Next Article in Journal
Effects of a 13-Week Personalized Lifestyle Intervention Based on the Diabetes Subtype for People with Newly Diagnosed Type 2 Diabetes
Previous Article in Journal
Glioblastoma in the Elderly: Review of Molecular and Therapeutic Aspects
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota

by
Lorena Elena Meliț
1,
Cristina Oana Mărginean
1,* and
Maria Oana Săsăran
2
1
Department of Pediatrics I, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
2
Department of Pediatrics III, George Emil Palade University of Medicine, Pharmacy, Science, and Technology of Târgu Mureș, Gheorghe Marinescu Street No 38, 540136 Târgu Mureș, Romania
*
Author to whom correspondence should be addressed.
Biomedicines 2022, 10(3), 645; https://doi.org/10.3390/biomedicines10030645
Submission received: 7 February 2022 / Revised: 5 March 2022 / Accepted: 7 March 2022 / Published: 10 March 2022
(This article belongs to the Section Microbiology in Human Health and Disease)

Abstract

:
The era of pediatric obesity is no longer a myth. Unfortunately, pediatric obesity has reached alarming incidence levels worldwide and the factors that contribute to its development have been intensely studied in multiple recent and emerging studies. Gut microbiota was recently included in the wide spectrum of factors implicated in the determination of obesity, but its role in pediatric obese patients is far from being fully understood. In terms of the infant gut microbiome, multiple factors have been demonstrated to shape its content, including maternal diet and health, type of delivery, feeding patterns, weaning and dietary habits. Nevertheless, the role of the intrauterine environment, such as the placental microbial community, cannot be completely excluded. Most studies have identified Firmicutes and Bacteroidetes as the most important players related to obesity risk in gut microbiota reflecting an increase of Firmicutes and a decrease in Bacteroidetes in the context of obesity; however, multiple inconsistencies between studies were recently reported, especially in pediatric populations, and there is a scarcity of studies performed in this age group.

1. Introduction

Pediatric obesity has reached epidemic proportions worldwide regardless of age. The prevalence of obesity varies by age, sex, ethnicity, race, and socio-economic status [1]. Obesity prevalence is higher in females of non-Hispanic black and of Hispanic origin when compared to non-Hispanic whites [1,2]. According to the World Health Organization, approximately 40 million children below the age of five years are diagnosed with obesity with particular prevalence in younger children [3]. The global prevalence of pediatric overweight and obesity was reported to vary from 5.7% to 40% depending on the studied population [4,5,6,7]. The multifactorial determination of this condition has been assessed in multiple studies which have shown that obesity occurs in genetically predisposed individuals who fulfill ‘obesogenic’ conditions, such as improper dietary habits, poverty or limited wealth, and even certain cultural traditions [1,8]. Nevertheless, no interdependence between genetic factors and ‘obesogenic’ factors has been established. Without doubt, genetic factors play a major role since multiple studies have shown that certain maternal gene polymorphisms are associated with increased risk for obesity in offspring [9,10,11,12], suggesting that predisposition to the development of obesity begins during intrauterine life. Moreover, the involvement of certain genes in weight gain by regulating feeding behavior, appetite, metabolism and energy expenditure has been clearly demonstrated in multiple studies [8]. Thirty-two loci of the human genome were found to be associated with increased body weight, but only 2% of the population was found to carry these genes [13]. In terms of the exogenous determination of obesity, energy intake that exceeds energy expenditure, along with a wide-spectrum of factors which interact with this process in multiple ways, represent the most important triggers [14]. The long term-persistence of obesity from childhood to adulthood is definitely a hallmark of decreased life expectancy taking into account the wide spectrum of complications related to this nutritional disorder, such as cardiovascular diseases, fatty liver disease or hepatocellular carcinoma [15]. Several human and animal studies have shown that obesity might be considered a major risk factor for carcinogenesis [16,17,18,19]. These findings were also supported by the relationship observed between pediatric obesity and primary liver carcinoma in later adulthood [20]. Most of these complications are directly related to the early inflammatory status identified in overweight and obese children [21]. Subclinical inflammatory status related to pediatric obesity was further demonstrated by the recently observed associations between increased serum levels of leptin and several pro-inflammatory cytokines, such as interleukin (IL) 6, IL 1β and tumor necrosis factor (TNF) α in obese children [22]. It has been suggested that adipose tissue serves not only a storage function, but also a secretory one. Thus, adipokines, when imbalanced, have the ability to stimulate not only a pro-inflammatory state by secreting multiple proteins, such as adiponectin or leptin, as well as cytokines with multiple biological activities, but also an insulin-resistant state, with both contributing to the pathogenesis of non-alcoholic fatty liver disease (NAFLD) and its future progression to non-alcoholic steatohepatitis [23,24,25]. The involvement of immune cells in this pro-inflammatory state is no longer a matter of debate. Macrophages, mast cells, neutrophils, and eosinophils, together with T and B lymphocytes, appear to form a complex array of cells involved in the pathogenesis of obesity [26]. Therefore, adipose tissue acts like a standalone organ, similar to endocrine or immune system organs.
The precise role of each factor involved in obesity etiology remains to be established. It is clear that genetic background and ‘obesogenic’ environment do not fully explain the etiology and variations between obesity prevalence among different populations. Thus, additional factors that might be related to obesity determinism should also be identified and thoroughly studied in order to develop effective strategies to prevent obesity development, and especially its long-term complications. One of these factors seems to be the intestinal microbiota, which has been intensely studied in a large body of emerging literature assuming the hypothesis that changes in its composition are related to both obesity determination and its associated complications [14]. The gut microbiota and humans evolved together and might be considered mutual partners; dysbiosis represents a life-threatening imbalance of this partnership since it has been demonstrated to be associated with several other conditions aside from obesity, such as diabetes, atherosclerosis, allergic and autoimmune disorders, gastrointestinal diseases and neoplasia [27,28,29,30].

