 69: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Nov 17.

Levels of endocannabinoids and palmitoylethanolamide and their pharmacologicalmanipulation in chronic granulomatous inflammation in rats.

Endocannabinoid Research Group(1), De Filippis D, D'Amico A, Cipriano M,Petrosino S, Orlando P, Di Marzo V, Iuvone T.

267. Gastroenterology. 2005 Sep;129(3):941-51.

Fatty acid amide hydrolase controls mouse intestinal motility in vivo.

Capasso R(1), Matias I, Lutz B, Borrelli F, Capasso F, Marsicano G, Mascolo N,Petrosino S, Monory K, Valenti M, Di Marzo V, Izzo AA.

268. PLoS One. 2015 Dec 3;10(12):e0144270. doi: 10.1371/journal.pone.0144270.eCollection 2015.

Prevention of Diet-Induced Obesity Effects on Body Weight and Gut Microbiota inMice Treated Chronically with Δ9-Tetrahydrocannabinol.

Cluny NL(1), Keenan CM(1), Reimer RA(2), Le Foll B(3), Sharkey KA(1).

269. Phytother Res. 2015 Nov;29(11):1707-13. doi: 10.1002/ptr.5414. Epub 2015 Jul 14.

Differential Inhibition of T Lymphocyte Proliferation and Cytokine Synthesis by[6]-Gingerol, [8]-Gingerol, and [10]-Gingerol.

Bernard M(1), Furlong SJ(1), Power Coombs MR(1), Hoskin DW(1,)(2,)(3).

270. Biochem Biophys Res Commun. 2009 Apr 24;382(1):134-9. doi:10.1016/j.bbrc.2009.02.160. Epub 2009 Mar 4.

6-Gingerol inhibits ROS and iNOS through the suppression of PKC-alpha andNF-kappaB pathways in lipopolysaccharide-stimulated mouse macrophages.

Lee TY(1), Lee KC, Chen SY, Chang HH.

271. Pharm Biol. 2016 Sep;54(9):1606-15. doi: 10.3109/13880209.2015.1110598. Epub 2016Mar 18.

6-Gingerol inhibits Vibrio cholerae-induced proinflammatory cytokines inintestinal epithelial cells via modulation of NF-κB.

Page 70: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Saha P(1), Katarkar A(1), Das B(1), Bhattacharyya A(1), Chaudhuri K(1).

Reduced ILs and COX2

272. J Agric Food Chem. 2014 Oct 29;62(43):10452-7. doi: 10.1021/jf5026086. Epub 2014 Oct 20.

Anti-inflammatory activity of grains of paradise (Aframomum melegueta Schum)extract.

Ilic NM(1), Dey M, Poulev AA, Logendra S, Kuhn PE, Raskin I.

273. Evid Based Complement Alternat Med. 2013;2013:146142. doi: 10.1155/2013/146142.Epub 2013 Jun 16.

Attenuation of Proinflammatory Responses by S-[6]-Gingerol via Inhibition ofROS/NF-Kappa B/COX2 Activation in HuH7 Cells.

Li XH(1), McGrath KC, Tran VH, Li YM, Duke CC, Roufogalis BD, Heather AK.

274. J Ethnopharmacol. 2010 Feb 3;127(2):515-20. doi: 10.1016/j.jep.2009.10.004. Epub 2009 Oct 13.

Comparative antioxidant and anti-inflammatory effects of [6]-gingerol,[8]-gingerol, [10]-gingerol and [6]-shogaol.

Dugasani S(1), Pichika MR, Nadarajah VD, Balijepalli MK, Tandra S, Korlakunta JN.

275. Nutrients. 2015 Feb 4;7(2):999-1020. doi: 10.3390/nu7020999.

6-gingerol protects against nutritional steatohepatitis by regulating key genesrelated to inflammation and lipid metabolism.

Tzeng TF(1), Liou SS(2), Chang CJ(3), Liu IM(4).

276. Food Funct. 2015 Oct;6(10):3334-41. doi: 10.1039/c5fo00513b.

6-Gingerol modulates proinflammatory responses in dextran sodium sulfate(DSS)-treated Caco-2 cells and experimental colitis in mice through adenosinemonophosphate-activated protein kinase (AMPK) activation.

Chang KW(1), Kuo CY.

Page 71: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

277. Evid Based Complement Alternat Med. 2013;2013:761852. doi: 10.1155/2013/761852.Epub 2013 Sep 24.

[6]-Gingerol Prevents Disassembly of Cell Junctions and Activities of MMPs inInvasive Human Pancreas Cancer Cells through ERK/NF- κ B/Snail SignalTransduction Pathway.

Kim SO(1), Kim MR.

278 Mol Nutr Food Res. 2016 Aug 4. doi: 10.1002/mnfr.201600274.

The ginger component6-shogaol prevents TNF-α-induced barrier loss via inhibition of PI3K/Akt andNF-κB signaling.

