ArticlePDF Available

Genetics of obesity

Authors:

Abstract and Figures

There is now widespread recognition that the continuing increase in the prevalence of obesity seen in many countries is likely to have major adverse effects on public health. The National Center for Health Statistics reports that 61% of adults in the United States are overweight and 26% are obese. Also The National Health and Nutrition Examination Survey IV, 1999–2002, documents that 16% of children are overweight and 31% are at risk of becoming overweight or are already overweight, representing nearly a 300% increase since the 1960s. The genetic influences are likely to be particularly powerful in people with severe and early-onset obesity, the group is most likely to suffer adverse clinical consequences. In this review we will discuss the Genetics of body weight regulation including genes encoding factors regulating food/energy intake, genes encoding factors implicated in energy expenditure, and genes encoding factors implicated in adipogenesis as well as syndromic forms of obesity.
Content may be subject to copyright.
REVIEW
Genetics of obesity
Rabah M. Shawky
a,b,
*, Doaa I. Sadik
b
a
Pediatrics Department, Faculty of Medicine, Ain Shams University, Egypt
b
Medical Genetics Centre, Ain Shams University, Cairo, Egypt
Received 15 May 2011; accepted 30 August 2011
Available online 9 December 2011
KEYWORDS
Obesity;
Body weight regulation;
Obesity syndromes
Abstract There is now widespread recognition that the continuing increase in the prevalence of
obesity seen in many countries is likely to have major adverse effects on public health. The National
Center for Health Statistics reports that 61% of adults in the United States are overweight and 26%
are obese. Also The National Health and Nutrition Examination Survey IV, 1999–2002, documents
that 16% of children are overweight and 31% are at risk of becoming overweight or are already
overweight, representing nearly a 300% increase since the 1960s. The genetic influences are likely
to be particularly powerful in people with severe and early-onset obesity, the group is most likely
to suffer adverse clinical consequences. In this review we will discuss the Genetics of body weight
regulation including genes encoding factors regulating food/energy intake, genes encoding factors
implicated in energy expenditure, and genes encoding factors implicated in adipogenesis as well
as syndromic forms of obesity.
Ó2012 Ain Shams University. Production and hosting by Elsevier B.V. All rights reserved.
*Corresponding author at: Medical Genetics Center, Ain Shams
University, Cairo, Egypt. Tel.: +20 2 26859398.
E-mail addresses: shawkyrabah@yahoo.com (R.M. Shawky),
doaadoaaibrahim@yahoo.com (D.I. Sadik).
1110-8630 Ó2012 Ain Shams University. Production and hosting by
Elsevier B.V. All rights reserved.
Peer review under responsibility of Ain Shams University.
doi:10.1016/j.ejmhg.2011.08.005
Production and hosting by Elsevier
The Egyptian Journal of Medical Human Genetics (2012) 13, 11–17
Ain Shams University
The Egyptian Journal of Medical Human Genetics
www.ejmhg.eg.net
www.sciencedirect.com
Contents
1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 12
2. Genetics of body weight regulation . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1. Genes encoding factors regulating food/energy intake . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.1. Mutations in genes encoding leptin and its receptors (LEP, LEPR). . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.2. Mutations in proopiomelanocortin (POMC) gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.3. Mutations in melanocortin 4 receptor (MC4R) gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.4. Mutations in proprotein convertase 1 (PC1) gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.5. Mutations in neuropeptide Y (NPY) gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 13
2.1.6. Mutations in ghrelin receptor gene. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.1.7. Mutations in genes related to food preferences . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2. Genes encoding factors implicated in energy expenditure . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2.1. Mutations in b2-adrenoceptor gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2.2. Mutations in b3-adrenoceptor gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2.3. Mutations in b1-adrenoceptor gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.2.4. Mutations in uncoupling proteins (UCPs) gene . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
2.3. Genes encoding factors implicated in adipogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 14
3. Syndromic forms of obesity . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1. Monogenic human obesity: pleiotropic syndromes . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.1. Bardet-Biedl syndrome (BBS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.2. Albright’s hereditary osteodystrophy syndrome. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.3. Borjeson, Forssman and Lehmann syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.4. Cohen syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.5. Alstro
¨m syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.6. Fragile X syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.7. Ulnar-mammary syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.8. Simpson-Golabi-Behmel, type 2 (SGBS) . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.9. Wilson–Turner syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.1.10. Mehmo syndrome. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.2. Obesity syndromes due to chromosomal rearrangements . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.2.1. Prader-Willi syndrome . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 15
3.2.2. Sim-1 . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
3.2.3. WAGR . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
Conflict of interest. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 16
1. Introduction
There is now widespread recognition that the continuing
increase in the prevalence of obesity seen in many countries
is likely to have major adverse effects on public health [1,2].