2. Gut Microbiota and Obesity—To Be or Not to Be?

The gut microbiota might be defined as a shield of protection for mucosal permeability which regulates the fermentation and absorption of dietary polysaccharides, accounting for its role in the regulation of fat accumulation and subsequent obesity development [31]. The gut microbiota and host are in a symbiotic relationship with the gut microbiota contributing to the host’s physiological homeostasis and impairment of the gut microbiota resulting in the development of a wide spectrum of disorders [32]. In terms of obesity, alteration in the gut microbiota leads to an increase in energy harvesting from the diet. Aside from its role in increasing energy harvesting and the regulation of fat storage [32,33], the gut microbiota has also been shown to contribute both to the modulation of substrate formation for storable fat synthesis [34], and the production of compounds that, once absorbed into the systemic circulation, will promote the development of obesity-associated complications by increasing tissue inflammatory injuries and insulin resistance [35].
The gut microbiota might be considered as a true standalone organ with each individual harboring hundreds of different species with only a few dozen conserved between individuals, forming a stable core in a normal healthy state [36]. Based on the definition of a standalone organ, the gut microbiota has multiple functions, including digestion of certain otherwise indigestible dietary fibers, and interaction with several cells, such as immune, metabolic or nervous system cells involved in protection against pathogens, contributing to creation of a barrier against infections [37]. It has long been known that the colonization process begins at birth and is strongly influenced by multiple factors up to the age of three years, such as delivery type, antibiotic administration during early childhood, feeding habits, time of weaning, food composition, and environmental hygiene, which will be discussed further.
Gut microbiota development continues during childhood and adolescence becoming increasingly stable over time, but it is generally accepted that it closely resembles that of an adult by around the age of three years [38,39]. The gastrointestinal tract of a healthy adult has been found to harbor approximately 102 microbial cells within the stomach, duodenum and jejunum, while the distal ileum hosts between 107 and 108 microbial cells, the highest proportion being found in the colon which contains around 1011–1012 microbial cells [32]. The human gut contains approximately 1–2 kg of microorganisms with >105-fold more genes compared to the human genome itself [32]. The gut microbiome is crucial in maintaining the host’s homeostasis by contributing to the digestive process and energy production, preventing pathogen colonization and regulating the activity of the immune system [40]. Studies have demonstrated that the gut microbiome has the ability to modulate the individual’s susceptibility to obesity by increasing caloric extraction from indigestible dietary fibers as well as by augmenting its storage in adipose tissue [40]. Studies have also revealed that the gut microbiome influences whole body metabolism since germ-free mice displayed alterations in liver, kidney and bowel homeostasis [41,42,43].
Diet is a crucial factor involved in both obesity development and gut microbiota regulation. Thus, it has been demonstrated that dietary patterns shape the appearance of distinct combinations of bacteria within the gut, defined as enterotypes [44], by providing different growth-promoting and growth-inhibiting factors for the development of specific phylotypes [45]. It is well-known that obesity is associated with subclinical inflammation, even in pediatric patients, which may lead to a wide spectrum of long-term complications [21,22]. The gut microbiota seems also to contribute to the development of this inflammatory profile. According to a study performed on overweight and obese subjects, individuals that consumed the most unhealthy food, consisting of confectionary and sugary drinks with low consumption of yoghurt, fruit and water, displayed a significantly more severe inflammatory profile when compared to those with the healthiest diet based on increased consumption of yoghurts, fruits and soups [46]. The same authors concluded that the healthiest dietary pattern was strongly related to the highest microbial diversity within the gut [46]. The complex interaction between obesity and gut microbiota is further supported by other findings that have shown a decrease in microbial community richness in the context of overweight and obesity [47]. As mentioned, not only does obesity seem to be shaped by the gut microbiota, but also obesity-associated inflammation. Thus, increased consumption of fat is associated with a more severe degree of white adipose tissue inflammation and its related complications, while polyunsaturated fatty acid-based diets were found to counteract this inflammatory state in overweight and obesity, promoting a metabolically healthy phenotype [48,49]. In terms of protein consumption, it was shown that, if coupled with exercise, this might exert a positive effect on gut microbiota development. Clarke et al. demonstrated a positive correlation between high protein consumption and gut microbial diversity, but they also showed that athletes with a low body mass index harbor significantly higher levels of Akkermansia, which is inversely correlated with obesity in both animals and humans [50,51]. Although the relative abundance of each member of the gut microbial community is highly dynamic during an individual’s lifespan, the overall composition of the gut microbiome can be stable for years [52,53]. Thus, we might assume that host genetic factors might also be involved in shaping the gut microbiota apart from dietary patterns, but the extent of this involvement remains unclear. Studies performed on monozygotic and dizygotic twins failed to demonstrate significant associations in gut microbiota profiles at various ages from infancy to adulthood, suggesting that genetic factors have only a minor role in shaping the human gut microbiota [54,55]. In addition, the influence of dietary patterns on gut microbiota has been found to be sex-dependent with diet affecting gut microbiota development differently in males and females [56]. These findings are in contradiction to the previously mentioned study indicating that host genetic factors remain active participants in shaping gut microbiota, and the precise role of genetic factors requires further clarification.
A wide spectrum of inconsistencies between studies is evident in terms of gut microbiota in obese individuals. Thus, although most studies have reported that obese individuals possess reduced richness of microbes or their genes within the gut [47,54,57,58], decreased diversity was not constant in all obese individuals [47]. Another inconsistency is related to the ratio of Bacteroidetes to Firmicutes in obese versus lean individuals, which was observed to increase, decrease or not change at all [59]. Ethnicity is also related to these inconsistencies based on two studies that assessed Prevotella-dominated communities showing a positive correlation of these communities with body mass index in HIV positive subjects and controls originating from Mexico City [60], but not in older Amish individuals [2]. Another recent study also suggested a role of race/ethnicity in triggering distinct gut microbiota subtypes in the context of obesity [61].

2.1. Gut Microbiota Composition and Obesity

The composition of the gut microbiota consists of the phyla Firmicutes, Bacteroidetes, Proteobacteria, Actinobacteria and Verrucomicrobia, with Firmicutes and Bacteroidetes accounting for more than 90% of total bacterial species [62]. The gut microbiota in healthy individuals comprises a high ratio of Bacteroidetes to Firmicutes, while this ratio is reversed in obese individuals with a higher abundance of Firmicutes [63]. Moreover, obese individuals were observed to possess a high level of Lactobacillus species and a relatively low abundance of Bacteroides vulgates [64]. Metagenomic studies have suggested that the ratio of Bacteroidetes to Firmicutes might be used as a biomarker of obesity susceptibility [54,65] (Figure 1). Metagenomic analysis of the gut microbiome revealed that 40% of the intestine microbial gene pool is common among all humans around the world, indicating the presence of a core microbiome [66]. Obesity metabolic changes were shown to be influenced by an increase in short-chain fatty acid producers (Actinobacteria) and pathogenic Proteobacteria, with increase in their abundance in the context of obesity [67]. Interestingly, these alterations in gut microbiota were found to be reversible with weight loss [68].
Several mechanisms have been proposed to account for the influence of the gut microbiota on obesity development. One suggested mechanism is related to the ability of microorganisms to ferment dietary polysaccharides otherwise not digested by humans [35]. Thus, the formation process of dietary fibers results in an increased production of short-chain fatty acids (e.g., butyrate, acetate, propionate) identified in fecal or cecal samples, which, after absorption, induce lipogenesis and promote triglyceride storage through various molecular pathways, such as the activation of carbohydrate responsive element-binding protein and sterol regulatory element-binding transcriptor 1 [35,69]. Another peculiarity of short-chain fatty acids is their ability to suppress the fasting-induced adipocyte factor leading to the inhibition of lipoprotein lipase and subsequent accumulation of triglycerides in host adipocytes [35]. Nevertheless, several positive effects of short-chain fatty acids on human metabolism were suggested by Morrison and Preston in a recent review, such as a potential role in maintaining the integrity of the gut mucosa (butyrate), beneficial effects on glucose homeostasis, a role in appetite regulation (propionate), and the regulation of immune system and inflammatory response [70]. Considering the inconsistencies reported in the literature related to this topic, further studies are needed to clarify the precise role of short-chain fatty acids in human homeostasis, especially in terms of lipid metabolism.

2.2. Maternal Factors

The gut microbiota shaping process begins in utero and the most important factor that influences fetal wellbeing is maternal diet [71,72]. According to Gibson et al., offspring born to mothers consuming fish oil were found to have a higher level of opportunistic bacteria, such as Enterococcus faecium, Bilophila wadsworthia and Bacteroides fragilis, with a negative impact on the infant’s immune responses [73]. In contrast, the study of Chu et al. [71] found a reduced level of Bacteroidetes in infants whose mothers consumed a high-fat diet during pregnancy. Habitual maternal intake of probiotic-containing food leads to a reduced risk for both spontaneous preterm delivery [74], and allergic disorders during childhood [75]. In addition, maternal alcohol consumption during pregnancy was demonstrated to negatively influence both maternal and neonatal gut microbiome, and, in terms of neonatal outcomes, it resulted in an increased risk for infections and other disorders later in life [76,77].
Maternal health during pregnancy is another key factor in infant wellbeing. Several studies aiming to identify the relationship between maternal health and infant wellbeing concluded that the microbial composition of infants born to obese mothers differs markedly from those born to lean mothers due to increased exposure of the offspring to oxidative stress [78,79,80]. It is well-known that infants born to mothers diagnosed with diabetes, or those with an increased body mass index, are more prone to developing diabetes and obesity in later life [81,82,83] due to alterations in the maternal gut microbiome during both pregnancy and lactation [84]. A recent study concluded that the microbiome in overweight or obese mothers is associated with an increased risk for early obesity in infants since early life microbial composition predicts obesity development later in life [85]. Nevertheless, the precise composition of pregnant women’s gut microbiota is not yet clearly established and reported findings remain contradictory. Collado et al. observed increased levels of Clostridium, Bacteroidetes and Staphylococcus, with a decreased level of Bifidobacterium, in mothers with overweight/obesity [78], while the study of Santacruz et al. found an abundance of Staphylococcus, Enterobacteriaceae and Staphylococcus, with a significantly lower level of Bifidobacterium and Bacteroides, when assessing a similar group [86].
Dysbiosis of the placental microbiota might represent another important factor for neonatal health. Despite the fast colonization during the first three years of life, the hypothesis that the fetus is seeded with bacteria during the intrauterine life from the maternal placental microbial community cannot be ruled out [87]. The few studies that have assessed placental microbial composition observed an increase in the order Pseudomonadales, and the genera Actinobacteria and Proteobacteria, along with lower levels of Bacteroidetes and Firmicutes in pregnant women with gestational diabetes mellitus [88,89]. Furthermore, studies that compared meconium samples of neonate subjects born to mothers with diabetes to those whose mothers were non-diabetic found an abundance of the Bacteroidetes and Lachnospiraceae families and the Parabacteriodes genus along with decreased levels of Proteobacteria in meconium samples from the diabetes group [90].
Based on the above, we might hypothesize a complex interaction between maternal diet and obesity, gestational diabetes mellitus, placental microbial composition and the risk of childhood obesity through important alterations in the newborn’s gut microbiome, beginning from intrauterine life.

