Submitted Successfully!
To reward your contribution, here is a gift for you: A free trial for our video production service.
Thank you for your contribution! You can also upload a video entry or images related to this topic.
Version Summary Created by Modification Content Size Created at Operation
1 + 3716 word(s) 3716 2021-08-13 05:26:38 |
2 format correction + 19 word(s) 3735 2021-08-25 03:00:36 |

Video Upload Options

Do you have a full video?

Confirm

Are you sure to Delete?
Cite
If you have any further questions, please contact Encyclopedia Editorial Office.
Fuke, N.; Nagata, N. Dietary Factors and Metabolic Endotoxemia. Encyclopedia. Available online: https://encyclopedia.pub/entry/13512 (accessed on 19 April 2024).
Fuke N, Nagata N. Dietary Factors and Metabolic Endotoxemia. Encyclopedia. Available at: https://encyclopedia.pub/entry/13512. Accessed April 19, 2024.
Fuke, Nobuo, Naoto Nagata. "Dietary Factors and Metabolic Endotoxemia" Encyclopedia, https://encyclopedia.pub/entry/13512 (accessed April 19, 2024).
Fuke, N., & Nagata, N. (2021, August 24). Dietary Factors and Metabolic Endotoxemia. In Encyclopedia. https://encyclopedia.pub/entry/13512
Fuke, Nobuo and Naoto Nagata. "Dietary Factors and Metabolic Endotoxemia." Encyclopedia. Web. 24 August, 2021.
Dietary Factors and Metabolic Endotoxemia
Edit

Metabolic endotoxemia is a condition in which blood lipopolysaccharide (LPS) levels are elevated, regardless of the presence of obvious infection. It has been suggested to lead to chronic inflammation-related diseases such as obesity, type 2 diabetes mellitus, non-alcoholic fatty liver disease (NAFLD), pancreatitis, amyotrophic lateral sclerosis, and Alzheimer’s disease.

metabolic endotoxemia lipopolysaccharide gut microbiota dietary factors

1. Introduction

Lipopolysaccharide (LPS) is a component of the outer membrane of gram-negative bacteria and is known to induce a variety of inflammatory reactions through Toll-like receptor 4 (TLR4). Injection of LPS into human blood elicits an inflammatory response [1][2], but it was thought that LPS is rarely detected in human blood, except under pathological conditions such as infection and colitis. However, in 2007, Cani et al. showed that mice fed with a high-fat diet had higher blood LPS levels than normal chow-fed mice, resulting in inflammation of the liver and adipose tissue, which led to the development of NAFLD and insulin resistance, and the authors defined this condition as metabolic endotoxemia [3]. Since then, studies on metabolic endotoxemia have been conducted for a variety of diseases. It has been reported that blood LPS levels are higher in humans with obesity [4], type 2 diabetes [5], NAFLD [6], pancreatitis [7], amyotrophic lateral sclerosis [8], and Alzheimer’s disease [8] than those in healthy individuals. Although the causal relationship between metabolic endotoxemia and disease onset is unclear, it is expected to be an interesting target in the future from the viewpoint of disease prevention and treatment. In recent years, the association between metabolic endotoxemia and dietary factors, and the mechanism by which fat intake induces metabolic endotoxemia have been actively studied. In contrast, dietary factors that suppress metabolic endotoxemia have also been explored.

2. Fat Intake and Metabolic Endotoxemia

2.1. Dysbiosis

As Cani et al. reported an increase in blood LPS levels due to a high-fat diet in mice, the mechanism of LPS influx by fat ingestion has been investigated. LPS content both in cecal contents and blood was concomitantly increased by fat ingestion [9], and this increase of LPS was suppressed with oral administration of intestinal alkaline phosphatase, a LPS inactivating enzyme [9]. Oral administration of ampicillin and neomycin, broad-spectrum antibiotics that are poorly absorbed, also suppressed the increase in blood LPS concentration induced by a high-fat diet [10]. These reports suggest that intestinal bacteria are an important source of LPS. In particular, Cani et al. demonstrated changes in intestinal flora (reduction in Bacteroides, Bifidobacterium, and Eubacterium) due to a high-fat diet. Thus, dysbiosis of the intestinal flora due to a high-fat diet has attracted attention as a possible cause for metabolic endotoxemia. Changes in the intestinal bacteria due to ingestion of a high-fat diet have been studied in animals and humans and have been summarized in a review by Netto Candido et al. [11]. In animals, it has been reported that a high-fat diet increases the proportion of Firmicutes, Proteobacteria, and the ratio of Firmicutes to Bacteroidetes. On the other hand, in humans, it has been reported that high-fat dietary intake increases the proportion of Bacteroidetes and decreases the proportion of Firmicutes and Proteobacteria. One possible cause of the different changes in the gut microbiota at the phylum level (e.g., Firmicutes, Bacteroidetes, Proteobacteria) in human and animal studies is the difference in the type of fat consumed. The high-fat diet used in animal experiments (e.g., Research Diets Inc., catalog# D12451) contains lard, while human studies assess fat intake in daily diets. Devkota et al. evaluated the gut microbiota in C57BL/6 mice fed a low-fat diet, a high-fat diet with lard, or a high-fat diet with milk fat for 21 days [12]. In this experiment, both high-fat diets were isocaloric, rich in saturated fatty acids, and 37% of the ingested kcal were from fat. As a result, the proportion of Firmicutes increased and that of Bacteroidetes decreased in the gut microbiota of mice fed a high-fat diet containing lard, compared to mice fed a low-fat diet. In contrast, in mice fed a high-fat diet containing milk fat, the proportion of Firmicutes decreased and that of Bacteroidetes increased compared to the low-fat diet fed mice. Interestingly, Devkota et al. also identified specific bacteria that increased only by ingestion of a high-fat diet containing milk fat [12]. Compared to mice fed a low-fat diet, or a high-fat diet containing lard, mice fed with a high-fat diet containing milk fat had increased proportions of Bilophila wadsworthia, a sulfite-reducing bacterium, in gut microbiota. They also elucidated the mechanism underlying this increase; intake of milk fat increased the level of taurocholic acid in bile. Bilophila wadsworthia populations increased by utilizing sulfur components in taurocholic acid, causing intestinal inflammation in mice. An increase in total fecal bile acid and a concomitant increase in Bilophila wadsworthia in the gut microbiota was also reported in humans upon dietary intake of animal fat [13]. Natividad et al. also showed that increased Bilophila wadsworthia in mice fed a high-fat diet contributed to increased blood LPS levels (they measured soluble CD14 as a surrogate marker), increased fasting blood glucose levels, and the development of a fatty liver [14]. As Helicobacter pylori was discovered as a pathogen in gastric cancer, some pathobionts may also exist for induction of metabolic endotoxemia (however, this cannot be detected by evaluating changes of the gut flora at the phylum levels). We further discuss the bacterial genera that are thought to be associated with metabolic endotoxemia in Section 4. It is also necessary to consider dietary LPS as a source of LPS. For example, milk has been reported to contain high concentrations of LPS in some commercial products [15]. Multiple animal studies have reported that ingested LPS may contribute to increased blood LPS levels. Specifically, Kaliannan et al. measured blood LPS levels 45 min after ingestion of LPS alone or corn oil and LPS in mice [9]. It showed that blood LPS levels were elevated when corn oil and LPS were co-administered. Lindenberg et al. reported that LPS concentrations in the blood were higher in mice fed a high-fat diet containing LPS than in mice fed a high-fat diet without LPS [16]. However, the effect of LPS levels in food on blood LPS levels has not been adequately studied in humans and further studies are needed.