Obesity in children is defined as a body mass index (BMI) at
or above the 95th percentile for children of the same age and
sex based on the Centers for Disease Control and Prevention
(CDC) growth charts for children in the United States [3].
With over 1 billion people now overweight or obese [4], the
World Health Organization has proclaimed this to be a global
epidemic. Particularly alarming is the explosion of childhood
obesity. The National Health and Nutrition Examination
Survey (NHANES) IV, 1999–2002, documents that 16% of
children are overweight and 31% are at risk for becoming
overweight or are already overweight, representing a nearly
300% increase since the 1960s and a 45% increase since the
last complete NHANES survey for 1988–1994 [5].
In the search for the environmental drivers of this epidemi-
ological phenomenon, there is some danger that we may
overlook the critical importance of inherited factors in the
determination of interindividual differences in fat mass. The
identification of such factors is of great clinical, as well as
theoretical importance for a number of reasons. Firstly, genet-
ic influences are likely to be particularly powerful in people
with severe and early-onset obesity, the group is most likely
to suffer adverse clinical consequences [6] (Fig. 1). Secondly,
the use of genetics to identify critical molecular components
Genetics of obesity
Non-syndromic Syndromes
Pleiotropic
syndromes
Chromosomal
rearrangement
Monogenic
obesity
Polygenic
obesity
ADRB1, ADRB2,
ADRB3
UCP1, UCP2, UCP3
LEP, LEPR, POMC,
MC4R, PC1, NPY,
SIM1
Figure 1 Genetics of obesity.
12 R.M. Shawky, D.I. Sadik
of the human control system for energy homoeostasis may
help to target safe and specific drug development. Finally, it
is known that diet and exercise programs, while frequently
effective in inducing weight loss, rarely maintain this. It is very
likely that the genetic makeup of an individual may influence
his/her response to particular measures. Ultimately, it should
be possible to identify genetic subgroups of subjects who might
be particularly responsive or resistant to specific environmen-
tal modulations [7].
Twin studies suggest a heritability of fat mass of between
40% and 70% with a concordance of 0.7–0.9 between mono-
zygotic twins compared with 0.35–0.45 between dizygotic twins
[8,9]. Correlation of monozygotic twins reared apart is virtu-
ally a direct estimate of the heritability (although monozygotic
twins do share the intrauterine environment, which may con-
tribute to lasting differences in body mass in later life). Esti-
mates vary from 40% to 70%, depending on the age of the
separation of twins and the length of follow-up [10].
2. Genetics of body weight regulation
Body weight regulation and stability depends upon an axis
with three interrelated components: food intake, energy expen-
diture and adipogenesis, although there are still many un-
known features concerning fuel homoeostasis and energy
balance. There are 358 studies on obese humans reporting po-
sitive associations with 113 candidate genes, among them, 18
genes are supported by at least five positive studies [6].
2.1. Genes encoding factors regulating food/energy intake
It was generally accepted that hypothalamic and brain stem
centres are involved in the regulation of food intake and en-
ergy balance but information on the relevant regulatory fac-
tors and their genes was scarce until the last decade [1].