2.3. Delivery Mode

It has been well-documented that the delivery mode shapes the neonatal gut microbiome. Thus, when born vaginally, the neonate comes in contact with the maternal vaginal and gut microbiota and the infant’s gut microbiome will be similar to the vaginal microbial composition, being dominated by the genera Lactobacillus, Sneathia and Prevotella [91]. Otherwise, if the baby is born by cesarean section, the major component of the neonate’s gut microbiome resembles the mother’s skin microbiome and nosocomial surroundings [92]. Recent studies have revealed an important relationship between cesarean section delivery and the risk of obesity during childhood [93,94]. It was found that infants born by cesarean section possess a lower diversity of the gut microbial community compared to vaginally delivered infants, with lower levels of both Bifidobacteria and Escherichia-Shigella, along with a complete absence of Bacteroides [95,96,97,98]. These infants display a gut microbial community mainly composed of the genera Staphylococcus, Propionibacterium and Corynebacterium, resembling the mother’s skin microbial community [91].

2.4. Feeding Patterns

Breastfeeding remains the most appropriate alimentation pattern for infants since human milk contains important bioactive compounds, such as oligosaccharides, which are essential for infant development and offer protection against several childhood disorders, such as obesity [99], type 2 diabetes mellitus [100], celiac disease [101], allergies [102], diarrhea [103], and other metabolic diseases [104]. Moreover, human oligosaccharides have the ability to promote Bifidobacteria and Bacteroides growth [105], and to regulate infant wellbeing due to their action as prebiotics, modulators of innate immune responses and their anti-inflammatory properties [106]. Additional evidence suggests that Bifidobacterium species, such as B. longum, B. breve, B. infantis and B. pseudocatenulatum, represent the most common Actinobacteria in breastfed infants [107,108,109], along with lactic acid bacteria, such as Enterococcus, Lactobacillus and Clostridium species [110]. Maternal health is another crucial modulator of human milk content resulting in alterations of the human milk microbiome and subsequent negative changes in the infant gut microbiome [92]. Maternal obesity was found to alter the human milk microbiome by decreasing bacterial community diversity, leading to lower levels of Bifidobacterium and higher levels of Staphylococcus [111,112]. Recent evidence has also underlined the role of human milk in maintaining infant protein balance [113]. In terms of formula-fed infants, a recent study, which assessed the composition of formula-fed and breast-fed infants gut microbiota at the age of 40 days, found that α diversity was lower in a breast-fed group compared to formula-fed infants [114]. These findings were supported by other studies which concluded that the gut microbiota of formula-fed infants is more diverse compared to that of breastfed infants, being similar to that of older children [115,116]. The most predominant genus in both breastfed and formula-fed infants are Bifidobacterium and Enterobacteriaceae which seem to decrease with age to almost zero by 18 months [114,117]. In addition, formula-fed infants were found to have increased levels of both Clostridia and Veillonella spp. compared to breastfed infants [108,114,116]. Considering that human milk is incontestably the most suitable feeding pattern for infants, further studies are required to identify the precise role of each bacterium within the gut microbial community and the differences between feeding patterns.

2.5. Weaning and Diet

Diet definitely plays a crucial role in modulating the microbial community with it being observed that, once weaned, infants begin to develop a similar microbial composition to that of adults with an increase in diversity and significant changes in taxonomic groups [108,109]. Most changes in gut microbiota composition are observed between the ages of 9 to 18 months, consisting of a decrease of Lactobacilli, Bifidobacteria and Enterobacteriaceae and an increase in Bacteroides and Clostridium species, reinforcing the major role of weaning in triggering these changes [110]. Moreover, the type of diet contributes to the composition of the gut bacterial community. Assessing the role of dietary habits in children with obesity, Nakayama et al. observed that a high-fat westernized diet results in a decrease in Prevotellaceae in the gut microbiota which might contribute to obesity development during childhood [118]. However, the authors stated that children with a carbohydrate-based diet showed an increase in Prevotellaceae and a decrease in Bacteroidaceae [118]. Therefore, weaning and diet probably represent decisive factors in gut microbiota development since they are the last factors shaping the microbial community before the age of three years, when this process achieves relative stability.
The influence of the above-mentioned factors involved in shaping gut microbiome during early childhood is summarized in Table 1. The precise role of each factor in the development of pediatric obesity is a topic that definitely requires further research (Figure 2).

3. Summary

The gut microbiota comprises a wide range of bacteria with complex physiological functions which extend to extraintestinal tissues and seem to be related to excessive weight gain [33]. It has been demonstrated that the gut microbial community might have a dichotomous role influenced by environmental factors, with the ability to promote energy storage or, in contrast, to favor leanness [110,119,120]. The shaping of the gut microbiota begins in utero and continues up to the age of 3–4 years being influenced by the maternal diet, amniotic fluid and placental microbial community, type of delivery, feeding practices and hygiene habits, such as brushing the teeth after meals [121]. The most important players related to obesity risk in the gut microbiota are the Firmicutes and Bacteroides. Studies on both animal and human subjects have found that obesity is associated with an increase in Firmicutes and a decrease in Bacteroidetes, independently of dietary caloric intake [34,122]. A recent systematic review [121] supported this statement, highlighting that several studies have found a positive relationship between higher levels of certain microbes belonging to the Firmicutes (e.g., Eubacterium halli, Clostridium leptum and Lactobacillus spp.) and overweight or obesity [110,119]. Nevertheless, the findings assessing the relationship between gut microbial composition and obesity remain contradictory, since other studies have found the contrary, with observed decrease in bacteria belonging to the same phyla (e.g., the genera Staphylococcus and Clostridium difficile [33,120]). Moreover, several studies have found higher levels of Bacteroides fragilis in children with overweight/obesity compared to normal-weight children [119,120,123], while others have found that overweight/obesity is associated with a decrease in the entire phylum Bacteroidetes, along with a decrease in the Bacteroides/Prevotella groups [124,125]. In addition, a meta-analysis that included mostly adults failed to find any association between the Bacteroidetes/Firmicutes ratio and obesity [65]. A recent study performed on 3-year-old children found inverse relationships between both Parabacteroidetes and Peptostrecoccae U and overweight/obesity [126]. The same study found a tendency of overweight/obese children to exhibit relative abundance of Ruminococcus and Akermansia [126]. Thus, the authors concluded that several differences in gut microbial composition between obese and lean adults are similar to the 3-year-old age group suggesting obesity predisposition has its origins in early childhood [126].
Regardless of the inconsistencies identified between studies, we must acknowledge the major role of gut microbiota in the development of childhood overweight/obesity. Additional studies to clarify the precise role of each bacterium from the gut microbiota would represent a major development in the era of pediatric obesity. Based on the wide spectrum of long-term complications associated with increased body mass index, filling the existing gaps regarding the relationship between gut microbial composition and pediatric obesity might be the cornerstone for the development of innovative prophylactic and/or therapeutic strategies targeted at decreasing the incidence of overweight/obesity in both children and adults.

Author Contributions

L.E.M., C.O.M. and M.O.S. conceptualized and designed the study, drafted the initial manuscript, and reviewed and revised the manuscript. All authors have read and agreed to the published version of the manuscript.

