Skip to content
Publicly Available Published by De Gruyter December 10, 2019

Childhood obesity: an overview of laboratory medicine, exercise and microbiome

  • Olga Scudiero EMAIL logo , Raffaela Pero , Annaluisa Ranieri , Daniela Terracciano ORCID logo , Fabio Fimiani , Arturo Cesaro , Luca Gentile , Eleonora Leggiero , Sonia Laneri , Elisabetta Moscarella , Cristina Mazzaccara , Giulia Frisso , Giovanni D’Alicandro , Giuseppe Limongelli , Lucio Pastore , Paolo Calabrò and Barbara Lombardo EMAIL logo

Abstract

In the last few years, a significant increase of childhood obesity incidence unequally distributed within countries and population groups has been observed, thus representing an important public health problem associated with several health and social consequences. Obese children have more than a 50% probability of becoming obese adults, and to develop pathologies typical of obese adults, that include type 2-diabetes, dyslipidemia and hypertension. Also environmental factors, such as reduced physical activity and increased sedentary activities, may also result in increased caloric intake and/or decreased caloric expenditure. In the present review, we aimed to identify and describe a specific panel of parameters in order to evaluate and characterize the childhood obesity status useful in setting up a preventive diagnostic approach directed at improving health-related behaviors and identifying predisposing risk factors. An early identification of risk factors for childhood obesity could definitely help in setting up adequate and specific clinical treatments.

Introduction

Obesity is a complex multifactorial disease characterized by irregular or excessive fat accumulation and is a result of an imbalance between energy intake and energy expenditure combined with a genetic predisposition for weight gain [1], [2].

In the last decade, a significant increase of childhood obesity incidence unequally distributed within countries and population groups has been observed. In 2010, 43 million children worldwide were obese, thus childhood obesity represents an important public health problem associated with several health and social consequences [3], [4].

The first clinical standard measure to estimate the level of adiposity in overweight children is the body mass index (BMI), that requires being adjusted for both age and gender [2].

Obese children show over 50% probability of becoming obese adults, while non-obese children show a probability of about 10% of developing pathologies typical of obese adults, that include type 2-diabetes, dyslipidemia and hypertension, and psychosocial and emotional issues [5], [6].

Reduced physical activity and increased sedentary activities, may also result in increased caloric intake and/or decreased caloric expenditure [7], [8].

In our review, we intende to identify and describe a specific panel of parameters that allows the evaluation and characterization of childhood obesity status (Figure 1) and, therefore, set up a preventive diagnostic approach directed to ameliorate health-related behaviors and identify predisposing risk factors.

Figure 1: 
Scheme of diagnostic parameters aimed at evaluating childhood obesity.
Figure 1:

Scheme of diagnostic parameters aimed at evaluating childhood obesity.

In particular, we focus our interest on epigenetic changes that may influence the process of adipogenesis and on genetic factors predisposing to obesity. Moreover, we also describe how the main biochemical parameters and cytokines levels are altered in obese children, and also identify the most specific markers of inflammatory status. We also underline the influence of obesity on the development of a number of pathological conditions including endocrine disorders and increased cardiovascular risk; in addition, we focus on how obesity modulates the gut microbiome. Finally, we described therapeutic interventions widely used to regulate body fat.

The evaluation and the understanding of the comprehensive obesity scenario and the identification of effective markers may lead to an early identification of risk factors for childhood obesity in order to set up adequate and specific clinical treatments.

miRNA

Recent studies showed that an altered regulation of microRNAs (miRNAs) causes their expression variations thus modifying the normal expression of genes involved in adipogenesis.

In particular, miRNAs represent key regulators of adipogenesis and metabolism processes and are involved in both differentiation of adipocytes and the regulation of mature adipocyte functions, including lipolysis, glucose uptake and insulin sensitivity [9], [10]. A large amount of evidence strongly suggest that dysregulation of miRNAs affects the status and function of different tissues and organs, probably contributing to metabolic abnormalities associated with obesity and obesity-related diseases [11].

MiRNAs are endogenous small non-coding RNAs 18–25 nucleotide-long that regulate gene expression through translation repression or degradation of target mRNAs at the post-transcriptional level, preferentially binding to the 3’ UTR regions, therefore contributing to the regulation of many biological processes. Each mature miRNA is partially complementary to multiple target mRNAs and through RNA-induced silencing complex (RISC) may identify the target mRNAs for inactivation [12], [13].

More than 2000 different miRNAs described in humans and many studies show that miRNAs may influence the expression of 30%–50% of protein encoded by murine and human genes [14]. Some miRNAs have a tissue-specific expression and mRNA may include multiple binding sites for different miRNAs originating an intricate regulatory network. In fact, miRNAs are involved in many cellular processes such as cell proliferation, differentiation, control of stem cell self-renewal, DNA repair, apoptosis, metabolism, development and tumor metastasis [13], [15].

Recent studies underline the role of miRNAs in obese adults and children (Table 1). Several miRNAs, such as miR-130 or miR-27b, regulate adipogenic differentiation by targeting the expression of peroxisome proliferator-activated receptor-γ (PPARγ), the master regulator of adipogenesis [16]. Upregulation of miR-125b-5p during human adipogenesis directly inhibits this biological process, downregulating the anti-adipogenic matrix metalloproteinase 11 (MMP-11) [17]. In addition, Jiang et al. observed that several adipokines, including interleukin (IL)-6, tumor necrosis factor (TNF)α, leptin, and free fatty acids, induce expression of miR-378 in human adipocytes [23]. miR-378 plays an important role in adipose differentiation, mitochondrial metabolism and systemic energy homeostasis. In particular, it stimulates the accumulation of triacylglycerol increasing the transcriptional activity of C/EBPα and C/EBPβ on adipocyte gene promoters; moreover, it is assumed that miR-378 being encoded within the PGC-1β (peroxisome proliferator-activated receptor γ coactivator 1β) gene, a transcriptional co-activator that regulates the metabolism and mitochondrial biogenesis, it can counterbalance the metabolic actions of PGC-1β [18]. Some evidence also shows that obesity induces over-expression of miR-14, which inhibits insulin-stimulated AKT activation leading to impairment of glucose metabolism [17].

Table 1:

Summary of circulating miRNAs involved in childhood obesity.

miRNAs Target/process References
miR-130, miR-27b Adipogenic differentiation targeting PPARγ [16]
miR-125b-5p MMP-11 [17]
miR-378 Adipocyte development and differentiation [18]
miR-14 Insulin-stimulated AKT [17]
miR-140-5p, miR-142-3p, miR-222 Morbidly obese patients [19]
miR-532-5p, miR-125b, miR-130b, miR-221, miR-15a, miR-423-5p, miR-520c-3p Morbidly obese patients [19]
miR-23a, miR-27a, miR-130, miR-195, miR-197, miR-320a, and miR-509-5p Metabolic syndrome [11]
miR-122, miR-324e3p, miR-375, and miR-652

miR-625
Pre-gestational and gestational obesity [20]
miR-335, miR-143, and miR-758, miR-27, miR-33, miR-378, and miR-370 Lipid metabolism [21]
miR-486-5p, miR-486-3p, miR-142-3p, miR-130b, miR-423-5p, miR-532-5p, miR-140-5p, miR-16-1, miR-222, miR-363, miR-122, miR-221, miR-28-3p, miR-125b and miR-328 Insulin resistance and lipid metabolism [22]

The attention of latest studies is on the identification of extracellular circulating miRNAs in blood, serum and plasma; alteration of expression of miRNAs in pathological conditions offers a great potential for the identification of novel biomarkers and candidate therapeutic targets [24]. In fact, differential profiles of circulating miRNAs are reported in subjects with obesity and different metabolic disorders [11]. A study conducted by Ortega et al. showed in obese patients a marked increase of circulating miR-140-5p, miR-142-3p, miR-222 and a decrease of miR-532-5p, miR-125b, miR-130b, miR-221, miR-15a, miR-423-5p, miR-520c-3p. In the same study changes in circulating miRNA levels in individuals undergoing bariatric surgery with subsequent weight loss were evaluated; in these subjects a significant decrease of circulating miR-140-5p, miR-122, miR-193a-5p, miR-16-1 and an increase of miR-221 and miR-199a-3p were observed [19].

In a different study altered levels in circulating miR-23a, miR-27a, miR-130, miR-195, miR-197, miR-320a and miR-509-5p have been associated with metabolic syndrome [11]. Different patterns of circulating miRNAs have also been described in pre-gestational and gestational obesity. In fact, significant reduction of miR-122, miR-324e3p, miR-375and miR-652 and increased levels of miR-625 in both pregestational obese and gestational obese pregnant women have been observed [20]. Recently, Iacomino et al. identified three differentially expressed miRNAs (miR-31-5p, miR-2355-5p and miR-206) in plasma samples of obese children compared to controls [25]. In a recent study, Can et al. reported that miR-335, miR-143, and miR-758 levels were lower in obese children, whereas miR-27, miR-33, miR-378 and miR-370 levels resulted in higher levels. The different expression of these circulating miRNAs may be responsible for the high triglycerides (TGs) and low-density lipoprotein-cholesterol (LDL-C) levels, and for the low level of high-density lipoprotein-cholesterol (HDL-C) [21]. In a study conducted by Prats-Puig et al. the changes in plasma circulating miRNA levels in prepuberal obese children were evaluated showing altered levels of specific miRNAs, i.e. an increase of miR-486-5p, miR-486-3p, miR-142-3p, miR-130b, miR-423-5p, miR-532-5p, miR-140-5p, miR-16-1, miR-222, miR-363 and miR-122 and a reduction of miR-221, miR-28-3p, miR-125b and miR-328. Moreover, the circulating levels of these miRNAs were significantly associated with BMI, percent fat mass and waist circumference (WC), parameters of fat distribution and with laboratory variables, such as homeostasis model assessment of insulin resistance (HOMA-IR), high-molecular-weight adiponectin, C-reactive protein (CRP), and circulating lipids. In the same study, during patient follow-up, changes in plasma concentrations of the same circulating miRNAs were observed as a result of BMI increase or decrease in obese children [22].

In addition, a large number of miRNAs are associated with pathogenesis of IR, endothelial dysfunction, serum lipid level alterations, inflammation and other metabolic dysfunctions in children. The discovery that some miRNAs are enriched in the hypothalamus suggest their primary role in the hypothalamic regulation of energy intake, expenditure and body weight control [17].

Interestingly, some studies also indicate that miRNAs may be regulated by diet and lifestyle factors [26], [27]. MiRNAs therefore represent an exciting, novel and promising class of markers that could improve the early diagnosis, stratification and therapy of metabolic diseases such as obesity.

The biological role of miRNAs in obesity could be well clarified through the use of bioinformatics tools such as miRBase, miRTarbase, miRanda and others that allow predicting their involvement in important pathways such as lipid metabolism and adipocytes differentiation and to identify their target [25].

The very early detection of changes in circulating miRNA levels represent a promising strategy for characterizing obese children and evaluating how dietary factors may influence obesity through the modulation of expression of miRNAs. In fact, the presence of risk factors at an early age is associated with the subsequent development of metabolic diseases in adulthood and the prevention of obesity in children can improve their health and reduce the development of obesity-related complications in adults.

Genetics and epigenetics of obesity

Genetic factors play a relevant role in obesity risk, with percentages ranging from 6 to 85% [28]. Studies on twins have been conducted to understand the genetic component of obesity showing that monozygotic twins subjected to excessive nutrition and regular exercises present similar changes in body weight, body composition and energy expenditure compared to dizygotic twins [29]. These results put forward the inheritance of obesity and adiposity [30]. Genetic predisposition of obesity has been classified into monogenic syndromic, monogenic non-syndromic and polygenic with main effects on body fat mass [31]. Monogenic obesity is a rare condition caused by recessive mutations in a single gene. In particular, monogenic obesity is divided into non-syndromic, when it is not associated with syndromes and it involved genes encoding for enzymes and receptors with a physiologic role in the development of the hypothalamus and the leptin melanocortin system, and syndromic when it is associated with different syndromes such as Bardet-Biedl, Alström, Prader-Willi and Carpenter syndromes, and microdeletion syndromes (e.g. 1p36, 2q37, 6q16 and 9q34) [31], [32]. Polygenic forms represent the majority of cases of obesity attributable to genetic interactions with environmental, cultural and lifestyle factors. In fact, obesity is rarely caused by alterations in single genes, being the result of a complex genetic background in which the effect of genes on the pathogenesis are stronger when combined with additional genetic alterations and environmental factors [32].

Genome-wide association studies (GWAS) have allowed the identification of more than 250 genes/loci associated with obesity, discovering new alterations with previously unknown effects.

Among these, variants of the fat mass and obesity-associated gene (FTO) gene showed an important role in obesity and pathogenesis developing type 2-diabetes, as these variations were associated with a higher BMI, fat mass index and leptin concentrations during puberty; in addition, they have a role in the regulation of appetite, probably through the influence of nearby genes such as retinitis pigmentosa GTPase regulator-interacting protein-1 like (RPGRIP1L) and Iroquois homeobox 3 (IRX3). GWAS studies allowed the identification of a locus near the gastric inhibitory polypeptide receptor (GIPR), the incretin receptor, that involves a variation in postprandial insulin secretion contributing to the development of obesity and sustaining the involvement of the gut microbiome in obesity [31].

In order to identify new genetic variations, high throughput molecular diagnostics tools, such as array-comparative genomic hybridization (a-CGH) and next generation sequencing (NGS) technology, are extremely useful tools [33], [34], [35], [36], [37]. These approaches have allowed identifying the association of many genes with obesity that play important biological roles in this disorder. A study conducted by Locke et al. identified 97 loci associated with BMI, 56 of which were previously unknown. Many of these loci were in or near genes that play a role in different biological processes, such as neuronal development: fas apoptotic inhibitory molecule 2 (FAIM2), polypyrimidine tract binding protein 2 (PTBP2) and suppression of tumorigenicity 5 (ST5), hypothalamic expression and regulatory function: transmembrane protein 18 (TMEM18), secretogranin III (SCG3) and gastrin releasing peptide (GRP); limb development: retinoic acid receptor-β (RARB), transcription factor AP-2β (TFAP2B) and LIM homeobox transcription factor 1β (LMX1B), lipid biosynthesis and metabolism cytochrome P450 family 27 subfamily A member 1 (CYP27A1), cytochrome P450 family 17 subfamily A member 1 (CYP17A1) and subfamily 1 group H member 3 nuclear receptor (NR1H3), cell proliferation and survival: parkin RBR E3 ubiquitin protein ligase (PARK2) and olfactomedin 4 (OLFM4) and immune system: interleukin 22 receptor subunit α2 (IL22RA2), mannosidase α class 1A member 1 (MAN1A1), interferon-γ receptor 1 (IFNGR1). Other genes have previously been associated with severe early-onset obesity: brain derived neurotrophic factor (BDNF), melanocortin 4 receptor (MC4R), SH2B adaptor protein 1 (SH2B1), TUB bipartite transcription factor (TUB) and proopiomelanocortin (POMC). It emerged that the common variants of these 97 loci affect almost 21% of cases of obesity [38]. Moreover, several studies have identified polymorphisms, within genes earlier known to be involved in monogenic obesity that also contribute to polygenic obesity. For example, mutations with homozygous loss of function in the leptin (LEP) gene or of its receptor LEPR are associated with hyperphagia and severe early-onset obesity in humans [39]. Proprotein convertase subtilisin/kexin type 1 gene (PCSK1) encodes the prohormone convertases 1 (PC1/3) expressed in neural and endocrine tissues and are highly expressed in the hypothalamus where they act on cleavage processing of POMC to α-melanocyte-stimulating hormone (α-MSH) [40]. PC1/3 deficiency is associated with hyperphagia, central diabetes insipidus, severe malabsorptive diarrhea and other endocrines dysfunctions while null mutations in PCSK1 cause a rare non-syndromic form of obesity [41]. POMC is a pro-peptide expressed in the hypothalamus, pituitary gland and brainstem and generates a family of melanocortin peptides such as the adrenocorticotropic hormone (ACTH) and α-, β-, and γ-melanocyte-stimulating hormones (MSH), with important roles in skin pigmentation, the control of adrenal growth and energy balance. Mutations that inactivate the POMC gene lead to severe early-onset obesity in children, adrenal insufficiency and pigmentation abnormalities [42].

Inactivating mutations at the heterozygote status in genes such as BDNF, neurotrophic receptor tyrosine kinase 2 (NTRK2), SIM bHLH transcription factor 1 (SIM1), MC4R, SH2B1, melanocortin 2 receptor accessory protein 2 (MRAP2) and LDL receptor related protein 2 (LRP2) have also been associated with severe early-onset obesity [32].