Insulin remained the only candidate for the key role in body
weight regulation for a long time.
2.1.1. Mutations in genes encoding leptin and its receptors
(LEP, LEPR)
This cytokine-like peptide mainly expressed by adipocytes is
believed to be a key regulator of fat metabolism and energy in-
take. Leptin is the product of human homologue of mouse ‘ob-
ese’ gene, whose homozygous mutation caused hereditary
obesity in mice (monogenic). Certain areas of the hypothala-
mus are rich in specific receptors binding regulatory peptides
and triggering central regulatory mechanisms. Studies in hu-
mans have failed to find leptin or any other mutant gene to
be the unique ‘obesity gene’. Conversely, multifactorial pat-
terns involving the actions of numerous polymorphic gene
products now look more likely, (proopiomelanocortin
(POMC), MC4R and proprotein convertase 1 (PC1) deficiency
[11]). Congenital human leptin deficiency has been identified in
subjects showing severe early-onset obesity (8 years and 86 kg,
or 2 years and 29 kg) with intense hyperphagia and undetect-
able levels of serum leptin due to a frame-shift mutation in
the ob gene (deletion G133) in a homozygosis, which resulted
in a truncated protein not secreted [12]. Children with leptin
deficiency had also profound abnormalities in T-cell number
and function consistent with the high rates of infection and
childhood mortality from infections. Leptin therapy in these
subjects has a major effect on appetite with normalisation of
hyperphagia and reductions in body weight. Leptin receptor-
deficient subjects were also found, with the phenotype being
similar to those with leptin deficiency. The birth weight was
normal, but a rapid weight gain was seen in the first months
of life, with severe hyperphagia and aggressive behaviour when
food was denied [11,12]. Basal temperature and resting meta-
bolic rate were normal and they were normoglycemic with
mildly elevated plasma insulin. They also had mild growth
retardation and impaired basal and stimulated growth hor-
mone secretion [12].
2.1.2. Mutations in proopiomelanocortin (POMC) gene
Homozygous and heterozygous subjects for mutations in
POMC have been found. In neonatal life these subjects showed
adrenocorticotropic hormone (ACTH) deficiency (the POMC
gene encoded ACTH and other peptides), the children have
red hair and pale skin due to the lack of melanocyte-stimulat-
ing hormone (MSH) action at the melanocortin-1 receptors in
skin and hair follicles [12]. The POMC deficiency is associated
with hyperphagia and early-onset obesity due to the lack of
activation of the melanocortin-4 receptor.
2.1.3. Mutations in melanocortin 4 receptor (MC4R) gene
Since 1998 many groups have reported at least 70 mutations in
MC4R that were associated with early onset obesity [13,14].
Other clinical features of MC4R mutation carriers are hyper-
phagia, accelerated linear growth in children and marked
increase in bone mineral density. Probands with homozygous
MC4R mutations show more severe obesity than their hetero-
zygous relatives; thus, the mode of inheritance is codominant
[12]. Severe obesity and early age of onset, may be markers
of MC4R mutations. It has been also shown that pathogenic
MC4R mutations are more prevalent in northern European
populations than in the Mediterranean or even Asian popula-
tions [13]. MC4R mutations have been extensively reported in
French, English, German, American, Italian and Spanish
populations [2,7–10]. It has been estimated that 1–6% of extre-
mely obese individuals harbour functionally relevant MC4R
mutations [7]. Functional analyses of MC4R mutations (mis-
sense, nonsense and frameshift mutations) allow us to classify
them on the basis of their effects on receptor signalling. Muta-
tions that caused intracellular retention of the receptor in vitro
were associated with earlier age of onset and greater severity of
obesity than other mutations [12]. Functional studies showed
that many of the missense mutations also lead to a loss-
of-function of the MC4R [2].