Funding

This article received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Wang, Y.; Beydoun, M.A. The obesity epidemic in the United States—Gender, age, socioeconomic, racial/ethnic, and geographic characteristics: A systematic review and meta-regression analysis. Epidemiol. Rev. 2007, 29, 6–28. [Google Scholar] [CrossRef] [PubMed]
  2. Zupancic, M.L.; Cantarel, B.L.; Liu, Z.; Drabek, E.F.; Ryan, K.A.; Cirimotich, S.; Jones, C.; Knight, R.; Walters, W.A.; Knights, D.; et al. Analysis of the gut microbiota in the old order Amish and its relation to the metabolic syndrome. PLoS ONE 2012, 7, e43052. [Google Scholar] [CrossRef] [PubMed]
  3. WHO. Overweight and Obesity. Available online: https://www.who.int/gho/ncd/risk_factors/overweight/en/ (accessed on 25 January 2019).
  4. Gulati, S.; Misra, A.; Colles, S.L.; Kondal, D.; Gupta, N.; Goel, K.; Bansal, S.; Mishra, M.; Madkaikar, V.; Bhardwaj, S. Dietary intakes and familial correlates of overweight/obesity: A four-cities study in India. Ann. Nutr. Metab. 2013, 62, 279–290. [Google Scholar] [CrossRef]
  5. Jafar, T.H.; Qadri, Z.; Islam, M.; Hatcher, J.; Bhutta, Z.A.; Chaturvedi, N. Rise in childhood obesity with persistently high rates of undernutrition among urban school-aged Indo-Asian children. Arch. Dis. Child. 2008, 93, 373–378. [Google Scholar] [CrossRef] [PubMed]
  6. Moraes, S.A.D.; Beltrán Rosas, J.; Mondini, L.; Freitas, I.C.M.D. Prevalence of overweight and obesity, and associated factors in school children from urban area in Chilpancingo, Guerrero, Mexico, 2004. Cad. Saude Publica 2006, 22, 1289–1301. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Ogden, C.L.; Carroll, M.D.; Kit, B.K.; Flegal, K.M. Prevalence of childhood and adult obesity in the United States, 2011–2012. JAMA 2014, 311, 806–814. [Google Scholar] [CrossRef] [Green Version]
  8. Mǎrginean, C.O.; Mǎrginean, C.; Meliţ, L.E. New Insights Regarding Genetic Aspects of Childhood Obesity: A Minireview. Front. Pediatr. 2018, 6, 271. [Google Scholar] [CrossRef] [Green Version]
  9. Mărginean, C.; Mărginean, C.O.; Iancu, M.; Szabo, B.; Cucerea, M.; Melit, L.E.; Crauciuc, A.; Bănescu, C. The role of TGF-β1 869 T > C and PPAR γ2 34 C > G polymorphisms, fat mass, and anthropometric characteristics in predicting childhood obesity at birth: A cross-sectional study according the parental characteristics and newborn’s risk for child obesity (the newborns obesity’s risk) NOR study. Medicine 2016, 95, e4265. [Google Scholar] [CrossRef]
  10. Mărginean, C.; Mărginean, C.O.; Bănescu, C.; Meliţ, L.E.; Tripon, F.; Iancu, M. The relationship among GNB3 rs5443, PNPLA3 rs738409, GCKR rs780094 gene polymorphisms, type of maternal gestational weight gain and neonatal outcomes (STROBE-compliant article). Medicine 2019, 98, e16414. [Google Scholar] [CrossRef]
  11. Mărginean, C.O.; Mărginean, C.; Bănescu, C.; Meliţ, L.E.; Tripon, F.; Iancu, M. The relationship between MMP9 and ADRA2A gene polymorphisms and mothers-newborns’ nutritional status: An exploratory path model (STROBE compliant article). Pediatr. Res. 2019, 85, 822–829. [Google Scholar] [CrossRef] [Green Version]
  12. Marginean, C.; Marginean, C.; Iancu, M.; Melit, L.; Tripon, F.; Banescu, C. MC4R and ENPP1 gene polymorphisms and their implication in maternal and neonatal risk for obesity. Sci. Rep. 2019, 9, 10858. [Google Scholar] [CrossRef] [PubMed]
  13. Speliotes, E.K.; Willer, C.J.; Berndt, S.I.; Monda, K.L.; Thorleifsson, G.; Jackson, A.U.; Lango Allen, H.; Lindgren, C.M.; Luan, J.; Mägi, R.; et al. Association analyses of 249,796 individuals reveal 18 new loci associated with body mass index. Nat. Genet. 2010, 42, 937–948. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  14. Arslan, N. Obesity, fatty liver disease and intestinal microbiota. World J. Gastroenterol. 2014, 20, 16452–16463. [Google Scholar] [CrossRef] [PubMed]
  15. Adams, L.A.; Lymp, J.F.; St Sauver, J.; Sanderson, S.O.; Lindor, K.D.; Feldstein, A.; Angulo, P. The natural history of nonalcoholic fatty liver disease: A population-based cohort study. Gastroenterology 2005, 129, 113–121. [Google Scholar] [CrossRef] [PubMed]
  16. Bugianesi, E. Non-alcoholic steatohepatitis and cancer. Clin. Liver Dis. 2007, 11, 191–207. [Google Scholar] [CrossRef] [PubMed]
  17. Kikuchi, L.; Oliveira, C.P.; Carrilho, F.J. Nonalcoholic fatty liver disease and hepatocellular carcinoma. Biomed. Res. Int. 2014, 2014, 106247. [Google Scholar] [CrossRef] [Green Version]
  18. Calle, E.E.; Rodriguez, C.; Walker-Thurmond, K.; Thun, M.J. Overweight, obesity, and mortality from cancer in a prospectively studied cohort of U.S. adults. N. Engl. J. Med. 2003, 348, 1625–1638. [Google Scholar] [CrossRef] [Green Version]
  19. Nair, S.; Mason, A.; Eason, J.; Loss, G.; Perrillo, R.P. Is obesity an independent risk factor for hepatocellular carcinoma in cirrhosis? Hepatology 2002, 36, 150–155. [Google Scholar] [CrossRef]
  20. Berentzen, T.L.; Gamborg, M.; Holst, C.; Sørensen, T.I.A.; Baker, J.L. Body mass index in childhood and adult risk of primary liver cancer. J. Hepatol. 2014, 60, 325–330. [Google Scholar] [CrossRef]
  21. Mărginean, C.O.; Meliţ, L.E.; Ghiga, D.V.; Mărginean, M.O. Early Inflammatory Status Related to Pediatric Obesity. Front. Pediatr. 2019, 7, 241. [Google Scholar] [CrossRef]
  22. Mărginean, C.O.; Meliţ, L.E.; Huțanu, A.; Ghiga, D.V.; Săsăran, M.O. The adipokines and inflammatory status in the era of pediatric obesity. Cytokine 2020, 126, 154925. [Google Scholar] [CrossRef] [PubMed]
  23. Gregor, M.F.; Hotamisligil, G.S. Thematic review series: Adipocyte Biology. Adipocyte stress: The endoplasmic reticulum and metabolic disease. J. Lipid Res. 2007, 48, 1905–1914. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  24. Machado, M.V.; Coutinho, J.; Carepa, F.; Costa, A.; Proença, H.; Cortez-Pinto, H. How adiponectin, leptin, and ghrelin orchestrate together and correlate with the severity of nonalcoholic fatty liver disease. Eur. J. Gastroenterol. Hepatol. 2012, 24, 1166–1172. [Google Scholar] [CrossRef] [PubMed]
  25. Mărginean, C.O.; Meliț, L.E.; Săsăran, M.O. Metabolic Associated Fatty Liver Disease in Children-From Atomistic to Holistic. Biomedicines 2021, 9, 1866. [Google Scholar] [CrossRef] [PubMed]
  26. Meliț, L.E.; Mărginean, C.O.; Mărginean, C.D.; Săsăran, M.O. The Peculiar Trialogue between Pediatric Obesity, Systemic Inflammatory Status, and Immunity. Biology 2021, 10, 512. [Google Scholar] [CrossRef]
  27. Chan, Y.K.; Estaki, M.; Gibson, D.L. Clinical consequences of diet-induced dysbiosis. Ann. Nutr. Metab. 2013, 63 (Suppl. S2), 28–40. [Google Scholar] [CrossRef]
  28. Sekirov, I.; Russell, S.L.; Antunes, L.C.M.; Finlay, B.B. Gut microbiota in health and disease. Physiol. Rev. 2010, 90, 859–904. [Google Scholar] [CrossRef] [Green Version]
  29. Clemente, J.C.; Ursell, L.K.; Parfrey, L.W.; Knight, R. The impact of the gut microbiota on human health: An integrative view. Cell 2012, 148, 1258–1270. [Google Scholar] [CrossRef] [Green Version]
  30. Knights, D.; Lassen, K.G.; Xavier, R.J. Advances in inflammatory bowel disease pathogenesis: Linking host genetics and the microbiome. Gut 2013, 62, 1505–1510. [Google Scholar] [CrossRef]
  31. Muscogiuri, G.; Cantone, E.; Cassarano, S.; Tuccinardi, D.; Barrea, L.; Savastano, S.; Colao, A.; on behalf of the Obesity Programs of nutrition, Education, Research and Assessment (OPERA) group. Gut microbiota: A new path to treat obesity. Int. J. Obes. Suppl. 2019, 9, 10–19. [Google Scholar] [CrossRef]