A study conducted by Lee showed that genetic and epigenetic modifications of protein tyrosine phosphatase, receptor type N2 (PTPRN2) gene could also play an important role in the development of obesity in childhood. PTPRN2 is localized on the membrane of insulin-containing dense-core vesicles and is involved in insulin secretion in response to glucose stimuli and in the metabolism including obesity and type 2-diabetes. Genome-wide copy number variation (CNV) analysis has led to the identification of novel candidate loci including PTPRN2 associated in pediatric obesity. The PTPRN2 gene is epigenetically regulated in several processes such as tumor pathogenesis and is significantly more hypermethylated in obese cases with respect to controls [43]. Chen et al. reported that the intrauterine environment in the human placenta influences the different gene expression and methylation of PTPRN2 and consequently the prenatal growth patterns and birthweight [44]. These results showed a correlation between genetic and epigenetic events of the PTPRN2 gene and childhood obesity. An additional array-CGH analysis of 100 children with syndromic obesity, defined the association with at least another feature such as intellectual disability (ID), facial dysmorphism or congenital malformations. Children between 1 and 18 years old all presented obesity and at least one other criterion such as ID, facial dysmorphism or a major malformation. Array-CGH performed in this cohort identified 60 CNVs in 42% of children and 22% of patients with pathogenic or potentially pathogenic CNVs. The CNVs identified in this study are: (a) deletion in 1p36.3 implicated in obesity found in the patient together with one duplication in 1q25.3 including the prostaglandin-endoperoxide synthase 2 gene (PTGS2), deficiency which was associated with reduced adiposity in mice; (b) 2p25.3 deletion associated with ID and obesity; (c) 1q21.1 susceptibility locus associated with several features, metabolic syndrome and obesity; (d) 16p11.2 distal deletion including SH2B1 associated with developmental delay and obesity; (e) 17p13.3 duplication associated with developmental delay, macrosomia, overweight and characteristic facial abnormalities. Additionally, other recurrent loci not always associated with obesity were found in this cohort and considered as clinically relevant: 16p13.1 duplication associated with neuropsychiatric disorders with a small 9p21.1 deletion including the leucine rich repeat and Ig domain containing 2 gene (LINGO2) potentially associated with obesity in the same patient, 3q29 locus associated with mild to moderate ID and sometimes with obesity and includes the 3-hydroxybutyrate dehydrogenase 1 gene (BDH1) encoding a protein which catalyzes the interconversion of two major ketone bodies produced during fatty acid catabolism together with one deletion in 4q32.2q32.3 encompassing the methylsterol monooxygenase 1 gene (MSMO1) linked to plasma lipid concentration and cholesterol biosynthesis, the minimal critical region of the 3q13.31 microdeletion syndrome associated with ID but also with obesity, 22q11.2 “distal” deletion associated in some cases with obesity and duplication in 15q11.2q13.1 containing the imprinted genes involved in Prader-Willi and Angelman syndromes in the same patient and finally a child with Asperger syndrome that had one deletion in 7p21.3 including mitochondrial complex associated NDUFA4 (NDUFA4), a gene highly expressed in brain and associated with autism. The following genes included in these CNVs were of particular interest: suppressor of cytokine signaling 6 (SOCS6), perilipin 2 (PLIN2), cadherin 13 (CDH13), contactin associated protein like 2 (CNTNAP2), SRY-box 3 (SOX3), and acyl-CoA oxidase like (ACOXL) [45].

These studies sustain an association between obesity and ID due to common genes involved in neurodevelopment that may affect the central circuits involved in the energy balance [45]. As obesity is characterized by genetic heterogeneity and multifactorial etiology, there are several causes that contribute to the pathogenicity of the disease, thus further studies are necessary to identify the correlation between new loci and obesity and to discover new variants. Furthermore, adequate molecular diagnosis of monogenic obesity is fundamental for patients and their relatives, while the clinicians could provide appropriate genetic counseling to the patient in order to improve their living conditions.

Biochemical profile

The evaluation of more biochemical parameters may be informative as early markers for the persistence of obesity from child to adult. Among other alterations, dyslipidemia is commonly found in children with obesity. Cook et al., reported that 45.8% of overweight children showed high levels of LDL and TGs and low levels of HDL with a higher prevalence in boys [46].

Non-HDL-C is a better indicator of persistent dyslipidemia and atherosclerosis in children and adults [47]. Typically, dyslipidemia in obesity is characterized by a high triglyceride level and low HDL level.

Obese children, who consume large amounts of sugar-sweetened beverages, show TG levels ranging from 150 to 400 mg/dL. These patients respond typically to dietary modification; if there is no response to diet, further evaluation is needed. The TG/HDL ratio is correlated to IR and early organ damage (heart, liver and carotid) [48], [49], [50].

Also, liver disease is a significant comorbidity for obesity at a young age. Elevated alanine aminotransferase (ALT) levels have been considered a marker for non-alcoholic fatty liver disease (NAFLD) in obese children. The prevalence of NAFLD in obese children is around 40% [51]. Bright liver on ultrasound examination, with or without elevation of ALT (>26 U/L in boys and >22 U/L in girls), suggests NAFLD [52], [53], [54]. However, determination of ALT alone may underestimate the extent of liver injury. Elevated levels of liver enzymes appear to be seen with increased age and WC. NAFLD is more prevalent in children with a higher WC and is directly proportional to age, weight and height [55], [56].

The patients’ family history is a significant predictor for a number of co-morbidities. Parental obesity could be considered a critical factor in the development of obesity due to genetic factors and family food choices. Evaluation of transaminases is suggested in all children and adolescents with obesity starting at the age of 6 years [57], [58], [59], [60]. NAFLD may also be screened in overweight children presenting the waist-to-height ratio >0.5, and the assessment yearly repeated [61].

Moreover, in obesity, the expanded and inflamed adipose tissue causes an imbalance in the secretion of cytokines, adipokines, growth factors and other biological mediators that have a particular role in liver fibrosis [62], [63], [64]. In particular, TGF-β1, PDGF-BB, leptin, and ferritin induce the production and accumulation of extracellular matrix (ECM) which has a crucial role in liver fibrogenesis [65] involving three major signaling pathways including MAP kinase, PI3K/Akt, and NF-κB. Liver fibrosis can be modulated by positively promoting resolution. The primary target for therapeutic strategy is to reduce chronic parenchymal injury induced by the primary etiology. For this purpose, several antioxidants and hepatoprotective agents have been reported to significantly increase liver fibrosis in rodent models (vitamin E, glutathione, N-acetylcysteine, S-adenosyl-methionine, resveratrol, curcumin, herbal supplements, inhibitors of NADPH oxidase isoforms and many others) [66], [67]. Several non-invasive tests have been made available for the assessment of liver fibrosis, such as the aspartate transaminase to platelet ratio index, enhanced liver fibrosis, FIB-4, FibroTest, Forns index and FibroScan [68], [69], [70].

Therefore, the identification of targets for anti-fibrotic therapy and non-invasive methodologies and biomarkers could be useful in monitoring the fibrotic progression of liver in obesity, and potentially the response to treatment [65], [71].

It is essential to evaluate a possible impairment in glucose metabolism in children with obesity. Fasting blood glucose determination is recommended in all overweight children and adolescents starting at the age of 6, as the first step for the identification of prediabetic conditions and type 2-diabetes. The oral glucose tolerance test (OGTT) is indicated after the age of 10 or at the onset of puberty. Impaired fasting glucose is determined as a fasting plasma glucose of ≥100 mg/dL but <126 mg/dL on repeated determinations. Impairment in glucose tolerance is defined as plasma glucose ≥140 mg/dL but <200 mg/dL on an OGTT at a 2-h timepoint [72].

Type 2-diabetes is the endpoint of metabolic decompensation that may evolve over months to years. In IR, the body can produce insulin but the muscle, fat and liver do not respond appropriately and thus cannot easily absorb glucose from the bloodstream, with progression from IR to prediabetes with impaired glucose tolerance (IGT) and impaired fasting glucose over time [73].

NAFLD, in combination with a high value of TGs, fasting blood glucose or TG to HDL-C ratio (TG/HDL-C) is associated with an increased risk of IGT and therefore, an OGTT may be considered in the latter cases [72], [74]. Besides, markers of hepatic fat content (serum γ-glutamyltransferase [GGT] activity and other liver enzymes) have been shown to predict the incidence of type 2-diabetes, IR and cardiovascular disease independently of obesity [75], [76].

Screening for a kidney profile is not recommended in non-diabetic and non-hypertensive children and adolescents with obesity. However, in adults, obesity is an independent risk factor for chronic kidney disease. Indeed, obesity complications (i.e. hypertension, dyslipidemia, IR, type 2-diabetes, inflammatory state, autonomous system dysfunction) can alter kidney function. The obesity-related glomerulopathy takes place in obese patients, and ameliorates after weight loss [77], [78]. Therefore, obesity is likely to be a risk factor for chronic renal disease in children too. Indeed, children with the renal disease have BMIs higher than the healthy population and kidneys transplanted from obese donors have reduced glomerular filtration and a higher rate of dysfunction than the kidneys obtained from regular weight donors [79], [80]. In the light of current evidence, assessment of microalbuminuria is not recommended in non-diabetic and non-hypertensive obese children. Cases of severe obesity that may be associated with proteinuria remain to be evaluated individually [81], [82], [83], [84], [85].

The most common metabolic complication in children with excess weight is IR. An association between obesity and IR and between obesity and diabetes has also been demonstrated in children, also IR has been recognized as a condition preceding the onset of type 2 diabetes (in adults). IR in humans does not seem to depend on obesity, as severe IR also characterizes patients lacking subcutaneous fat, such as those with lipodystrophy [86]. A linear correlation has been found between liver fat content and direct measures of hepatic IR independent of obesity. Thus, fatty liver might help to explain why some, but not all, obese individuals have IR and why even lean individuals may be IR and thereby at risk of developing diabetes-related metabolic disorders. Growing evidence suggests that GGT is not only a marker of fatty liver but also a marker of oxidative stress. Probably the occurrence of GGT-mediated redox reactions plays a direct role in the pathogenesis of atherogenic dyslipidemia and poor glycemic control, independently of the presence of fatty liver, possibly through the induction of chronic inflammation and IR. It is therefore possible that obesity itself may not be a sufficient risk factor for atherogenic dyslipidemia or poor glycemic control in type 2 diabetes [75], [87].

Insulin metabolism is tightly linked with the insulin-like growth factor (IGF) system, an evolutionarily conserved group of factors exerting long-term effects on growth. IGF-1 regulates cell proliferation, differentiation, migration and survival in cells.

Hyperinsulinemia leads to higher bioavailability of free, active IGF-1 by downregulating the synthesis of IGF binding proteins (IGFBP-1, IGFBP-2) and by upregulating hepatic IGF-1 synthesis. Different biomarkers, including fasting insulin, C-peptide, which is cleaved from proinsulin and is considered an indicator of endogenous insulin secretion with a longer half-life than insulin itself and IGF-1 as well as IGF-binding proteins have been investigated. Fasting insulin and C-peptide have been shown to correlate positively with BMI. However, the relationship of IGF-1 with obesity is less evident because of the above-described effects of obesity on IGF-1 synthesis on the one hand, and IGF-1 bioavailability on the other hand. Studies relating to total IGF-1 to obesity found inverse associations with BMI. Few studies investigated obesity or BMI as a determinant of free IGF-1, but a small cross-sectional study showed that free IGF-1 but not total IGF-1 was higher in obese than in normal-weight individuals. In a meta-analysis of prospective cohort studies, higher fasting insulin concentrations were associated with higher risk of hypertension and coronary heart disease (CHD) but not with stroke. C-peptide has been shown to predict total and cardiovascular mortality in non-diabetic individuals better than other measures of IR including fasting insulin, blood glucose and the HOMA-IR [88]. The biochemical parameters mentioned should be useful to characterize the childhood condition, to evaluate the risk of the disease and also follow-up the pathology progression.

Cytokines

Adipose tissue is a complex and dynamic endocrine organ active in cellular reactions and metabolic homeostasis that synthesize and release hormones, cytokines, extracellular matrix proteins and growth and vasoactive factors, collectively termed adipokines. Mechanisms underlying adipose tissue dysfunction include: adipocyte hyperplasia and hypertrophy with adipose tissue expansion, immune cells infiltration into the adipose tissue and ECM remodeling allowing adequate tissue expansion to adapt the excessive caloric intake [89]. The adipose tissue expansion induces activation of the immune response both innate and adaptive. Several studies revealed an infiltration of different immune cells such as lymphocytes, eosinophils, neutrophils, mast cells, foam cells [90] and in particular an increase in the number of macrophages in adipose tissue of obese subjects [91]. These changes in number and function of immune cells promote adipose tissue inflammation in obesity. The innate immune system is the first line of defense in response to injury or pathogens and comprises principally monocytes and macrophages [92]. In obese subjects, macrophages are increased in adipose tissues; in these cells a polarization towards M1-type macrophages with a pro-inflammatory phenotype is observed together with the secretion of pro-inflammatory cytokines. Adipose tissue macrophages are classified into two types: M1 or “classically activated” macrophages and M2 or “alternatively activated” macrophages. M1 macrophages are the first line of defense against intracellular pathogens and secretes inflammatory cytokines including interleukins IL-1, IL-6, IL-12, IL-1β, monocyte chemoattractant protein (MCP)-1, inducible NOS (iNOS) and TNF-α. M2 macrophages are implicated in inflammation resolution and secretes interleukins IL-4, IL-10, and transforming growth factor-α (TGF-α). Multiple studies have shown that inflammation in obesity is associated with an increase of M1 macrophages and a decrease of M2 macrophages in adipose tissue. Macrophages are characterized by plasticity and change rapidly in response to external stimuli. In fact, humans and murine models with a dominant M2 phenotype show a “phenotypic switch” towards an M1 phenotype under the stress of obesity [89], [93]. The adaptive immune system, that include T cells, B cells, natural killer cells (NK), natural killer T cells (NKT) and type 2 innate lymphoid cells (ILC2), plays an important role in the obesity, and is critical for specific immune responses and the development of immune response memory. T lymphocytes consist mainly of CD4+ T cells and CD8+ T cells. In adipose tissue, CD4+ T cells are generally classified into the T helper 1 (Th1) and T helper 2 (Th2) but also include T helper 17 (Th17), induced T regulatory cells (iTreg) and regulatory type 1 cells (Tr1). Th1 and Th17 cells release interferon-γ (IFN-γ) and IL-17 stimulating pro-inflammatory M1 macrophage functions and the release of IL-6 and TNF-α. On the other hand, anti-inflammatory Th2 cells and Treg cells produce IL-4, IL-10 and IL-13 promoting macrophage differentiation towards an M2 phenotype. During adipose tissue expansion the imbalance between the relations of Th1:Treg and Th1:Th2 cells are responsible for a shift of macrophages to M1 macrophage phenotype; this contributes to start and perpetuate adipose tissue inflammation [89], [94]. Secretion of proinflammatory cytokines from adipose tissue causes a persistent inflammatory state associate with the risk of developing adverse outcomes in obesity-linked complications. Studies conducted on obese people and animals show a higher level of serum TNF-α, IL-1β and IL-6 produced by macrophages of adipose tissue. These cytokines regulate proliferation and apoptosis of adipocytes, promote lipolysis, inhibit lipid synthesis and decrease blood lipids through autocrine and paracrine mechanisms. The mentioned mechanisms will be predisposed to decrease lipid accumulation in the adipose tissue. Circulating concentrations of TNF-α are greater in obese subjects while concentrations of TNF-α and IL-6 are reduced following weight loss. TNF-α induce apoptosis in preadipocytes and adipocytes, block differentiation and induce de-differentiation in preadipocytes reducing adiposity [95]. On the other hand, an increase of IL-6 in obese subjects stimulates liver secretion of CRP, an acute phase protein of inflammation that increases in obese individuals. CRP represents a marker for the early diagnosis of metabolic syndrome and cardiovascular risk in obese children [96]. However, studies investigating the links between TNF-α, IL-6 and obesity in children, have shown inconsistent findings. For example, a study conducted on obese Caucasian subjects showed that IL-6 levels were not increased [97] and another study in overweight/obese Hispanic children reported raised levels of CRP and TNF-α, but not IL-6. Being that IL-6 is secreted from adipose tissue and induces the hepatic production of CRP, both values should increase in obese children [98]. Moreover, high levels of IL-6 have been found in overweight/obese 15-year-old girls compared with healthy weight girls, but there was no differences observed in 8-year-old subjects and in boys [99]. Discrepancies between CRP and IL-6 might originate from a non-strictly rigorous pre-analytical phase or inaccurate analytical phase, thus underestimating the measurement of IL-6.