2.1.4. Mutations in proprotein convertase 1 (PC1) gene
Subject carriers of PC1 mutations mainly have severe early-
onset obesity, impaired prohormone processing and hypocort-
isolaemia. Another clinical feature is small intestine
dysfunction, which may result from an erroneous maturation
of propeptides within the PC1-secreting cells along the gut [12].
2.1.5. Mutations in neuropeptide Y (NPY) gene
NPY is released from the arcuate hypothalamic nucleus in fast-
ing or in hypoglycaemia situations, its secretion being inhibited
after food intake. The Leu7Pro polymorphism in the NPY
gene appears to be implicated in lipid metabolism regulation.
Some works reported that carriers of the Pro7 allele had higher
NPY levels and also body fatness [15].
Genetics of obesity 13
2.1.6. Mutations in ghrelin receptor gene
For the ghrelin receptor gene, two SNPs were reported:
Ala204Glu and Phe279Leu, which selectively impair the
constitutive activity of the receptor in humans leading to short
stature and obesity that apparently develop during puberty
[16].
2.1.7. Mutations in genes related to food preferences
The identification of relevant genes related to food prefer-
ences has just started. A novel family of 40–80 human
and rodent G protein-coupled receptors expressed in taste
receptor cells of tongue and palate epithelia has been identi-
fied. Taste 2 receptors (T2Rs) have been shown to function
as bitter taste receptor and T1Rs as putative receptor for
sweet taste. There is no information on polymorphism in
the T1R family genes while some SNPs in T2R have been
reported [17,18]. Rapid progress has been made in this field
to elucidate the genetic mechanism controlling formation of
food preferences.
2.2. Genes encoding factors implicated in energy expenditure
The adaptive thermogenesis in humans is closely related to the
active mobilization of lipids from fat tissues and demands
special interest in relation to obesity. Central neural pathways
responsible for the food intake and energy expenditure
regulation are tightly interconnected. The peripheral transmis-
sion of central commands to the fat stores is mediated by the
sympathetic nervous system. The b-adrenoceptor gene families
(ADRB2, ADRB3, ADRB1) are intensively studied candidate
genes in the obesity field for their participation in energy
expenditure regulation.
2.2.1. Mutations in b2-adrenoceptor gene
The b2-adrenergic receptor gene (ADRB2) encodes a major
lipolytic receptor protein in human fat cells. Two common
polymorphisms of the ADRB2 gene, characterised by an
amino acid replacement of arginine by glycine in codon 16
(Arg16Gly) and glutamine by glutamic acid in codon 27
(Gln27Glu), have been explored in several diseases such as
hypertension and obesity [19–23]. A relationship between
the Arg16Gly polymorphism and an altered function of
the ADBR2 has been reported leading to decreased agonist
sensitivity. Meanwhile, the Gln27Glu variant was also found
to be linked to obesity in some populations. In men, the
27Glu allele has been associated with increased BMI and
subcutaneous fat and with elevated leptin and triglyceride
levels, while in women, the 27Glu variant was reported to
be linked to increased BMI, body fat mass and waist to
hip ratio [23]. However, other studies in Caucasians (Danish
men, Austrian women and German subjects) found no asso-
ciation between the Gln27Glu variant of the ADRB2 gene
and obesity [24].
2.2.2. Mutations in b3-adrenoceptor gene
The b3-adrenergic receptor (ADRB3) protein plays a role in
adipocyte metabolism. It mediates the rate of lipolysis in re-
sponse to catecholamines and their agonists which have poten-
tial anti-diabetes and anti-obesity properties [25,26].A
common polymorphism in this gene, characterised by an ami-
no acid replacement of tryptophan by arginine at position 64
(Trp64Arg), has been identified and may be linked to lower
lipolytic activity and account for lipid accumulation in the adi-
pose tissue [27]. This polymorphism has been associated with
abdominal/visceral fat obesity in several populations such as
Caucasians and Japanese subjects. Similarly, several studies
carried out among Mexican American, Japanese and Cauca-
sian women have shown that carriers of the Arg allele had a
higher BMI and lower reduction in visceral fat after weight
loss [27]. Some authors, however, failed to reproduce the find-
ing on b-adrenoceptors gene variants and further confirmation
is required.
2.2.3. Mutations in b1-adrenoceptor gene
ADRB1 is considered a potential candidate gene for obesity
because of its role in catecholamine-induced energy homoeo-
stasis. Stimulation of ADRB1, a member of G-protein-coupled
receptors, mediates energy expenditure and lipolysis in adipose
tissue [28,29].
2.2.4. Mutations in uncoupling proteins (UCPs) gene
Uncoupling proteins (UCPs) are involved in the modulation
of heat-generating uncoupled respiration at the mitochon-
drial level. They represent a family of carrier proteins local-
ised in the inner layer of mitochondrial membranes [26].
There are different members: UCP1, mostly expressed in
brown adipose tissue, and has a role in thermogenesis,
UCP2 is ubiquitously present in any tissue and UCP3 is
mainly expressed in skeletal muscle and brown adipose tis-
sue. UCP1, UCP2 mediate mitochondrial proton leak releas-
ing energy stores as heat and thereby affecting energy
metabolism efficiency [26]. The actual functions for UCP3
proteins are still under investigation. It has been proposed
that uncoupling proteins act as regulators of energy metab-
olism. They are fatty acid transmembrane transporters in the
mitochondria facilitating proton exchange [26]. UCP2 and
UCP3 are considered as candidate genes for obesity, given
their function in the regulation of fuel metabolism. Several
UCP2 gene variants have been described: a G/A mutation
in the promoter region 2866G/A, a valine for alanine substi-
tution at amino acid 55 in exon 4 (Ala55Val) and a 45 base
pair insertion/deletion in the untranslated region of exon 8
[26,30]. From the literature, it seems that allele G in the
promoter region of UCP2 increases obesity risk while it af-
fords relative protection for type 2 diabetes [26]. Meanwhile
the Ala55Val polymorphism has shown to be associated
with increased exercise efficiency [31,32].
2.3. Genes encoding factors implicated in adipogenesis
The last group of genes acting in connection with the
peripheral regulation of energy expenditure comprises the
transcription factors leading to adipogenesis and adipocytes
differentiation. The key factor is peroxisome proliferator-
activated receptor g, particularly the adipose specific isoform
PPARG2. In a meta-analysis examining the Pro12Ala poly-
morphism in 19 136 subjects, a positive association with
BMI was found [26]. In this study, the frequency of the Ala
allele, similar to other Caucasian populations, was higher in
obese subjects (allelic frequency 0.13) than in controls (0.08),
suggesting that this polymorphism was associated with obesity
[33]. There is also information on the functional role of
PPARG gene variants. Some mutant proteins appear to have
reduced activity [26].
14 R.M. Shawky, D.I. Sadik
3. Syndromic forms of obesity
3.1. Monogenic human obesity: pleiotropic syndromes
There are about 30 Mendelian disorders with obesity as a
clinical feature, often in association with mental retardation,
dysmorphic features and organ specific developmental abnor-
malities (i.e., pleiotropic syndromes) [12,34]. Positional genetic
strategies have led to the recent identification of several differ-
ent causative mutations underlying such syndromes; however,
in most cases the defective gene product is an intracellular pro-
tein that is expressed throughout the body and its function is
unknown. These syndromes include:
3.1.1. Bardet-Biedl syndrome (BBS)
The origin of obesity is more complex in Bardet-Biedl syn-
drome (prevalence of BBS, 1/100 000). It is an autosomal
recessive syndrome characterised by central obesity (75%),
polydactyly, learning disabilities, rod–cone dystrophy, hypo-
gonadism and renal abnormalities. It is a genetically heteroge-
nous disorder that is known to map to at least eight loci, seven
of which have now been identified at the molecular level
(mutations in BBS1–BBS11 genes) [12,34,35].
3.1.2. Albright’s hereditary osteodystrophy syndrome
Albright’s hereditary osteodystrophy is an autosomal domi-
nant disorder due to mutations in GNASI, which encodes
for a-subunit of the stimulatory G protein (Gs a). Maternal
transmission of GNASI mutations leads to Albright’s heredi-
tary osteodystrophy (obesity, short stature, round facies, ecto-
pic tissue ossification) plus resistance to several hormones,
such as parathyroid hormone which activate Gs in their target
tissues, while paternal transmission leads only to pseudo-
hypoparathyroidism [12,34].
3.1.3. Borjeson, Forssman and Lehmann syndrome
Borjeson, Forssman and Lehmann is described as a syndrome
characterized by obesity, moderate to severe mental retarda-
tion, epilepsy, hypogonadism, and with marked gynaecomas-
tia. Mutations in a novel, widely expressed zinc-finger gene
plant homeodomain (PHD)-like finger (PHF6) have been iden-
tified in affected families [36], although the functional proper-
ties of this protein remain unclear.
3.1.4. Cohen syndrome
Cohen syndrome is an autosomal recessive disorder charac-
terized by obesity, mental retardation, microcephaly, promi-
nent upper central incisors and progressive retinochoroidal
dystrophy that is over-represented in the Finnish population,
although cases have been reported worldwide [37]. The ge-
netic locus for Cohen syndrome was mapped to chromo-
some 8q, and a novel gene, COH1, in this locus was
shown to carry mutations in many patients from different
ethnic groups [38].
3.1.5. Alstro
¨m syndrome
Alstro
¨m syndrome is another autosomal recessive disorder
that is characterized by childhood obesity associated with
hyperinsulinaemia, chronic hyperglycaemia and neurosensory
deficits [39]. Subsets of affected individuals present with
additional features such as dilated cardiomyopathy, hepatic
dysfunction, hypothyroidism, male hypogonadism, short
stature and mild to moderate developmental delay [40].
Mutations in a single gene, ALMS1, have been found to
be responsible for all cases of Alstro
¨m syndrome so far
characterized [41]. The ALMS1 protein has no signal se-
quences or transmembrane regions, suggesting an intracellu-
lar localization.
3.1.6. Fragile X syndrome
Fragile X syndrome is characterized by moderate to severe
mental retardation, macroorchidism, large ears, macroceph-
aly, prominent jaw (mandibular prognathism), high-pitched
jocular speech and mild obesity. In 1991, the molecular clon-
ing of the fragile X locus revealed unstable expansions of a
CGG trinucleotide repeat located in the FMR1 (fragile X
mental retardation) gene [42]. The CGG repeat is polymor-
phic in normal population, with alleles of 6 to about 50
CGGs. Large expansions of the repeat (from 230 to >1000
CGGs) are seen in affected patients, with moderate expan-
sions (from 60 to about 200 CGGs) that are unmethylated
are found in normal transmitting males and in the majority
of clinically normal carrier females. Although the exact func-
tion of FMR protein is not known, it may play a role in the
regulation of transport, stability or translation of some mes-
senger RNAs [43].
3.1.7. Ulnar-mammary syndrome
This syndrome characterized by ulnar defects, delayed pub-
erty, and hypoplastic nipples, is due to defect in the gene
TBX3 located in 12q24.1 [44]
3.1.8. Simpson-Golabi-Behmel, type 2 (SGBS)
It is an X-linked overgrowth syndrome with associated visceral
and skeletal abnormalities. Alterations in the glypican-3 gene
(GPC3), which is located on Xq26, have been implicated in
the aetiology of relatively milder cases of this disorder. Not
all individuals with SGBS have demonstrated disruptions of
the GPC3 locus, which raises the possibility that other loci
on the X chromosome could be responsible for some cases of
this syndrome [45].
3.1.9. Wilson–Turner syndrome
This syndrome characterized by X-linked mental retardation
(XLMR), obesity, gynaecomastia, speech difficulties, emo-
tional lability, tapering fingers, and small feet. [46].
3.1.10. Mehmo syndrome
MEHMO (Mental retardation, Epileptic seizures, Hypogeni-
talism, Microcephaly and Obesity) is an X-linked disorder
characterised by mental retardation, epileptic seizures, hypo-
genitalism, microcephaly and obesity. It was recently assigned
to the locus Xp21.1-p22.13 [47].
3.2. Obesity syndromes due to chromosomal rearrangements
3.2.1. Prader-Willi syndrome
The most frequent syndrome is the Prader-Willi syndrome with
a prevalence of about one in 25 000 births and a population prev-
alence of one in 50 000 [48], and is characterised by obesity,
hyperphagia, hypotonia, mental retardation, short stature and
hypogonadiotropic hypogonadism. It is usually caused by lack
of the paternal segment 15q11.2-q12, either through deletion
Genetics of obesity 15
of the paternal critical segment (75%) or through loss of the en-
tire paternal chromosome 15 with the presence of two maternal
homologues in 22% of patients (uniparental maternal disomy).
One suggested mediator of the obesity phenotype is ghrelin, the
stomach-secreted peptide that increases appetite by interacting
with POMC/CART (cocaine- and amphetamine-regulated tran-
script) and NPY hypothalamic neurons whose levels are high in
Prader-Willi syndrome patients.
3.2.2. Sim-1
A girl has been reported with hyperphagia and early-onset
obesity and a balanced translocation between 1p22.1 and
6q16.2, which would be predicted to disrupt the SIM-1 gene
on 6q. The Drosophila single-minded (sim) gene is a regulator
of neurogenesis, and in mouse Sim-1 is expressed in the devel-
oping central nervous system, and is essential for formation of
the supraoptic and paraventricular (PVN) nuclei which express
the melanocortin-4 receptor. Mice heterozygous for loss-
of-function mutations in Sim-1 are obese [48,49]. In humans,
deletion or disruption of the SIM1 region results in either
‘Prader-Willi-like’ phenotype or an early-onset obesity linked
to hyperphagia. A number of patients with obesity, hypotonia
and developmental delay in association with interstitial chro-
mosome 6q deletions have been described [50], although
whether this syndrome can be attributed to SIM-1 is unclear.
3.2.3. WAGR
The WAGR syndrome (Wilms tumour, anorexia, ambiguous
genitalia and mental retardation) is one of the best-studied
contiguous gene syndromes associated with chromosomal
deletions at 11p13, the location of the WT1 gene. [51]. Some
patients with WAGR syndrome and obesity have been
reported with the deletions of chromosome 11p14-p12 [52].
In conclusion, obesity is a complex phenotype, and the
assessment of obese patients should be directed at identifying
genetic conditions so that appropriate genetic counselling
and in some cases treatment can be instituted. An increasing
number of genes linked to human obesity are being identified.
This knowledge will increase our understanding of the causes
of obesity and the ways in which children respond to their
environments and become obese.
Conflict of interest
The author declares that there is no conflict of interest
References
[1] Kopelman PG. Obesity as a medical problem. Nature
2000;404:635–43.
[2] Friedrich MJ. Epidemic of obesity expands its spread to devel-
oping countries. JAMA 2002;287:1382–6.
[3] Centers for Disease Control and Prevention. National Health and
Nutrition Examination Survey, 2006. Retrieved from <http://
www.cdc.gov/nchs/nhanes.htm>.
[4] Froguel P, Boutin P. Genetics of pathways regulating body weight
in the development of obesity in humans. Exp Biol Med
2001;226:991–6.
[5] Joseph A, Skelton Colin D, Rudolph. Overweight and obesity.
Kliegman: Nelson Textbook of Pediatrics, 18th ed. 2007; vol.
1(44).
[6] Maillard G, Charles MA, Tibult N. Trends in the prevalence of
obesity in children and adolescents in France between 1980 and
1991. Int J Obes 2000;24:1608–17.
[7] Stephen O’Rahilly I, Sadaf Farooqi, Giles S, Yeo H, Challis
Benjamin G. Minireview: human obesity––lessons from mono-
genic disorders. Endocrinology 2003;144(9):3757–64.
[8] Stunkard AJ, Foch TT, Hrubec Z. A twin study of human obesity.
JAMA 1986;256:51–4.
[9] Stunkard AJ, Harris JR, Pedersen NL, McClearn GE. The body-
mass index of twins who have been reared apart. N Engl J Med
1990;322:1483–7.
[10] Maes HH, Neale MC, Eaves LJ. Genetic and environmental
factors in relative body weight and human adiposity. Behav Genet
1997;27:325–51.
[11] Zurbano R, Ochoa MC, Moreno-Aliaga MJ, Marti A. Estudios
sobre obesidad de origen monoge ´nico en humanos. Revista
Espan ~ola de Obesidad 2004;2:269–78.
[12] Farooqi IS. Genetic and hereditary aspects of childhood obesity.
Best Pract Res Clin Endocrinol Metab 2005;19:359–74.
[13] Kublaoui BM, Zinn AR. MC4R mutations–weight before screen-
ing! J Clin Endocrinol Metab 2006;91:1671–2.
[14] Lubrano-Berthelier C, Dubern B, Lacorte JM, Picard F, Shapiro
A, Zhang S, et al. Melanocortin 4 receptor mutations in a large
cohort of severely obese adults: prevalence, functional classifica-
tion, genotype-phenotype relationship, and lack of association
with binge eating. J Clin Endocrinol Metab 2006;91:1811–8.
[15] Van Rossum CT, Pijl H, Adan RA, Hoebee B, Seidell JC.
Polymorphisms in the NPY and AGRP genes and body fatness in
Dutch adults. Int J Obes (London) 2006;30:1522–8.
[16] Holst B, Schwartz TW. Ghrelin receptor mutations – too little
height and too much hunger. J Clin Invest 2006;116:637–41.
[17] Hinrichs AL, Wang JC, Bufe B, Kwon JM, Budde J, Allen R,
et al. Functional variant in a bitter-taste receptor (hTAS2R16)
influences risk of alcohol dependence. Am J Hum Genet
2006;78:103–11.
[18] Kim UK, Jorgenson E, Coon H, Leppert M, Risch N, Drayna D.
Positional cloning of the human quantitative trait locus underly-
ing taste sensitivity to phenylthiocarbamide. Science
2003;299:1221–5.
[19] Macho-Azcarate T, Marti A, Gonzalez A, Martinez JA, Ibanez J.
Gln27Glu polymorphism in the beta2 adrenergic receptor gene
and lipid metabolism during exercise in obese women. Int J Obes
Relat Metab Disord 2002;26:1434–41.
[20] Macho-Azcarate T, Calabuig J, Marti A, Martı ´nez JA. A
maximal effort trial in obese women carrying the beta2- adreno-
ceptor Gln27Glu polymorphism. J Physiol Biochem
2002;58:103–8.
[21] Corbala ´n MS, Marti A, Forga L, Martinez-Gonzalez MA,
Martinez JA. The 27Glu polymorphism of the beta2-adrenergic
receptor interacts with physical activity influencing obesity risk
among female subjects. Clin Genet 2002;61:305–7.
[22] Corbalan MS, Marti A, Forga L, Martinez-Gonzalez MA,
Martinez JA. Beta(2)-Adrenergic receptor mutation and abdom-
inal obesity risk: effect modification by gender and HDL-
cholesterol. Eur J Nutr 2002;41:114–8.
[23] Marti A, Corbalan MS, Martinez-Gonzalez MA, Martinez JA.
TRP64ARG polymorphism of the beta3-adrenergic receptor gene
and obesity risk: effect modification by a sedentary lifestyle.
Diabetes Obes Metab 2002;4:428–30.
[24] Marti A, Martinez JA. