Cervantes-Tolentino M, Beltrán-Campos V, Padilla-Raygoza N. Influence of Lactobacillus Casei Shirota Strain on Body Composition: A Review. Biomed Pharmacol J 2020;13(1).
Manuscript received on :07-01-2020
Manuscript accepted on :26-02-2020
Published online on: 13-03-2020
Plagiarism Check: Yes
Reviewed by: Rajendra Kumar Jangde
Second Review by: Daya Shankar Gautam orcid
How to Cite    |   Publication History
Views Views: (Visited 1,118 times, 1 visits today)   Downloads PDF Downloads: 440

Miriam Cervantes-Tolentino1, Vicente Beltrán-Campos2 and Nicolás Padilla-Raygoza3*

1Division of Health Sciences and Engineering, Campus Celaya-Salvatierra, University of Guanajuato, Celaya, Mexico

2Department of Clinical Nursing, Division of Health Sciences and Engineering, Campus Celaya-Salvatierra, University of Guanajuato, Celaya, México

3School of Medicine, University of Celaya, Celaya, México

Corresponding Author E-mail : npadilla@udec.edu.mx

DOI : https://dx.doi.org/10.13005/bpj/1865

Abstract

A review of the literature is made about the most relevant aspects of the effects of the administration of prebiotics and probiotics on body composition, mainly on fat mass and on the Body Mass Index, as well as the effect they have on the inflammation that causes obesity in humans and that results in unfavorable health outcomes. There is evidence that suggests that the administration of prebiotics and probiotics, support the fight against obesity.

Keywords

Inflammation; Obesity; Prebiotics

Download this article as: 
Copy the following to cite this article:

Cervantes-Tolentino M, Beltrán-Campos V, Padilla-Raygoza N. Influence of Lactobacillus Casei Shirota Strain on Body Composition: A Review. Biomed Pharmacol J 2020;13(1).

Copy the following to cite this URL:

Cervantes-Tolentino M, Beltrán-Campos V, Padilla-Raygoza N. Influence of Lactobacillus Casei Shirota Strain on Body Composition: A Review. Biomed Pharmacol J 2020;13(1). Available from: https://bit.ly/2xzHVDu

Introduction

Obesity

Obesity is defined as “an abnormal or excessive accumulation of fat that poses a health risk”, which can be determined through the calculation of the Body Mass Index (BMI), and must be ≥30 kg / m2 [1].

Because obesity represents an imbalance between intake and energy expenditure, this is therefore a consequence of the combination of diets with high caloric density, as well as zero or low physical activity [2], leading to an increase in adipose tissue, which in turn generates a state of mild chronic inflammation [3].

Afshin et al. [4], indicate that obesity represents a public health problem,  as it is considered a chronic disease [3], with a prevalence that is constantly increasing; according to Ng et al. [5], of 857 million people with obesity and overweight to 2.1 billion. It should also be noted that this is associated with other diseases such as insulin resistance, type 2 diabetes mellitus, heart disease and cancer [6,7].

According to data from the National Survey of Health and Nutrition (ENSANUT) 2016, in Mexico, the prevalence of overweight and obesity in adults 20 years of age or older, went from 71.2% in 2012 to 72.5% in 2016. However, this increase in 1.3 percentage points was not statistically significant. Meanwhile, the prevalence of overweight, obesity and morbid obesity were higher in the female sex. Similarly, although the combined prevalence of overweight and obesity does not differ much in urban areas (72.9%) than in rural areas (71.6%), the prevalence of overweight was 4.5 percentage points higher in rural areas, while the prevalence of obesity it was 5.8 percentage points higher in urban areas [8].

The inflammatory process in obesity

Adipose tissue, in addition to having thermoregulatory function, giving mechanical protection to organs and storing triglycerides during caloric excesses, or, releasing them under fasting conditions, also acts as an endocrine organ [9,10], because it secretes peptides called adipokines, which are cytosines that when synthesized by this type of tissue receive that name [11]. In addition, they can be pro-inflammatory in nature such as leptin [12], monocyte chemoattractant protein 1 (MCP-1), tumour necrosis factor alpha (TNF-α) and interleukin (IL) -6, causing in the organism a low-grade inflammatory state that favors the progression of obesity; while on the other, that same tissue, inhibits the secretion of anti-inflammatory adipokines, such as adiponectin, whose function is to protect from the complications caused by such disease [13-16].

In contrast, the considerable increase in adipose tissue that results from obesity [17], leads to deterioration of its function, since the adipose hypertrophy coming from [9], generates an increase in the levels of pro-inflammatory adipokines [18], while adiponectin and other anti-inflammatory adipokines, such as interleukin (IL) -10, are reduced [9,18].

It should also be mentioned that adipokines also intervene in other specific processes, such as leptin, which regulates appetite and satiety, glucose metabolism, insulin sensitivity and atherogenesis [19,20]. However, because in obesity serum concentrations of proinflammatory factors increase, (IL) -6 and (TNF-α) decrease insulin sensitivity [12]; while in the case of leptin, although at the level of the hypothalamus it increases the synthesis of anorexigenic peptides and decreases that of orexigenic peptides, this serum increase prevents food intake and hyperglycaemia from being reduced [21], so that these changes explain the link between obesity and other metabolic and cardiovascular comorbidities in addition to inflammation [22,23].

Regarding adiponectin, beyond having anti-inflammatory action, they also have anti-apoptotic and insulin sensitization action [24]. However, several studies indicate that in people with obesity their plasma levels are low [25-27], since it has been seen that in fat cells in vitro , some hormones associated with insulin resistance, such as TNF-α and IL-6, regulate their expression and secretion [28], so, with based on Ryo et al. [29], have a negative relationship with the accumulation of visceral fat [30].

Thus, the fact of having a reduction in the body weight of people with obesity, has been associated with an increase in plasma levels of adiponectin, as well as a plasma reduction in markers of inflammation such as IL-6 [31].

Signalling Mechanisms Associated with Obesity Inflammation

The mechanism of inflammatory signalling may arise due to the presence of extracellular mediators such as adipokines associated with inflammation and excess lipids, or they may be the product of some intracellular mediators such as endoplasmic reticulum stress or the abundant production of Species Oxygen Reagents (ERO). Meanwhile, regardless of the type, in the end both mediators lead to two paths, because on the one hand they activate inflammatory mediators through transcriptional regulations, and on the other they directly inhibit insulin signalling [32].

Although, at the level of the endoplasmic reticulum, obesity overloads the ability of cells to perform protein folding, generating a stress mechanism that involves the activation of the N-terminal c-Jun kinase 1 (JNK1) and the inhibitor of kappa kinase β (IKK2) [33, 34].

In turn, Koop et al. [35], mention that IKK2 is activated and phosphorylates the inhibitor of NF-κB (IκB), leading to its degradation and the inhibition of nuclear factor Kapa B (NF-κB) in basal conditions . However, the latter, when released into the cell nucleus, stimulates the transcription of some inflammation mediators, such as TNF-α and IL-6 [36], which, according to Ferrer et al. [37], are also known as immunological mediators. Also, in addition to intervening in the inflammatory response, the transcription factor NF-κB also participates in the immune response [36], while suppressing adiponectins and type 4 glucose transporter proteins (GLUT-4) [38].

On the other hand, there are several experiments with mice in which it has been seen that, at the level of the hypothalamus, the activation of JNK1 in Agouti-related Peptides (AgRP) produces neuronal and systemic conditions in leptin, as well as induces weight gain and adiposity after hyperphagia. In contrast, the activation of IKK2 reduces insulin signaling in AgRP neuropeptides, leading to an alteration in glucose homeostasis, which is why both kinases play an important role in the cellular and systemic resistance of insulin (IKK2) and leptin (JNK1) associated with obesity [39].

Another mechanism that initially leads to the activation of inflammatory pathways is oxidative stress [40], which, at the mitochondrial level, generates an increase in the production of reactive oxygen species, therefore, in people with obesity , the obtaining of ERO is elevated as a result of the increase in glucose metabolism {41, 42], since the endothelial cells of adipose tissue introduce large amounts of glucose through their respective glucose transporters, GLUT 4 [40].

Thus, in these hyperglycaemic conditions, excessive production of ROS causes oxidative damage in endothelial cells that activates inflammatory signalling [40], allowing adipose tissue to attract other inflammatory cells such as macrophages [41], which are also part of it [43], and whose function under normal conditions is to participate in the innate immune response [32], and express certain adipocyte genetic products such as the receptor Nuclear Peroxisome-Proliferator-Activated Gamma (PPARγ) and Fatty Acid Conveyor Protein (FABP), especially those of the aP2 type (FABP-aP2) [44,45], said proteins regulating the accumulation of cholesterol in macrophages and the accumulation of lipids in adipocytes, which modulates therefore, atherosclerosis and insulin resistance [32]. However, the consequent inflammation of adipose tissue associated with obesity causes macrophages to absorb and store lipids to become atherosclerotic foam cells as well as contribute, by themselves or in conjunction with adipocytes, to the production of inflammation mediators [32].

Finally, other pathways that are also associated with insulin inhibition as a consequence of inflammation, are those regulated by the iNOS proteins (nitric oxide synthetase) and those that belong to the family known as Cytosine Signaling Suppressor (SOCS) [32], which means that expression is mediated by cytosines [46]). So that, in the case of iNOS, this is induced in skeletal muscle and fat after the action of pro-inflammatory cytosines, so that excess nitric oxide impairs the function of the β cells of the pancreas and the action of insulin in muscle cells [47, 48].

In contrast, SOCS, including SOCS1, SOCS3 and SOCS6, inhibit insulin signaling through proteosomal degradation of insulin receptors (IR) such as IRS-1 and IRS-2 [46,49], or else, inhibit it by hindering tyrosine phosphorylation in such receptors. However, the increase in SOCS is not only linked to obesity, but also due to endotoxemia induced by a compound called lipopolysaccharide (LPS) [50], thus being an endotoxin that It constitutes the cell membrane of most Gram-negative bacteria [7].

In this regard, it is known that LPS activates Toll-like receptors (TLR), which participate in the innate immune response. As a result of the union between LPS and TLR-4, an intense inflammatory response is obtained that leads the organism to a state of inflammation [51,52.53], because, based on the provisions of Muzio, et al. [54], this activation leads to the translocation of NF-κB in the cell nucleus, thus initiating the transcription of IL-6 and TNF- α, since TLRs also induce degradation of IκB once they trigger NF-κB [52].

Similarly, TLR-4 receptors can also be activated by saturated fatty acids that are ingested through the diet, thereby inducing inflammation signalling after allowing the synthesis of the same pro-inflammatory adipokines (IL-6 and TNF-α) associated with insulin resistance and increased adiposity, both in macrophages and in fat cells [55].

In summary, it could be said that the intestinal microbiota has the ability to promote a low-grade inflammatory state, favour insulin resistance and increase cardiovascular risk through the mechanisms described above, including its exposure to LPS [51].

The Role of Lipids in the Regulation of Inflammatory Signalling Pathways

Although the consequent obesity-associated hyperlipidaemia leads to the production of fatty acid metabolites that activate the inflammatory response and inhibit insulin signalling [56], some intracellular lipids such as liver X receptor (LXR) and PPAR nuclear receptor families counteract inflammatory processes, since they inhibit the expression of genes associated with inflammation in macrophages and adipocytes through the suppression of NF-κB that occurs in the nucleus. In addition, both transcription factors are responsible for promoting metabolism, nutrient transport and cholesterol and lipid flow stored in macrophages and adipocytes, respectively [32].

Despite the above, once the inflammatory pathways are underway, the activity of LXR and PPAR is influenced by the aP2 fatty acid transporter protein (FABP-aP2) found in the adipocyte cytoplasm [57], which means that, far from exerting its beneficial action, this type of sequestration protein is probably linked to said transcription factors to favour the state of inflammation [32].

Intestinal Microbiota and its Relationship with Obesity

Although they seem indistinct terms, there is a difference between the definition of microbiota and the microbiome. Although, the first refers to a community made up of several types of organisms that are present in an environmental habitat, and may be of the Archaea, Bacteria or Eukarya domain, as well as viruses. Meanwhile, the microbiome constitutes the total of microorganisms that are found in an environmental habitat, including their functions, genes and metabolites, therefore, the composition of the microbiome depends on the region of the body in which it is present [7,58,59].

Thus, the intestinal microbiota is the result of a symbiosis between microbial species of any domain [58]. Meanwhile, the intestinal microbiome is made up of more than 100 billion bacteria that colonize the human intestine, predominantly the Firmicutes and Bacteroidetes [60,61], which represent those anaerobic bacteria whose purpose is to ferment non-digestible carbohydrates [62].

Among the benefits provided by the intestinal microbiota to the host is to regulate its immune function, as it provides protection against pathogenic bacteria and chronic inflammation [63].

On the other hand, the intestinal microbiota also regulates energy homeostasis, as it is involved in the extraction of energy from food, obtaining compounds that can cross the intestinal barrier and the production of vitamins and hormones [58, 64, 65]. However, both the function and the microbial composition can be impaired (dysbiosis) by intrinsic factors, such as intestinal motility, pH, antibacterial proteins and mucus, as well as by extrinsic factors, some of these being the genetic determinants of host, medications, such as antibiotics, and diet [62, 66-68].

Meanwhile, when energy homeostasis is affected, the development of obesity is encouraged [64], which is why Parséus et al. [69] point to the intestinal microbiota as another environmental factor that triggers this disease, since there are several studies that have shown that in people with obesity, microbial diversity is reduced, which leads them to suffer metabolic complications that depend on both genetic effects and dietary patterns [60]. However, eating a high-fat diet not only modulates the intestinal microbiota, but also the plasma concentration of LPS [70], since following a diet with these characteristics increases the amount of bacterial LPS, modifying in turn the composition of the microbiota [71].

Regarding the fermentation metabolites produced by Firmicutes and Bacteroidetes in the lumen of the colon, it should be noted that these are known as short chain fatty acids (SCFA) and include acetate, propionate and butyrate, absorbing the first two directly to the portal circulation; while the third is used by the colonocytes as a source of energy. In fact, it is estimated that 10% of the energy used by the body comes from said fermentation [62, 72,73], meanwhile, how these metabolites are involved in the release of ghrelin, a molecule that acts at the brain level in the regulation of appetite and insulin action, the reduction in bacterial taxa that produce SCFA is associated with the development of obesity [58].

On the other hand, with regard to the microbial composition of the intestine, it is known that it differs between healthy individuals and those with excess adipose tissue, insulin resistance and dyslipidemia, and there may even be a decrease in its richness [74]. In this regard, scientific evidence indicates that the prevalence of the Bacteroidetes edge is lower in people with obesity. However, this proportion may increase as weight is lost after following a hypocaloric diet. Meanwhile, the Firmicutes edge increases proportionally, which is associated with a greater presence of enzymes for the fermentation of non-digestible carbohydrates [75], relating the above with higher levels of energy collection through diet [76].

After all of the above, it is likely that the low-grade inflammatory state attributed to obesity is due to changes in the microbial constitution, increased intestinal permeability and metabolic endotoxemia [59], which is why which scientific evidence has revealed that probiotics could be the organic component that helps regulate the intestinal microbiota [7, 77], because through them it is possible to increase the amount of bacteria that produce short chain fatty acids, adiposity is reduced and the production of some metabolites such as lipopolysaccharides decreases, as well as the inflammation that the latter cause in the body [78].

The use of Probiotics and Prebiotics as a Modulating Therapy of the Intestinal Microbiota in the Treatment of Obesity

So far we know that, because the diet is a factor that determines the composition of the intestinal microbiota [58], ingest large amounts of fats, in addition to increasing the edge Firmicutes and decrease Bacteroidetes, increases the permeability of the intestine, resulting in a bacterial translocation and an increase in LPS, which affects weight gain, morphological alterations of adipose tissue, insulin resistance and inflammation, all of which is characteristic of obesity [79-82]. After the above, it has been shown that after colonizing the intestine of germ-free mice with intestinal microbiota of conventionally raised mice, their body fat increases by up to 60% compared to germ-free control mice, which implies greater absorption of monosaccharides that induce de novo lipogenesis in the liver [83].

In this regard, it could be said that, in addition to following a diet high in fats and sugars, colonizing the intestine with conventional microbiota promotes obesity, since a study in mice deduced that those that were free of germs were protected against the development of said disease despite following a diet with such characteristics [84]. This could be due to elevated levels of Fasting Induced Adipose Factor (FIAF) [84], also known as angiopoietin type 4 protein, which is produced in the liver, intestine, white fat and brown fat, and inhibits lipoprotein lipase (LPS) [83], whose function is to regulate the release of fatty acids belonging to lipoproteins rich in triglycerides of muscle, heart and fat [85]. However, Bergö, et al. [86], mention that, because LPS is inhibited, this translates into a greater absorption of fatty acids, as well as a greater accumulation of adipocyte triglycerides. However, it should be noted that everything described above is only carried out in the intestinal epithelium and not in the liver or in both types of fat [84].

Thus, there are several studies that have shown that, in contrast to the control groups, those who have received probiotic supplementation have presented significant reductions in anthropometric measurements, such as body weight, BMI, waist circumference, Fat mass and fat percentage, so these could be used both in the prevention and treatment of obesity [78]. because the intestinal microbiota is an environmental factor that promotes fat storage [83].

In particular, those bacteria that have a beneficial effect on health are known as probiotics, provided they are administered in adequate amounts [87]. In fact, according to O’Toole, et al. [88], among the most frequently used species are the genus Lactobacillus, Bifidobacterium and Saccharomyces, although other genera such as Bacillus, Propionibacterium, StreptococYYcus and Escherichia, are used.

On the other hand, it has been established that prebiotics are a type of non-digestible dietary fiber that includes oligosaccharides or short polysaccharides with inulin, oligofructose, galactofructose, galacto-oligosaccharides and xyl-oligosaccharide, which after fermentation generate an increase in the amount of beneficial bacteria [7]. In this regard, it is believed that especially inulin-type fructans, could contribute to the treatment of obesity after fermentation and promote the growth of beneficial bacteria, resulting in a change in the composition and / or activity of the intestinal microbiota in favor of health [7, 89, 90], such as weight reduction and improvement in lipid and glucose levels after studies in rats [91].

As a result, inulin-type fructans containing short chain oligosaccharides increase the levels of proglucagon mRNA and glucagon-1-like peptide (GLP-1) in the proximal colon, allowing such prebiotics to ferment and reduce intake of food, fat mass, body weight and homeostasis of inulin, as long as there is a functional GLP-1 receptor (GLP-1R), which in turn also depends on the type of bacteria that colonize the intestine [91,92]. In this regard, the increase in the amount and release of GLP-1 is mediated by intestinal L cells through the factor NGN-3 and NeuroD [93]. These cells allow the expression of the proglucagon gene, which could occur due to the action of SCFA, especially butyrate [91], thus synthesizing the active form of GLP-1, as well as PYY, another peptide that is also secreted by L cells and that is involved in the regulation of food intake by being an anorexinogenic hormone that reduces weight gain after inhibiting such intake [58, 91]. In fact, in a study with experimental animals, it was confirmed that after administering high-fat diets with oligofructose, a kind of inulin fructane, proglucagon expression increased in the proximal colon, favoring the beneficial effect that the prebiotic has on glycemia, the development of fat mass and weight gain [91, 94]. On the other hand, it was observed that, after following an oligofructose treatment, the number of Kupffer cells increased, thereby increasing the ability to eliminate proinflammatory agents such as LPS [95]. Likewise, it was also shown that the PYY portal increased, while serum ghrelin levels decreased, which could be related to the satiating effect of the prebiotic [96], remember that ghrelin, when synthesized mainly in the stomach, increases appetite and stimulates food intake, favoring weight gain and adiposity ( Santos-Marcos, Perez-Jimenez, & Camargo, 2019).

In addition to the above, prebiotics are also related to the increase of glucagon-2-like peptide (GLP-2), allowing the intestinal barrier to be restored, which translates into improvements in its permeability, which is also associated with a decrease in proinflammatory cytosine levels, such as IL-6 and MCP-1 [58].

Finally, it should be mentioned that in another study conducted with rats, where in addition to administering the same type of diet and prebiotic, Bifidobacterium strains were added, a positive correlation was obtained with the improvement in glucose tolerance and the normalization of inflammatory tone , which included both reductions in endotoxemia and in proinflammatory cytokines of adipose tissue and plasma [81], although ultimately, both Lactobacilli and Bifidobacteria can have the same beneficial effects on the metabolism of glucose and entotoxemia after these increase after following a prebiotic treatment [60].

Conclusion

With the results detected in this review, it is concluded that the field of probiotics and prebiotics is promising for the management of body composition and obesity.

Now, the way is to carry out multiple clinical studies in different populations to show if they have clinical efficacy in reducing body composition and decreasing the percentage of obesity in the population.

References

  1. World Health Organization. Health topics. Obesity. 2018. Available in: https://www.who.int/topics/obesity/en/
  2. Hill JO, Wyatt HR, Peters JC. Energy and Balance and Obesity. Circulation, 2012;126(1): 126-132. doi: https://doi.org/10.1161/CIRCULATIONAHA.111.087213.
  3. Avolio E, Gualtieri P, Romano LP, Ferrano S, Di Renzo L, De Lorenzo A. Obesity and body composition in man and woman: Associated diseases and the new role of gut microbiota. Current Medical Chemistry, 2019; 26(0):1-13. doi: https://doi.org/10.2174/092986732666619032611360
  4. Afshin A, Forouzanfar MH, Reitsma MB, Sur P, Estep K, Lee A, et al. Health effects of overweight and obesity in 195 countries over 25 years. N Engl J Med. 2017;377:13–27. doi: https://doi.org/10.1056/NEJMoa1614362.
  5. Ng M, Fleming T, Robinson M, Thomson B, Graetz N, Margono C, et al. Global, regional, and national prevalence of overweight and obesity in children and adults during 1980-2013: A systematic analysis for the Global Burden of Disease Study 2013. Lancet. 2014;384:766–81. doi: https://doi.org/10.1016/s0140-6736(14)60460-8.
  6. Jung UJ, Choi MS. Obesity and Its Metabolic Complications: The Role of Adipokines and the Relationship between Obesity, Inflammation, Insulin Resistance, Dyslipidemia and Nonalcoholic Fatty Liver Disease. International Journal of Molecular Sciences, 2014;15(4): 6184-223. doi: https://doi.org/10.3390/ijms15046184
  7. Marchesi JR, Adams DH, Fava F, Hermes GD, Hirschfield GM, Hold G, et al. The gut microbiota and host health: a new clinical frontier. Gut, 2016; 65(2): 330-9. doi: https://doi.org/10.1136/gutjnl-2015-309990
  8. National Institute of Public Health., Ministry of Health. Mid-Way Health and Nutrition Survey. Final Results Report. Cuernavaca, National Institute of Public Health. Available in: http://transparencia.insp.mx/2017/auditorias-insp/12701_Resultados_Encuesta_ENSANUT_MC2016.pdf
  9. Klöting N, Blüher M. Adipocyte dysfunction, inflammation and metabolic syndrome. Reviews in Endocrine and Metabolic Disorders, 2014; 15(4): 277-87. doi: https://doi.org/10.1007/s11154-014-9301-0
  10. Blüher M. Adipose tissue dysfunction contributes to obesity related metabolic diseases. Best Practice and Research: Clinical Endocrinology and Metabolism, 2013; 27(2): 163-77. doi: https://doi.org/10.1016/j.beem.2013.02.005
  11. Toan NL, Van Hoan N, Cuong DV, Dung NV, Dung PT, Hang NT, et al. Adipose tissue-derived cytokines and their correlations with clinical characteristics in Vietnamese patients with type 2 diabetes mellitus. Diabetology & Metabolic Syndrome, 2018; 10(1041): 1-14. doi: https://doi.org/10.1186/s13098-018-0343-4
  12. Fasshauer M, Blüher M. Adipokines in health and disease. Trends in Pharmatological Sciences, 2015; 36(7): 461-70. doi: https://doi.org/10.1016/j.tips.2015.04.014
  13. Ouchi N, Parker JL, Lugus JJ, Walsh K. Adipokines in inflammation and metabolic disease. Nature Reviews Immunology, 2011; 11(2): 85-97. doi: https://doi.org/10.1038/nri2921
  14. Ouchi N, Kihara S, Funahashi T, Matsuzawa Y, Walsh K. Obesity, adiponectin and vascular inflammatory disease. Current Opinion in Lipidology, 2003; 14(6): 561-6. doi: https://doi.org/10.1097/00041433-200312000-00003
  15. Ouchi N, Higuchi A, Ohashi K, Oshima Y, Gokce N, Shibata R, et al. Sfrp5 is an anti-inflammatory adipokine that modulates metabolic dysfunction in obesity. Science, 2010; 329(5990): 454-7. doi: https://doi.org/10.1126/science.1188280
  16. Enomoto T, Ohashi K, Shibata, R, Higuchi A, Maruyama S, Izumiya Y, et al. Adipolin/C1qdc2/CTRP12 protein functions as an adipokine that improves glucose metabolism. Journal of Biological Chemistry, 2011; 286: 34552-8. doi: https://doi.org/10.1074/jbc.M111.277319
  17. Tenorio-Jiménez C, Martínez-Ramírez MJ, Del Castillo-Códigos I, Arraiza-Irigoyen C, Tercero-Lozano M, Camacho J, et al. Lactobacillus reuteri V3401 Reduces Inflammatory Biomarkers and Modifies the Gastrointestinal Microbiome in Adults with Metabolic Syndrome: The PROSIR Study. Nutrients, 2019; 11(8): 1-14. doi: https://doi.org/10.3390/nu11081761
  18. Skurk T, Alberti-Huber C, Herder C, Hauner H. Relationship between adipocyte size and adipokine expression and secretion. Journal of Clinical Endocrinology and Metabolism, 2007; 92(3): 1023-33. doi: https://doi.org/10.1210/jc.2006-1055
  19. Blüher, M. (2014). Adipokines – removing road blocks to obesity and diabetes therapy. Molecular Metabolism, 2014;3(3): 230-40. doi: https://doi.org/10.1016/j.molmet.2014.01.005
  20. Rexford SA, Flier JS. Leptin. Annual Review of Physiology, 2000; 62: 413-37. doi: https://doi.org/10.1146/annurev.physiol.62.1.413
  21. Ahima RS, Flier JS. Leptin. Annual Review of Physiology, 2000; 62: 413-37. doi: https://doi.org/10.1146/annurev.physiol.62.1.413
  22. Blüher M. Importance of adipokines in glucose. Diabetes Manage, 2013; 3(5), 389-400. doi: https://doi.org/10.2217/DMT.13.35
  23. Blüher M, Mantzoros CS. From leptin to other adipokines in health and disease: Facts and expectations at the beginning of the 21st century. Metabolism: Clinical and Experimental, 2015; 64(1): 131-45. doi: https://doi.org/10.1016/j.metabol.2014.10.016
  24. Turer AT, Scherer PE. Adiponectin: Mechanistic insights and clinical implications. Diabetologia, 2012; 55(9), 2319-26. doi: https://doi.org/10.1007/s00125-012-2598-x
  25. Hotta K, Funahashi T, Bodkin NL, Ortmeyer HK, Arita Y, Hansen B, et al. Circulating concentrations of the adipocyte protein adiponectin are decreased in parallel with reduced insulin sensitivity during the progression to type 2 diabetes in rhesus monkeys. Diabetes, 2001; 50(5), 1126-33. doi: https://doi.org/10.2337/diabetes.50.5.1126
  26. Kumada MK. Association of hypoadiponectinemia with coronary artery disease in men. Arteriosclerosis, Thrombosis, and Vascular Biology, 2003; 23(1): 85-9. doi: https://doi.org/10.1161/01.ATV.0000048856.22331.50
  27. Iwashima Y, Katsuya T, Ishikawa K, Ouchi N, Ohishi M, Sugimoto K, et al. Hypoadiponectinemia is an independent risk factor for hypertension. Hypertension, 2004; 43(6): 1318-23. doi: https://doi.org/10.1161/01.HYP.0000129281.03801.4b
  28. Fasshauera, M, Klein J, Neumann S, Eszlinger M, Paschke R. Hormonal Regulation of Adiponectin Gene Expression in 3T3-L1 Adipocytes. Scienece Direct, 2002; 290(3): 1084-89. doi: https://doi.org/10.1006/bbrc.2001.6307
  29. Ryo M, Nakamura T, Kihara S, Kumada M, Shibazaki S, Takahashi M, et al. Adiponectin as a biomarker of the metabolic syndrome. Circ. J. 2004; 68: 975-981 doi: https://doi.org/10.1253/circj.68.975
  30. Ohashi K, Shibata R, Murohara T, Ouchi N. Role of anti-inflammatory adipokines in obesity-related diseases. Trends in Endocrinology & Metabolism,2014; 25(7): 348-55. doi: https://doi.org/10.1016/j.tem.2014.03.009
  31. Esposito K, Pontillo A, Di Palo C, Giugliano G, Masella M, Marfella R, et al. Effect of Weight Loss and Lifestyle Changes on Vascular Inflammatory Markers in Obese Women: A Randomized Trial. Journal of the American Medical Association, 2003; 289(14): 1799-804. doi: https://doi.org/10.1001/jama.289.14.1799
  32. Wellen KE, Hotamisligil GS. Inflammation, stress, and diabetes. The Journal of Clinical Investigation, 2005; 115(5): 1111-19. doi: https://doi.org/10.1172/JCI25102
  33. Özcan U, Cao Q, Yilmaz E, Lee AH, Iwakoshi N, Özdelen E, et al. Endoplasmic reticulum stress links obesity, insulin action, and type 2 diabetes. Science, 2004; 306(5695): 457-61. doi: https://doi.org/10.1126/science.1103160
  34. Özcan U, Yilmaz E, Özcan L, Furuhashi M, Vaillancourt E, Smith R, et al. Chemical Chaperones Reduce ER Stress and Restore Glucose Homeostasis in a Mouse Model of Type 2 Diabetes. Science, 2006; 313(5790): 1137-40. doi: https://doi.org/10.1126/science.1128294
  35. Kopp E, Ghosh S. Inhibition of NF-kappa B by sodium salicylate and aspirin. Science 1994; 265(5174): 956–959. doi: https://doi.org/10.1126/science.8052854
  36. Shoelson S, Lee J, Yuan M. Inflammation and the IKKβ/IκB/NF-κB axis in obesity- and diet-induced insulin resistance. International Journal of Obesity, 2003; 27: S49-S52. doi: https://doi.org/10.1038/sj.ijo.0802501
  37. Ferrer B, Dalmau J. Reflexiones sobre el síndrome metabólico. Acta Pediatr Esp. 2008; 66(3): 77-82. Available in: https://www.actapediatrica.com/index.php/secciones/nutricion-infantil/566-reflexiones-sobre-el-s%C3%ADndrome-metab%C3%B3lico#.Xe_w2uhKjIU
  38. Blancas-Flores G, Almanza-Pérez JC, López-Roa RI, Javier AAF, García-Macedo R, Cruz M. Obesity as an inflammatory process. Bol. Med. Hosp Infant Mex, 2010; 67: 88-97. Available in: https://pdfs.semanticscholar.org/1464/662e8efd2ff53f1405892ed0a14cfa4a7267.pdf
  39. Tsaousidou E, Paeger L, Belgardt BF, Pal M, Wunderlich CM, Brönneke H, et al. Distinct Roles for JNK and IKK Activation in Agouti-Related Peptide Neurons in the Development of Obesity and Insulin Resistance. Cell Reports, 2014; 9(4): 1495-506. doi: https://doi.org/10.1016/j.celrep.2014.10.045
  40. Brownlee M. Biochemistry and molecular cell biology of diabetic complications. Nature, 2001; 414(6865): 813-20. doi: https://doi.org/10.1038/414813a
  41. Lin Y, Berg AH, Iyengar P, Lam TK, Giacca A, Combs TP, et al. (2005). The Hyperglycemia-induced Inflammatory Response in Adipocytes. Journal of Biological Chemistry, 2005; 280(6): 4617-26. doi: https://doi.org/10.1074/jbc.M411863200
  42. Furukawa S, Fujita T, Shimabukuro M, Iwaki M, Yamada Y, Nakajima Y, et al. Increased oxidative stress in obesity and its impact on metabolic syndrome. The Journal of Clinical Investigation, 2004; 114(12): 1752-61. doi: https://doi.org/10.1172/JCI200421625
  43. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. The Journal of Clinical Investigation, 2003; 112(12): 1796-808. doi: https://doi.org/10.1172/JCI200319246
  44. Makowski L, Boord JB, Maeda K, Babaev VR, Uysal KT, Morgan MA, et al. Lack of macrophage fatty-acid–binding protein aP2 protects mice deficient in apolipoprotein E against atherosclerosis. Nature Medicine, 2001; 7(6): 699-705. doi: https://doi.org/10.1038/89076
  45. Tontonoz P, Nagy L, Alvarez JG, Thomazy VA, Evans RM. (1998). PPARγ Promotes Monocyte/Macrophage Differentiation and Uptake of Oxidized LDL. Cell, 1998; 93(2): 241-52. doi: https://doi.org/10.1016/S0092-8674(00)81575-5
  46. Mooney RA, Senn J, Cameron S, Inamdar N, Boivin LM, Shang Y, et al. Suppressors of Cytokine Signaling-1 and -6 Associate with and Inhibit the Insulin Receptor. A potential Mechanism for Cytokine-Mediated Insulin Resistance. Journal of Biological Chemistry, 2001; 276: 25889-93. doi: https://doi.org/10.1074/jbc.M010579200
  47. Shimabukuro M, Ohneda M, Lee Y, Unger RH. Role of Nitric Oxide in Obesity-induced b Cell Disease. The Journal of Clinical Investigation, 1997; 100(2): 290-5. doi: https://doi.org/10.1172/JCI119534
  48. Perreault M, Marette A. Targeted disruption of inducible nitric oxide synthase protects against obesity-linked insulin resistance in muscle. Nature Medicine, 2001; 7(10): 1138-43. doi: https://doi.org/10.1038/nm1001-1138
  49. Rui L, Yuan M, Frantz D, Shoelson S, White MF. SOCS-1 and SOCS-3 Block Insulin Signaling by Ubiquitin-mediated Degradation of IRS1 and IRS2. Journal of Biological Chemistry, 2002; 277: 42394-8. doi: https://doi.org/10.1074/jbc.C200444200
  50. Ueki K, Kondo T, Kahn CR. (2004). Suppressor of Cytokine Signaling 1 (SOCS-1) and SOCS-3 Cause Insulin Resistance through Inhibition of Tyrosine Phosphorylation of Insulin Receptor Substrate Proteins by Discrete Mechanisms. Molecular and Cellular Biology,2004; 24(12): 5434-46. doi: https://doi.org/10.1128/MCB.24.12.5434-5446.2004
  51. Creely SJ, McTernan PG, Kusminski CM, Fisher M, Da Silva NF, Khanolkar M, et al. Lipopolysaccharide activates an innate immune system response in human adipose tissue in obesity and type 2 diabetes. American Journal of Physiology – Endocrinology and Metabolism, 2007; 292(3): E740-E747. doi: https://doi.org/10.1152/ajpendo.00302.2006
  52. Kaisho T, Akira S. Toll-like receptors as adjuvant receptors. Biochimica et Biophysica Acta (BBA) – Molecular Cell Research, 2002; 1589(1): 1-13. doi: https://doi.org/10.1016/S0167-4889(01)00182-3
  53. Amar J, Burcelin R, Ruidavets JB, Cani PD, Fauvel J, Alessi MC, et al. Energy intake is associated with endotoxemia in apparently healthy men. The Americal Journal of Clinical Nutrition, 2008; 87(5): 1219-23. doi: https://doi.org/10.1093/ajcn/87.5.1219
  54. Muzio M, Polentarutti N, Bosisio D, Manoj Kumar PP, Mantovani A. Toll-like receptor family and signalling pathway. Biochem Soc Trans, 2000; 28(5): 563–6. doi: https://doi.org/10.1042/bst0280563
  55. Tsukumo DM, Carvalho-Filho MA, Carvalheira JB, Prada PO, Hirabara SM, Schenka AA, et al. Loss-of-Function Mutation in Toll-Like Receptor 4 Prevents Diet-Induced Obesity and Insulin Resistance. Diabetes, 2007; 56(8): 1986-98. doi: https://doi.org/10.2337/db06-1595
  56. Yu C, Chen Y, Cline GW, Zhang D, Zong H, Wang Y, et al. Mechanism by Which Fatty Acids Inhibit Insulin Activation of Insulin Receptor Substrate-1 (IRS-1)-associated Phosphatidylinositol 3-Kinase Activity in Muscle. The Journal of Biological Chemistry, 2002; 277(52): 50230-6 doi: https://doi.org/10.1074/jbc.M200958200
  57. Maeda K, Cao H, Kono K, Gorgun CZ, Furuhashi M, Uysal KT, et al. (2005). Adipocyte/macrophage fatty acid binding proteins control integrated metabolic responses in obesity and diabetes. Cell Metabolism, 2005; 1(2): 107-19. doi: https://doi.org/10.1016/j.cmet.2004.12.008
  58. Santos-Marcos JA, Perez-Jimenez F, Camargo A. The role of diet and intestinal microbiota in the development of metabolic syndrome. The Journal of Nutricional Biochemistry, 2019; 70: 1-27. doi: https://doi.org/10.1016/j.jnutbio.2019.03.017
  59. Icaza-Chávez M. Gut microbiota in health and disease. Revista de Gastroenterología de México (English Edition), 2013; 78(4): 240-80. doi: https://doi.org/10.1016/j.rgmxen.2014.02.009
  60. Vallianou N, Stratigou T, Christodoulatos GS, Dalamaga M. Understanding the Role of the Gut Microbiome and Microbial Metabolites in Obesity and Obesity-Associated Metabolic Disorders: Current Evidence and Perspectives. Current Obesity Reports, 2019; 8(3): 317-32. doi: https://doi.org/10.1007/s13679-019-00352-2
  61. Johnson EL, Heaver SL, Walters WA, Ley RE. Microbiome and metabolic disease: revisiting the bacterial phylum Bacteroidetes. Journal of Molecular Medicine (Berlin, Germany), 2017; 95(1): 1-8. doi: https://doi.org/10.1007/s00109-016-1492-2
  62. Andoh A. Physiological Role of Gut Microbiota for Maintaining Human Health. Digestion, 2016; 93: 176-81. doi: https://doi.org/10.1159/000444066
  63. Sanz Y, Nadal I, Sánchez E. Probiotics as Drugs Against Human Gastrointestinal Infections. Recent Patents on Anti-Infective Drug Discovery, 2007; 2(2): 148-56. doi: https://doi.org/10.2174/157489107780832596
  64. Moreno-Indias I, Cardona F, Tinahones FJ, Queipo-Ortuño MI. (2014). Impact of the gut microbiota on the development of obesity and type 2 diabetes mellitus. Frontiers in Microbiology, 2014; 5: 1-10. doi: https://doi.org/10.3389/fmicb.2014.00190
  65. Reid G, Sanders M, Gaskins HR, Gibson GR, Mercenier A, Rastall R et al. New scientific paradigms for probiotics and prebiotics. Journal of Clinical Gastroenterology, 2003; 37(2): 105-18. doi: https://doi.org/10.1097/00004836-200308000-00004
  66. Goodrich JK, Waters JL, Poole AC, Sutter JL, Koren O, Blekhman R, et al. Human Genetics Shape the Gut Microbiome. Cell, 2014; 159(4): 789-99. doi: https://doi.org/10.1016/j.cell.2014.09.053
  67. Faith JJ, Colombel JF, Gordon JI. Identifying strains that contribute to complex diseases through the study of microbial inheritance. Proceedings of the National Academy of Sciences of the United States of America, 2015; 112(3): 633-40. doi: https://doi.org/10.1073/pnas.1418781112
  68. Tamburini S, Shen N, Wu HC, Clemente JC. (2016). The microbiome in early life: implications for health outcomes. Nature medicine, 2016; 22(7): 713-22. doi: https://doi.org/10.1038/nm.4142
  69. Parséus A, Sommer N, Sommer F, Caesar R, Molinaro A, Ståhlman M et al (2016) Microbiota-induced obesity requires farnesoid X receptor. Gut, 2016; 66:1–9. doi: https://doi.org/10.1136/gutjnl-2015-310283
  70. Cani PD, Bibiloni R, Knauf C, Waget A, Neyrinck AM, Delzenne NM, Burcelin R. Changes in Gut Microbiota Control Metabolic Endotoxemia-Induced Inflammation in High-Fat Diet–Induced Obesity and Diabetes in Mice. Diabetes, 2008; 57(6): 1482-14980. doi: https://doi.org/10.2337/db07-1403
  71. Cani PD, Amar J, Iglesias MA, Poggi M, Knauf C, Bastelica D, et al. Metabolic Endotoxemia Initiates Obesity and Insulin Resistance. Diabetes, 2007; 56(7):1761-72. doi: http://doi.org/10.2337/db06-1491
  72. Donohoe DR, Garge N, Zhang X, Sun W, O’Connell TM, Bunger MK, et al. The Microbiome and Butyrate Regulate Energy Metabolism and Autophagy in the Mammalian Colon. Cell Metabolism, 2012;13(5): 517-26. doi: http://doi.org/10.1016/j.cmet.2011.02.018
  73. Turnbaugh RE, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature, 2006; 444(7122): 1027-31. doi: http://doi.org/10.1038/nature05414
  74. Le Chatelier E, Nielsen T, Qin J, Prifti E, Hildebrand F, Falony G, et al. Richness of human gut microbiome correlates with metabolic markers. Nature, 2013; 500(7464): 541-6. doi: http://doi.org/10.1038/nature12506
  75. Ley RE, Turnbaugh PJ, Klein S, Gordon JI. Human gut microbes associated with obesity. Nature, 2006; 444(7122): 1022-23. doi: http://doi.org/10.1038/4441022a
  76. Jumpertz R, Le DS, Turnbaugh PJ, Trinidad C, Bogardus C, Gordon JI, et al. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. The American Journal of Clinical Nutrition, 2011; 94(1): 58-65. doi: http://doi.org/10.3945/ajcn.110.010132
  77. Cani PD, Delzenne NM. The Role of the Gut Microbiota in Energy Metabolism and Metabolic Disease. Current Pharmaceutical Design, 2009; 15(13): 1546–58. doi: http://doi.org/10.2174/138161209788168164
  78. Wang ZB, Xin SS, Ding LN, Ding WY, Hou YL, Liu CQ, et al. The Potential Role of Probiotics in Controlling Overweight/Obesity and Associated Metabolic Parameters in Adults: A Systematic Review and Meta-Analysis. Evidence-based complementary and alternative medicine, 2019; 3862971: 1-14. doi: http://doi.org/10.1155/2019/3862971
  79. Garidou L, Pomié C, Klopp P, Waget A, Charpentier J, Aloulou M, et al. The Gut Microbiota Regulates Intestinal CD4 T Cells Expressing RORγt and Controls Metabolic Disease. Cell Matabolism, 2015; 22(1): 100-12. doi: http://doi.org/10.1016/j.cmet.2015.06.001
  80. Lam YY, Ha CW, Campbell CR, Mitchell AJ, Dinudom A, Oscarsson J, et al. (2012). Increased Gut Permeability and Microbiota Change Associate with Mesenteric Fat Inflammation and Metabolic Dysfunction in Diet-Induced Obese Mice. PLoS One, 2012; 7(3): e34233. doi: http://doi.org/10.1371/journal.pone.0034233
  81. Cani PD, Neyrinck AM, Fava F, Knauf C, Burcelin RG, Tuohy KM, et al. Selective increases of bifidobacteria in gut microflora improve high-fat-diet-induced diabetes in mice through a mechanism associated with endotoxaemia. Diabetologia, 2007; 50(11): 2374–83. Doi: https://doi.org/10.1007/s00125-007-0791-0
  82. Amar J, Chabo C, Waget A, Klopp P, Vachoux C, Bermúdez-Humarán L G, et al. Intestinal mucosal adherence and translocation of commensal bacteria at the early onset of type 2 diabetes: molecular mechanisms and probiotic treatment. EMBO Molecular Medicine, 2011; 3(9): 559-72. doi: http://doi.org/10.1002/emmm.201100159
  83. Bäckhed F, Ding H, Wang T, Hooper LV, Koh GY, Nagy, A, et al. The gut microbiota as an environmental factor that regulates fat storage. Proceedings of the National Academy of Sciences of the United States of America, 2004;101(44): 15718-23. doi: http://doi.org/10.1073/pnas.0407076101
  84. Bäckhed F, Manchester JK, Semenkovich CF, Gordon JI. Mechanisms underlying the resistance to diet-induced obesity in germ-free mice. Proceedings of the National Academy of Sciences of the United States of America, 2007; 104(3): 979-89. doi: http://doi.org/10.1073/pnas.0605374104
  85. Preiss-Landl K, Zimmermann R, Hämmerle G, Zechner R. Lipoprotein lipase: the regulation of tissue specific expression and its role in lipid and energy metabolism. Current Opinion in Lipidology, 2002; 13(5): 471-81. doi: https://doi.org/10.1097/00041433-200210000-00002
  86. Bergo M, Olivecrona G, Olivecrona T. Forms of lipoprotein lipase in rat tissues: in adipose tissue the proportion of inactive lipase increases on fasting. Biochem J, 1996; 313: 893-8 doi: https://doi.org/10.102/bj3130893
  87. International Scientific Association for Probiotics and Prebiotics. (2019). What are probiotics? 2019. Avialable in: https://isappscience.org/for-consumers/learn/probiotics/
  88. O’Toole PW, Marchesi JR, Hill C. Next‐generation probiotics: the spectrum from probiotics to live biotherapeutics. Nature Microbiology, 2017; 2: 17057. doi: http://doi.org/10.1038/nmicrobiol.2017.57
  89. Englyst KN, Englyst HN. Carbohydrate bioavailability. British Journal of Nutrition, 2005; 94(11): 1-11. doi: http://doi.org/10.1079/BJN20051457
  90. Vallianou NG, Stratigou T, Tsagarakis S. Microbiome and diabetes: Where are we now? Diabetes Research and Clinical Practice, 2018; 146: 111-8. doi: http://doi.org/10.1016/j.diabres.2018.10.008
  91. Delzenne NM, Cani PD, Neyrinck AM. Modulation of Glucagon-like Peptide 1 and Energy Metabolism by Inulin and Oligofructose: Experimental Data. The Journal of Nutrition, 2007; 137(11): 2547S-551S. doi: http://doi.org/10.1093/jn/137.11.2547S
  92. Cani PD, Knauf C, Iglesias MA, Drucker DJ, Delzenne NM, Burcelin R. Improvement of Glucose Tolerance and Hepatic Insulin Sensitivity by Oligofructose Requires a Functional Glucagon-Like Peptide 1 Receptor. Diabetes, 2006; 5: 1484-90. doi: http://doi.org/10.2337/db05-1360
  93. Fujita Y, Cheung AT, Kieffer TJ. Harnessing the gut to treat diabetes. Pediatric Diabetes, 2004; 5: 57-69. doi: https://doi.org/10.1111/j.1399-543X.2004.00080.x
  94. Delmée E, Cani PD, Gual G, Knauf C, Burcelin R, Maton N, et al. Relation between colonic proglucagon expression and metabolic response to oligofructose in high fat diet-fed mice. Life Sciences, 2006; 79(10): 1007-13. doi: http://doi.org/10.1016/j.lfs.2006.05.013
  95. Neyrinck AM, Alexiou H, Delzenne NM. Kupffer Cell Activity Is Involved in the Hepatoprotective Effect of Dietary Oligofructose in Rats with Endotoxic Shock. The Journal of Nutrition, 2004; 134(5): 1124-29. doi: http://doi.org/10.1093/jn/134.5.1124
  96. Cani PD, Dewever C, Delzenne NM. Inulin-type fructans modulate gastrointestinal peptides involved in appetite regulation (glucagon-like peptide-1 and ghrelin) in rats. British Journal of Nutrition, 2004; 92(3): 521-526. doi: https://doi.org/10.1079/bjn20041225
Share Button
(Visited 1,118 times, 1 visits today)

Creative Commons License
This work is licensed under a Creative Commons Attribution 4.0 International License.