2.2. Mechanisms of the Influx of LPS into the Bloodstream

The gut is protected by a barrier consisting of a mucin layer and epithelial cells. Thus, even if the number of gram-negative bacteria that produce LPS increases in the gut, it is unlikely that the bacterium itself will invade the body. The limulus amebocyte lysate assay used to measure LPS recognizes lipid A, a glycolipid moiety of LPS [17], but because lipid A is embedded in the outer membrane of gram-negative bacteria [18], elevated blood LPS levels suggest that LPS released from gram-negative bacteria is flowing into the blood. In an in vitro study with Escherichia coli, the concentration of free LPS in the culture medium increased with bacterial growth, but the addition of antibiotics stimulated further LPS release [19]. In addition, Jin et al. suggested that treatment with penicillin and erythromycin killed the gram-negative bacteria, Bacteroides and γ-Proteobacteria, leading to increased blood LPS levels in mice [20]. Radilla-Vázquez et al. conducted a correlation analysis of blood LPS levels with fecal Escherichia coli, Prevotella, and Bacteroides fragilis counts in humans and reported that the lower the number of gram-negative bacteria Escherichia coli, the higher the risk of increased blood LPS levels [21]. These reports suggest that LPS release by lysis as well as the increase in gram-negative bacteria may be important factors in increasing blood LPS levels, which may contribute to the inconsistent relationship between changes in intestinal flora and blood LPS levels described above.
With respect to the influx of free LPS, Laugerette et al. reported that in an in vitro assay system using the intestinal epithelial cell line caco-2, LPS permeability to the basal side was increased in the presence of oleic acid, 2-oleoylglycerol, soybean lecithin, cholesterol, and sodium taurocholate [22]. In addition, Clement-Postigo et al. reported a positive correlation between increased LPS levels in the chylomicron fraction and increased triglyceride concentration in serum up to 3 h after a high-fat meal [23]. LPS uptake in chylomicrons has been observed by immunoelectron microscopy [22]. These results suggest that released-LPS in the intestine is taken up into micelles during lipid absorption, and then LPS is absorbed from the intestine together with lipids. In mice, ingestion of a high-fat diet has been reported to increase intestinal permeability by inhibiting the mRNA expression of tight junction-related factors, zonula occludens-1 (ZO-1) and occludin in intestinal epithelial cells [10]. This increase in intestinal permeability is markedly inhibited by antibiotic administration [10], suggesting that it is not the direct effect of lipids but rather a change in intestinal flora. Indeed, secondary bile acids metabolized by enteric bacteria are known to inhibit expression of intestinal tight junction proteins [24][25]. Increased intestinal LPS has been reported to destroy the tight junction of intestinal epithelial cells through TLR4 [26]. Although ingestion of a high-fat diet broadly enhances intestinal and colonic permeability [27], permeability in the colon is closely related to increased blood LPS levels [28][29]. Therefore, disruption of the barrier function by a high-fat diet may have also contributed to the LPS inflow, and the colon may be important as a site of the absorption. The transit time of colonic contents is also probably important. In mice, Anitha et al. suggested that saturated fatty acids induced apoptosis of neurons in the large intestine, reduced peristalsis, induced constipation, and increased blood LPS levels [30]. On the other hand, Reichardt et al. similarly evaluated peristalsis of the large intestine by ingestion of a high-fat diet, but did not observe a clear decrease in peristalsis and an increase in blood LPS levels [31]. Anitha et al. and Reichardt et al. used high-fat diets where either 60% or 30%, respectively of ingested kcal came from fat. Although the ratio of fat to energy intake varied, it has been reported that blood LPS levels increased by consumption of a high-fat diet with 30% of kcal ingested being from fat [32][33]. Therefore, the reason for the lack of increase in blood LPS levels in the study of Reichardt et al. is not considered to be a difference in the fat content of the diet. Ingestion of a high-fat diet does not simply increase blood LPS levels, and retention time of colonic contents due to constipation may also contribute to absorption of LPS.

2.3. Kinetics and Activity of LPS

The LPS concentration in the portal blood is approximately 10 times higher than the LPS concentration in the peripheral blood [34], suggesting that a part of the LPS released in the intestinal tract is flowing from the portal vein. On the other hand, LPS which is concomitantly absorbed with lipids binds to lipoproteins in chylomicrons via LPS-binding protein (LBP) [35], and is thought to pass through the lymphatic system, flow into the blood stream from the left subclavian vein, and then circulate throughout the body. It is reported that blood LPS is bound to various lipoproteins, with plasma LPS concentrations of 31%, 30%, 29%, and 10% for the very low-density lipoprotein (VLDL) fraction, low-density lipoprotein (LDL) fraction, high-density lipoprotein (HDL) fraction, and free LPS, respectively [36]. In addition, LPS bound to lipoproteins of HDL has been reported to be transferred to VLDL and LDL by LBP and phospholipid transfer protein [37], suggesting that the LPS concentration of each lipoprotein fraction changes actively. There are several reports that bioactivity of LPS bound to lipoprotein varies with the type of lipoprotein. First, Vreugdenhil et al. evaluated the effect of chylomicrons, HDL, LDL, and VLDL on the production of tumor necrosis factor-α (TNF-α) from human peripheral blood mononuclear cells on LPS stimulation and showed that chylomicrons inhibited TNF-α production the most [35]. Emansipator et al. reported that a mix of LPS with LDL or HDL decreased the spike recovery of LPS activity in the limulus amebocyte lysate test, and that incubation of LPS with apo A1 decreased the febrile response of rabbits when injected compared to those without apo A1 [38]. In a study using human mononuclear cells [39] and the mouse macrophage cell line Raw 264.7 [40], it was reported that LPS bound to HDL showed reduced interleukin-6 (IL-6) and TNF-α production. VLDL has also been reported to inhibit LPS-induced activation of nuclear factor κB (NF-κB) [41]. On the other hand, oxidized LDL has been shown to promote NFκB activation with LPS in macrophages [42], suggesting that binding to lipoproteins not only decreases LPS activity but also may promote inflammatory responses.
Increased LPS content has been reported in the livers of mice fed a high-fat diet [43], suggesting that the liver is an important site for LPS clearance. Ninety percent of the free LPS that entered the bloodstream is captured by liver resident macrophages (i.e., Kupffer cells) within 1 h [44]. LPS bound to HDL attaches primarily to sinusoidal epithelial cells of the liver [40][44], but it shows slower blood kinetics than free LPS, with 50% present in plasma even 1 h after administration and the amount accumulated in the liver accounted for only 15% of the dose [44]. LPS bound to HDL on the other hand is distributed widely to organs other than the liver, such as the kidney and adipose tissue [44]. LPS accumulated in the liver is inactivated by acyloxyacyl hydroxylase produced by Kupffer cells regardless of free or HDL-bound form [44]. Previously, in a mouse model of high-fat diet plus streptozotocin-induced non-alcoholic steatohepatitis-hepatocellular carcinoma, fecal LPS levels were continuously elevated from six weeks, while liver LPS levels were transiently elevated at eight weeks, followed by increased plasma LPS levels [45]. This report suggests that the liver acts as the first barrier against LPS entering from the intestinal tract and that liver dysfunction leads to elevated blood LPS levels. Interestingly, LPS administration in mice increased the expression of apolipoprotein AIV in the liver via TLR4, suggesting that the liver has a mechanism to increase HDL production and protect itself against LPS stimulation [46].

3. Dietary Factors that Decrease Blood LPS Levels

Previous reports investigating the effects of dietary factors on blood LPS levels are summarized in Table 1 (human interventional studies), Table 2 (human epidemiological studies) and Table 3 (animal studies). The findings about representative food categories are reviewed in the following sections.
Table 1. Dietary factors that have been evaluated for efficacy on blood lipopolysaccharide (LPS) levels in human interventional studies.

Category

Dietary Factor

Dose

Consumption Period

Subject

LPS

LBP

Gut Microbes with Significant Changes in Proportion **

Increase

Decrease

Probiotics/

Prebiotics

Yakult light

(Lactobacillus casei Shirota 1 × 108 CFU/mL) [47]

195 mL

3 months

Metabolic syndrome

ND

Low-fat yogurt

[48]

339 g

9 weeks

Healthy subject or Obesity

Low-fat yogurt

[49]

226 g

Premeal

Healthy subject or Obesity

(postprandial endotoxemia was assessed)

Oligofructose

[50]

21 g

12 weeks

Overweight/

Obesity

Oligofructose-

enriched inulin

[51]

10 g

8 weeks

Type 2 diabetes

Inulin +

Oligofructose

[52]

8 g

8 g

3 months

Obesity

Bifidobacterium,

Faecalibacterium prausnitzii

Bacteroides intestinalis,

Bacteroides vulgatus,

Propionibacterium

Galacto-

oligosaccharide

[53]

5.5 g

12 weeks

Type 2 diabetes

none

none

Galacto-

oligosaccharide

[54]

15 g

12 weeks

Overweight/

Obesity

Bifidobacterium spp.

none

α-Galacto-

oligosaccharide

[55]

6–18 g

14 days

Overweight

Bifidobacteria

none

Resistant dextrin

[56]

10 g

8 weeks

Type 2 diabetes

Insoluble dietary fiber

[from Fiber One Original cereal

(General mills)]

[57]

30 g

With high-fat, high-calorie meal

Healthy subject

(postprandial endotoxemia was assessed)

↓*

Whole grains

[58]

3 servings

6 weeks

Overweight/

Obesity

none

none

Probiotics/

Prebiotics

Bifidobacterium longum +

Oligofructose +

Life style modification

[59]

24 weeks

Non-alcoholic steatohepatitis

Polyphenol

Resveratrol +

Polyphenol

[60]

100 mg

75 mg

10 minutes before intake of high-fat high-carbohydrate meal

Healthy subjects

(postprandial endotoxemia was assessed)

↓ *

Red wine

[61]

272 mL

With high-fat meal

Healthy subjects

(postprandial endotoxemia was assessed)

Dietary habits

Fruits +

Vegetables

[58]

3 servings

6 weeks

Overweight/

Obesity

α-diversity

(No significant change in bacterial genera was found)

none

Caloric restriction

[62]

800 kcal

4 weeks

Obesity

Anaerostipes hadrus,

Blautia sp.,

Ruminococcus faecis,

Bifidodbacterium sp.