  32. Patterson, E.; Ryan, P.M.; Cryan, J.F.; Dinan, T.G.; Ross, R.P.; Fitzgerald, G.F.; Stanton, C. Gut microbiota, obesity and diabetes. Postgrad. Med. J. 2016, 92, 286–300. [Google Scholar] [CrossRef]
  33. Bäckhed, F.; Ding, H.; Wang, T.; Hooper, L.V.; Koh, G.Y.; Nagy, A.; Semenkovich, C.F.; Gordon, J.I. The gut microbiota as an environmental factor that regulates fat storage. Proc. Natl. Acad. Sci. USA 2004, 101, 15718–15723. [Google Scholar] [CrossRef] [Green Version]
  34. Turnbaugh, P.J.; Ley, R.E.; Mahowald, M.A.; Magrini, V.; Mardis, E.R.; Gordon, J.I. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006, 444, 1027–1031. [Google Scholar] [CrossRef]
  35. Khan, M.J.; Gerasimidis, K.; Edwards, C.A.; Shaikh, M.G. Role of Gut Microbiota in the Aetiology of Obesity: Proposed Mechanisms and Review of the Literature. J. Obes. 2016, 2016, 7353642. [Google Scholar] [CrossRef] [Green Version]
  36. Blottière, H.M.; de Vos, W.M.; Ehrlich, S.D.; Doré, J. Human intestinal metagenomics: State of the art and future. Curr. Opin. Microbiol. 2013, 16, 232–239. [Google Scholar] [CrossRef]
  37. Blottière, H.M. The gut microbiota and obesity. In Energy Balance and Obesity; Romieu, I., Dossus, L., Willett, W.C., Eds.; IARC Working Group Reports; International Agency for Research on Cancer: Lyon, France, 2017; ISBN 978-92-832-2519-5. [Google Scholar]
  38. Palmer, C.; Bik, E.M.; DiGiulio, D.B.; Relman, D.A.; Brown, P.O. Development of the human infant intestinal microbiota. PLoS Biol. 2007, 5, e177. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Borre, Y.E.; O’Keeffe, G.W.; Clarke, G.; Stanton, C.; Dinan, T.G.; Cryan, J.F. Microbiota and neurodevelopmental windows: Implications for brain disorders. Trends Mol. Med. 2014, 20, 509–518. [Google Scholar] [CrossRef] [PubMed]
  40. Sivamaruthi, B.S.; Kesika, P.; Suganthy, N.; Chaiyasut, C. A Review on Role of Microbiome in Obesity and Antiobesity Properties of Probiotic Supplements. Biomed. Res. Int. 2019, 2019, 3291367. [Google Scholar] [CrossRef]
  41. Zhao, L.; Shen, J. Whole-body systems approaches for gut microbiota-targeted, preventive healthcare. J. Biotechnol. 2010, 149, 183–190. [Google Scholar] [CrossRef]
  42. Claus, S.P.; Tsang, T.M.; Wang, Y.; Cloarec, O.; Skordi, E.; Martin, F.-P.; Rezzi, S.; Ross, A.; Kochhar, S.; Holmes, E.; et al. Systemic multicompartmental effects of the gut microbiome on mouse metabolic phenotypes. Mol. Syst. Biol. 2008, 4, 219. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  43. Swann, J.R.; Want, E.J.; Geier, F.M.; Spagou, K.; Wilson, I.D.; Sidaway, J.E.; Nicholson, J.K.; Holmes, E. Systemic gut microbial modulation of bile acid metabolism in host tissue compartments. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4523–4530. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  44. Graf, D.; Di Cagno, R.; Fåk, F.; Flint, H.J.; Nyman, M.; Saarela, M.; Watzl, B. Contribution of diet to the composition of the human gut microbiota. Microb. Ecol. Health Dis. 2015, 26, 26164. [Google Scholar] [CrossRef] [PubMed]
  45. Flint, H.J.; Scott, K.P.; Louis, P.; Duncan, S.H. The role of the gut microbiota in nutrition and health. Nat. Rev. Gastroenterol. Hepatol. 2012, 9, 577–589. [Google Scholar] [CrossRef] [PubMed]
  46. Kong, L.C.; Holmes, B.A.; Cotillard, A.; Habi-Rachedi, F.; Brazeilles, R.; Gougis, S.; Gausserès, N.; Cani, P.D.; Fellahi, S.; Bastard, J.-P.; et al. Dietary patterns differently associate with inflammation and gut microbiota in overweight and obese subjects. PLoS ONE 2014, 9, e109434. [Google Scholar] [CrossRef] [PubMed]
  47. Le Chatelier, E.; Nielsen, T.; Qin, J.; Prifti, E.; Hildebrand, F.; Falony, G.; Almeida, M.; Arumugam, M.; Batto, J.-M.; Kennedy, S.; et al. Richness of human gut microbiome correlates with metabolic markers. Nature 2013, 500, 541–546. [Google Scholar] [CrossRef] [PubMed]
  48. Oh, D.Y.; Talukdar, S.; Bae, E.J.; Imamura, T.; Morinaga, H.; Fan, W.; Li, P.; Lu, W.J.; Watkins, S.M.; Olefsky, J.M. GPR120 is an omega-3 fatty acid receptor mediating potent anti-inflammatory and insulin-sensitizing effects. Cell 2010, 142, 687–698. [Google Scholar] [CrossRef] [Green Version]
  49. Caesar, R.; Tremaroli, V.; Kovatcheva-Datchary, P.; Cani, P.D.; Bäckhed, F. Crosstalk between Gut Microbiota and Dietary Lipids Aggravates WAT Inflammation through TLR Signaling. Cell Metab. 2015, 22, 658–668. [Google Scholar] [CrossRef] [Green Version]
  50. Everard, A.; Belzer, C.; Geurts, L.; Ouwerkerk, J.P.; Druart, C.; Bindels, L.B.; Guiot, Y.; Derrien, M.; Muccioli, G.G.; Delzenne, N.M.; et al. Cross-talk between Akkermansia muciniphila and intestinal epithelium controls diet-induced obesity. Proc. Natl. Acad. Sci. USA 2013, 110, 9066–9071. [Google Scholar] [CrossRef] [Green Version]
  51. Karlsson, C.L.J.; Onnerfält, J.; Xu, J.; Molin, G.; Ahrné, S.; Thorngren-Jerneck, K. The microbiota of the gut in preschool children with normal and excessive body weight. Obesity 2012, 20, 2257–2261. [Google Scholar] [CrossRef]
  52. 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. [Google Scholar] [CrossRef] [Green Version]
  53. Faith, J.J.; Guruge, J.L.; Charbonneau, M.; Subramanian, S.; Seedorf, H.; Goodman, A.L.; Clemente, J.C.; Knight, R.; Heath, A.C.; Leibel, R.L.; et al. The long-term stability of the human gut microbiota. Science 2013, 341, 1237439. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Turnbaugh, P.J.; Hamady, M.; Yatsunenko, T.; Cantarel, B.L.; Duncan, A.; Ley, R.E.; Sogin, M.L.; Jones, W.J.; Roe, B.A.; Affourtit, J.P.; et al. A core gut microbiome in obese and lean twins. Nature 2009, 457, 480–484. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  55. Yatsunenko, T.; Rey, F.E.; Manary, M.J.; Trehan, I.; Dominguez-Bello, M.G.; Contreras, M.; Magris, M.; Hidalgo, G.; Baldassano, R.N.; Anokhin, A.P.; et al. Human gut microbiome viewed across age and geography. Nature 2012, 486, 222–227. [Google Scholar] [CrossRef]
  56. Bolnick, D.I.; Snowberg, L.K.; Hirsch, P.E.; Lauber, C.L.; Org, E.; Parks, B.; Lusis, A.J.; Knight, R.; Caporaso, J.G.; Svanbäck, R. Individual diet has sex-dependent effects on vertebrate gut microbiota. Nat. Commun. 2014, 5, 4500. [Google Scholar] [CrossRef] [PubMed]
  57. Sze, M.A.; Schloss, P.D. Looking for a Signal in the Noise: Revisiting Obesity and the Microbiome. mBio 2016, 7, e01018-16. [Google Scholar] [CrossRef] [Green Version]
  58. Peters, B.A.; Shapiro, J.A.; Church, T.R.; Miller, G.; Trinh-Shevrin, C.; Yuen, E.; Friedlander, C.; Hayes, R.B.; Ahn, J. A taxonomic signature of obesity in a large study of American adults. Sci. Rep. 2018, 8, 9749. [Google Scholar] [CrossRef] [Green Version]
  59. Hermes, G.D.A.; Zoetendal, E.G.; Smidt, H. Molecular ecological tools to decipher the role of our microbial mass in obesity. Benef. Microbes 2015, 6, 61–81. [Google Scholar] [CrossRef]
  60. Pinto-Cardoso, S.; Lozupone, C.; Briceño, O.; Alva-Hernández, S.; Téllez, N.; Adriana, A.; Murakami-Ogasawara, A.; Reyes-Terán, G. Fecal Bacterial Communities in treated HIV infected individuals on two antiretroviral regimens. Sci. Rep. 2017, 7, 43741. [Google Scholar] [CrossRef] [Green Version]
  61. Stanislawski, M.A.; Dabelea, D.; Lange, L.A.; Wagner, B.D.; Lozupone, C.A. Gut microbiota phenotypes of obesity. NPJ Biofilms Microbiomes 2019, 5, 18. [Google Scholar] [CrossRef]
  62. Castaner, O.; Goday, A.; Park, Y.-M.; Lee, S.-H.; Magkos, F.; Shiow, S.-A.T.E.; Schröder, H. The Gut Microbiome Profile in Obesity: A Systematic Review. Int. J. Endocrinol. 2018, 2018, 4095789. [Google Scholar] [CrossRef]
  63. Jumpertz, R.; Le, D.S.; Turnbaugh, P.J.; Trinidad, C.; Bogardus, C.; Gordon, J.I.; Krakoff, J. Energy-balance studies reveal associations between gut microbes, caloric load, and nutrient absorption in humans. Am. J. Clin. Nutr. 2011, 94, 58–65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  64. Karlsson, F.; Tremaroli, V.; Nielsen, J.; Bäckhed, F. Assessing the human gut microbiota in metabolic diseases. Diabetes 2013, 62, 3341–3349. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  65. Walters, W.A.; Xu, Z.; Knight, R. Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett. 2014, 588, 4223–4233. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  66. Qin, J.; Li, R.; Raes, J.; Arumugam, M.; Burgdorf, K.S.; Manichanh, C.; Nielsen, T.; Pons, N.; Levenez, F.; Yamada, T.; et al. A human gut microbial gene catalogue established by metagenomic sequencing. Nature 2010, 464, 59–65. [Google Scholar] [CrossRef] [Green Version]
  67. Ojeda, P.; Bobe, A.; Dolan, K.; Leone, V.; Martinez, K. Nutritional modulation of gut microbiota—the impact on metabolic disease pathophysiology. J. Nutr. Biochem. 2016, 28, 191–200. [Google Scholar] [CrossRef] [Green Version]
  68. Shen, W.; Gaskins, H.R.; McIntosh, M.K. Influence of dietary fat on intestinal microbes, inflammation, barrier function and metabolic outcomes. J. Nutr. Biochem. 2014, 25, 270–280. [Google Scholar] [CrossRef]
  69. Al-Assal, K.; Martinez, A.C.; Torrinhas, R.S.; Cardinelli, C.; Waitzberg, D. Gut microbiota and obesity. Clin. Nutr. Exp. 2018, 20, 60–64. [Google Scholar] [CrossRef] [Green Version]
  70. Morrison, D.J.; Preston, T. Formation of short chain fatty acids by the gut microbiota and their impact on human metabolism. Gut Microbes 2016, 7, 189–200. [Google Scholar] [CrossRef] [Green Version]
  71. Chu, D.M.; Meyer, K.M.; Prince, A.L.; Aagaard, K.M. Impact of maternal nutrition in pregnancy and lactation on offspring gut microbial composition and function. Gut Microbes 2016, 7, 459–470. [Google Scholar] [CrossRef] [Green Version]
  72. Lundgren, S.N.; Madan, J.C.; Emond, J.A.; Morrison, H.G.; Christensen, B.C.; Karagas, M.R.; Hoen, A.G. Maternal diet during pregnancy is related with the infant stool microbiome in a delivery mode-dependent manner. Microbiome 2018, 6, 109. [Google Scholar] [CrossRef] [Green Version]
  73. Gibson, M.K.; Wang, B.; Ahmadi, S.; Burnham, C.-A.D.; Tarr, P.I.; Warner, B.B.; Dantas, G. Developmental dynamics of the preterm infant gut microbiota and antibiotic resistome. Nat. Microbiol. 2016, 1, 16024. [Google Scholar] [CrossRef] [PubMed]
  74. Myhre, R.; Brantsæter, A.L.; Myking, S.; Gjessing, H.K.; Sengpiel, V.; Meltzer, H.M.; Haugen, M.; Jacobsson, B. Intake of probiotic food and risk of spontaneous preterm delivery. Am. J. Clin. Nutr. 2011, 93, 151–157. [Google Scholar] [CrossRef] [Green Version]
  75. Bertelsen, R.J.; Brantsæter, A.L.; Magnus, M.C.; Haugen, M.; Myhre, R.; Jacobsson, B.; Longnecker, M.P.; Meltzer, H.M.; London, S.J. Probiotic milk consumption in pregnancy and infancy and subsequent childhood allergic diseases. J. Allergy Clin. Immunol. 2014, 133, e1–e8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  76. Dubinkina, V.B.; Tyakht, A.V.; Odintsova, V.Y.; Yarygin, K.S.; Kovarsky, B.A.; Pavlenko, A.V.; Ischenko, D.S.; Popenko, A.S.; Alexeev, D.G.; Taraskina, A.Y.; et al. Links of gut microbiota composition with alcohol dependence syndrome and alcoholic liver disease. Microbiome 2017, 5, 141. [Google Scholar] [CrossRef] [PubMed]
  77. Labrecque, M.T.; Malone, D.; Caldwell, K.E.; Allan, A.M. Impact of Ethanol and Saccharin on Fecal Microbiome in Pregnant and Non-Pregnant Mice. J. Pregnancy Child Health 2015, 2, 1000193. [Google Scholar] [CrossRef] [Green Version]
  78. Collado, M.C.; Isolauri, E.; Laitinen, K.; Salminen, S. Effect of mother’s weight on infant’s microbiota acquisition, composition, and activity during early infancy: A prospective follow-up study initiated in early pregnancy. Am. J. Clin. Nutr. 2010, 92, 1023–1030. [Google Scholar] [CrossRef]
  79. Galley, J.D.; Bailey, M.; Kamp Dush, C.; Schoppe-Sullivan, S.; Christian, L.M. Maternal obesity is associated with alterations in the gut microbiome in toddlers. PLoS ONE 2014, 9, e113026. [Google Scholar] [CrossRef]
  80. Gallardo, J.M.; Gómez-López, J.; Medina-Bravo, P.; Juárez-Sánchez, F.; Contreras-Ramos, A.; Galicia-Esquivel, M.; Sánchez-Urbina, R.; Klünder-Klünder, M. Maternal obesity increases oxidative stress in the newborn. Obesity 2015, 23, 1650–1654. [Google Scholar] [CrossRef]
  81. Deierlein, A.L.; Siega-Riz, A.M.; Chantala, K.; Herring, A.H. The association between maternal glucose concentration and child BMI at age 3 years. Diabetes Care 2011, 34, 480–484. [Google Scholar] [CrossRef] [Green Version]
  82. Mehta, S.H.; Kruger, M.; Sokol, R.J. Is maternal diabetes a risk factor for childhood obesity? J. Matern.-Fetal Neonatal Med. 2012, 25, 41–44. [Google Scholar] [CrossRef]
  83. Gaillard, R.; Durmuş, B.; Hofman, A.; Mackenbach, J.P.; Steegers, E.A.P.; Jaddoe, V.W.V. Risk factors and outcomes of maternal obesity and excessive weight gain during pregnancy. Obesity 2013, 21, 1046–1055. [Google Scholar] [CrossRef] [PubMed]
  84. Singh, S.; Karagas, M.R.; Mueller, N.T. Charting the Maternal and Infant Microbiome: What Is the Role of Diabetes and Obesity in Pregnancy? Curr. Diab. Rep. 2017, 17, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Stanislawski, M.A.; Dabelea, D.; Wagner, B.D.; Iszatt, N.; Dahl, C.; Sontag, M.K.; Knight, R.; Lozupone, C.A.; Eggesbø, M. Gut Microbiota in the First 2 Years of Life and the Association with Body Mass Index at Age 12 in a Norwegian Birth Cohort. mBio 2018, 9, e01751-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Santacruz, A.; Collado, M.C.; García-Valdés, L.; Segura, M.T.; Martín-Lagos, J.A.; Anjos, T.; Martí-Romero, M.; Lopez, R.M.; Florido, J.; Campoy, C.; et al. Gut microbiota composition is associated with body weight, weight gain and biochemical parameters in pregnant women. Br. J. Nutr. 2010, 104, 83–92. [Google Scholar] [CrossRef] [Green Version]
  87. Aagaard, K.; Ma, J.; Antony, K.M.; Ganu, R.; Petrosino, J.; Versalovic, J. The placenta harbors a unique microbiome. Sci. Transl. Med. 2014, 6, 237ra65. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  88. Bassols, J.; Serino, M.; Carreras-Badosa, G.; Burcelin, R.; Blasco-Baque, V.; Lopez-Bermejo, A.; Fernandez-Real, J.-M. Gestational diabetes is associated with changes in placental microbiota and microbiome. Pediatr. Res. 2016, 80, 777–784. [Google Scholar] [CrossRef] [Green Version]
  89. Zheng, J.; Xiao, X.; Zhang, Q.; Mao, L.; Yu, M.; Xu, J.; Wang, T. The Placental Microbiota Is Altered among Subjects with Gestational Diabetes Mellitus: A Pilot Study. Front. Physiol. 2017, 8, 675. [Google Scholar] [CrossRef] [Green Version]
  90. Hu, J.; Nomura, Y.; Bashir, A.; Fernandez-Hernandez, H.; Itzkowitz, S.; Pei, Z.; Stone, J.; Loudon, H.; Peter, I. Diversified microbiota of meconium is affected by maternal diabetes status. PLoS ONE 2013, 8, e78257. [Google Scholar] [CrossRef] [Green Version]
  91. Dominguez-Bello, M.G.; Blaser, M.J.; Ley, R.E.; Knight, R. Development of the human gastrointestinal microbiota and insights from high-throughput sequencing. Gastroenterology 2011, 140, 1713–1719. [Google Scholar] [CrossRef]
  92. Kumbhare, S.V.; Patangia, D.V.V.; Patil, R.H.; Shouche, Y.S.; Patil, N.P. Factors influencing the gut microbiome in children: From infancy to childhood. J. Biosci. 2019, 44, 49. [Google Scholar] [CrossRef]
  93. Kuhle, S.; Woolcott, C.G. Caesarean section is associated with offspring obesity in childhood and young adulthood. Evid. Based Med. 2017, 22, 111. [Google Scholar] [CrossRef] [PubMed]
  94. Rutayisire, E.; Wu, X.; Huang, K.; Tao, S.; Chen, Y.; Tao, F. Cesarean section may increase the risk of both overweight and obesity in preschool children. BMC Pregnancy Childbirth 2016, 16, 338. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  95. Azad, M.B.; Konya, T.; Maughan, H.; Guttman, D.S.; Field, C.J.; Chari, R.S.; Sears, M.R.; Becker, A.B.; Scott, J.A.; Kozyrskyj, A.L.; et al. Gut microbiota of healthy Canadian infants: Profiles by mode of delivery and infant diet at 4 months. CMAJ 2013, 185, 385–394. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Rutayisire, E.; Huang, K.; Liu, Y.; Tao, F. The mode of delivery affects the diversity and colonization pattern of the gut microbiota during the first year of infants’ life: A systematic review. BMC Gastroenterol. 2016, 16, 86. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  97. Song, S.J.; Dominguez-Bello, M.G.; Knight, R. How delivery mode and feeding can shape the bacterial community in the infant gut. CMAJ 2013, 185, 373–374. [Google Scholar] [CrossRef] [Green Version]
  98. Jakobsson, H.E.; Abrahamsson, T.R.; Jenmalm, M.C.; Harris, K.; Quince, C.; Jernberg, C.; Björkstén, B.; Engstrand, L.; Andersson, A.F. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by caesarean section. Gut 2014, 63, 559–566. [Google Scholar] [CrossRef] [Green Version]
  99. Miralles, O.; Sánchez, J.; Palou, A.; Picó, C. A physiological role of breast milk leptin in body weight control in developing infants. Obesity 2006, 14, 1371–1377. [Google Scholar] [CrossRef]
  100. Pereira, P.F.; Alfenas, R.D.C.G.; Araújo, R.M.A. Does breastfeeding influence the risk of developing diabetes mellitus in children? A review of current evidence. J. Pediatr. 2014, 90, 7–15. [Google Scholar] [CrossRef] [Green Version]
  101. Palma, G.D.; Capilla, A.; Nova, E.; Castillejo, G.; Varea, V.; Pozo, T.; Garrote, J.A.; Polanco, I.; López, A.; Ribes-Koninckx, C.; et al. Influence of milk-feeding type and genetic risk of developing coeliac disease on intestinal microbiota of infants: The PROFICEL study. PLoS ONE 2012, 7, e30791. [Google Scholar] [CrossRef] [Green Version]
  102. Oddy, W.H. Breastfeeding, Childhood Asthma, and Allergic Disease. Ann. Nutr. Metab. 2017, 70 (Suppl. S2), 26–36. [Google Scholar] [CrossRef]
  103. Strand, T.A.; Sharma, P.R.; Gjessing, H.K.; Ulak, M.; Chandyo, R.K.; Adhikari, R.K.; Sommerfelt, H. Risk factors for extended duration of acute diarrhea in young children. PLoS ONE 2012, 7, e36436. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  104. Zivkovic, A.M.; German, J.B.; Lebrilla, C.B.; Mills, D.A. Human milk glycobiome and its impact on the infant gastrointestinal microbiota. Proc. Natl. Acad. Sci. USA 2011, 108 (Suppl. S1), 4653–4658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  105. Marcobal, A.; Sonnenburg, J.L. Human milk oligosaccharide consumption by intestinal microbiota. Clin. Microbiol. Infect. 2012, 18 (Suppl. S4), 12–15. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  106. Thurl, S.; Munzert, M.; Henker, J.; Boehm, G.; Müller-Werner, B.; Jelinek, J.; Stahl, B. Variation of human milk oligosaccharides in relation to milk groups and lactational periods. Br. J. Nutr. 2010, 104, 1261–1271. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  107. Jost, T.; Lacroix, C.; Braegger, C.P.; Chassard, C. New insights in gut microbiota establishment in healthy breast fed neonates. PLoS ONE 2012, 7, e44595. [Google Scholar] [CrossRef]
  108. Bäckhed, F.; Roswall, J.; Peng, Y.; Feng, Q.; Jia, H.; Kovatcheva-Datchary, P.; Li, Y.; Xia, Y.; Xie, H.; Zhong, H.; et al. Dynamics and Stabilization of the Human Gut Microbiome during the First Year of Life. Cell Host Microbe 2015, 17, 690–703. [Google Scholar] [CrossRef] [Green Version]
  109. Stewart, C.J.; Ajami, N.J.; O’Brien, J.L.; Hutchinson, D.S.; Smith, D.P.; Wong, M.C.; Ross, M.C.; Lloyd, R.E.; Doddapaneni, H.; Metcalf, G.A.; et al. Temporal development of the gut microbiome in early childhood from the TEDDY study. Nature 2018, 562, 583–588. [Google Scholar] [CrossRef]
  110. Bergström, A.; Skov, T.H.; Bahl, M.I.; Roager, H.M.; Christensen, L.B.; Ejlerskov, K.T.; Mølgaard, C.; Michaelsen, K.F.; Licht, T.R. Establishment of intestinal microbiota during early life: A longitudinal, explorative study of a large cohort of Danish infants. Appl. Environ. Microbiol. 2014, 80, 2889–2900. [Google Scholar] [CrossRef] [Green Version]
  111. Cabrera-Rubio, R.; Collado, M.C.; Laitinen, K.; Salminen, S.; Isolauri, E.; Mira, A. The human milk microbiome changes over lactation and is shaped by maternal weight and mode of delivery. Am. J. Clin. Nutr. 2012, 96, 544–551. [Google Scholar] [CrossRef] [Green Version]
  112. Collado, M.C.; Laitinen, K.; Salminen, S.; Isolauri, E. Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr. Res. 2012, 72, 77–85. [Google Scholar] [CrossRef] [Green Version]
  113. Baumann-Dudenhoeffer, A.M.; D’Souza, A.W.; Tarr, P.I.; Warner, B.B.; Dantas, G. Infant diet and maternal gestational weight gain predict early metabolic maturation of gut microbiomes. Nat. Med. 2018, 24, 1822–1829. [Google Scholar] [CrossRef] [PubMed]
  114. Ma, J.; Li, Z.; Zhang, W.; Zhang, C.; Zhang, Y.; Mei, H.; Zhuo, N.; Wang, H.; Wang, L.; Wu, D. Comparison of gut microbiota in exclusively breast-fed and formula-fed babies: A study of 91 term infants. Sci. Rep. 2020, 10, 15792. [Google Scholar] [CrossRef] [PubMed]
  115. Roger, L.C.; Costabile, A.; Holland, D.T.; Hoyles, L.; McCartney, A.L. Examination of faecal Bifidobacterium populations in breast- and formula-fed infants during the first 18 months of life. Microbiology 2010, 156, 3329–3341. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. Bridgman, S.L.; Azad, M.B.; Field, C.J.; Haqq, A.M.; Becker, A.B.; Mandhane, P.J.; Subbarao, P.; Turvey, S.E.; Sears, M.R.; Scott, J.A.; et al. Fecal Short-Chain Fatty Acid Variations by Breastfeeding Status in Infants at 4 Months: Differences in Relative versus Absolute Concentrations. Front. Nutr. 2017, 4, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  117. Yassour, M.; Vatanen, T.; Siljander, H.; Hämäläinen, A.-M.; Härkönen, T.; Ryhänen, S.J.; Franzosa, E.A.; Vlamakis, H.; Huttenhower, C.; Gevers, D.; et al. Natural history of the infant gut microbiome and impact of antibiotic treatment on bacterial strain diversity and stability. Sci. Transl. Med. 2016, 8, 343ra81. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  118. Nakayama, J.; Yamamoto, A.; Palermo-Conde, L.A.; Higashi, K.; Sonomoto, K.; Tan, J.; Lee, Y.-K. Impact of Westernized Diet on Gut Microbiota in Children on Leyte Island. Front. Microbiol. 2017, 8, 197. [Google Scholar] [CrossRef] [PubMed]
  119. Ignacio, A.; Fernandes, M.R.; Rodrigues, V.a.A.; Groppo, F.C.; Cardoso, A.L.; Avila-Campos, M.J.; Nakano, V. Correlation between body mass index and faecal microbiota from children. Clin. Microbiol. Infect. 2016, 22, e1–e8. [Google Scholar] [CrossRef] [Green Version]
  120. Scheepers, L.E.J.M.; Penders, J.; Mbakwa, C.A.; Thijs, C.; Mommers, M.; Arts, I.C.W. The intestinal microbiota composition and weight development in children: The KOALA Birth Cohort Study. Int. J. Obes. 2015, 39, 16–25. [Google Scholar] [CrossRef]