Luettig J, Rosenthal R, Lee IM, Krug SM, Schulzke JD

279. Phytother Res. 2015 Apr;29(4):566-72. doi: 10.1002/ptr.5286. Epub 2015 Jan 29.

Pharmacological activity of 6-gingerol in dextran sulphate sodium-inducedulcerative colitis in BALB/c mice.

Ajayi BO(1), Adedara IA, Farombi EO.

280. Biomaterials. 2016 Sep;101:321-40. doi: 10.1016/j.biomaterials.2016.06.018. Epub 2016 Jun 9.

Edible ginger-derived nanoparticles: A novel therapeutic approach for theprevention and treatment of inflammatory bowel disease and colitis-associatedcancer.

Zhang M(1), Viennois E(2), Prasad M(3), Zhang Y(2), Wang L(4), Zhang Z(2), HanMK(2), Xiao B(5), Xu C(6), Srinivasan S(3), Merlin D(4).

281. Int J Food Sci Nutr. 2006 Feb-Mar;57(1-2):65-73.

Species differences in the prokinetic effects of ginger.

Ghayur MN(1), Gilani AH.

282. Int J Food Sci Nutr. 2011 Mar;62(2):106-10. doi: 10.3109/09637486.2010.515565.Epub 2010 Sep 28.

Page 72: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Inhibitory potential of ginger extracts against enzymes linked to type 2diabetes, inflammation and induced oxidative stress.

Rani MP(1), Padmakumari KP, Sankarikutty B, Cherian OL, Nisha VM, Raghu KG.

283. J Trace Elem Med Biol. 2013 Jul;27(3):184-90. doi: 10.1016/j.jtemb.2012.11.003.Epub 2013 Jan 16.

Enhanced intestinal uptake of iron, zinc and calcium in rats fed pungent spiceprinciples--piperine, capsaicin and ginger (Zingiber officinale).

Prakash UN(1), Srinivasan K.

Supplementary

S1. PLoS One. 2012;7(4):e35240. doi: 10.1371/journal.pone.0035240. Epub 2012 Apr 10.

Butyrate and propionate protect against diet-induced obesity and regulate guthormones via free fatty acid receptor 3-independent mechanisms.

Lin HV(1), Frassetto A, Kowalik EJ Jr, Nawrocki AR, Lu MM, Kosinski JR, HubertJA, Szeto D, Yao X, Forrest G, Marsh DJ.

S2. Nat Commun. 2013;4:1829. doi: 10.1038/ncomms2852.

The gut microbiota suppresses insulin-mediated fat accumulation via theshort-chain fatty acid receptor GPR43.

Kimura I(1), Ozawa K, Inoue D, Imamura T, Kimura K, Maeda T, Terasawa K,Kashihara D, Hirano K, Tani T, Takahashi T, Miyauchi S, Shioi G, Inoue H,Tsujimoto G.

S3. Cell. 2014 Jan 16;156(1-2):84-96. doi: 10.1016/j.cell.2013.12.016. Epub 2014 Jan 9.

Microbiota-generated metabolites promote metabolic benefits via gut-brain neural circuits.

De Vadder F(1), Kovatcheva-Datchary P(2), Goncalves D(1), Vinera J(1), ZitounC(1), Duchampt A(1), Bäckhed F(3), Mithieux G(4).

Page 73: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

S4. Diabetes Res Clin Pract. 2014 Sep;105(3):295-301. doi:10.1016/j.diabres.2014.04.008. Epub 2014 Jun 11.

Intestinal glucose metabolism revisited.

Mithieux G(1), Gautier-Stein A(2).

S5. Am J Physiol Gastrointest Liver Physiol. 2013 Dec;305(12):G900-10. doi:10.1152/ajpgi.00265.2013. Epub 2013 Oct 17.

Gut-derived short-chain fatty acids are vividly assimilated into hostcarbohydrates and lipids.

den Besten G(1), Lange K, Havinga R, van Dijk TH, Gerding A, van Eunen K, Müller M, Groen AK, Hooiveld GJ, Bakker BM, Reijngoud DJ.

S6. Int J Obes (Lond). 2015 Mar;39(3):424-9. doi: 10.1038/ijo.2014.153. Epub 2014 Aug11.

The short chain fatty acid propionate stimulates GLP-1 and PYY secretion via freefatty acid receptor 2 in rodents.

Psichas A(1), Sleeth ML(1), Murphy KG(2), Brooks L(2), Bewick GA(3), HanyalogluAC(4), Ghatei MA(2), Bloom SR(2), Frost G(1).

S7. Diabetes. 2012 Feb;61(2):364-71. doi: 10.2337/db11-1019. Epub 2011 Dec 21.

Short-chain fatty acids stimulate glucagon-like peptide-1 secretion via theG-protein-coupled receptor FFAR2.

Tolhurst G(1), Heffron H, Lam YS, Parker HE, Habib AM, Diakogiannaki E, CameronJ, Grosse J, Reimann F, Gribble FM.

S8. Br J Nutr. 2013 May 28;109(10):1755-64. doi: 10.1017/S0007114512003923. Epub 2012Oct 31.

Male mice that lack the G-protein-coupled receptor GPR41 have low energyexpenditure and increased body fat content.

Bellahcene M(1), O'Dowd JF, Wargent ET, Zaibi MS, Hislop DC, Ngala RA, Smith DM, Cawthorne MA, Stocker CJ, Arch JR.

Page 74: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

S9. Proc Natl Acad Sci U S A. 2011 May 10;108(19):8030-5. doi:10.1073/pnas.1016088108. Epub 2011 Apr 25.

Short-chain fatty acids and ketones directly regulate sympathetic nervous system via G protein-coupled receptor 41 (GPR41).

Kimura I(1), Inoue D, Maeda T, Hara T, Ichimura A, Miyauchi S, Kobayashi M,Hirasawa A, Tsujimoto G.

S10. Endocrinology. 2016 May;157(5):1881-94. doi: 10.1210/en.2015-1944. Epub 2016 Mar 18.

Short-Chain Fatty Acid Acetate Stimulates Adipogenesis and MitochondrialBiogenesis via GPR43 in Brown Adipocytes.

Hu J(1), Kyrou I(1), Tan BK(1), Dimitriadis GK(1), Ramanjaneya M(1), TripathiG(1), Patel V(1), James S(1), Kawan M(1), Chen J(1), Randeva HS(1).

S11. J Nutr. 2013 Dec;143(12):1872-81. doi: 10.3945/jn.113.179549. Epub 2013 Oct 16.

Short-chain fatty acids activate AMP-activated protein kinase and ameliorateethanol-induced intestinal barrier dysfunction in Caco-2 cell monolayers.

Elamin EE(1), Masclee AA, Dekker J, Pieters HJ, Jonkers DM.

S12. PLoS One. 2013 May 16;8(5):e63388. doi: 10.1371/journal.pone.0063388. Print 2013.

Butyrate-producing probiotics reduce nonalcoholic fatty liver disease progressionin rats: new insight into the probiotics for the gut-liver axis.

Endo H(1), Niioka M, Kobayashi N, Tanaka M, Watanabe T.

S13. J Nutr. 2009 Sep;139(9):1619-25. doi: 10.3945/jn.109.104638. Epub 2009 Jul 22.

Butyrate enhances the intestinal barrier by facilitating tight junction assembly via activation of AMP-activated protein kinase in Caco-2 cell monolayers.

Peng L(1), Li ZR, Green RS, Holzman IR, Lin J.

S14. Int J Mol Sci. 2016 Oct 10;17(10). pii: E1696.

Sodium Butyrate Promotes Reassembly of Tight Junctions in Caco-2 MonolayersInvolving Inhibition of MLCK/MLC2 Pathway and Phosphorylation of PKCβ2.

Page 75: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

Miao W(1,)(2,)(3), Wu X(4,)(5,)(6), Wang K(7,)(8,)(9), Wang W(10,)(11,)(12), WangY(13,)(14,)(15), Li Z(16,)(17,)(18), Liu J(19,)(20,)(21), Li L(22,)(23,)(24),Peng L(25,)(26,)(27).

S15. Nutr Res Pract. 2015 Apr;9(2):117-22. doi: 10.4162/nrp.2015.9.2.117. Epub 2015Mar 6.

Curcumin utilizes the anti-inflammatory response pathway to protect the intestineagainst bacterial invasion.

Cho JA(1), Park E(2).

S16. PLoS One. 2014 Sep 24;9(9):e108577. doi: 10.1371/journal.pone.0108577.eCollection 2014.

Oral supplementation with non-absorbable antibiotics or curcumin attenuateswestern diet-induced atherosclerosis and glucose intolerance in LDLR-/-mice--role of intestinal permeability and macrophage activation.

Ghosh SS(1), Bie J(1), Wang J(1), Ghosh S(1).

S17. 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.

S18. Appl Environ Microbiol. 2014 Oct;80(19):5935-43. doi: 10.1128/AEM.01357-14. Epub 2014 Jul 18.

Effect of metformin on metabolic improvement and gut microbiota.

Lee H(1), Ko G(2).

S19. Acta Pharmacol Sin. 2016 Aug;37(8):1063-75. doi: 10.1038/aps.2016.21. Epub 2016May 16.

Metformin exerts glucose-lowering action in high-fat fed mice via attenuatingendotoxemia and enhancing insulin signaling.

Zhou ZY(1), Ren LW(1), Zhan P(1), Yang HY(1), Chai DD(1), Yu ZW(1).

Page 76: neobium.orgneobium.org/wp-content/uploads/SupraBiotic Full Write-Up.docx · Web viewneobium.org

S20. 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.