Agathobacter rectalis

Others

Glutamine

[63]

30 g

14 weeks

Overweight/

Obesity

 
ND: Not detected, —: No data, ↑: Significantly increased, →: Not significantly changed, ↓: Significantly decreased, *: Attenuation of postprandial endotoxemia, **: The bacteria mentioned by the author in the paper are listed.
Table 2. Correlation of dietary factors, gut microbes, and blood LPS levels in human epidemiological studies.

Subject

Number of Subject

Correlation of

Dietary Factor and Gut Microbe *

Correlation of

Blood LPS and Gut Microbe

Correlation of

Blood LPS and Dietary Factor

Over-

weight pregnant women

[64]

88

P

Dietary fiber vs.

diversity, richness, Firmicutes in unidentified family of order Clostridiales, Barnciellaceae family belonging to the phylum Bacteroidetes

P

none

P

none

Vitamin A,

β-Carotene vs.

Firmicutes

N

Fat vs.

diversity, richness, Barnsiellaceae

N

none

N

none

Healthy subjects

[65]

150

N

25-Hydroxy vitamin D vs.

Coprococcus, Bifdobacterium

N

LPS vs.

Faecalibacterium

N

LPS

vs.

25-Hydroxy vitamin D

Type 1 diabetes

[66]

668

N

LPS

vs.

Dietary pattern; “Fish”(frequently eat fish dishes), “Healthy snack” (frequently eat fruits, berries, fresh vegetable, yoghurt, low-fat cheese, and do not drink much soft drinks), “Modern”(frequently eat poultry, pasta, rice, meat dishes, fried and grilled foods, and fresh vegetables)

 
—: No data, P: Positive correlation, N: Negative correlation, LPS: lipopolysaccharide, *: The bacteria mentioned by the author in the paper are listed.
Table 3. Dietary factors that have been evaluated for efficacy on blood LPS levels in animal interventional studies.

Category

Dietary Factor

Dose

Administration Period

Model

LPS

LBP

Significant Change in

Gut Microbiota

Probiotics/

Prebiotics

Lactobacillus rhamnosus GG

[67]

1 × 108 CFU/day

12 weeks

HFD-fed ApoE KO mouse

no

Lactobacillus rhamnosus CNCM I-4036

[68]

1 × 1010 CFU/day

30 days

Chow diet-fed

Zucker-Lepfa/fa rat

Lactobacillus sakei OK67 +/−

Lactobacillus sakei PK16

[69]

1 × 109 CFU/day

1 × 109 CFU/day

4 weeks

HFD-fed

C57BL/6 mouse

yes

Bifidobacterium longum BR-108

(sterilized)

[70]

200, 400 mg/kg/day

4 weeks

HFD-fed

C57BL/6J mouse

yes

Bifidobacterium infantis +

Lactobacillus acidophilus +

Bacillus cereus

[71]

0.5 × 106 CFU/day

0.5 × 106 CFU/day

0.5 × 105 CFU/day

12 weeks

HFHSD-fed

SD rat

yes

Lactobacillus plantarum LC27 +/−

Bifidobacterium longum LC67

[43]

1 × 109 CFU/day each

(or 0.75 × 109 (LC27) + 0.25×109 (LC67) CFU/day in mix)

4 weeks

HFD-fed

C57BL/6 mouse

yes

Oligofructose

[72]

10% (mixed in diet)

12 weeks

HFHSD-fed

SD rat

yes

Galacto-

oligosaccharide

[73]

800 mg/kg/day

8 weeks

HFD-fed

SD rat

yes

Inulin

[74]

5% (intragastric administration, sample volume was not described)

6 weeks

standardized diet (kcal %: 10% fat, 20% protein, and 70% carbo- hydrate; 3.85 kcal g−1)-fed db/db mouse

yes

Wheat-derived arabinoxylan

[75]

7.5% (mixed in diet)

8 weeks

HFD-fed

C57BL/6J mouse

Polyphenols

Grape seed proanthocyanidin

[33]

500 mg/kg/day

10 days (prophylactic) or

17 weeks (with cafeteria diet)

Cafeteria diet

(high-fat/high carbohydrate diet)-fed Wistar rat

Grape-seed proanthocyanidin

[29]

100, 500 mg/kg/day

2 weeks

Cafeteria diet

(high saturated-fat/high refined-carbohydrate diet)-fed Wistar rat

Resveratrol

[76]

50, 75, 100 mg/kg/day

16 weeks

HFD-fed

C57BL/6 mouse

yes

Apple-derived polymeric procyanidins

[77]

0.5% (administration route was not described)

20 weeks

HFHSD-fed

C57BL/6J mouse

yes

Genistein

[78]

0.2% (mixed in diet)

6 months

HFD-fed

C57BL/6 mouse

yes

Isoflavone

[79]

0.1% (mixed in diet)

5 weeks

HFD-fed

C57BL/6 mouse

yes

Syringaresinol

[80]

50 mg/kg/day

10 weeks

40-week-old

C57BL/6 mouse

yes

Sulfated polysaccharide

Sea cucumber-derived sulfated polysaccharide

[81]

300 mg/kg/day

8 weeks

HFD-fed

BALB/c mouse

yes

Sea cucumber-derived sulfated polysaccharide

[82]

300 mg/kg/day

42 days

Chow-fed

BALB/c mouse

yes

Acaudina molpadioides-derived fucosylated chondroitin sulfate

[83]

80 mg/kg/day

10 weeks

HFD-fed

C57BL/6J mouse

yes

Chicken-derived chondroitin sulfate

[84]

150 mg/kg/day

16 days

Exhaustive exercise stress model

BALB/c mouse

yes

Fucoidan

[73]

100 mg/kg/day

8 weeks

HFD-fed

SD rat

yes

Other dietary components

Tetrahydro iso-alpha acid

(included in hops)

[85]

0.1% (mixed in diet)

8 weeks

HFD-fed

C57BL/6J mouse

Rhein (included in rhubarb)

[86]

120 mg/kg/day

6 weeks

HFD-fed

C57BL/6J mouse

yes

Phlorizin (included in apple)

[87]

20 mg/kg/day

10 weeks

Chow-fed

db/db mouse

yes

Capsaicin

[88]

0.01% (mixed in diet)

12 weeks

HFD-fed

C57BL/6J mouse

yes

Rutin

[89]

0.64% (mixed in diet)

20 weeks

HFD-fed

C57BL/6J mouse

yes

Lycopene

[90]

0.03% (mixed in diet)

10 weeks

HFD and fructose-fed C57BL/6 J mouse

Other extracts/dietary components

Broccoli sprout extract

[91]

2.2% (mixed in diet)

14 weeks

HFD-fed

C57BL/6JSlc mouse

yes

Camu camu extract

[92]

200 mg/kg/day

8 weeks

HFHSD-fed

C57BL/6J mouse

yes

Other extracts/dietary components

Cranberry extract

[93]

200 mg/kg/day

8 weeks

HFHSD-fed

C57BL/6J mouse

yes

Green tea extract

[34]

2% (mixed in diet)

8 weeks

HFD-fed

C57BL/6J mouse

yes

Tartary buckwheat protein

[32]

23.5% (mixed in diet)

6 weeks

HFD-fed

C57BL/6 mouse

yes

Foods

Cocoa

[94]

8% (mixed in diet)

18 weeks

HFD-fed

C57BL/6J mouse

Nopal

[95]

5% of dietary fiber was replaced with those of nopal-derived (mixed in diet)

1 month

HFHSD-fed

Wistar rat

yes

Steamed fish meat

[96]

Ad libitum

(9:00–12:00 and 18:00–21:00)

8 weeks

Chow-fed

C57BL/6 mouse

yes

Chinese medicines

Geniposide +

Chlorogenic acid

[97]

90 mg/kg/day

1.34 mg/kg/day

4 weeks

HFD-fed

C57BL/6 mouse

Potentilla discolor Bunge water extract

[98]

400 mg/kg/day

8 weeks

HFD-fed, streptozotocin-

injected

C57BL/6J mouse

yes

Ganoderma lucidum mycelium water extract

[99]

2–8 mg/day

8 weeks

HFD-fed

C57BL/6NCrlBltw mouse

yes

Semen hoveniae extract

[100]

300, 600 mg/kg/day

8 weeks

Alcohol-containing Lieber-DeCarli diet-fed SD rat (Alcoholic liver disorder model)

yes

Shenling Baizhu powder

[101]

30 g/kg/day

16 weeks

HFD-fed

SD rat

yes

Caloric restriction

30% caloric restriction

[102]

62–141 weeks

HFD, LFD-fed

C57BL/6J mouse

yes

40% caloric restriction

[103]

30 days

Chow-fed C57BL/6J mouse

yes

 

—: No data, HFD: High-fat diet, HFHSD: High-fat high-sucrose diet, ↑: Significantly increased, →: Not significantly changed, ↓: Significantly decreased.

4. Association of Dietary Factor-Induced Reduction of Blood LPS and Modulation of Gut Microbiota

Although few studies have evaluated the relationship between the effect of dietary factors on blood LPS and intestinal flora in humans, several studies have evaluated intestinal flora in oligosaccharide intervention studies (Table 1). A common finding in these reports is an increase in Bifidobacterium. Bifidobacterium has been reported to enhance the intestinal tight junction by preserving claudin 4 and occludin localization at tight junctions, and inhibit permeability in mice with colitis [104]. Similarly, in human colonic epithelial cell line T84, the addition of culture supernatant of Bifidobacterium has been reported to enhance barrier function through increased expression of tight junction protein, suggesting that some humoral factors contribute to improved intestinal barrier function [105]. Increased expression of tight junction protein in Bifidobacterium-treated mice has been reported to be associated with increased short-chain fatty acids (acetic acid, butyric acid, and propionic acid) in the intestinal tract [106]. These short-chain fatty acids have been reported in the human colonic epithelial cell line caco-2 to act as an energy source for epithelial cells to protect themselves, and also act as a histone deacetylase inhibitor which inhibit Nod-like receptor P3 inflammasomes to maintain the barrier function of epithelial cells [107]. These results suggest that the increase in Bifidobacterium induced by oligosaccharide intake decreases blood LPS levels through the improvement of the barrier function of the intestinal tract. In addition, dietary factors that increase Bifidobacterium are expected to reduce blood LPS levels.
All of the dietary factors commonly lowered blood LPS or LBP levels in animals, as described in Table 3. In other words, by finding bacteria that have decreased or increased in many dietary factor intervention studies, we can find specific bacteria that contribute to the increase or decrease in blood LPS levels. To this end, we have organized the number of reports that show increases or decreases of each bacterial genus (Figure 1). We selected eight of these genera (Lactobacillus, Bacteroides, Akkermansia, Clostridium, Escherichia, Roseburia, Prevotella and Desulfovibrio) as bacteria included in a sufficient number (five or more) of reports, and a biased number of reports (Bifidobacterium was excluded because it was discussed above. Faecalibacterium was also excluded because there is almost no bias in the number of reports).
Figure 1. The number of reported changes of intestinal bacterial genera in dietary factor intervention studies in animals.

References

  1. Kiers, D.; Leijte, G.P.; Gerretsen, J.; Zwaag, J.; Kox, M.; Pickkers, P. Comparison of different lots of endotoxin and evaluation of in vivo potency over time in the experimental human endotoxemia model. Innate Immun. 2019, 25, 34–45.
  2. Benson, S.; Engler, H.; Wegner, A.; Rebernik, L.; Spreitzer, I.; Schedlowski, M.; Elsenbruch, S. What makes you feel sick after inflammation? Predictors of acute and persisting physical sickness symptoms induced by experimental endotoxemia. Clin. Pharmacol. Ther. 2017, 102, 141–151.
  3. Cani, P.D.; Amar, J.; Iglesias, M.A.; Poggi, M.; Knauf, C.; Bastelica, D.; Neyrinck, A.M.; Fava, F.; Tuohy, K.M.; Chabo, C.; et al. Metabolic endotoxemia initiates obesity and insulin resistance. Diabetes 2007, 56, 1761–1772.
  4. Liang, H.; Hussey, S.E.; Sanchez-Avila, A.; Tantiwong, P.; Musi, N. Effect of lipopolysaccharide on inflammation and insulin action in human muscle. PLoS ONE 2013, 8, e63983.
  5. Pussinen, P.J.; Havulinna, A.S.; Lehto, M.; Sundvall, J.; Salomaa, V. Endotoxemia is associated with an increased risk of incident diabetes. Diabetes Care 2011, 34, 392–397.
  6. Jin, R.; Willment, A.; Patel, S.S.; Sun, X.; Song, M.; Mannery, Y.O.; Kosters, A.; McClain, C.J.; Vos, M.B. Fructose induced endotoxemia in pediatric nonalcoholic fatty liver disease. Int. J. Hepatol. 2014, 2014, 560620.
  7. Jandhyala, S.M.; Madhulika, A.; Deepika, G.; Rao, G.V.; Reddy, D.N.; Subramanyam, C.; Sasikala, M.; Talukdar, R. Altered intestinal microbiota in patients with chronic pancreatitis: Implications in diabetes and metabolic abnormalities. Sci. Rep. 2017, 7, 43640.
  8. Zhang, R.; Miller, R.G.; Gascon, R.; Champion, S.; Katz, J.; Lancero, M.; Narvaez, A.; Honrada, R.; Ruvalcaba, D.; McGrath, M.S. Circulating endotoxin and systemic immune activation in sporadic amyotrophic lateral sclerosis (sALS). J. Neuroimmunol. 2009, 206, 121–124.
  9. Kaliannan, K.; Hamarneh, S.R.; Economopoulos, K.P.; Nasrin Alam, S.; Moaven, O.; Patel, P.; Malo, N.S.; Ray, M.; Abtahi, S.M.; Muhammad, N.; et al. Intestinal alkaline phosphatase prevents metabolic syndrome in mice. Proc. Natl. Acad. Sci. USA 2013, 110, 7003–7008.
  10. Cani, P.D.; Bibiloni, R.; Knauf, C.; Waget, A.; Neyrinck, A.M.; Delzenne, N.M.; Burcelin, R. Changes in gut microbiota control metabolic endotoxemia-induced inflammation in high-fat diet-induced obesity and diabetes in mice. Diabetes 2008, 57, 1470–1481.
  11. Candido, T.L.N.; Bressan, J.; Alfenas, R.d.C.G. Dysbiosis and metabolic endotoxemia induced by high-fat diet. Nutr. Hosp. 2018, 35, 1432–1440.
  12. Devkota, S.; Wang, Y.; Musch, M.W.; Leone, V.; Fehlner-Peach, H.; Nadimpalli, A.; Antonopoulos, D.A.; Jabri, B.; Chang, E.B. Dietary-fat-induced taurocholic acid promotes pathobiont expansion and colitis in Il10-/- mice. Nature 2012, 487, 104–108.
  13. David, L.A.; Maurice, C.F.; Carmody, R.N.; Gootenberg, D.B.; Button, J.E.; Wolfe, B.E.; Ling, A.V.; Devlin, A.S.; Varma, Y.; Fischbach, M.A.; et al. Diet rapidly and reproducibly alters the human gut microbiome. Nature 2014, 505, 559–563.
  14. Natividad, J.M.; Lamas, B.; Pham, H.P.; Michel, M.-L.; Rainteau, D.; Bridonneau, C.; da Costa, G.; van Hylckama Vlieg, J.; Sovran, B.; Chamignon, C.; et al. Bilophila wadsworthia aggravates high fat diet induced metabolic dysfunctions in mice. Nat. Commun. 2018, 9, 2802.
  15. Gehring, U.; Spithoven, J.; Schmid, S.; Bitter, S.; Braun-Fahrländer, C.; Dalphin, J.-C.; Hyvärinen, A.; Pekkanen, J.; Riedler, J.; Weiland, S.K.; et al. Endotoxin levels in cow’s milk samples from farming and non-farming families - the PASTURE study. Environ. Int. 2008, 34, 1132–1136.
  16. Lindenberg, F.C.B.; Ellekilde, M.; Thörn, A.C.; Kihl, P.; Larsen, C.S.; Hansen, C.H.F.; Metzdorff, S.B.; Aalbæk, B.; Hansen, A.K. Dietary LPS traces influences disease expression of the diet-induced obese mouse. Res. Vet. Sci. 2019, 123, 195–203.
  17. Gutsmann, T.; Howe, J.; Zähringer, U.; Garidel, P.; Schromm, A.B.; Koch, M.H.J.; Fujimoto, Y.; Fukase, K.; Moriyon, I.; Martínez-de-Tejada, G.; et al. Structural prerequisites for endotoxic activity in the limulus test as compared to cytokine production in mononuclear cells. Innate Immun. 2010, 16, 39–47.
  18. Bishop, R.E. Structural biology of membrane-intrinsic beta-barrel enzymes: Sentinels of the bacterial outer membrane. Biochim. Biophys. Acta 2008, 1778, 1881–1896.
  19. Van Den Berg, C.; de Neeling, A.J.; Schot, C.S.; Hustinx, W.N.M.; Wemer, J.; de Wildt, D.J. Delayed antibiotic-induced lysis of escherichia coli in vitro is correlated with enhancement of LPS release. Scand. J. Infect. Dis. 1992, 24, 619–627.
  20. Jin, Y.; Wu, Y.; Zeng, Z.; Jin, C.; Wu, S.; Wang, Y.; Fu, Z. From the cover: Exposure to oral antibiotics induces gut microbiota dysbiosis associated with lipid metabolism dysfunction and low-grade inflammation in mice. Toxicol. Sci. 2016, 154, 140–152.
  21. Radilla-Vázquez, R.B.; Parra-Rojas, I.; Martínez-Hernández, N.E.; Márquez-Sandoval, Y.F.; Illades-Aguiar, B.; Castro-Alarcón, N. Gut microbiota and metabolic endotoxemia in young obese mexican subjects. Obes. Facts 2016, 9, 1–11.
  22. Laugerette, F.; Vors, C.; Géloën, A.; Chauvin, M.; Soulage, C.; Lambert-Porcheron, S.; Peretti, N.; Alligier, M.; Burcelin, R.; Laville, M.; et al. Emulsified lipids increase endotoxemia: Possible role in early postprandial low-grade inflammation. J. Nutr. Biochem. 2011, 22, 53–59.
  23. Clemente-Postigo, M.; Queipo-Ortuño, M.I.; Murri, M.; Boto-Ordoñez, M.; Perez-Martinez, P.; Andres-Lacueva, C.; Cardona, F.; Tinahones, F.J. Endotoxin increase after fat overload is related to postprandial hypertriglyceridemia in morbidly obese patients. J. Lipid Res. 2012, 53, 973–978.
  24. Murakami, Y.; Tanabe, S.; Suzuki, T. High-fat diet-induced intestinal hyperpermeability is associated with increased bile acids in the large intestine of mice. J. Food Sci. 2016, 81, H216–H222.
  25. Ahmad, R.; Rah, B.; Bastola, D.; Dhawan, P.; Singh, A.B. Obesity-induces organ and tissue specific tight junction restructuring and barrier deregulation by claudin switching. Sci. Rep. 2017, 7, 5125.
  26. Guo, S.; Nighot, M.; Al-Sadi, R.; Alhmoud, T.; Nighot, P.; Ma, T.Y. Lipopolysaccharide regulation of intestinal tight junction permeability is mediated by TLR4 signal transduction pathway activation of FAK and MyD88. J. Immunol. 2015, 195, 4999–5010.
  27. Fang, W.; Xue, H.; Chen, X.; Chen, K.; Ling, W. Supplementation with sodium butyrate modulates the composition of the gut microbiota and ameliorates high-fat diet-induced obesity in mice. J. Nutr. 2019, 149, 747–754.
  28. Blanchard, C.; Moreau, F.; Chevalier, J.; Ayer, A.; Garcon, D.; Arnaud, L.; Pais de Barros, J.-P.; Gautier, T.; Neunlist, M.; Cariou, B.; et al. Sleeve gastrectomy alters intestinal permeability in diet-induced obese mice. Obes. Surg. 2017, 27, 2590–2598.
  29. González-Quilen, C.; Gil-Cardoso, K.; Ginés, I.; Beltrán-Debón, R.; Pinent, M.; Ardévol, A.; Terra, X.; Blay, M.T. Grape-Seed proanthocyanidins are able to reverse intestinal dysfunction and metabolic endotoxemia induced by a cafeteria diet in wistar rats. Nutrients 2019, 11, 979.
  30. Anitha, M.; Reichardt, F.; Tabatabavakili, S.; Nezami, B.G.; Chassaing, B.; Mwangi, S.; Vijay-Kumar, M.; Gewirtz, A.; Srinivasan, S. Intestinal dysbiosis contributes to the delayed gastrointestinal transit in high-fat diet fed mice. Cell. Mol. Gastroenterol. Hepatol. 2016, 2, 328–339.
  31. Reichardt, F.; Chassaing, B.; Nezami, B.G.; Li, G.; Tabatabavakili, S.; Mwangi, S.; Uppal, K.; Liang, B.; Vijay-Kumar, M.; Jones, D.; et al. Western diet induces colonic nitrergic myenteric neuropathy and dysmotility in mice via saturated fatty acid- and lipopolysaccharide-induced TLR4 signalling. J. Physiol. 2017, 595, 1831–1846.
  32. Zhou, X.-L.; Yan, B.; Xiao, Y.; Zhou, Y.; Liu, T. Tartary buckwheat protein prevented dyslipidemia in high-fat diet-fed mice associated with gut microbiota changes. Food Chem. Toxicol. 2018, 119, 296–301.
  33. Gil-Cardoso, K.; Ginés, I.; Pinent, M.; Ardévol, A.; Blay, M.; Terra, X. The co-administration of proanthocyanidins and an obesogenic diet prevents the increase in intestinal permeability and metabolic endotoxemia derived to the diet. J. Nutr. Biochem. 2018, 62, 35–42.
  34. Dey, P.; Sasaki, G.Y.; Wei, P.; Li, J.; Wang, L.; Zhu, J.; McTigue, D.; Yu, Z.; Bruno, R.S. Green tea extract prevents obesity in male mice by alleviating gut dysbiosis in association with improved intestinal barrier function that limits endotoxin translocation and adipose inflammation. J. Nutr. Biochem. 2019, 67, 78–89.
  35. Vreugdenhil, A.C.E.; Rousseau, C.H.; Hartung, T.; Greve, J.W.M.; van ’t Veer, C.; Buurman, W.A. Lipopolysaccharide (LPS)-binding protein mediates LPS detoxification by chylomicrons. J. Immunol. 2003, 170, 1399–1405.
  36. Vergès, B.; Duvillard, L.; Lagrost, L.; Vachoux, C.; Garret, C.; Bouyer, K.; Courtney, M.; Pomié, C.; Burcelin, R. Changes in lipoprotein kinetics associated with type 2 diabetes affect the distribution of lipopolysaccharides among lipoproteins. J. Clin. Endocrinol. Metab. 2014, 99, E1245–E1253.
  37. Levels, J.H.M.; Marquart, J.A.; Abraham, P.R.; van den Ende, A.E.; Molhuizen, H.O.F.; van Deventer, S.J.H.; Meijers, J.C.M. Lipopolysaccharide is transferred from high-density to low-density lipoproteins by lipopolysaccharide-binding protein and phospholipid transfer protein. Infect. Immun. 2005, 73, 2321–2326.
  38. Emancipator, K.; Csako, G.; Elin, R.J. In vitro inactivation of bacterial endotoxin by human lipoproteins and apolipoproteins. Infect. Immun. 1992, 60, 596–601.
  39. Correa, W.; Brandenburg, K.; Zähringer, U.; Ravuri, K.; Khan, T.; von Wintzingerode, F. Biophysical analysis of lipopolysaccharide formulations for an understanding of the low endotoxin recovery (LER) phenomenon. Int. J. Mol. Sci. 2017, 18, 2737.
  40. Yao, Z.; Mates, J.M.; Cheplowitz, A.M.; Hammer, L.P.; Maiseyeu, A.; Phillips, G.S.; Wewers, M.D.; Rajaram, M.V.S.; Robinson, J.M.; Anderson, C.L.; et al. Blood-Borne lipopolysaccharide is rapidly eliminated by liver sinusoidal endothelial cells via high-density lipoprotein. J. Immunol. 2016, 197, 2390–2399.
  41. van Bergenhenegouwen, J.; Kraneveld, A.D.; Rutten, L.; Garssen, J.; Vos, A.P.; Hartog, A. Lipoproteins attenuate TLR2 and TLR4 activation by bacteria and bacterial ligands with differences in affinity and kinetics. BMC Immunol. 2016, 17, 42.
  42. Wiesner, P.; Choi, S.; Almazan, F.; Benner, C.; Huang, W.; Diehl, C.J.; Gonen, A.; Butler, S.; Witztum, J.L.; Glass, C.K.; et al. Low doses of lipopolysaccharide and minimally oxidized low-density lipoprotein cooperatively activate macrophages via nuclear factor kappa B and activator protein-1: Possible mechanism for acceleration of atherosclerosis by subclinical endotoxemia. Circ. Res. 2010, 107, 56–65.
  43. Kim, H.I.; Kim, J.-K.; Kim, J.-Y.; Jang, S.E.; Han, M.J.; Kim, D.-H. Lactobacillus plantarum LC27 and Bifidobacterium longum LC67 simultaneously alleviate high-fat diet-induced colitis, endotoxemia, liver steatosis, and obesity in mice. Nutr. Res. 2019, 67, 78–89.
  44. Shao, B.; Munford, R.S.; Kitchens, R.; Varley, A.W. Hepatic uptake and deacylation of the LPS in bloodborne LPS-lipoprotein complexes. Innate Immun. 2012, 18, 825–833.
  45. Xie, G.; Wang, X.; Liu, P.; Wei, R.; Chen, W.; Rajani, C.; Hernandez, B.Y.; Alegado, R.; Dong, B.; Li, D.; et al. Distinctly altered gut microbiota in the progression of liver disease. Oncotarget 2016, 7, 19355–19366.
  46. Dandekar, A.; Qiu, Y.; Kim, H.; Wang, J.; Hou, X.; Zhang, X.; Zheng, Z.; Mendez, R.; Yu, F.-S.; Kumar, A.; et al. Toll-like receptor (TLR) signaling interacts with CREBH to modulate high-density lipoprotein (HDL) in response to bacterial endotoxin. J. Biol. Chem. 2016, 291, 23149–23158.
  47. Leber, B.; Tripolt, N.J.; Blattl, D.; Eder, M.; Wascher, T.C.; Pieber, T.R.; Stauber, R.; Sourij, H.; Oettl, K.; Stadlbauer, V. The influence of probiotic supplementation on gut permeability in patients with metabolic syndrome: An open label, randomized pilot study. Eur. J. Clin. Nutr. 2012, 66, 1110–1115.
  48. Pei, R.; DiMarco, D.M.; Putt, K.K.; Martin, D.A.; Gu, Q.; Chitchumroonchokchai, C.; White, H.M.; Scarlett, C.O.; Bruno, R.S.; Bolling, B.W. Low-fat yogurt consumption reduces biomarkers of chronic inflammation and inhibits markers of endotoxin exposure in healthy premenopausal women: A randomised controlled trial. Br. J. Nutr. 2017, 118, 1043–1051.
  49. Pei, R.; DiMarco, D.M.; Putt, K.K.; Martin, D.A.; Chitchumroonchokchai, C.; Bruno, R.S.; Bolling, B.W. Premeal low-fat yogurt consumption reduces postprandial inflammation and markers of endotoxin exposure in healthy premenopausal women in a randomized controlled trial. J. Nutr. 2018, 148, 910–916.
  50. Parnell, J.A.; Klancic, T.; Reimer, R.A. Oligofructose decreases serum lipopolysaccharide and plasminogen activator inhibitor-1 in adults with overweight/obesity. Obesity 2017, 25, 510–513.
  51. Dehghan, P.; Pourghassem Gargari, B.; Asghari Jafar-abadi, M. Oligofructose-enriched inulin improves some inflammatory markers and metabolic endotoxemia in women with type 2 diabetes mellitus: A randomized controlled clinical trial. Nutrition 2014, 30, 418–423.
  52. Dewulf, E.M.; Cani, P.D.; Claus, S.P.; Fuentes, S.; Puylaert, P.G.B.; Neyrinck, A.M.; Bindels, L.B.; de Vos, W.M.; Gibson, G.R.; Thissen, J.; et al. Insight into the prebiotic concept: Lessons from an exploratory, double blind intervention study with inulin-type fructans in obese women. Gut 2013, 62, 1112–1121.
  53. Pedersen, C.; Gallagher, E.; Horton, F.; Ellis, R.J.; Ijaz, U.Z.; Wu, H.; Jaiyeola, E.; Diribe, O.; Duparc, T.; Cani, P.D.; et al. Host-microbiome interactions in human type 2 diabetes following prebiotic fibre (galacto-oligosaccharide) intake. Br. J. Nutr. 2016, 116, 1869–1877.
  54. Canfora, E.E.; van der Beek, C.M.; Hermes, G.D.A.; Goossens, G.H.; Jocken, J.W.E.; Holst, J.J.; van Eijk, H.M.; Venema, K.; Smidt, H.; Zoetendal, E.G.; et al. Supplementation of diet with Galacto-oligosaccharides increases bifidobacteria, but not insulin sensitivity, in obese prediabetic individuals. Gastroenterology 2017, 153, 87–97.
  55. Morel, F.B.; Dai, Q.; Ni, J.; Thomas, D.; Parnet, P.; Fança-Berthon, P. α-Galacto-oligosaccharides dose-dependently reduce appetite and decrease inflammation in overweight adults. J. Nutr. 2015, 145, 2052–2059.
  56. Farhangi, M.A.; Javid, A.Z.; Sarmadi, B.; Karimi, P.; Dehghan, P. A randomized controlled trial on the efficacy of resistant dextrin, as functional food, in women with type 2 diabetes: Targeting the hypothalamic-pituitary-adrenal axis and immune system. Clin. Nutr. 2018, 37, 1216–1223.
  57. Ghanim, H.; Batra, M.; Abuaysheh, S.; Green, K.; Makdissi, A.; Kuhadiya, N.D.; Chaudhuri, A.; Dandona, P. Antiinflammatory and ROS suppressive effects of the addition of fiber to a high-fat high-calorie meal. J. Clin. Endocrinol. Metab. 2017, 102, 858–869.
  58. Kopf, J.C.; Suhr, M.J.; Clarke, J.; Eyun, S.; Riethoven, J.M.; Ramer-Tait, A.E.; Rose, D.J. Role of whole grains versus fruits and vegetables in reducing subclinical inflammation and promoting gastrointestinal health in individuals affected by overweight and obesity: A randomized controlled trial. Nutr. J. 2018, 17, 72.
  59. Malaguarnera, M.; Vacante, M.; Antic, T.; Giordano, M.; Chisari, G.; Acquaviva, R.; Mastrojeni, S.; Malaguarnera, G.; Mistretta, A.; Li Volti, G.; et al. Bifidobacterium longum with fructo-oligosaccharides in patients with non alcoholic steatohepatitis. Dig. Dis. Sci. 2012, 57, 545–553.
  60. Ghanim, H.; Sia, C.L.; Korzeniewski, K.; Lohano, T.; Abuaysheh, S.; Marumganti, A.; Chaudhuri, A.; Dandona, P. A resveratrol and polyphenol preparation suppresses oxidative and inflammatory stress response to a high-fat, high-carbohydrate meal. J. Clin. Endocrinol. Metab. 2011, 96, 1409–1414.
  61. Clemente-Postigo, M.; Queipo-Ortuño, M.I.; Boto-Ordoñez, M.; Coin-Aragüez, L.; Roca-Rodriguez, M.D.M.; Delgado-Lista, J.; Cardona, F.; Andres-Lacueva, C.; Tinahones, F.J. Effect of acute and chronic red wine consumption on lipopolysaccharide concentrations. Am. J. Clin. Nutr. 2013, 97, 1053–1061.
  62. Ott, B.; Skurk, T.; Hastreiter, L.; Lagkouvardos, I.; Fischer, S.; Büttner, J.; Kellerer, T.; Clavel, T.; Rychlik, M.; Haller, D.; et al. Effect of caloric restriction on gut permeability, inflammation markers, and fecal microbiota in obese women. Sci. Rep. 2017, 7, 11955.
  63. Abboud, K.Y.; Reis, S.K.; Martelli, M.E.; Zordão, O.P.; Tannihão, F.; de Souza, A.Z.Z.; Assalin, H.B.; Guadagnini, D.; Rocha, G.Z.; Saad, M.J.A.; et al. Oral glutamine supplementation reduces obesity, pro-inflammatory markers, and improves insulin sensitivity in DIO wistar rats and reduces waist circumference in overweight and obese humans. Nutrients 2019, 11, 536.
  64. Röytiö, H.; Mokkala, K.; Vahlberg, T.; Laitinen, K. Dietary intake of fat and fibre according to reference values relates to higher gut microbiota richness in overweight pregnant women. Br. J. Nutr. 2017, 118, 343–352.
  65. Luthold, R.V.; Fernandes, G.R.; Franco-de-Moraes, A.C.; Folchetti, L.G.D.; Ferreira, S.R.G. Gut microbiota interactions with the immunomodulatory role of vitamin D in normal individuals. Metabolism. 2017, 69, 76–86.
  66. Ahola, A.J.; Lassenius, M.I.; Forsblom, C.; Harjutsalo, V.; Lehto, M.; Groop, P.-H. Dietary patterns reflecting healthy food choices are associated with lower serum LPS activity. Sci. Rep. 2017, 7, 6511.
  67. Chan, Y.K.; Brar, M.S.; Kirjavainen, P.V.; Chen, Y.; Peng, J.; Li, D.; Leung, F.C.; El-Nezami, H. High fat diet induced atherosclerosis is accompanied with low colonic bacterial diversity and altered abundances that correlates with plaque size, plasma A-FABP and cholesterol: A pilot study of high fat diet and its intervention with Lactobacillus rhamno. BMC Microbiol. 2016, 16, 264.
  68. Plaza-Díaz, J.; Robles-Sánchez, C.; Abadía-Molina, F.; Morón-Calvente, V.; Sáez-Lara, M.J.; Ruiz-Bravo, A.; Jiménez-Valera, M.; Gil, Á.; Gómez-Llorente, C.; Fontana, L. Adamdec1, Ednrb and Ptgs1/Cox1, inflammation genes upregulated in the intestinal mucosa of obese rats, are downregulated by three probiotic strains. Sci. Rep. 2017, 7, 1939.
  69. Jang, H.-M.; Han, S.-K.; Kim, J.-K.; Oh, S.-J.; Jang, H.-B.; Kim, D.-H. Lactobacillus sakei alleviates high-fat-diet-induced obesity and anxiety in mice by inducing AMPK activation and SIRT1 expression and inhibiting gut microbiota-mediated NF-κB activation. Mol. Nutr. Food Res. 2019, 63, e1800978.
  70. Kikuchi, K.; Ben Othman, M.; Sakamoto, K. Sterilized bifidobacteria suppressed fat accumulation and blood glucose level. Biochem. Biophys. Res. Commun. 2018, 501, 1041–1047.
  71. Xue, L.; He, J.; Gao, N.; Lu, X.; Li, M.; Wu, X.; Liu, Z.; Jin, Y.; Liu, J.; Xu, J.; et al. Probiotics may delay the progression of nonalcoholic fatty liver disease by restoring the gut microbiota structure and improving intestinal endotoxemia. Sci. Rep. 2017, 7, 45176.
  72. Rios, J.L.; Bomhof, M.R.; Reimer, R.A.; Hart, D.A.; Collins, K.H.; Herzog, W. Protective effect of prebiotic and exercise intervention on knee health in a rat model of diet-induced obesity. Sci. Rep. 2019, 9, 3893.
  73. Chen, Q.; Liu, M.; Zhang, P.; Fan, S.; Huang, J.; Yu, S.; Zhang, C.; Li, H. Fucoidan and galactooligosaccharides ameliorate high-fat diet-induced dyslipidemia in rats by modulating the gut microbiota and bile acid metabolism. Nutrition 2019, 65, 50–59.
  74. Li, K.; Zhang, L.; Xue, J.; Yang, X.; Dong, X.; Sha, L.; Lei, H.; Zhang, X.; Zhu, L.; Wang, Z.; et al. Dietary inulin alleviates diverse stages of type 2 diabetes mellitus via anti-inflammation and modulating gut microbiota in db/db mice. Food Funct. 2019, 10, 1915–1927.
  75. Neyrinck, A.M.; Van Hée, V.F.; Piront, N.; De Backer, F.; Toussaint, O.; Cani, P.D.; Delzenne, N.M. Wheat-derived arabinoxylan oligosaccharides with prebiotic effect increase satietogenic gut peptides and reduce metabolic endotoxemia in diet-induced obese mice. Nutr. Diabetes 2012, 2, e28.
  76. Campbell, C.L.; Yu, R.; Li, F.; Zhou, Q.; Chen, D.; Qi, C.; Yin, Y.; Sun, J. Modulation of fat metabolism and gut microbiota by resveratrol on high-fat diet-induced obese mice. Diabetes. Metab. Syndr. Obes. 2019, 12, 97–107.
  77. Masumoto, S.; Terao, A.; Yamamoto, Y.; Mukai, T.; Miura, T.; Shoji, T. Non-absorbable apple procyanidins prevent obesity associated with gut microbial and metabolomic changes. Sci. Rep. 2016, 6, 31208.
  78. López, P.; Sánchez, M.; Perez-Cruz, C.; Velázquez-Villegas, L.A.; Syeda, T.; Aguilar-López, M.; Rocha-Viggiano, A.K.; Del Carmen Silva-Lucero, M.; Torre-Villalvazo, I.; Noriega, L.G.; et al. Long-term genistein consumption modifies gut microbiota, improving glucose metabolism, metabolic endotoxemia, and cognitive function in mice fed a high-fat diet. Mol. Nutr. Food Res. 2018, 62, e1800313.
  79. Kaliannan, K.; Robertson, R.C.; Murphy, K.; Stanton, C.; Kang, C.; Wang, B.; Hao, L.; Bhan, A.K.; Kang, J.X. Estrogen-mediated gut microbiome alterations influence sexual dimorphism in metabolic syndrome in mice. Microbiome 2018, 6, 205.
  80. Cho, S.-Y.; Kim, J.; Lee, J.H.; Sim, J.H.; Cho, D.; Bae, I.; Lee, H.; Seol, M.A.; Shin, H.M.; Kim, T.-J.; et al. Modulation of gut microbiota and delayed immunosenescence as a result of syringaresinol consumption in middle-aged mice. Sci. Rep. 2016, 6, 39026.
  81. Zhu, Z.; Zhu, B.; Sun, Y.; Ai, C.; Wang, L.; Wen, C.; Yang, J.; Song, S.; Liu, X. Sulfated polysaccharide from sea cucumber and its depolymerized derivative prevent obesity in association with modification of gut microbiota in high-fat diet-fed mice. Mol. Nutr. Food Res. 2018, 62, e1800446.
  82. Zhu, Z.; Zhu, B.; Sun, Y.; Ai, C.; Wu, S.; Wang, L.; Song, S.; Liu, X. Sulfated polysaccharide from sea cucumber modulates the gut microbiota and its metabolites in normal mice. Int. J. Biol. Macromol. 2018, 120, 502–512.
  83. Hu, S.; Wang, J.; Xu, Y.; Yang, H.; Wang, J.; Xue, C.; Yan, X.; Su, L. Anti-inflammation effects of fucosylated chondroitin sulphate from acaudina molpadioides by altering gut microbiota in obese mice. Food Funct. 2019, 10, 1736–1746.
  84. Liu, F.; Zhang, N.; Li, Z.; Wang, X.; Shi, H.; Xue, C.; Li, R.W.; Tang, Q. Chondroitin sulfate disaccharides modified the structure and function of the murine gut microbiome under healthy and stressed conditions. Sci. Rep. 2017, 7, 6783.
  85. Everard, A.; Geurts, L.; Van Roye, M.; Delzenne, N.M.; Cani, P.D. Tetrahydro iso-alpha acids from hops improve glucose homeostasis and reduce body weight gain and metabolic endotoxemia in high-fat diet-fed mice. PLoS ONE 2012, 7, e33858.
  86. Wang, S.; Huang, X.; Zhang, P.; Wang, H.; Zhang, Q.; Yu, S.; Yu, Y. Chronic rhein treatment improves recognition memory in high-fat diet-induced obese male mice. J. Nutr. Biochem. 2016, 36, 42–50.
  87. Mei, X.; Zhang, X.; Wang, Z.; Gao, Z.; Liu, G.; Hu, H.; Zou, L.; Li, X. Insulin sensitivity-enhancing activity of phlorizin Is associated with lipopolysaccharide decrease and gut microbiota changes in obese and type 2 diabetes (db/db) mice. J. Agric. Food Chem. 2016, 64, 7502–7511.
  88. Kang, C.; Wang, B.; Kaliannan, K.; Wang, X.; Lang, H.; Hui, S.; Huang, L.; Zhang, Y.; Zhou, M.; Chen, M.; et al. Gut microbiota mediates the protective effects of dietary capsaicin against chronic low-grade Inflammation and associated obesity induced by high-fat diet. MBio 2017, 8, 1–14.
  89. Guo, X.; Tang, R.; Yang, S.; Lu, Y.; Luo, J.; Liu, Z. Rutin and its combination with Inulin attenuate gut dysbiosis, the inflammatory status and endoplasmic reticulum stress in paneth cells of obese mice induced by high-fat diet. Front. Microbiol. 2018, 9, 2651.
  90. Wang, J.; Wang, Z.; Li, B.; Qiang, Y.; Yuan, T.; Tan, X.; Wang, Z.; Liu, Z.; Liu, X. Lycopene attenuates western-diet-induced cognitive deficits via improving glycolipid metabolism dysfunction and inflammatory responses in gut-liver-brain axis. Int. J. Obes. 2018.
  91. Nagata, N.; Xu, L.; Kohno, S.; Ushida, Y.; Aoki, Y.; Umeda, R.; Fuke, N.; Zhuge, F.; Ni, Y.; Nagashimada, M.; et al. Glucoraphanin ameliorates obesity and insulin resistance through adipose tissue browning and reduction of metabolic endotoxemia in mice. Diabetes 2017, 66, 1222–1236.
  92. Anhê, F.F.; Nachbar, R.T.; Varin, T.V.; Trottier, J.; Dudonné, S.; Le Barz, M.; Feutry, P.; Pilon, G.; Barbier, O.; Desjardins, Y.; et al. Treatment with camu camu (Myrciaria dubia) prevents obesity by altering the gut microbiota and increasing energy expenditure in diet-induced obese mice. Gut 2018, 1–12.
  93. Anhê, F.F.; Roy, D.; Pilon, G.; Dudonné, S.; Matamoros, S.; Varin, T.V.; Garofalo, C.; Moine, Q.; Desjardins, Y.; Levy, E.; et al. A polyphenol-rich cranberry extract protects from diet-induced obesity, insulin resistance and intestinal inflammation in association with increased Akkermansia spp. population in the gut microbiota of mice. Gut 2015, 64, 872–883.
  94. Gu, Y.; Yu, S.; Park, J.Y.; Harvatine, K.; Lambert, J.D. Dietary cocoa reduces metabolic endotoxemia and adipose tissue inflammation in high-fat fed mice. J. Nutr. Biochem. 2014, 25, 439–445.
  95. Sánchez-Tapia, M.; Aguilar-López, M.; Pérez-Cruz, C.; Pichardo-Ontiveros, E.; Wang, M.; Donovan, S.M.; Tovar, A.R.; Torres, N. Nopal (Opuntia ficus indica) protects from metabolic endotoxemia by modifying gut microbiota in obese rats fed high fat/sucrose diet. Sci. Rep. 2017, 7, 4716.
  96. Zhang, Z.; Li, D.; Tang, R. Changes in mouse gut microbial community in response to the different types of commonly consumed meat. Microorganisms 2019, 7, 76.
  97. Peng, J.; Leng, J.; Tian, H.; Yang, T.; Fang, Y.; Feng, Q.; Zhao, Y.; Hu, Y.-Y. Geniposide and chlorogenic acid combination ameliorates non-alcoholic steatohepatitis involving the protection on the gut barrier function in mouse induced by high-fat diet. Front. Pharmacol. 2018, 9, 1399.
  98. Han, L.; Li, T.; Du, M.; Chang, R.; Zhan, B.; Mao, X. Beneficial effects of potentilla discolor bunge water extract on inflammatory cytokines release and gut microbiota in high-fat diet and streptozotocin-induced type 2 diabetic mice. Nutrients 2019, 11, 670.
  99. Chang, C.; Lin, C.; Lu, C.; Martel, J.; Ko, Y.; Ojcius, D.M.; Tseng, S.; Wu, T.; Chen, Y.M.; Young, J.D.; et al. Ganoderma lucidum reduces obesity in mice by modulating the composition of the gut microbiota. Nat. Commun. 2015, 6, 7489.
  100. Qiu, P.; Dong, Y.; Zhu, T.; Luo, Y.-Y.; Kang, X.-J.; Pang, M.-X.; Li, H.-Z.; Xu, H.; Gu, C.; Pan, S.-H.; et al. Semen hoveniae extract ameliorates alcohol-induced chronic liver damage in rats via modulation of the abnormalities of gut-liver axis. Phytomedicine 2019, 52, 40–50.
  101. Zhang, Y.; Tang, K.; Deng, Y.; Chen, R.; Liang, S.; Xie, H.; He, Y.; Chen, Y.; Yang, Q. Effects of shenling baizhu powder herbal formula on intestinal microbiota in high-fat diet-induced NAFLD rats. Biomed. Pharmacother. 2018, 102, 1025–1036.
  102. Zhang, C.; Li, S.; Yang, L.; Huang, P.; Li, W.; Wang, S.; Zhao, G.; Zhang, M.; Pang, X.; Yan, Z.; et al. Structural modulation of gut microbiota in life-long calorie-restricted mice. Nat. Commun. 2013, 4, 2163.
  103. Fabbiano, S.; Suárez-Zamorano, N.; Chevalier, C.; Lazarević, V.; Kieser, S.; Rigo, D.; Leo, S.; Veyrat-Durebex, C.; Gaïa, N.; Maresca, M.; et al. Functional gut microbiota remodeling contributes to the caloric restriction-induced metabolic improvements. Cell Metab. 2018, 28, 907–921.
  104. Bergmann, K.R.; Liu, S.X.L.; Tian, R.; Kushnir, A.; Turner, J.R.; Li, H.-L.; Chou, P.M.; Weber, C.R.; De Plaen, I.G. Bifidobacteria stabilize claudins at tight junctions and prevent intestinal barrier dysfunction in mouse necrotizing enterocolitis. Am. J. Pathol. 2013, 182, 1595–1606.
  105. Ewaschuk, J.B.; Diaz, H.; Meddings, L.; Diederichs, B.; Dmytrash, A.; Backer, J.; Looijer-van Langen, M.; Madsen, K.L. Secreted bioactive factors from Bifidobacterium infantis enhance epithelial cell barrier function. Am. J. Physiol. Gastrointest. Liver Physiol. 2008, 295, G1025–G1034.
  106. Wang, H.; Zhang, W.; Zuo, L.; Zhu, W.; Wang, B.; Li, Q.; Li, J. Bifidobacteria may be beneficial to intestinal microbiota and reduction of bacterial translocation in mice following ischaemia and reperfusion injury. Br. J. Nutr. 2013, 109, 1990–1998.
  107. Feng, Y.; Wang, Y.; Wang, P.; Huang, Y.; Wang, F. Short-Chain fatty acids manifest stimulative and protective effects on intestinal barrier function through the inhibition of NLRP3 inflammasome and autophagy. Cell. Physiol. Biochem. 2018, 49, 190–205.
More
Information
Contributors MDPI registered users' name will be linked to their SciProfiles pages. To register with us, please refer to https://encyclopedia.pub/register : ,
View Times: 542
Revisions: 2 times (View History)
Update Date: 25 Aug 2021
1000/1000