The evaluation of different cytokine levels in obese children considering age, sex and ethnicity could better define the relationship between obesity and inflammation.

Furthermore, it has been shown that adipokines and myokine hormones, such as irisine, produced by adipose tissue and skeletal muscle have a critical role in the regulation of energy metabolism and inflammation [100]. In fact, the adiponectin Acrp30 has anti-diabetic, anti-atherogenic and anti-inflammatory properties [101], [102] while irisin plays a role in the maintenance of energy balance increasing energy expenditure and improving glucose homeostasis [103]. Dysregulation of Acrp30, its high molecular weight (HMW) oligomers, and irisin may be involved in the development and progression of obesity and IR in adults and children.

Moreover, as in adults, concentrations of leptin results are higher in the serum of obese children and its increase is related to the body fat and BMI. Leptin is a hormone of a protein nature that acts on receptors present at the hypothalamic level by regulating body weight; in fact, it causes a decrease in appetite and an increase in energy expenditure. It was hypothesized that there is a leptin resistance in prepubertal children due to an higher energy requirements and only at the end of puberty phase the leptin resistance return to normal level. This data makes leptin resistance in the prepubertal era, necessary for normal growth and development, allowing greater energy storage essential for growth. Elevated leptin levels in obese children could represent a consequence of a leptin resistance alteration necessary for a normal development [104].

All these data could improve our knowledge of the physiopathology of this complex and multifactorial disease thus leading to a new scenario for a more accurate diagnostic approach.

Hormones

In a small number of children showing a sudden onset of obesity, an endocrinopathy should be suspected [105], [106]. In particular, hypothalamic obesity, hypothyroidism, hypercortisolism, growth hormone deficiency (GHD) or pseudohypoparathyroidism could be hypothesized. Early recognition of an endocrine disease allows physicians to start effective therapies with relevant benefits for the patients. The following clinical scenarios are commonly found [107]:

  1. Hypothalamic obesity. Congenital defects or injury to the hypothalamus (i.e. surgery for craniopharyngioma, trauma, pituitary tumors) which damage ventromedial, arcuate, paraventricular and dorsomedial nuclei of the hypothalamus can cause obesity. These hypothalamic nuclei release neuropeptides which regulate appetite and energy consumption. In these patients, other endocrine diseases are commonly found such as GHD, hypothyroidism and precocious or late puberty [106], [108].

  2. Hypothyroidism. Mild BMI gain and linear growth decrease associated with high thyroid-stimulating hormone (TSH) and low thyroxine (T4) levels are suggestive of hypothyroidism. In such a case diminished energy consumption and water retention lead to weight increase [109]. Moreover, TSH is able to bind its receptor on fat tissue, promoting adipogenesis and increase in size of adipose tissue [110], [111]. In obese children, higher triiodothyronine (T3), but not T4 circulating levels were found. Free T4 and free T3 were normal or slightly elevated, suggesting an adaptive response of thyroid function to increased BMI in order to reduce further weight gain [112], [113]. However, T3 increase was not associated with pathological conditions such as hypothyroidism, iodine deficiency and autoimmune thyroiditis [114].

  3. Hypercortisolism. Children that present central obesity with growth deficiency, hirsutism and hypertension are suspected of hypercortisolism. An endogenous increase of cortisol in children is frequently a consequence of pituitary microadenoma in children over 6 years of age and of adrenal hyperplasia or tumor in younger patients. Moreover, long-term treatments with cortisone (as in the case of autoimmune, dermatological or respiratory diseases and, rarely, in pediatric cancer) may lead to iatrogenic hypercortisolism [115]. In children, high glucocorticoids levels have a significant impact on bone, leading to a decrease in linear growth [116]. This clinical feature allows early suspicion of the underlying disorder.

  4. GHD. Children with GHD exhibit mild truncal obesity, decreased growth rate and short stature. To confirm GHD, provocative tests have to be performed, after exclusion of thyroid dysfunction, sex steroid alterations and hypercortisolism [117]. Because of their low specificity, these tests have to be performed only in those children where the probability of finding GHD is high [118]. Low circulating levels of IGF-1 and insulin-like growth factor binding protein-3 (IGFBP3) are strongly suggestive of GHD [119], [120].

  5. Albright hereditary osteodystrophy (AHO). Mutations inactivating the guanine nucleotide-binding protein G(s) subunit (GNAS), the gene coding for α chain of Gs, cause AHO. These patients present short stature, round facies, early-onset obesity, developmental delay and mental disabilities, multihormone resistance (e.g. PTH, TSH, gonadotropins, and glucagon) [121] and pseudohypoparathyroidism types 1a [106], [108], [122], [123], [124]. The unexpected weight gain in these children seems to be caused by mild hyperphagia and increased food intake [125]. In addition, some authors reported that these patients have a reduced energy consumption [126], [127].

Furthermore, GHD (caused by GHRH resistance) [128] and hypothyroidism (from TSH resistance) [129] also contribute to weight gain in PHP1a. Laboratory tests in these patients show a picture of PTH resistance: elevated PTH and phosphate circulating level and hypocalcemia [130], [131]. Collectively, there are several alerts that have to be taken into account to hypothesize an endocrine cause of childhood obesity: in fact, stature and height increase are slowed down and bone age is delayed [132]. A regular growth rate generally is not related to endocrine causes [133], [134]. A short stature suggests GHD, central obesity arising in early childhood (<6 years) associated with growth deficiency may indicate hypercortisolism, mild hyperphagia associated with short stature, and early-onset obesity, may be associated with AHO [135].

In obese children presenting these clinical features, an appropriate laboratory work-up including thyroid function evaluation, ACTH stimulation testing, dexamethasone stimulus and serum, salivary, 24-h urine free cortisol need to be performed [136]. This kind of approach can identify specific endocrine disorders. Consequently, an adequate treatment strategy can be adopted and a physiological condition including normal growth can be restored.

Cardiovascular risk

There is robust evidence that adult obesity is a risk factor for atherosclerotic diseases and cardiovascular events [137], [138], mainly as a result of its strong association with the risk of developing arterial hypertension, dyslipidemia and type 2-diabetes [139], [140], [141], [142]. Obesity, indeed, is also associated with morbidity during developmental age: arterial hypertension, dyslipidemia, glucose intolerance and diabetes; they are not exclusive for adult obesity, but can also begin to increase cardiovascular risk from childhood in children with severe obesity [2], [143].

The combination of obesity and hypertension is a complex and multifactorial condition that seems to involve IR, activation of the sympathetic nervous system, the renin-angiotensin system, abnormal renal sodium retention, possible resistance to leptin and altered vascular reactivity, and alterations of the hypothalamus-pituitary-adrenal axis [140], [144], [145]. The most common metabolic complication in children with excess weight is IR [141]. An association between obesity and IR and between obesity and diabetes has also been demonstrated in children, IR has also been recognized as a condition preceding the onset of type 2 diabetes (in adults).

To date, a great deal of scientific evidence supports the close association between childhood obesity and the development of increased cardiovascular risk, with long-term follow-up [146], [147], [148], [149]. Data from a broad meta-analysis [146], show that childhood obesity is significantly and positively associated with adult systolic blood pressure (Zr=0.11; 95% CI: 0.07, 0.14), diastolic blood pressure (Zr=0.11; 95% CI: 0.07, 0.14), high TG levels (Zr=0.08; 95% CI: 0.03, 0.13), and significantly and inversely associated with adult HDL (Zr=−0.06; 95% CI: −0.10, −0.02) [146]. Juonala et al. [150] analyzed data from four prospective cohorts (two North Americans, one Australian and one Finnish). They measured BMI both in childhood and in adulthood, to establish whether a change from obesity during childhood to a non-obese BMI in adulthood, as compared with obesity during childhood that persists into adulthood, would be correlated to a reduced risk of cardiovascular and diabetes diseases. The study cohort consisted of 6328 subjects, and the mean length of follow-up was 23.1±3.3 years. Only 14.6% of children with normal weight have developed adult obesity, while 65% of overweight children and 82% of obese children have developed adult obesity. Patients with a persistent high adiposity status from childhood to adulthood, showed an increased risk of type 2 diabetes (DM2), as compared with normal BMI subjects both in childhood and in adulthood (relative risk [RR]: 5.4; range 95% confidence interval [CI]: 3.4–8.5, p<0.001). They showed an increase risk of arterial hypertension (RR 2.7, 95% CI 2.2–3.3, p<0.001), high LDL-C (RR 1.8, 95% CI 1.4–2.3, p<0.001), low HDL-C (RR 2.1, 95% CI 1.8–2.5, p<0.001), elevated TG levels (RR 3.0, 95% CI 2.4–3.8, p<0.001), and increased carotid-artery intima-media thickness (RR 1.7, 95% CI 1.4–2.2, p=0.002). Adult subjects with normal BMI but childhood obesity, had a similar cardiovascular risk as compared with subjects with normal BMI in both childhood and adulthood [150]. The increased incidence of development of cardiovascular risk factors is expressed in an increased incidence of cardiovascular events, mainly concerning CHD [149]. Ohlsson et al. [148] conducted a study exclusively on male subjects; overweight boys during adolescence and overweight boys during childhood and youth, which showed they had an increased risk of cardiovascular mortality compared with normal-weight subjects (hazard ratio [HR] 2.39, 95% CI 1.86–3.09; HR 1.85, 95% CI 1.28–2.67; respectively).

Childhood obesity involves an increased risk of CHD, which may be mediated by cardiovascular risk factors [151]; in fact, the cumulative burden of this conventional risk factors in this setting patients may alter premature steps in atherosclerosis [152]. Moreover, the development of these risk factors begins very early and also in the initial degrees of obesity. In a large prospective cohort study, data from 5235 children aged 9–12 years at baseline, were analyzed [153]. The prevalence of cardiovascular risk factors at age 15–16 were higher in obese subjects group at baseline [153]. De Kroon et al. [154], have analyzed data from 642 subjects from the Terneuzen Birth Cohort. Results showed that BMI change from 2 to 18 years are related to increased cardiometabolic risk at young adulthood, the age interval 2–6 year being the most predictive [154]. Zabarsky et al. [155], have investigated obesity-associated morbidity incidence in severely obese youth. They divided a childhood obesity cohort into degrees of obesity (classes I, II, III and IV), based on the percentiles of BMI by age and sex. Baseline classes III and IV obesity compared with overweight were related with increased risk of having obesity-associated morbidity at follow-up (OR=5.76, p=0.001; OR=5.36, p=0.001, respectively). The results show that the metabolic risk associated with childhood obesity precedes advanced obesity degree, suggesting to use a complication-oriented approach and management [155]. Even though there is evidence in obese children of a worse outcome in adulthood, a mild reduction in body weight before the adolescence, can considerably reduce the risk for cardiovascular and metabolic disease later in life [150].

The relationship between obesity, hypercholesterolemia and lipoprotein profile modifications is well known [156], [157], [158]. Additional and independent risk factor for CHD is represented by high lipoprotein (a) [Lp(a)] levels, studied in several patient groups [159], [160], [161]. The association between its plasma levels and the condition of childhood obesity is unclear [162]. Glowinska et al. [163] studied lipid profiles in obese, hypertensive and diabetic young patients showing significant changes in lipid metabolism, including Lp(a), apolipoprotein A (Apo A) and Apo B levels. The association between Lp(a) levels and obesity is unclear probably because Lp(a) circulating levels are mainly genetically determined, but there are several studies that show a significant decrease in Lp(a) levels proportional to weight loss and Lp(a) levels reduction with physical activity [161], [164].

In conclusion, childhood obesity has both significant short-term and long-term consequences on the cardiovascular system, which may be mediated through the development of cardiovascular risk factors and circulating mediators produced by adipose tissue [152], [165]. Appropriate strategies should be implemented to counteract the progress of childhood obesity, considering that its cardiovascular effects may be reversible if children normalize their weight before adulthood.

Exercise

It has been suggested that exercise plays a pivotal role in treating overweight and obese children and adolescents [8], [166]. Exercise seems to favor a reduction in body weight, BMI, central obesity, fat mass percentage, TGs, IR markers, and cardiorespiratory fitness. Exercise interventions help to regulate body fat; however, in the young population, it is unknown to what extent more significant volumes of exercise influence body fat. Current findings showed overall a significant medium reduction in BMI and a small effect size in fat mass percentage and a small but significant reduction in central obesity [167], [168]. Regarding the dose-response effects of exercise on measures of adiposity, a meta-analysis conducted by Atlantis et al. including 481 overweight boys and girls (aged ~12 years) concluded that 155–180 min per week of moderate to high-intensity exercise was effective in reducing body fat and central obesity in overweight and obese children and adolescents [169]. Besides, studies with overweight and obese participants concluded that aerobic plus resistance exercise interventions (8–24 weeks’ duration) produced decreases in body weight, BMI and fat mass, but no changes in fat-free mass and WC [170], [171]. Stoner et al. found that exercise intervention reduced BMI (moderate effect), body weight and body fat percentage (all with small effects) and noted that central obesity reduction in response to weight loss might be conditioned by obesity phenotype (BMI vs. fat mass) [172]. Exercise can be therapeutic in reducing body fat; in fact, it is possible to observe an increase of energy expenditure, lipid oxidation and lipid synthesis inhibition by the activation of the AMP-activated protein kinase pathway and free fatty acid flux to the liver [173]. Mainly abdominal obesity excess is linked to a raised lipid profile and a risk of developing IR and metabolic syndrome, constituting the basis for impaired vascular function [174].

Supervised training is a more potent stimulus than non-supervised training in enhancing (by ~1.5%) flow-mediated dilation (FMD). Previous prognostic studies had suggested the clinical relevance of this improvement in FMD. A meta-analysis of 5547 adults was associated with a 1% increase in FMD with a 13% decrease in cardiometabolic events, so that the magnitude of FMD improvement found following exercise can favorably affect endothelial function in healthy young adults, indicative of another cardioprotective effect of exercise against the progression of atherosclerosis. In this context, several biologically plausible mechanisms could explain the effects of exercise in modulating endothelial function and arterial stiffness [175]. The primary physiological mechanisms involved up-regulate endothelial nitric oxide synthase activity with a subsequent decreased expression of nicotinamide adenine dinucleotide phosphate oxidase (NADPH), and stimulation of radical scavenging systems that include copper/zinc-containing superoxide dismutase (Cu/ZnSOD), extracellular superoxide dismutase, glutathione peroxidase, and glutathione [175]. Also, aerobic exercise performed for 60 min, thrice a week, at ≤75% maximum heart rate improves LDL-C and TG concentrations in obese children, and combined exercise increases HDL-C concentrations [176]. Marson et al. demonstrated that in overweight adolescents the improvements in the insulin sensitivity index (IR, HOMA) were similar for aerobic and for strength training, and neither were there differences in the improvements between the exercise training groups and those with combined training. The findings overall showed a significant reduction in fasting glucose and fasting insulin, and these findings have important health implications and provide health care professionals with therapeutic strategies for the treatment of childhood obesity and the reduction of IR in the young population [177]. There is evidence [178] that exercise improves various parameters related to health (body mass, BMI, fat mass, TGs, fasting glucose, fasting insulin). Surprisingly, relative to those of over 12 weeks, the 2- to 4-week programs showed more significant improvements in BMI, visceral fat, and subcutaneous fat. Probably, the shorter programs do not need the strict control of the progression of exercise intensity that the more extended programs do so that their results would be more pronounced. However, in general, for the cardiometabolic and vascular parameters it seems programs of 4–12 weeks, or of more than 60 min per session, or with a total exercise time of 1500 min or more were effective in improving HDL-C levels, fasting glucose, fasting insulin, HOMA-IR, intrahepatic fat, systolic blood pressure and carotid intima-media thickness. This phenomenon may be due to the organism’s rapid adaptation to the load represented by the physical exercise program, so that an increase in the intensity may be needed to produce a new adaptation. Although in the field of sports the precise quantification of training programs is the norm, in the field of health, quantification of exercise programs is at best only very general in form, and sometimes almost non-existent [179]. A dose-response relationship for the different health parameters could be applied to obtain benefits in health and to develop more effective lifestyle interventions in the obese young population. Giving the positive effects of physical exercise, efforts should be made to increase physical activity adherence in overweight children. Indeed, current adherence of children to lifestyle recommendations to prevent childhood obesity is low. In the Identification and prevention of Dietary- and lifestyle-induced health Effects in Children and infants (IDEFICS) study only 1.1% of children observed five out of six recommendations, with differences by country and age (children in northern countries and younger children showed better adherence to the recommendations) [180].

Microbiota

The intestinal microbiota is composed of many different microorganisms mostly from the Bacteria kingdom [181]. About 90% belong to the phyla Firmicutes, and Bacteroidetes and the most abundant genera are Bacteroides, Faecalibacterium and Bifidobacterium the proportions of which vary between individuals. At the species level, there is a great variety which can originate a unique profile for each host [182], [183].

Gut microbiotas in childhood are affected by geography and food culture, in fact in children living in advanced countries their guts are mainly constituted by Bacteroides-Bifidobacterium-dominate microbiotas (BB-type) whereas children in developing countries are represented by Prevotella-dominated microbiotas (P-type) [184], [185], [186].

Furthermore, these two microbiota-types are often observed within the same country but are associated with the level of development in an area. Nakayama et al. recently found that P- and BB-type microbiotas were represented in children living in rural and urban sites and that a type shift was associated with modernization of consumed foods, and notably a change to high-fat diets. These studies suggest that diet had a more significant influence on gut microbial communities than host genetics. Indeed, the microbiotas were enriched in bacteria with genes encoding bile acids that aid in lipid absorption or oligosaccharide-degrading enzymes involved in plant digestion in BB-type and P-type children, respectively [187].

The composition of gut microbiota is defined at birth and evolves until about 3 years old, then it remains constant during the lifetime; however, a number of factors, such as antibiotics and diet, can change it [188], [189]. In human twins, the similarity of the microbiota is within the same family members and not only in monozygotic twins, indicating that environmental exposure has more impact than genotype in microbiota development [190], [191].

Cho et al. demonstrated that low dose antibiotic exposure in young mice led to increased adiposity, metabolic hormone levels, and short-chain fatty acids (SCFA) levels, as well as changes to the hepatic metabolism of lipids and cholesterol [192]. The antibiotic exposition has an overpowering impact on gut microbiota [193]. Epidemiological studies have demonstrated that the administration of antibiotics during childhood is correlated with a higher risk of obesity [194], [195]. Early life exposition to antibiotics has also been proposed to drive to obesity [196]. In neonates, after repeated exposure to antibiotics, has been shown a decrease of anti-obesogenic bacteria (bifidobacteria and Bacteroides) [197]. The common exposure to antibiotics, in the perinatal period, exerts a wide effect on the gut microbiota. After the neonatal period, diet is the most important factor regulating the gut microbiota with breast milk promoting the ascendancy of bifidobacteria in the infant gut [198].

In addition, Cox et al. found that low dose penicillin given at birth can induce sustained effects on body composition and enhance high fat diet-induced obesity in mice. Furthermore, the obese phenotype was transferable to germ-free mice by transfer of low-dose penicillin microbiome, implicating the microbiome as the driver of this phenotype as opposed to antibiotics [199]. High fat and protein diets significantly reduced SCFA and altered intestinal bacteria composition. The supplementation with prebiotics promotes the growth of specific gut microbiota species, and the use of probiotics has benefits in host health, but the impact on large-term gut microbiota remains to be proven [200], [201], [202].

In obesity, gut microbiota is known to be altered by a decreased ratio of Bacteroidetes to Firmicutes, with a superior capacity to harvest energy from a diet [203], [204], [205].

Besides, long-term diet habits influence the composition of gut microbiota. Intestinal bacteria react to daily dietary fat and carbohydrates and change its metabolic pattern [206], [207], [208].

A very recent study explored the impact of gut microbiota and diet on visceral fat mass accumulation (VFM) in 1760 older female twins [209].

In this large study it was shown that gut microbiota composition and diet are related to VFM accumulation and that these two factors are closely linked. Moreover, operational taxonomic units (OUTs) relative abundance, even after revision for nutrient inlet, is strongly associated with VFM indicating that bacteria may not require the presence of nutrients to affect host phenotypes. However, the influence of nutrients in improving the growth of beneficial microbes and their consequence on host health is not ruled out [209].

In this same twins UK cohort, Le roy et al., demonstrated novel evidence of association between six adiposity phenotypes and fecal microbial OTUs. Most of these results involve visceral fat associations, with the strongest associations between visceral fat and Oscillospira members suggesting that this genus if replicated could be a key player of the host-GM mutualism in relation to fat deposition. While several genera appear as protective factors in VFM, only Blautia stood out as a risk factor that is in part heritable [210]. When neonatal microbiota starts is until now a matter of great debate. The idea that, under physiological status, the human fetal environment is sterile and microbial colonization develops with birth, has been in dispute for decades. Recently, there has been different evidence that have described a lowly bacterial colonization in the placenta, endometrium, amniotic fluid and meconium in healthy, full-term pregnancies [211]. These findings influenced some researchers about the seeding of the microbiota before birth. Moreover, different scientists have pointed out the necessary to lend part of attention when working on samples with a low microbial biomass due to the contamination issues with such samples, when using PCR amplification and NGS techniques [211], [212], [213].

Preterm birth can be considered a polymicrobial disease [214] and it has been reported that adults who were born prematurely present increased risk for obesity [215], [216] in comparison with individuals who were born after a full-term pregnancy. Moreover, the incidence of several prenatal and early postnatal factors associated with the development of infant adiposity (such as prematurity and low birth weight, gestational diabetes, excess body mass gain during gestation and infant formula feeding) has also increased [217]. In this scenario, preterm birth can be considered a further risk factor for obesity.

As mentioned earlier, the intestinal microbiota colonization occurs at birth, or before in uterus, and it is the perinatal period that is the most important one to define gut microbiota in later ages. Early life environment factors involved are:

  1. host genetics [218], [219];

  2. in uterus colonization [191], [220], [221];

  3. maternal lifestyle [222];

  4. birth delivery mode [223], [224], [225];

  5. breastfeeding vs. formula [225];

  6. antibiotics exposure [226], [227];

  7. hygiene level [228], [229];

  8. prebiotics and probiotics administration to pregnant women and neonates [230], [231].

At 3–5 years old, the gut microbiota composition is similar to adults and remains very stable [220]. A large amount of studies reveals differences between gut microbiota of obese and lean children. In obese children with ages between 4 and 5 years old, Karlsson et al. found significant differences in the abundance of bacteria but only a tendency in their diversity. The abundance of Enterobacteriaceae was significantly higher in obese children, whereas there was a significantly lower amount of Desulfovibrio and Akkermansia muciniphila-like bacteria. No substantial differences were found in the content of Lactobacillus, Bifidobacterium or the Bacteroides fragilis spp. [232]. In another study comparing children between 6 and 16 years of age, the authors found elevated Firmicutes-to-Bacteroidetes ratio in obese children compared with thin ones. Additionally, low relative proportions of Bacteroides vulgatus and high levels of Lactobacillus spp. were observed in the obese children [233]. Also, gut microbiota changes were found in obese adolescents when they altered their lifestyle, mainly diet and exercise, suggesting interactions between diet, gut microbiota and host metabolism and immunity in obesity [234]. Several recent studies suggest the positive role of gut microbiota modulation in adult obesity treatment. However, there are no sufficient data to state that child dysbiosis increases the risk of subsequent obesity. Primary prevention strategies may include modeling maternal and infants gut microbiota to break the obesity cycle. The use of prebiotics, probiotics and changes in maternal lifestyle may be successful interventions to avoid childhood dysbiosis and consequent obesity [235].

Nutraceuticals and probiotics

Pharmacological therapy should only be used after failure of multidisciplinary lifestyle interventions. When substantial weight loss cannot be achieved through lifestyle-based interventions, the use of drugs is justified, mainly in obese individual affected by serious different disorders [236], [237], [238], [239], [240]. Orlistat (tetrahydro-lipstinate) is the single drug available for the treatment of children and adolescents with severe obesity. Few studies are available on the effects of anti-obesity pharmacological therapy in pediatric ages [241], [242], [243], [244]; it seems to produce significant weight loss and favoring behavioral changes. There is no implications on the mineral balance if a low-calorie diet is associated with usual mineral intake; in contrast, it must prevent liposoluble vitamins deficiency [245], [246], [247], [248].

Obesity has been correlated with a specific profile of the gut microbiota characterized by lower levels of bacteria belonging to the Bacteroides and Bifidobacterium genera compared to that of lean individuals [249]. Besides, bifidobacteria have been shown to be higher in children maintaining normal weight at 7 years old than in children developing overweight, and their administration was able to reduce serum and liver TG levels and to decrease hepatic adiposity [250], [251]. The mixture of B. breve (BR03 and B632) was used in a cross-over double-blind, randomized controlled trial to re-establish metabolic homeostasis and reduce chronic inflammation in obese children. Preliminary results demonstrated that a B. breve administration in obese children is promising: 8 weeks of treatment seems to ameliorate glucose metabolism and could help in weight management by reducing BMI, waist to height ratio and WC [252].

Intake of fiber supplements is inversely associated with body weight, body fat and BMI [253], thus promoting satiation, reducing macronutrients absorption, and modifying the production of gut hormones. Although increasing consumption of fibers (e.g. fruits, vegetables, grains, legumes) with diet is a crucial step to curb the obesity, fiber addiction should also be considered. Glucomannan, a soluble and viscous dietary fiber derived from the konjac plant, can cause weight loss and improve lipid and lipoprotein parameters and glycemic status with minimal gastrointestinal side effects [254]. Even though the weight reduction by psyllium is weakly supported, it has been showed to be able to improve glucose homeostasis, and the lipid and lipoprotein profile in obese children. Inulin (from chicory root) is being sold in many mainstream stores as a fiber supplement, including for use in children; however, its weight-lowering effect has not been studied [255]. Additional studies are necessary to gain definitive conclusions.

Conclusions

Childhood obesity represents one of the most pressing medical and public health problems of our day. The prevalence is unacceptably high, and the rate of increase in severe obesity continues to climb. Obesity is classified into three classes: class I (95th percentile to <120% of the 95th percentile), class II (120%–<140% of the 95th percentile), and class III (≥140% of the 95th percentile).

The multifactorial etiology of obesity is related to genetics and epigenetic predisposition, nutrition, environment, lifestyle and hormone dysfunction. This review intended to underline the role of the main factors influenced by the gain weight and also those that may modulate and predispose to childhood obesity.

There is increasing evidence for the role of epigenetic factors in the development of obesity. In particular, miRNAs represent relevant regulators of processes related to adipogenesis contributing to metabolic abnormalities associated with childhood obesity and obesity-related diseases.

Heritable factors appear to be responsible for 30%–50% of the variation in adiposity. It was well demonstrated that obese children with genetic syndromes typically show early-onset obesity and characteristics and also pathological phenotypes.

The most common single gene defects currently identified in children with obesity are mutations in the melanocortin 4 receptor. Other gene defects include those in leptin, leptin receptor, proopiomelanocortin and proprotein convertase.

Furthermore, the screening of biochemical parameters can well define the organ’s metabolic status and characterize any modification and/or organ damage. We also focused on the regulation of different inflammatory obesity-related cytokines that modulate the proliferation and apoptosis of adipocytes, promoting lipolysis and inhibit lipid synthesis through autocrine and paracrine mechanisms.

Most children with endocrine disorders resulting in weight gain show poor linear growth, short stature and/or hypogonadism. This status includes either endogenous or exogenous glucocorticoid excess, hypothyroidism, growth hormone deficiency, and pseudohypoparathyroidism type 1a.

In addition, we underlined the increased risk of hyperinsulinemia, IR, prediabetes, and also the high prevalence of other cardiometabolic risk factors including elevated blood pressure, low levels of HDL-C, and elevated levels of TGs. In particular, Lp(a) evaluation is strongly recommended as an independent causal risk factor for cardiovascular disease.

Moreover, we also considered the benefits of physical activity necessary to avoid the energy imbalances strictly related to overweight and to contribute to achieve healthy habits and adequate life style that improves lower levels of overweight. In this contest gut microbiota may also play a pivotal role in the early diagnosis of childhood obesity as it has been correlated with a specific profile of the gut microbiota. Furthermore, a brief outline was made on the possibility of designing a therapy intervention strategy by using the intake of fiber supplements together with life style interventions.

Each factor described in this review can contribute to define an early diagnostic tool and highlight the importance to adopt early interventions that target both weight and mental health in childhood to minimize negative outcomes later in adulthood. The design of specific and individualized diagnostic models, based on the degree of obesity, is strictly necessary to devise a suitable multidisciplinary medical intervention.

  1. Author contributions: All the authors have accepted responsibility for the entire content of this submitted manuscript and approved submission.

  2. Research funding: None declared.

  3. Employment or leadership: None declared.

  4. Honorarium: None declared.

  5. Competing interests: The funding organization(s) played no role in the study design; in the collection, analysis, and interpretation of data; in the writing of the report; or in the decision to submit the report for publication.

References

1. Carnovali M, Luzi L, Terruzzi I, Banfi G, Mariotti M. Metabolic and bone effects of high-fat diet in adult zebrafish. Endocrine 2018;61:317–26.10.1007/s12020-017-1494-zSearch in Google Scholar

2. Kumar S, Kelly AS. Review of childhood obesity: from epidemiology, etiology, and comorbidities to clinical assessment and treatment. Mayo Clin Proc 2017;92:251–65.10.1016/j.mayocp.2016.09.017Search in Google Scholar

3. Hruby A, Hu FB. The epidemiology of obesity: a big picture. Pharmacoeconomics 2015;33:673–89.10.1007/s40273-014-0243-xSearch in Google Scholar

4. Abarca-Gómez L, Abdeen ZA, Hamid ZA, Abu-Rmeileh NM, Acosta-Cazares B, Acuin C, et al. Worldwide trends in body-mass index, underweight, overweight, and obesity from 1975 to 2016: a pooled analysis of 2416 population-based measurement studies in 128·9 million children, adolescents, and adults. Lancet 2017;390:2627–42.10.1016/S0140-6736(17)32129-3Search in Google Scholar

5. Parsons TJ, Power C, Logan S, Summerbell CD. Childhood predictors of adult obesity: a systematic review. Int J Obes Relat Metab Disord 1999;23(Suppl 8):S1–107.10.1038/sj/ijo/0800851Search in Google Scholar

6. Kopelman PG. Obesity as a medical problem. Nature 2000;404:635–43.10.1038/35007508Search in Google Scholar PubMed

7. Katzmarzyk PT, Chaput JP, Fogelholm M, Hu G, Maher C, Maia J, et al. International Study of Childhood Obesity, Lifestyle and the Environment (ISCOLE): contributions to understanding the global obesity epidemic. Nutrients 2019;11:848.10.3390/nu11040848Search in Google Scholar PubMed PubMed Central

8. Lombardi G, Sanchis-Gomar F, Perego S, Sansoni V, Banfi G. Implications of exercise-induced adipo-myokines in bone metabolism. Endocrine 2016;54:284–305.10.1007/s12020-015-0834-0Search in Google Scholar PubMed

9. Zaiou M, El Amri H, Bakillah A. The clinical potential of adipogenesis and obesity-related microRNAs. Nutr Metab Cardiovasc Dis 2018;28:91–111.10.1016/j.numecd.2017.10.015Search in Google Scholar PubMed

10. Goody D, Pfeifer A. MicroRNAs in brown and beige fat. BBA – Mol Cell Biol Lipids 2019;1864:29–36.10.1016/j.bbalip.2018.05.003Search in Google Scholar PubMed

11. Iacomino G, Siani A. Role of microRNAs in obesity and obesity-related diseases. Genes Nutr 2017;12:23.10.1186/s12263-017-0577-zSearch in Google Scholar PubMed PubMed Central

12. Bartel DP. MicroRNAs: genomics, biogenesis, mechanism, and function. Cell 2004;116:281–97.10.1016/S0092-8674(04)00045-5Search in Google Scholar

13. Ha M, Kim VN. Regulation of microRNA biogenesis. Nat Rev Mol Cell Biol 2014;15:509–24.10.1038/nrm3838Search in Google Scholar PubMed

14. Zhong H, Ma M, Liang T, Guo L. Role of MicroRNAs in obesity-induced metabolic disorder and immune response. J Immunol Res 2018;2018:1–8.10.1155/2018/2835761Search in Google Scholar PubMed PubMed Central

15. Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215–33.10.1016/j.cell.2009.01.002Search in Google Scholar PubMed PubMed Central

16. Lee EK, Lee MJ, Abdelmohsen K, Kim W, Kim MM, Srikantan S, et al. miR-130 Suppresses adipogenesis by inhibiting peroxisome proliferator-activated receptor expression. Mol Cell Biol 2011;31:626–38.10.1128/MCB.00894-10Search in Google Scholar PubMed PubMed Central

17. Bussler S, Penke M, Flemming G, Elhassan YS, Kratzsch J, Sergeyev E, et al. Novel insights in the metabolic syndrome in childhood and adolescence. Horm Res Paediatr 2017;88:181–93.10.1159/000479510Search in Google Scholar PubMed

18. Carrer M, Liu N, Grueter CE, Williams AH, Frisard MI, Hulver MW, et al. Control of mitochondrial metabolism and systemic energy homeostasis by microRNAs 378 and 378*. Proc Natl Acad Sci U S A 2012;109:15330–5.10.1073/pnas.1207605109Search in Google Scholar PubMed PubMed Central

19. Ortega FJ, Mercader JM, Catalán V, Moreno-Navarrete JM, Pueyo N, Sabater M, et al. Targeting the circulating microRNA signature of obesity. Clin Chem 2013;59:781–92.10.1373/clinchem.2012.195776Search in Google Scholar PubMed

20. Carreras-Badosa G, Bonmatí A, Ortega FJ, Mercader JM, Guindo-Martínez M, Torrents D, et al. Altered circulating miRNA expression profile in pregestational and gestational obesity. J Clin Endocrinol Metab 2015;100:E1446–56.10.1210/jc.2015-2872Search in Google Scholar PubMed

21. Can U, Buyukinan M, Yerlikaya FH. The investigation of circulating microRNAs associated with lipid metabolism in childhood obesity. Pediatr Obes 2016;11:228–34.10.1111/ijpo.12050Search in Google Scholar PubMed

22. Prats-Puig A, Ortega FJ, Mercader JM, Moreno-Navarrete JM, Moreno M, Bonet N, et al. Changes in circulating MicroRNAs are associated with childhood obesity. J Clin Endocrinol Metab 2013;98:E1655–60.10.1210/jc.2013-1496Search in Google Scholar PubMed

23. Jiang X, Xue M, Fu Z, Ji C, Guo X, Zhu L, et al. Insight into the effects of adipose tissue inflammation factors on miR-378 expression and the underlying mechanism. Cell Physiol Biochem 2014;33:1778–88.10.1159/000362957Search in Google Scholar PubMed

24. Turchinovich A, Samatov T, Tonevitsky A, Burwinkel B. Circulating miRNAs: cell-cell communication function? Front Genet 2013;4:119.10.3389/fgene.2013.00119Search in Google Scholar PubMed PubMed Central

25. Iacomino G, Russo P, Stillitano I, Lauria F, Marena P, Ahrens W, et al. Circulating microRNAs are deregulated in overweight/obese children: preliminary results of the I. Family study. Genes Nutr 2016;11:7.10.1186/s12263-016-0525-3Search in Google Scholar PubMed PubMed Central

26. Slattery ML, Herrick JS, Mullany LE, Stevens JR, Wolff RK. Diet and lifestyle factors associated with miRNA expression in colorectal tissue. Pharmgenomics Pers Med 2017;10:1–16.10.2147/PGPM.S117796Search in Google Scholar PubMed PubMed Central

27. Palmer JD, Soule BP, Simone BA, Zaorsky NG, Jin L, Simone NL. MicroRNA expression altered by diet: Can food be medicinal? Ageing Res Rev 2014;17:16–24.10.1016/j.arr.2014.04.005Search in Google Scholar PubMed

28. Yang W, Kelly T, He J. Genetic epidemiology of obesity. Epidemiol Rev 2007;29:49–61.10.1093/epirev/mxm004Search in Google Scholar PubMed

29. Bouchard C, Tremblay A, Després J-P, Thériault G, Nadeauf A, Lupien PJ, et al. The response to exercise with constant energy intake in identical twins. Obes Res 1994;2:400–10.10.1002/j.1550-8528.1994.tb00087.xSearch in Google Scholar PubMed

30. Stunkard AJ, Sørensen TI, Hanis C, Teasdale TW, Chakraborty R, Schull WJ, et al. An adoption study of human obesity. N Engl J Med 1986;314:193–8.10.1056/NEJM198601233140401Search in Google Scholar PubMed

31. Sanghera DK, Bejar C, Sharma S, Gupta R, Blackett PR. Obesity genetics and cardiometabolic health: potential for risk prediction. Diabetes Obes Metab 2019;21:1088–100.10.1111/dom.13641Search in Google Scholar PubMed PubMed Central

32. da Fonseca AC, Mastronardi C, Johar A, Arcos-Burgos M, Paz-Filho G. Genetics of non-syndromic childhood obesity and the use of high-throughput DNA sequencing technologies. J Diabetes Complications 2017;31:1549–61.10.1016/j.jdiacomp.2017.04.026Search in Google Scholar PubMed

33. Sanna V, Ceglia C, Tarsitano M, Lombardo B, Coppola A, Zarrilli F, et al. Aberrant F8 gene intron 1 inversion with concomitant duplication and deletion in a severe hemophilia A patient from Southern Italy. J Thromb Haemost 2013;11:195–7.10.1111/jth.12061Search in Google Scholar PubMed

34. Zebisch A, Schulz E, Grosso M, Lombardo B, Acierno G, Sill H, et al. Identification of a novel variant of epsilon-gamma-delta-beta thalassemia highlights limitations of next generation sequencing. Am J Hematol 2015;90:E52–4.10.1002/ajh.23913Search in Google Scholar PubMed

35. Nunziato M, Starnone F, Lombardo B, Pensabene M, Condello C, Verdesca F, et al. Fast detection of a BRCA2 large genomic duplication by next generation sequencing as a single procedure: a case report. Int J Mol Sci 2017;18:E2487.10.3390/ijms18112487Search in Google Scholar PubMed PubMed Central

36. Lombardo B, Ceglia C, Tarsitano M, Pierucci I, Salvatore F, Pastore L. Identification of a deletion in the NDUFS4 gene using array-comparative genomic hybridization in a patient with suspected mitochondrial respiratory disease. Gene 2014;535:376–9.10.1016/j.gene.2013.10.074Search in Google Scholar PubMed

37. Iossa S, Costa V, Corvino V, Auletta G, Barruffo L, Cappellani S, et al. Phenotypic and genetic characterization of a family carrying two Xq21.1-21.3 interstitial deletions associated with syndromic hearing loss. Mol Cytogenet 2015;8:18.10.1186/s13039-015-0120-0Search in Google Scholar PubMed PubMed Central

38. Locke AE, Kahali B, Berndt SI, Justice AE, Pers TH, Day FR, et al. Genetic studies of body mass index yield new insights for obesity biology. Nature 2015;518:197–206.10.1038/nature14177Search in Google Scholar PubMed PubMed Central

39. Clément K, Vaisse C, Lahlou N, Cabrol S, Pelloux V, Cassuto D, et al. A mutation in the human leptin receptor gene causes obesity and pituitary dysfunction. Nature 1998;392:398–401.10.1038/32911Search in Google Scholar PubMed

40. Hoshino A, Lindberg I. Peptide biosynthesis: prohormone convertases 1/3 and 2. Colloq Ser Neuropeptides 2012;1:1–112.10.4199/C00050ED1V01Y201112NPE001Search in Google Scholar

41. Frank GR, Fox J, Candela N, Jovanovic Z, Bochukova E, Levine J, et al. Severe obesity and diabetes insipidus in a patient with PCSK1 deficiency. Mol Genet Metab 2013;110:191–4.10.1016/j.ymgme.2013.04.005Search in Google Scholar PubMed PubMed Central

42. Krude H, Biebermann H, Luck W, Horn R, Brabant G, Grüters A. Severe early-onset obesity, adrenal insufficiency and red hair pigmentation caused by POMC mutations in humans. Nat Genet 1998;19:155–7.10.1038/509Search in Google Scholar PubMed

43. Lee S. The association of genetically controlled CpG methylation (cg158269415) of protein tyrosinephosphatase, receptor type N2 (PTPRN2) with childhood obesity. Sci Rep 2019;9:4855.10.1038/s41598-019-40486-wSearch in Google Scholar PubMed PubMed Central

44. Chen PY, Chu A, Liao WW, Rubbi L, Janzen C, Hsu FM, et al. Prenatal growth patterns and birthweight are associated with differential DNA methylation and gene expression of cardiometabolic risk genes in human placentas: a discovery-based approach. Reprod Sci 2018;25:523–39.10.1177/1933719117716779Search in Google Scholar PubMed PubMed Central

45. Vuillaume ML, Naudion S, Banneau G, Diene G, Cartault A, Cailley D, et al. New candidate loci identified by array-CGH in a cohort of 100 children presenting with syndromic obesity. Am J Med Genet Part A 2014;164:1965–75.10.1002/ajmg.a.36587Search in Google Scholar PubMed

46. Cook S, Kavey RE. Dyslipidemia and pediatric obesity. Pediatr Clin North Am 2011;58:1363–73.10.1016/j.pcl.2011.09.003Search in Google Scholar PubMed PubMed Central

47. Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, et al. Expert Panel on Integrated Guidelines for Cardiovascular Health and risk reduction in children and adolescents: summary report. Pediatrics 2011;128:S213–56.10.1542/peds.2009-2107CSearch in Google Scholar PubMed PubMed Central

48. Pacifico L, Bonci E, Andreoli G, Romaggioli S, Di Miscio R, Lombardo CV, et al. Association of serum triglyceride-to-HDL cholesterol ratio with carotid artery intima-media thickness, insulin resistance and nonalcoholic fatty liver disease in children and adolescents. Nutr Metab Cardiovasc Dis 2014;24:737–43.10.1016/j.numecd.2014.01.010Search in Google Scholar PubMed

49. Di Bonito P, Valerio G, Grugni G, Licenziati MR, Maffeis C, Manco M, et al. Comparison of non-HDL-cholesterol versus triglycerides-to-HDL-cholesterol ratio in relation to cardiometabolic risk factors and preclinical organ damage in overweight/obese children: the CARITALY study. Nutr Metab Cardiovasc Dis 2015;5:489–94.10.1016/j.numecd.2015.01.012Search in Google Scholar PubMed

50. Di Bonito P, Moio N, Scilla C, Cavuto L, Sibilio G, Sanguigno E, et al. Usefulness of the high triglyceride-to-HDL cholesterol ratio to identify cardiometabolic risk factors and preclinical signs of organ damage in outpatient children. Diabetes Care 2012;35:158–62.10.2337/dc11-1456Search in Google Scholar PubMed PubMed Central

51. Sharma V, Coleman S, Nixon J, Sharples L, Hamilton-Shield J, Rutter H, et al. A systematic review and meta-analysis estimating the population prevalence of comorbidities in children and adolescents aged 5 to 18years. Obes Rev 2019;20:1341–9.10.1111/obr.12904Search in Google Scholar PubMed PubMed Central

52. Schwimmer JB, Deutsch R, Kahen T, Lavine JE, Stanley C, Behling C. Prevalence of fatty liver in children and adolescents. Pediatrics 2006;118:1388–93.10.1542/peds.2006-1212Search in Google Scholar PubMed

53. Feldstein AE, Charatcharoenwitthaya P, Treeprasertsuk S, Benson JT, Enders FB, Angulo P. The natural history of non-alcoholic fatty liver disease in children: a follow-up study for up to 20 years. Gut 2009;58:1538–44.10.1136/gut.2008.171280Search in Google Scholar PubMed PubMed Central

54. Schwimmer JB, Dunn W, Norman GJ, Pardee PE, Middleton MS, Kerkar N, et al. SAFETY study: alanine aminotransferase cutoff values are set too high for reliable detection of pediatric chronic liver disease. Gastroenterology 2010;138:1357–64.10.1053/j.gastro.2009.12.052Search in Google Scholar PubMed PubMed Central

55. Deeb A, Attia S, Mahmoud S, Elhaj G, Elfatih A. Dyslipidemia and fatty liver disease in overweight and obese children. J Obes 2018;2018:8626818.10.1155/2018/8626818Search in Google Scholar PubMed PubMed Central

56. D’Adamo E, Castorani V, Nobili V. The liver in children with metabolic syndrome. Front Endocrinol 2019;10:514.10.3389/fendo.2019.00514Search in Google Scholar PubMed PubMed Central

57. Nobili V, Alkhouri N, Alisi A, Ottino S, Lopez R, Manco M, et al. Retinol-binding protein 4: a promising circulating marker of liver damage in pediatric nonalcoholic fatty liver disease. Clin Gastroenterol Hepatol 2009;7:575–9.10.1016/j.cgh.2008.12.031Search in Google Scholar PubMed

58. Lebensztejn DM, Wierzbicka A, Socha P, Pronicki M, Skiba E, Werpachowska I, et al. Cytokeratin-18 and hyaluronic acid levels predict liver fibrosis in children with non-alcoholic fatty liver disease. Acta Biochim Pol 2011;58:563–6.10.18388/abp.2011_2225Search in Google Scholar

59. Alkhouri N, Mansoor S, Giammaria P, Liccardo D, Lopez R, Nobili V. The development of the Pediatric NAFLD Fibrosis Score (PNFS) to predict the presence of advanced fibrosis in children with nonalcoholic fatty liver disease. PLoS One 2014;9:e104558.10.1371/journal.pone.0104558Search in Google Scholar PubMed PubMed Central

60. Marzuillo P, Grandone A, Perrone L, Miraglia Del Giudice E. Controversy in the diagnosis of pediatric non-alcoholic fatty liver disease. World J Gastroenterol 2015;21:6444–50.10.3748/wjg.v21.i21.6444Search in Google Scholar PubMed PubMed Central

61. Maffeis C, Banzato C, Rigotti F, Nobili V, Valandro S, Manfredi R, et al. Biochemical parameters and anthropometry predict NAFLD in obese children. J Pediatr Gastroenterol Nutr 2011;53:590–3.10.1097/MPG.0b013e31822960beSearch in Google Scholar PubMed

62. Marra F, Lotersztajn S. Pathophysiology of NASH: perspectives for a targeted treatment. Curr Pharm Des 2013;19:5250–69.10.2174/13816128113199990344Search in Google Scholar PubMed PubMed Central

63. Schuppan D, Ashfaq-Khan M, Yang AT, Kim YO. Liver fibrosis: direct antifibrotic agents and targeted therapies. Matrix Biol 2018;68–69:435–51.10.1016/j.matbio.2018.04.006Search in Google Scholar PubMed

64. Marra F, Svegliati-Baroni G. Lipotoxicity and the gut-liver axis in NASH pathogenesis. J Hepatol 2018;68:280–95.10.1016/j.jhep.2017.11.014Search in Google Scholar PubMed

65. Cong M, Iwaisako K, Jiang C, Kisseleva T. Cell signals influencing hepatic fibrosis. Int J Hepatol 2012;2012:1–18.10.1155/2012/158547Search in Google Scholar PubMed PubMed Central

66. Weiskirchen R, Tacke F. Liver fibrosis: which mechanisms matter? Clin Liver Dis 2016;8:94–9.10.1002/cld.581Search in Google Scholar PubMed PubMed Central

67. Luangmonkong T, Suriguga S, Mutsaers HA, Groothuis GM, Olinga P, Boersema M. Targeting oxidative stress for the treatment of liver fibrosis. Rev Physiol Biochem Pharmacol 2018;175:71–102.10.1007/112_2018_10Search in Google Scholar PubMed

68. Chou R, Wasson N. Blood tests to diagnose fibrosis or cirrhosis in patients with chronic hepatitis C virus infection: a systematic review. Ann Intern Med 2013;158:807–20.10.7326/0003-4819-158-11-201306040-00005Search in Google Scholar PubMed

69. Arslan FD, Karakoyun I, Tatar B, Pala EE, Yıldırım M, Ulasoglu C, et al. SHFI: a novel noninvasive predictive model for significant fibrosis in patients with chronic hepatitis B. Hepat Mon 2018;18:e63310.10.5812/hepatmon.63310Search in Google Scholar

70. Korkmaz P, Demirturk N, Batırel A, Cem Yardimci A, Cagir U, Nemli SA, et al. Noninvasive models to predict liver fibrosis in patients with chronic hepatitis B: a study from Turkey. Hepat Mon 2017;17:e60266.10.5812/hepatmon.60266Search in Google Scholar

71. Hagan M, Asrani SK, Talwalkar J. Non-invasive assessment of liver fibrosis and prognosis. Expert Rev Gastroenterol Hepatol 2015;9:1251–60.10.1586/17474124.2015.1075391Search in Google Scholar PubMed

72. American Diabetes Association/ADA. Classification and diagnosis of diabetes: standards of medical care in diabetes – 2018. Diabetes Care 2018;41:S13–27.10.2337/dc18-S002Search in Google Scholar PubMed

73. Zhang X, Gregg EW, Williamson DF, Barker LE, Thomas W, Bullard KM, et al. A1C level and future risk of diabetes: a systematic review. Diabetes Care 2010;33:1665–73.10.2337/dc09-1939Search in Google Scholar PubMed PubMed Central

74. Targher G, Bertolini L, Padovani R, Rodella S, Tessari R, Zenari L, et al. Prevalence of nonalcoholic fatty liver disease and its association with cardiovascular disease among type 2. Diabetes Care 2007;30:1212–8.10.2337/dc06-2247Search in Google Scholar PubMed

75. Zoppini G, Targher G, Trombetta M, Lippi G, Muggeo M. Relationship of serum γ-glutamyltransferase to atherogenic dyslipidemia and glycemic control in tdype 2 diabetes. Obesity 2009;17:370–4.10.1038/oby.2008.544Search in Google Scholar PubMed

76. Targher G, Bertolini L, Rodella S, Lippi G, Franchini M, Zoppini G, et al. NASH predicts plasma inflammatory biomarkers independently of visceral fat in men. Obesity 2008;16:1394–9.10.1038/oby.2008.64Search in Google Scholar PubMed

77. Springer SC, Silverstein J, Copeland K, Moore KR, Prazar GE, Raymer T, et al. Management of type 2 diabetes mellitus in children and adolescents. Pediatrics 2013;131:e648–64.10.1542/9781581108613-part05-managementSearch in Google Scholar

78. Kapadia CR. Are the ADA hemoglobin A1c criteria relevant for the diagnosis of type 2 diabetes in youth? Curr Diab Rep 2013;13:51–5.10.1007/s11892-012-0343-ySearch in Google Scholar PubMed

79. Wang Y, Chen X, Song Y, Caballero B, Cheskin LJ. Association between obesity and kidney disease: a systematic review and meta-analysis. Kidney Int 2008;73:19–33.10.1038/sj.ki.5002586Search in Google Scholar PubMed

80. Savino A, Pelliccia P, Chiarelli F, Mohn A. Obesity-related renal injury in childhood. Horm Res Paediatr 2010;73:303–11.10.1159/000308161Search in Google Scholar PubMed

81. Filler G, Reimão SM, Kathiravelu A, Grimmer J, Feber J, Drukker A. Pediatric nephrology patients are overweight: 20 years’ experience in a single Canadian tertiary pediatric nephrology clinic. Int Urol Nephrol 2007;39:1235–40.10.1007/s11255-007-9258-ySearch in Google Scholar PubMed

82. Espinoza R, Gracida C, Cancino J, Ibarra A. Effect of obese living donors on the outcome and metabolic features in recipients of kidney transplantation. Transplant Proc 2006;38:888–9.10.1016/j.transproceed.2006.02.044Search in Google Scholar PubMed

83. Burgert TS, Dziura J, Yeckel C, Taksali SE, Weiss R, Tamborlane W, et al. Microalbuminuria in pediatric obesity: prevalence and relation to other cardiovascular risk factors. Int J Obes 2006;30:273–80.10.1038/sj.ijo.0803136Search in Google Scholar PubMed

84. Hirschler V, Molinari C, Maccallini G, Aranda C. Is albuminuria associated with obesity in school children? Pediatr Diabetes 2010;11:322–30.10.1111/j.1399-5448.2009.00599.xSearch in Google Scholar PubMed

85. Savino A, Pelliccia P, Giannini C, De Giorgis T, Cataldo I, Chiarelli F, et al. Implications for kidney disease in obese children and adolescents. Pediatr Nephrol 2011;26:749–58.10.1007/s00467-010-1659-ySearch in Google Scholar PubMed

86. Czech MP. Insulin action and resistance in obesity and type 2 diabetes. Nat Med 2017;23:804–14.10.1038/nm.4350Search in Google Scholar PubMed PubMed Central

87. Lippi G, Targher G, Montagnana M, Salvagno GL, Guidi GC. Relationship between γ-glutamyltransferase, lipids and lipoprotein(a) in the general population. Clin Chim Acta 2007;384:163–6.10.1016/j.cca.2007.06.005Search in Google Scholar PubMed

88. Nimptsch K, Konigorski S, Pischon T. Diagnosis of obesity and use of obesity biomarkers in science and clinical medicine. Metabolism 2019;92:61–70.10.1016/j.metabol.2018.12.006Search in Google Scholar PubMed

89. Unamuno X, Gómez-Ambrosi J, Rodríguez A, Becerril S, Frühbeck G, Catalán V. Adipokine dysregulation and adipose tissue inflammation in human obesity. Eur J Clin Invest 2018;48:e12997.10.1111/eci.12997Search in Google Scholar PubMed

90. Ferrante AW. The immune cells in adipose tissue. Diabetes, Obes Metab 2013;15:34–8.10.1111/dom.12154Search in Google Scholar PubMed PubMed Central

91. Weisberg SP, McCann D, Desai M, Rosenbaum M, Leibel RL, Ferrante AW. Obesity is associated with macrophage accumulation in adipose tissue. J Clin Invest 2003;112:1796–808.10.1172/JCI200319246Search in Google Scholar

92. Lumeng CN. Innate immune activation in obesity. Mol Aspects Med 2013;34:12–29.10.1016/j.mam.2012.10.002Search in Google Scholar PubMed PubMed Central

93. Lumeng CN, Bodzin JL, Saltiel AR. Obesity induces a phenotypic switch in adipose tissue macrophage polarization. J Clin Invest 2007;117:175–84.10.1172/JCI29881Search in Google Scholar PubMed PubMed Central

94. Magrone T, Jirillo E. Childhood obesity: immune response and nutritional approaches. Front Immunol 2015;6:76.10.3389/fimmu.2015.00076Search in Google Scholar PubMed PubMed Central

95. Coppack SW. Pro-inflammatory cytokines and adipose tissue. Proc Nutr Soc 2008;60:349–56.10.1079/PNS2001110Search in Google Scholar

96. Wisse BE. The inflammatory syndrome: the role of adipose tissue cytokines in metabolic disorders linked to obesity. J Am Soc Nephrol 2004;15:2792–800.10.1097/01.ASN.0000141966.69934.21Search in Google Scholar PubMed

97. Nagel G, Rapp K, Wabitsch M, Büchele G, Kroke A, Zöllner I, et al. Prevalence and cluster of cardiometabolic biomarkers in overweight and obese schoolchildren: results from a large survey in Southwest Germany. Clin Chem 2008;54:317–25.10.1373/clinchem.2007.094821Search in Google Scholar PubMed

98. Caballero AE, Bousquet-Santos R, Robles-Osorio L, Montagnani V, Soodini G, Porramatikul S, et al. Overweight latino children and adolescents have marked endothelial dysfunction and subclinical vascular inflammation in association with excess body fat and insulin resistance. Diabetes Care 2008;31:576–82.10.2337/dc07-1540Search in Google Scholar PubMed

99. Tam CS, Garnett SP, Cowell CT, Heilbronn LK, Lee JW, Wong M, et al. IL-6, IL-8 and IL-10 levels in healthy weight and overweight children. Horm Res Paediatr 2010;73:128–34.10.1159/000277632Search in Google Scholar PubMed

100. Trayhurn P, Drevon CA, Eckel J. Secreted proteins from adipose tissue and skeletal muscle – adipokines, myokines and adipose/muscle cross-talk. Arch Physiol Biochem 2011;17:47–56.10.3109/13813455.2010.535835Search in Google Scholar PubMed

101. Achari AE, Jain SK. Adiponectin, a therapeutic target for obesity, diabetes, and endothelial dysfunction. Int J Mol Sci 2017;18:1321.10.3390/ijms18061321Search in Google Scholar PubMed PubMed Central

102. Bianco A, Nigro E, Monaco ML, Matera MG, Scudiero O, Mazzarella G, et al. The burden of obesity in asthma and COPD: role of adiponectin. Pulm Pharmacol Ther 2017;43:20–5.10.1016/j.pupt.2017.01.004Search in Google Scholar PubMed

103. Perakakis N, Triantafyllou GA, Fernández-Real JM, Huh JY, Park KH, Seufert J, et al. Physiology and role of irisin in glucose homeostasis. Nat Rev Endocrinol 2017;13:324–37.10.1038/nrendo.2016.221Search in Google Scholar PubMed PubMed Central

104. Hassink SG, Sheslow DV, De Lancey E, Opentanova I, Considine RV, Caro JF. Serum leptin in children with obesity: relationship to gender and development. Pediatrics 1996;98:201–3.10.1542/peds.98.2.201Search in Google Scholar

105. Crinò A, Greggio NA, Beccaria L, Schiaffini R, Pietrobelli A, Maffeis C. Diagnosis and differential diagnosis of obesity in childhood. Minerva Pediatr 2003;55:461–70.Search in Google Scholar

106. Crocker MK, Yanovski JA. Pediatric obesity: etiology and treatment. Pediatr Clin N Am 2011;58:1217–40.10.1016/j.pcl.2011.07.004Search in Google Scholar PubMed PubMed Central

107. Styne DM, Arslanian SA, Connor EL, Farooqi IS, Murad MH, Silverstein JH, et al. Pediatric obesity-assessment, treatment, and prevention: an endocrine society clinical practice guideline. J Clin Endocrinol Metab 2017;102:709–57.10.1210/jc.2016-2573Search in Google Scholar PubMed PubMed Central

108. Mason K, Page L, Balikcioglu PG. Screening for hormonal, monogenic, and syndromic disorders in obese infants and children. Pediatr Ann 2014;43:e218–24.10.3928/00904481-20140825-08Search in Google Scholar PubMed PubMed Central

109. Niranjan U, Wright NP. Should we treat subclinical hypothyroidism in obese children? Br Med J 2016;352:i941.10.1136/bmj.i941Search in Google Scholar PubMed

110. Sorisky A, Bell A, Gagnon A. TSH receptor in adipose cells. Horm Metab Res 2008;32:468–74.10.1055/s-2007-978672Search in Google Scholar PubMed

111. Valyasevi RW, Harteneck DA, Dutton CM, Bahn RS. Stimulation of adipogenesis, peroxisome proliferator-activated receptor-γ (PPARγ), and thyrotropin receptor by PPARγ agonist in human orbital preadipocyte fibroblasts. J Clin Endocrinol Metab 2002;87:2352–8.10.1210/jcem.87.5.8472Search in Google Scholar PubMed

112. Reinehr T. Thyroid function in the nutritionally obese child and adolescent. Curr Opin Pediatr 2011;23:415–20.10.1097/MOP.0b013e328344c393Search in Google Scholar PubMed

113. Reinehr T. Obesity and thyroid function. Mol Cell Endocrinol 2010;316:165–71.10.1016/j.mce.2009.06.005Search in Google Scholar PubMed

114. Stichel H, L’allemand D, Grüters A. Thyroid function and obesity in children and adolescents. Horm Res 2000;54:14–9.10.1159/000063431Search in Google Scholar PubMed

115. Verma N, Jain V. Iatrogenic Cushing syndrome. Indian Pediatr 2012;49:765.10.1007/s13312-012-0147-9Search in Google Scholar PubMed

116. Magiakou MA. Growth in disorders of adrenal hyperfunction. Pediatr Endocrinol Rev 2004;1:484–9.Search in Google Scholar

117. Vyas V, Kumar A, Jain V. Growth hormone deficiency in children: from suspecting to diagnosing. Indian Pediatr 2017;54:955–60.10.1007/s13312-017-1190-3Search in Google Scholar PubMed

118. Grimberg A, DiVall SA, Polychronakos C, Allen DB, Cohen LE, Quintos JB, et al. Guidelines for growth hormone and insulin-like growth factor-I treatment in children and adolescents: growth hormone deficiency, idiopathic short stature, and primary insulin-like growth factor-I deficiency. Horm Res Paediatr 2017;86:361–97.10.1159/000452150Search in Google Scholar PubMed

119. Cianfarani S, Tondinelli T, Spadoni GL, Scirè G, Boemi S, Boscherini B. Height velocity and IGF-I assessment in the diagnosis of childhood onset GH insufficiency: do we still need a second GH stimulation test? Clin Endocrinol (Oxf) 2002;57:161–7.10.1046/j.1365-2265.2002.01591.xSearch in Google Scholar PubMed

120. Cianfarani S, Liguori A, Boemi S, Maghnie M, Iughetti L, Wasniewska M, et al. Inaccuracy of insulin-like growth factor (IGF) binding protein (IGFBP)-3 assessment in the diagnosis of growth hormone (GH) deficiency from childhood to young adulthood: association to low GH dependency of IGF-II and pre sence of circulating IGFBP-3 18-ki. J Clin Endocrinol Metab 2005;90:6028–34.10.1210/jc.2005-0721Search in Google Scholar PubMed

121. Gelfand IM, Eugster EA, DiMeglio LA. Presentation and clinical progression of pseudohypoparathyroidism with multi-hormone resistance and Albright hereditary osteodystrophy: a case series. J Pediatr 2006;149:877–80.10.1016/j.jpeds.2006.08.050Search in Google Scholar PubMed

122. Perez KM, Lee EB, Kahanda S, Duis J, Reyes M, Jüppner H, et al. Cognitive and behavioral phenotype of children with pseudohypoparathyroidism type 1A. Am J Med Genet Part A 2018;176:283–9.10.1002/ajmg.a.38534Search in Google Scholar PubMed PubMed Central

123. Bakker B, Sonneveld LJ, Woltering MC, Bikker H, Kant SG. A girl with beckwith-wiedemann syndrome and pseudohypoparathyroidism type 1B due to multiple imprinting defects. J Clin Endocrinol Metab 2015;100:3963–6.10.1210/jc.2015-2260Search in Google Scholar PubMed

124. de Lange IM, Verrijn Stuart AA, van der Luijt RB, Ploos van Amstel HK, van Haelst MM. Macrosomia, obesity, and macrocephaly as first clinical presentation of PHP1b caused by STX16 deletion. Am J Med Genet Part A 2016;170:2431–5.10.1002/ajmg.a.37818Search in Google Scholar

125. Chen M, Wang J, Dickerson KE, Kelleher J, Xie T, Gupta D, et al. Central nervous system imprinting of the G protein Gsα and its role in metabolic regulation. Cell Metab 2009;9:548–55.10.1016/j.cmet.2009.05.004Search in Google Scholar

126. Shoemaker AH, Lomenick JP, Saville BR, Wang W, Buchowski MS, Cone RD. Energy expenditure in obese children with pseudohypoparathyroidism type 1a. Int J Obes 2013;37:1147–53.10.1038/ijo.2012.200Search in Google Scholar

127. Roizen JD, Danzig J, Groleau V, McCormack S, Casella A, Harrington J, et al. Resting energy expenditure is decreased in pseudohypoparathyroidism type 1A. J Clin Endocrinol Metab 2016;101:880–8.10.1210/jc.2015-3895Search in Google Scholar

128. Mantovani G, Maghnie M, Weber G, De Menis E, Brunelli V, Cappa M, et al. Growth hormone-releasing hormone resistance in pseudohypoparathyroidism type Ia: new evidence for imprinting of the Gsα gene. J Clin Endocrinol Metab 2003;88:4070–4.10.1210/jc.2002-022028Search in Google Scholar

129. Germain-Lee EL, Ding CL, Deng Z, Crane JL, Saji M, Ringel MD, et al. Paternal imprinting of Gαs in the human thyroid as the basis of TSH resistance in pseudohypoparathyroidism type 1a. Biochem Biophys Res Commun 2002;296:67–72.10.1016/S0006-291X(02)00833-1Search in Google Scholar

130. Shoemaker AH, Jüppner H. Nonclassic features of pseudohypoparathyroidism type 1A. Curr Opin Endocrinol Diabetes Obes 2017;24:33–8.10.1097/MED.0000000000000306Search in Google Scholar PubMed PubMed Central

131. Germain-Lee EL. Short stature, obesity, and growth hormone deficiency in pseudohypoparathyroidism type 1a. Pediatr Endocrinol Rev 2006;3:318–27.Search in Google Scholar

132. Greydanus DE, Agana M, Kamboj MK, Shebrain S, Soares N, Eke R, et al. Pediatric obesity: current concepts. Dis Mon 2018;64:98–156.10.1016/j.disamonth.2017.12.001Search in Google Scholar PubMed

133. Mamun AA, Hayatbakhsh MR, O’Callaghan M, Williams G, Najman J. Early overweight and pubertal maturation – pathways of association with young adults’ overweight: a longitudinal study. Int J Obes 2009;33:14–20.10.1038/ijo.2008.220Search in Google Scholar PubMed

134. Johnson W, Stovitz SD, Choh AC, Czerwinski SA, Towne B, Demerath EW. Patterns of linear growth and skeletal maturation from birth to 18 years of age in overweight young adults. Int J Obes 2012;36:535–41.10.1038/ijo.2011.238Search in Google Scholar PubMed PubMed Central

135. Reinehr T, Hinney A, de Sousa G, Austrup F, Hebebrand J, Andler W. Definable somatic disorders in overweight children and adolescents. J Pediatr 2007;150:618–22.10.1016/j.jpeds.2007.01.042Search in Google Scholar PubMed

136. Kamboj MK, Patel DR. Polycystic Ovarian Syndrome: a diagnostic and therapeutic challenge. J Pediatr Sci 2010;2:e4.Search in Google Scholar

137. Ortega FB, Lavie CJ, Blair SN. Obesity and cardiovascular disease. Circ Res 2016;118:1752–70.10.1161/CIRCRESAHA.115.306883Search in Google Scholar PubMed

138. Bastien M, Poirier P, Lemieux I, Després JP. Overview of epidemiology and contribution of obesity to cardiovascular disease. Prog Cardiovasc Dis 2014;56:369–81.10.1016/j.pcad.2013.10.016Search in Google Scholar PubMed

139. Jiang S-Z, Lu W, Zong X-F, Ruan H-Y, Liu Y. Obesity and hypertension. Exp Ther Med 2016;12:2395–9.10.3892/etm.2016.3667Search in Google Scholar PubMed PubMed Central

140. Hall JE, do Carmo JM, da Silva AA, Wang Z, Hall ME. Obesity-induced hypertension. Circ Res 2015;116:991–1006.10.1161/CIRCRESAHA.116.305697Search in Google Scholar PubMed PubMed Central

141. Ye J. Mechanisms of insulin resistance in obesity. Front Med China 2013;7:14–24.10.1007/s11684-013-0262-6Search in Google Scholar PubMed PubMed Central

142. Noakes TD. So what comes first: the obesity or the insulin resistance? And which is more important? Clin Chem 2018;64:7–9.10.1373/clinchem.2017.282962Search in Google Scholar PubMed

143. Skinner AC, Perrin EM, Moss LA, Skelton JA. Cardiometabolic risks and severity of obesity in children and young adults. N Engl J Med 2015;373:1307–17.10.1056/NEJMoa1502821Search in Google Scholar PubMed

144. Modan M, Halkin H, Almog S, Lusky A, Eshkol A, Shefi M, et al. Hyperinsulinemia. A link between hypertension obesity and glucose intolerance. J Clin Invest 1985;75:809–17.10.1172/JCI111776Search in Google Scholar PubMed PubMed Central

145. Querques F, Cantilena B, Cozzolino C, Esposito MT, Passaro F, Parisi S, et al. Angiotensin receptor I stimulates osteoprogenitor proliferation through TGFβ-mediated signaling. J Cell Physiol 2015;230:1466–74.10.1002/jcp.24887Search in Google Scholar PubMed

146. Lilly CL, Cottrell LE, Giacobbi P, Innes KE, Umer A, Kelley GA. Childhood obesity and adult cardiovascular disease risk factors: a systematic review with meta-analysis. BMC Public Health 2017;17:1–24.10.1186/s12889-017-4691-zSearch in Google Scholar

147. Barker DJ, Osmond C, Forsén TJ, Kajantie E, Eriksson JG. Trajectories of growth among children who have coronary events as adults. N Engl J Med 2005;353:1802–9.10.1056/NEJMoa044160Search in Google Scholar

148. Ohlsson C, Bygdell M, Sondén A, Rosengren A, Kindblom JM. Association between excessive BMI increase during puberty and risk of cardiovascular mortality in adult men: a population-based cohort study. Lancet Diabetes Endocrinol 2016;4:1017–24.10.1016/S2213-8587(16)30273-XSearch in Google Scholar

149. Zheng Y, Song M, Manson JE, Giovannucci EL, Hu FB. Group-based trajectory of body shape from ages 5 to 55 years and cardiometabolic disease risk in 2 US cohorts. Am J Epidemiol 2017;186:1246–55.10.1093/aje/kwx188Search in Google Scholar PubMed PubMed Central

150. Juonala M, Magnussen CG, Berenson GS, Venn A, Burns TL, Sabin MA, et al. Childhood adiposity, adult aadiposity, and cardiovascular risk factors. N Engl J Med 2011;365:1876–85.10.1056/NEJMoa1010112Search in Google Scholar PubMed

151. Bjerregaard LG, Adelborg K, Baker JL. Change in body mass index from childhood onwards and risk of adult cardiovascular disease. Trends Cardiovasc Med 2019;13:29.10.1016/j.tcm.2019.01.011Search in Google Scholar PubMed

152. Ayer J, Charakida M, Deanfield JE, Celermajer DS. Lifetime risk: childhood obesity and cardiovascular risk. Eur Heart J 2015;36:1371–6.10.1093/eurheartj/ehv089Search in Google Scholar PubMed

153. Lawlor DA, Benfield L, Logue J, Tilling K, Howe LD, Fraser A, et al. Association between general and central adiposity in childhood, and change in these, with cardiovascular risk factors in adolescence: prospective cohort study. Br Med J 2010;341:c6224.10.1136/bmj.c6224Search in Google Scholar PubMed PubMed Central

154. de Kroon ML, Renders CM, van Wouwe JP, van Buuren S, Hirasing RA. The terneuzen birth cohort: BMI change between 2 and 6 years is most predictive of adult cardiometabolic risk. PLoS One 2010;5:e13966.10.1371/journal.pone.0013966Search in Google Scholar PubMed PubMed Central

155. Zabarsky G, Beek C, Hagman E, Pierpont B, Caprio S, Weiss R. Impact of severe obesity on cardiovascular risk factors in youth. J Pediatr 2018;192:105–14.10.1016/j.jpeds.2017.09.066Search in Google Scholar PubMed

156. Klop B, Elte JW, Cabezas MC. Dyslipidemia in obesity: mechanisms and potential targets. Nutrients 2013;5:1218–40.10.3390/nu5041218Search in Google Scholar PubMed PubMed Central

157. Gostynski M, Gutzwiller F, Kuulasmaa K, Döring A, Ferrario M, Grafnetter D, et al. Analysis of the relationship between total cholesterol, age, body mass index among males and females in the WHO MONICA Project. Int J Obes 2004;28:1082–90.10.1038/sj.ijo.0802714Search in Google Scholar

158. Yu BL, Zhao SP, Hu JR. Cholesterol imbalance in adipocytes: a possible mechanism of adipocytes dysfunction in obesity. Obes Rev 2010;11:560–7.10.1111/j.1467-789X.2009.00699.xSearch in Google Scholar

159. Burgess S, Ference BA, Staley JR, Freitag DF, Mason AM, Nielsen SF, et al. Association of LPA variants with risk of coronary disease and the implications for lipoprotein(a)-lowering therapies. J Am Med Assoc Cardiol 2018;3:619.10.1001/jamacardio.2018.1470Search in Google Scholar

160. Banach M. Lipoprotein (a) – we know so much yet still have much to learn …. J Am Hear Assoc 2016;5:e003597.10.1161/JAHA.116.003597Search in Google Scholar

161. Lombardo B, Izzo V, Terracciano D, Ranieri A, Mazzaccara C, Fimiani F, et al. Laboratory medicine: health evaluation in elite athletes. Clin Chem Lab Med 2019;57:1450–73.10.1515/cclm-2018-1107Search in Google Scholar

162. Palmeira ÁC, Leal AA, Ramos Nde M, Neto Jde A, Simões MO, Medeiros CC. Lipoprotein (a) and cardiovascular risk factors in children and adolescents. Rev Paul Pediatr 2013;31:531–7.10.1590/S0103-05822013000400017Search in Google Scholar

163. Glowinska B, Urban M, Koput A, Galar M. New atherosclerosis risk factors in obese, hypertensive and diabetic children and adolescents. Atherosclerosis 2003;167:275–86.10.1016/S0021-9150(03)00003-0Search in Google Scholar

164. Brandstätter A, Lingenhel A, Zwiauer K, Strobl W, Kronenberg F. Decrease of Lp(a) during weight reduction in obese children is modified by the apo(a) kringle-IV copy number variation. Int J Obes 2009;33:1136–42.10.1038/ijo.2009.144Search in Google Scholar PubMed

165. Ho HC, Maddaloni E, Buzzetti R. Risk factors and predictive biomarkers of early cardiovascular disease in obese youth. Diabetes Metab Res Rev 2019;35:e3134.10.1002/dmrr.3134Search in Google Scholar PubMed

166. Watts K, Jones TW, Davis EA, Green D. Exercise training in obese children and adolescents: current concepts. Sport Med 2005;35:375–92.10.2165/00007256-200535050-00002Search in Google Scholar PubMed

167. Kelley GA, Kelley KS, Pate RR. Exercise and BMI in overweight and obese children and adolescents: a systematic review and trial sequential meta-analysis. Biomed Res Int 2015;2015:1–17.10.1155/2015/704539Search in Google Scholar PubMed PubMed Central

168. McGovern L, Johnson JN, Paulo R, Hettinger A, Singhal V, Kamath C, et al. Treatment of pediatric obesity: a systematic review and meta-analysis of randomized trials. J Clin Endocrinol Metab 2008;93:4600–5.10.1210/jc.2006-2409Search in Google Scholar PubMed

169. Atlantis E, Barnes EH, Singh MA. Efficacy of exercise for treating overweight in children and adolescents: a systematic review. Int J Obes 2006;30:1027–40.10.1038/sj.ijo.0803286Search in Google Scholar PubMed

170. García-Hermoso A, Sánchez-López M, Martínez-Vizcaíno V. Effects of aerobic plus resistance exercise on body composition related variables in pediatric obesity: a systematic review and meta-analysis of randomized controlled trials. Pediatr Exerc Sci 2015;27:431–40.10.1123/pes.2014-0132Search in Google Scholar PubMed

171. Ruotsalainen H, Kyngäs H, Tammelin T, Kääriäinen M. Systematic review of physical activity and exercise interventions on body mass indices, subsequent physical activity and psychological symptoms in overweight and obese adolescents. J Adv Nurs 2015;71:2461–77.10.1111/jan.12696Search in Google Scholar PubMed

172. Stoner L, Rowlands D, Morrison A, Credeur D, Hamlin M, Gaffney K, et al. Efficacy of exercise intervention for weight loss in overweight and obese adolescents: meta-analysis and implications. Sport Med 2016;46:1737–51.10.1007/s40279-016-0537-6Search in Google Scholar PubMed

173. Lavoie JM, Gauthier MS. Regulation of fat metabolism in the liver: link to non-alcoholic hepatic steatosis and impact of physical exercise. Cell Mol Life Sci 2006;63:1393–409.10.1007/s00018-006-6600-ySearch in Google Scholar PubMed

174. Cayres SU, Agostinete RR, de Moura Mello Antunes B, Lira FS, Fernandes R. Impact of physical exercise/activity on vascular structure and inflammation in pediatric populations: a literature review. J Spec Pediatr Nurs 2016;21:99–108.10.1111/jspn.12149Search in Google Scholar PubMed

175. Dias KA, Green DJ, Ingul CB, Pavey TG, Coombes JS. Exercise and vascular function in child obesity: a meta-analysis. Pediatrics 2015;136:e648–59.10.1542/peds.2015-0616Search in Google Scholar PubMed

176. Escalante Y, Saavedra JM, García-Hermoso A, Domínguez AM. Improvement of the lipid profile with exercise in obese children: a systematic review. Prev Med (Baltim) 2012;54:293–301.10.1016/j.ypmed.2012.02.006Search in Google Scholar PubMed

177. Marson EC, Delevatti RS, Prado AK, Netto N, Kruel LF. Effects of aerobic, resistance, and combined exercise training on insulin resistance markers in overweight or obese children and adolescents: a systematic review and meta-analysis. Prev Med (Baltim) 2016;93:211–8.10.1016/j.ypmed.2016.10.020Search in Google Scholar PubMed

178. Ashor AW, Lara J, Siervo M, Celis-Morales C, Mathers JC. Effects of exercise modalities on arterial stiffness and wave reflection: a systematic review and meta-analysis of randomized controlled trials. PLoS One 2014;9:e110034.10.1371/journal.pone.0110034Search in Google Scholar PubMed PubMed Central

179. Mujika Ĩ. The alphabet of sport science research starts with Q. Int J Sports Physiol Perform 2013;8:465–6.10.1123/ijspp.8.5.465Search in Google Scholar PubMed

180. Kovács E, Siani A, Konstabel K, Hadjigeorgiou C, de Bourdeaudhuij I, Eiben G, et al. Adherence to the obesity-related lifestyle intervention targets in the IDEFICS study. Int J Obes 2014;38:S144–51.10.1038/ijo.2014.145Search in Google Scholar PubMed PubMed Central

181. Rinninella E, Raoul P, Cintoni M, Franceschi F, Miggiano G, Gasbarrini A, et al. What is the healthy gut microbiota composition? A changing ecosystem across age, environment, diet, and diseases. Microorganisms 2019;7:14.10.3390/microorganisms7010014Search in Google Scholar PubMed PubMed Central

182. Almeida A, Mitchell AL, Boland M, Forster SC, Gloor GB, Tarkowska A, et al. A new genomic blueprint of the human gut microbiota. Nature 2019;568:499–504.10.1038/s41586-019-0965-1Search in Google Scholar PubMed PubMed Central

183. Magnúsdóttir S, Heinken A, Kutt L, Ravcheev DA, Bauer E, Noronha A, et al. Generation of genome-scale metabolic reconstructions for 773 members of the human gut microbiota. Nat Biotechnol 2017;35:81–9.10.1038/nbt.3703Search in Google Scholar PubMed

184. Nakayama J, Watanabe K, Jiang J, Matsuda K, Chao SH, Haryono P, et al. Diversity in gut bacterial community of school-age children in Asia. Sci Rep 2015;5:8397.10.1038/srep08397Search in Google Scholar PubMed PubMed Central

185. Costea PI, Hildebrand F, Manimozhiyan A, Bäckhed F, Blaser MJ, Bushman FD, et al. Enterotypes in the landscape of gut microbial community composition. Nat Microbiol 2017;3:8–16.10.1038/s41564-017-0072-8Search in Google Scholar PubMed PubMed Central

186. Christensen L, Roager HM, Astrup A, Hjorth MF. Microbial enterotypes in personalized nutrition and obesity management. Am J Clin Nutr 2018;108:645–51.10.1093/ajcn/nqy175Search in Google Scholar PubMed

187. Nakayama J, Yamamoto A, Palermo-Conde LA, Higashi K, Sonomoto K, Tan J, et al. Impact of westernized diet on gut microbiota in children on Leyte island. Front Microbiol 2017;8:1e18.10.3389/fmicb.2017.00197Search in Google Scholar PubMed PubMed Central

188. Soderborg TK, Borengasser SJ, Barbour LA, Friedman JE. Microbial transmission from mothers with obesity or diabetes to infants: an innovative opportunity to interrupt a vicious cycle. Diabetologia 2016;59:895–906.10.1007/s00125-016-3880-0Search in Google Scholar PubMed PubMed Central

189. Bokulich NA, Chung J, Battaglia T, Henderson N, Jay M, Li H, et al. Antibiotics, birth mode, and diet shape microbiome maturation during early life. Sci Transl Med 2016;8:343ra82.10.1126/scitranslmed.aad7121Search in Google Scholar PubMed PubMed Central

190. Turnbaugh PJ, Ley RE, Hamady M, Fraser-Liggett CM, Knight R, Gordon JI. The Human Microbiome Project. Nature 2007;449:804–10.10.1038/nature06244Search in Google Scholar PubMed PubMed Central

191. Aagaard K, Ma J, Antony KM, Ganu R, Petrosino J, Versalovic J. The placenta harbors a unique microbiome. Sci Transl Med 2014;6:237ra65.10.1126/scitranslmed.3008599Search in Google Scholar PubMed PubMed Central

192. Cho I, Yamanishi S, Cox L, Methé BA, Zavadil J, Li K, et al. Antibiotics in early life alter the murine colonic microbiome and adiposity. Nature 2012;488:621–6.10.1038/nature11400Search in Google Scholar PubMed PubMed Central

193. Turta O, Rautava S. Antibiotics, obesity and the link to microbes – what are we doing to our children? BMC Med 2016;14:57.10.1186/s12916-016-0605-7Search in Google Scholar PubMed PubMed Central

194. Trasande L, Blustein J, Liu M, Corwin E, Cox LM, Blaser MJ. Infant antibiotic exposures and early-life body mass. Int J Obes 2013;37:16–23.10.1038/ijo.2012.132Search in Google Scholar PubMed PubMed Central

195. Ajslev TA, Andersen CS, Gamborg M, Sørensen TI, Jess T. Childhood overweight after establishment of the gut microbiota: the role of delivery mode, pre-pregnancy weight and early administration of antibiotics. Int J Obes 2011;35:522–9.10.1038/ijo.2011.27Search in Google Scholar PubMed

196. Yallapragada SG, Nash CB, Robinson DT. Early-life exposure to antibiotics, alterations in the intestinal microbiome, and risk of metabolic disease in children and adults. Pediatr Ann 2015;44:e265–9.10.3928/00904481-20151112-09Search in Google Scholar PubMed

197. Reinhardt C, Reigstad CS, Bäckhed F. Intestinal microbiota during infancy and its implications for obesity. J Pediatr Gastroenterol Nutr 2009;48:249–56.10.1097/MPG.0b013e318183187cSearch in Google Scholar

198. Isolauri E, Salminen S, Rautava S. Early microbe contact and obesity risk: evidence of causality? Pediatr Gastroenterol Nutr 2016;63:S3–5.10.1097/MPG.0000000000001220Search in Google Scholar PubMed

199. Cox LM, Yamanishi S, Sohn J, Alekseyenko AV, Leung JM, Cho I, et al. Altering the intestinal microbiota during a critical developmental window has lasting metabolic consequences. Cell 2014;158:705–21.10.1016/j.cell.2014.05.052Search in Google Scholar PubMed PubMed Central

200. Brinkworth GD, Noakes M, Clifton PM, Bird AR. Comparative effects of very low-carbohydrate, high-fat and high-carbohydrate, low-fat weight-loss diets on bowel habit and faecal short-chain fatty acids and bacterial populations. Br J Nutr 2009;101:1493–502.10.1017/S0007114508094658Search in Google Scholar PubMed

201. Russell WR, Gratz SW, Duncan SH, Holtrop G, Ince J, Scobbie L, et al. High-protein, reduced-carbohydrate weight-loss diets promote metabolite profiles likely to be detrimental to colonic health. Am J Clin Nutr 2011;93:1062–72.10.3945/ajcn.110.002188Search in Google Scholar PubMed

202. Scott KP, Gratz SW, Sheridan PO, Flint HJ, Duncan SH. The influence of diet on the gut microbiota. Pharmacol Res 2013;69:52–60.10.1016/j.phrs.2012.10.020Search in Google Scholar PubMed

203. Sze MA, Schloss PD. Looking for a signal in the noise: revisiting obesity and the microbiome. MBio 2016;7:e01018-16.10.1128/mBio.01018-16Search in Google Scholar PubMed PubMed Central

204. Walters WA, Xu Z, Knight R. Meta-analyses of human gut microbes associated with obesity and IBD. FEBS Lett 2014;588:4223–33.10.1016/j.febslet.2014.09.039Search in Google Scholar PubMed PubMed Central

205. Finucane MM, Sharpton TJ, Laurent TJ, Pollard KS. A taxonomic signature of obesity in the microbiome? Getting to the guts of the matter. PLoS One 2014;9:e84689.10.1371/journal.pone.0084689Search in Google Scholar PubMed PubMed Central

206. Turnbaugh PJ, Ley RE, Mahowald MA, Magrini V, Mardis ER, Gordon JI. An obesity-associated gut microbiome with increased capacity for energy harvest. Nature 2006;444: 1027–31.10.1038/nature05414Search in Google Scholar PubMed

207. López-Cepero AA, Palacios C. Association of the intestinal microbiota and obesity. P R Heal Sci J 2015;34:60–4.Search in Google Scholar

208. Tremaroli V, Bäckhed F. Functional interactions between the gut microbiota and host metabolism. Nature 2012;489:242–9.10.1038/nature11552Search in Google Scholar PubMed

209. Le Roy CI, Bowyer RC, Castillo-Fernandez JE, Pallister T, Menni C, Steves CJ, et al. Dissecting the role of the gut microbiota and diet on visceral fat mass accumulation. Sci Rep 2019;9:9758.10.1038/s41598-019-46193-wSearch in Google Scholar PubMed PubMed Central

210. Le Roy CI, Beaumont M, Jackson MA, Steves CJ, Spector TD, Bell JT. Heritable components of the human fecal microbiome are associated with visceral fat. Gut Microbes 2018;9:61–7.10.1080/19490976.2017.1356556Search in Google Scholar PubMed PubMed Central

211. Willyard C. Could baby’s first bacteria take root before birth? Nature 2018;553:264–6.10.1038/d41586-018-00664-8Search in Google Scholar PubMed

212. Lauder AP, Roche AM, Sherrill-Mix S, Bailey A, Laughlin AL, Bittinger K, et al. Comparison of placenta samples with contamination controls does not provide evidence for a distinct placenta microbiota. Microbiome 2016;4:29.10.1186/s40168-016-0172-3Search in Google Scholar PubMed PubMed Central

213. Perez-Muñoz ME, Arrieta MC, Ramer-Tait AE, Walter J. A critical assessment of the “sterile womb” and “in utero colonization” hypotheses: implications for research on the pioneer infant microbiome. Microbiome 2017;5:48–67.10.1186/s40168-017-0268-4Search in Google Scholar PubMed PubMed Central

214. Payne MS, Bayatibojakhi S. Exploring preterm birth as a polymicrobial disease: an overview of the uterine microbiome. Front Immunol 2014;5:595.10.3389/fimmu.2014.00595Search in Google Scholar PubMed PubMed Central

215. Thomas EL, Parkinson JR, Hyde MJ, Yap IK, Holmes E, Doré CJ, et al. Aberrant adiposity and ectopic lipid deposition characterize the adult phenotype of the preterm infant. Pediatr Res 2011;70:507–12.10.1203/PDR.0b013e31822d7860Search in Google Scholar PubMed

216. Breukhoven PE, Kerkhof GF, Willemsen RH, Hokken-Koelega AC. Fat mass and lipid profile in young adults born preterm. J Clin Endocrinol Metab 2012;97:1294–302.10.1210/jc.2011-2621Search in Google Scholar PubMed

217. Hobel CJ, Dolan SM, Hindoyan NA, Zhong N, Menon R. History of the establishment of the Preterm Birth international collaborative (PREBIC). Placenta 2019;79:3–20.10.1016/j.placenta.2019.03.008Search in Google Scholar PubMed

218. Spor A, Koren O, Ley R. Unravelling the effects of the environment and host genotype on the gut microbiome. Nat Rev Microbiol 2011;9:279–90.10.1038/nrmicro2540Search in Google Scholar PubMed

219. Zhang C, Zhang M, Wang S, Han R, Cao Y, Hua W, et al. Interactions between gut microbiota, host genetics and diet relevant to development of metabolic syndromes in mice. ISME J 2010;4:232–41.10.1038/ismej.2009.112Search in Google Scholar PubMed

220. Rodríguez JM, Murphy K, Stanton C, Ross RP, Kober OI, Juge N, et al. The composition of the gut microbiota throughout life, with an emphasis on early life. Microb Ecol Health Dis 2015;26:26050.10.3402/mehd.v26.26050Search in Google Scholar PubMed PubMed Central

221. de Goffau MC, Lager S, Sovio U, Gaccioli F, Cook E, Peacock SJ, et al. Human placenta has no microbiome but can contain potential pathogens. Nature 2019;572:329–34.10.1038/s41586-019-1451-5Search in Google Scholar PubMed PubMed Central

222. Collado MC, Laitinen K, Salminen S, Isolauri E. Maternal weight and excessive weight gain during pregnancy modify the immunomodulatory potential of breast milk. Pediatr Res 2012;72:77–85.10.1038/pr.2012.42Search in Google Scholar PubMed

223. Jakobsson HE, Abrahamsson TR, Jenmalm MC, Harris K, Quince C, Jernberg C, et al. Decreased gut microbiota diversity, delayed Bacteroidetes colonisation and reduced Th1 responses in infants delivered by Caesarean section. Gut 2014;63:559–66.10.1136/gutjnl-2012-303249Search in Google Scholar PubMed

224. Bäckhed F, Roswall J, Peng Y, Feng Q, Jia H, Kovatcheva-Datchary P, et al. Dynamics and stabilization of the human gut microbiome during the first year of life. Cell Host Microbe 2015;17:690–703.10.1016/j.chom.2015.04.004Search in Google Scholar PubMed

225. Dominguez-Bello MG, De Jesus-Laboy KM, Shen N, Cox LM, Amir A, Gonzalez A, et al. Partial restoration of the microbiota of cesarean-born infants via vaginal microbial transfer. Nat Med 2016;22:250–3.10.1038/nm.4039Search in Google Scholar PubMed PubMed Central

226. Francino MP. Antibiotics and the human gut microbiome: dysbioses and accumulation of resistances. Front Microbiol 2016;6:1543.10.3389/fmicb.2015.01543Search in Google Scholar PubMed PubMed Central

227. Kumar H, Rautava S, Collado M, Borzykh N, Loyttyniemi E, Isolauri E, et al. Neonatal antibiotic exposure alters compositional gut microbiota development during the first 6 months of life. FASEB Journal Conf Exp Biol 2015;29:1.10.1096/fasebj.29.1_supplement.575.3Search in Google Scholar

228. Ege MJ, Mayer M, Normand A-C, Genuneit J, Cookson WO, Braun-Fahrländer C, et al. Exposure to environmental microorganisms and childhood asthma. N Engl J Med 2011;364:701–9.10.1056/NEJMoa1007302Search in Google Scholar PubMed

229. Zhou D. Impact of sanitary living environment on gut microbiota. Precis Med 2016;2:e1161.Search in Google Scholar

230. Rautava S, Collado MC, Salminen S, Isolauri E. Probiotics modulate host-microbe interaction in the placenta and fetal gut: a randomized, double-blind, placebo-controlled trial. Neonatology 2012;102:178–84.10.1159/000339182Search in Google Scholar PubMed

231. Luoto R, Laitinen K, Nermes M, Isolauri E. Impact of maternal probiotic-supplemented dietary counselling on pregnancy outcome and prenatal and postnatal growth: a double-blind, placebo-controlled study. Br J Nutr 2010;103:1792–9.10.1017/S0007114509993898Search in Google Scholar PubMed

232. Karlsson CL, Önnerfält 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;20:2257–61.10.1038/oby.2012.110Search in Google Scholar PubMed

233. Bervoets L, Van Hoorenbeeck K, Kortleven I, Van Noten C, Hens N, Vael C, et al. Differences in gut microbiota composition between obese and lean children: a cross-sectional study. Gut Pathog 2013;5:10.10.1186/1757-4749-5-10Search in Google Scholar PubMed PubMed Central

234. Zhang C, Yin A, Li H, Wang R, Wu G, Shen J, et al. Dietary modulation of gut microbiota contributes to alleviation of both genetic and simple obesity in children. EBioMedicine 2015;2:968–84.10.1016/j.ebiom.2015.07.007Search in Google Scholar PubMed PubMed Central

235. Dahiya DK, Renuka, Puniya M, Shandilya UK, Dhewa T, Kumar N, et al. Gut microbiota modulation and its relationship with obesity using prebiotic fibers and probiotics: a review. Front Microbiol 2017;8:563.10.3389/fmicb.2017.00563Search in Google Scholar PubMed PubMed Central

236. Speiser PW, Rudolf MC, Anhalt H, Camacho-Hubner C, Chiarelli F, Eliakim A, et al. Childhood obesity. J Clin Endocrinol Metab 2005;90:1871–87.10.1210/jc.2004-1389Search in Google Scholar PubMed

237. August GP, Caprio S, Fennoy I, Freemark M, Kaufman FR, Lustig RH, et al. Prevention and treatment of pediatric obesity: an Endocrine Society Clinical Practice Guideline based on expert opinion. J Clin Endocrinol Metab 2008;93:4576–99.10.1210/jc.2007-2458Search in Google Scholar PubMed PubMed Central

238. Lau DC, Douketis JD, Morrison KM, Hramiak IM, Sharma AM, Ur E, et al. 2006 Canadian clinical practice guidelines on the management and prevention of obesity in adults and children. CMAJ 2007;176:S1–13.10.1503/cmaj.061409Search in Google Scholar PubMed PubMed Central

239. National Health and Medical Research Council. Clinical practice guidelines for the management of overweight and obesity in adults, adolescents and children in Australia. Natl Heal Med Res 2013.Search in Google Scholar

240. Sherafat-Kazemzadeh R, Yanovski SZ, Yanovski JA. Pharmacotherapy for childhood obesity: present and future prospects. Int J Obes 2013;37:1–15.10.1038/ijo.2012.144Search in Google Scholar PubMed PubMed Central

241. Padwal R, Li SK, Lau DC. Long-term pharmacotherapy for overweight and obesity: a systematic review and meta-analysis of randomized controlled trials. Int J Obes 2003;27:1437–46.10.1038/sj.ijo.0802475Search in Google Scholar PubMed

242. Franz MJ, VanWormer JJ, Crain AL, Boucher JL, Histon T, Caplan W, et al. {A figure is presented}Weight-loss outcomes: a systematic review and meta-analysis of weight-loss clinical trials with a minimum 1-year follow-up. J Am Diet Assoc 2007;107:1755–67.10.1016/j.jada.2007.07.017Search in Google Scholar PubMed

243. Iughetti L, Berri R, China M, Predieri B. Current and future drugs for appetite regulation and obesity treatment. Recent Pat Endocr Metab Immune Drug Discov 2009;3:102–28.10.2174/187221409788452345Search in Google Scholar

244. Iughetti L, China M, Berri R, Predieri B. Pharmacological treatment of obesity in children and adolescents: present and future. J Obes 2011;2011:928165.10.1155/2011/928165Search in Google Scholar PubMed PubMed Central

245. Norgren S, Danielsson P, Jurold R, Lötborn M, Marcus C. Orlistat treatment in obese prepubertal children: a pilot study. Acta Paediatr Int J Paediatr 2003;92:666–70.10.1111/j.1651-2227.2003.tb00596.xSearch in Google Scholar

246. McDuffie JR, Calis KA, Uwaifo GI, Sebring NG, Fallon EM, Frazer TE, et al. Efficacy of orlistat as an adjunct to behavioral treatment in overweight African American and Caucasian adolescents with obesity-related co-morbid conditions. J Pediatr Endocrinol Metab 2004;17:307–19.10.1515/JPEM.2004.17.3.307Search in Google Scholar

247. Chanoine J-P, Hampl S, Jensen C, Boldrin M, Hauptman J. Effect of Orlistat on weight and body composition in obese adolescents. J Am Med Assoc 2005;293:2873–83.10.1001/jama.293.23.2873Search in Google Scholar PubMed

248. Zhi J, Moore R, Kanitra L. The effect of short-term (21-day) Orlistat treatment on the physiologic balance of six selected macrominerals and microminerals in obese adolescents. J Am Coll Nutr 2003;22:357–62.10.1080/07315724.2003.10719318Search in Google Scholar PubMed

249. Angelakis E, Armougom F, Million M, Raoult D. The relationship between gut microbiota and weight gain in humans. Future Microbiol 2012;7:91–109.10.2217/fmb.11.142Search in Google Scholar PubMed

250. Kalliomäki M, Collado MC, Salminen S, Isolauri E. Early differences in faecal microbiota composition in children may predict later weight gain. Am J Clin Nutr 2008;7:534–8.10.1093/ajcn/87.3.534Search in Google Scholar PubMed

251. Yin YN, Yu QF, Fu N, Liu XW, Lu FG. Effects of four Bifidobacteria on obesity in high-fat diet induced rats. World J Gastroenterol 2010;16:3394–401.10.3748/wjg.v16.i27.3394Search in Google Scholar PubMed PubMed Central

252. Prodam F, Archero F, Aloisio I, Solito A, Ricotti R, Giglione E, et al. Efficacy of the treatment with Bifidobacterium breve B632 and Bifidobacterium breve BR03 on endocrine response to the oral glucose tolerance test in pediatric obesity: a cross-over double blind randomized controlled trial; Proceedings of the 39° Congresso 2017.Search in Google Scholar

253. Slavin JL. Dietary fiber and body weight. Nutrition 2005;21: 411–8.10.1016/j.nut.2004.08.018Search in Google Scholar PubMed

254. Keithley J, Swanson B. Glucomannan and obesity: a critical review. Altern Ther Health Med 2005;11:30–4.Search in Google Scholar

255. Moreno LA, Tresaco B, Bueno G, Fleta J, Rodríguez G, Garagorri JM, et al. Psyllium fibre and the metabolic control of obese children and adolescents. Utilidad del psyllium para el control metabólico de niños y adolescentes obesos (minirrevisión). J Physiol Biochem 2010;59:235–42.10.1007/BF03179920Search in Google Scholar PubMed

Received: 2019-07-30
Accepted: 2019-11-12
Published Online: 2019-12-10
Published in Print: 2020-08-27

©2019 Walter de Gruyter GmbH, Berlin/Boston

Downloaded on 23.4.2024 from https://www.degruyter.com/document/doi/10.1515/cclm-2019-0789/html
Scroll to top button