  121. Indiani, C.M.D.S.P.; Rizzardi, K.F.; Castelo, P.M.; Ferraz, L.F.C.; Darrieux, M.; Parisotto, T.M. Childhood Obesity and Firmicutes/Bacteroidetes Ratio in the Gut Microbiota: A Systematic Review. Child. Obes. 2018, 14, 501–509. [Google Scholar] [CrossRef]
  122. Ley, R.E.; Turnbaugh, P.J.; Klein, S.; Gordon, J.I. Microbial ecology: Human gut microbes associated with obesity. Nature 2006, 444, 1022–1023. [Google Scholar] [CrossRef]
  123. Vael, C.; Verhulst, S.L.; Nelen, V.; Goossens, H.; Desager, K.N. Intestinal microflora and body mass index during the first three years of life: An observational study. Gut Pathog. 2011, 3, 8. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  124. Xu, P.; Li, M.; Zhang, J.; Zhang, T. Correlation of intestinal microbiota with overweight and obesity in Kazakh school children. BMC Microbiol. 2012, 12, 283. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  125. Borgo, F.; Verduci, E.; Riva, A.; Lassandro, C.; Riva, E.; Morace, G.; Borghi, E. Relative Abundance in Bacterial and Fungal Gut Microbes in Obese Children: A Case Control Study. Child. Obes. 2017, 13, 78–84. [Google Scholar] [CrossRef] [PubMed]
  126. Karvonen, A.M.; Sordillo, J.E.; Gold, D.R.; Bacharier, L.B.; O’Connor, G.T.; Zeiger, R.S.; Beigelman, A.; Weiss, S.T.; Litonjua, A.A. Gut microbiota and overweight in 3-year old children. Int. J. Obes. 2019, 43, 713–723. [Google Scholar] [CrossRef] [PubMed]
Figure 1. The ratio of Bacteroides/Firmicutes—a biomarker of obesity succeptibility (created with www.BioRender.com, accessed on 6 February 2022).
Figure 1. The ratio of Bacteroides/Firmicutes—a biomarker of obesity succeptibility (created with www.BioRender.com, accessed on 6 February 2022).
Biomedicines 10 00645 g001
Figure 2. Factors influencing gut microbiome in children (created with www.BioRender.com, accessed on 6 February 2022).
Figure 2. Factors influencing gut microbiome in children (created with www.BioRender.com, accessed on 6 February 2022).
Biomedicines 10 00645 g002
Table 1. The factors influencing gut microbiome in children with obesity.
Table 1. The factors influencing gut microbiome in children with obesity.
FactorsInfluence on Infant’s Gut Microbiota, Authors, Year
Maternal factorsMaternal Dietfish oil consumptionhigher level of Enterococcus faecium, Bilophila wadsworthia and Bacteroides fragilis—a negative impact on the infant’s immune responses (Gibson et al., 2016 [73]).
high-fat dietreduced level of Bacteroidetes (Chu et al., 2016 [71])
probiotic-containing foodreduced risk of preterm delivery (Myhre et al., 2011 [74]), and allergic disorders during childhood (Bertelsen et al., 2014 [75]).
alcohol consumption
  • negative influence on both maternal and neonatal gut microbiome (Dubinkina et al. 2017, Labrecque et al., 2015, [76,77]).
  • increased risk for infections and other disorders later in life (Dubinkina et al. 2017, Labrecque et al., 2015, [76,77]).
Maternal healthoverweight and obesityincreased exposure to oxidative stress (Collado et al., 2010, Galley et al., 2014, Gallardo et al., 2015, [78,79,80]).
diabetes or increased body mass indexhigher risk of developing diabetes and obesity (Deierlein et al., 2011, Mehta et al., 2012, Gaillard et al., 2013, [81,82,83]).
microbiome in overweight or obese mothersincreased risk for early obesity in infant => their microbial composition predicts obesity development later in life (Stanislawski et al., 2018 [85]).
Dysbiosis of the placental microbiotamothers with diabetesabundance of Bacteroidetes, Lachnospiraceae family and Parabacteriodes and decreased levels of Proteobacteria in meconium samples (Hu et al., 2013 [90]).
Delivery modeVaginallydirect contact with maternal vaginal and gut microbiotainfant’s gut microbiome is similar to the vaginal microbial composition being dominated by the genera Lactobacillus, Sneathia and Prevotella (Dominguez-Bello et al., 2011 [91]).
Cesarean sectioncontact with maternal skin microbiome
  • neonate’s gut microbiome resembles the mother’s skin microbiome and nosocomial surroundings (Kumbhare et al., 2019 [92]).
  • risk for obesity during childhood (Kuhle et al., 2017, Rutayisire et al., 2016 [93,94]).
  • lower diversity of the gut microbial produce lower levels of both Bifidobacteria and Escherichia-Shigella and absence of Bacteroides (Azad et al., 2013; Rutayisire et al., 2016; Song et al., 2013; Jakobsson et al., 2014 [95,96,97,98]).
  • gut microbial community mainly composed by the genera Staphylococcus, Propionibacterium and Corynebacterium (Dominguez-Bello et al., 2011 [91]).
Feeding patternsBreastfeeding
  • promotes Bifidobacteria and Bacteorides growth (Marcobal et al., 2012 [105]) and regulate infant’s wellbeing modulators of innate immune responses and anti-inflammatory properties (Thurl et al., 2010 [106]).
  • B. longum, B. breve, B. infantis and B. pseudocatenulatum (Jost et al., 2012; Bäckhed et al., 2015; Stewart et al., 2018; [107,108,109]) along with lactic acid bacteria, such as Enterococcus, Lactobacillus and Clostridium species (Bergström et al., 2014 [110]).
  • maternal health is a modulator of human milk content influencing the infant’s gut microbiome (Kumbhare et al., 2019 [92]).
  • maternal obesity alter human milk microbiome determining lower levels of Bifidobacterium levels and higher levels of Staphylococcus (Cabrera-Rubio et al., 2012; Collado et al., 2012 [111,112]).
Formula-feeding
  • diversity of gut microbiota at the age of 40 days was higher than in breast-fed group (Ma et al., 2020 [114]), similar to the gut microbiota of older children (Roger et al., 2010 [115]; Bridgman et al., 2017 [116]).
  • the predominant genus in both breastfed and formula-fed infants are Bifidobacterium and Enterobacteriaceae which decrease with age (Ma et al., 2020 [114], Yassour et al., 2016 [117]
  • increased levels of both Clostridia and Veillonella spp. Bäckhed et al., 2015 [108]; Ma et al., 2020 [114]; Bridgman et al., 2017 [116]).
Weaning and diet influence on gut microbiomeWeaning
  • modulating microbial community determine similar microbial composition to that of adults (Bäckhed et al., 2015; Stewart et al., 2018 [108,109]).
  • at 9 to 18 months will decrease of Lactobacilli, Bifidobacteria and Enterobacteriaceae and an increase in Bacteroides and Clostridium species (Cabrera-Rubio et al., 2012 [110]).
Type of dietchildren with high-fat westernized dietdecrease in Prevotellaceae-obesity development during childhood (Nakayama et al., 2017 [118]).
children with carbohydrate-based dietincrease in Prevotellaceae and a decrease in Bacteroidaceae (Nakayama et al., 2017 [118]).
Publisher’s Note: MDPI stays neutral with regard to jurisdictional claims in published maps and institutional affiliations.

Share and Cite

MDPI and ACS Style

Meliț, L.E.; Mărginean, C.O.; Săsăran, M.O. The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota. Biomedicines 2022, 10, 645. https://doi.org/10.3390/biomedicines10030645

AMA Style

Meliț LE, Mărginean CO, Săsăran MO. The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota. Biomedicines. 2022; 10(3):645. https://doi.org/10.3390/biomedicines10030645

Chicago/Turabian Style

Meliț, Lorena Elena, Cristina Oana Mărginean, and Maria Oana Săsăran. 2022. "The Yin-Yang Concept of Pediatric Obesity and Gut Microbiota" Biomedicines 10, no. 3: 645. https://doi.org/10.3390/biomedicines10030645

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop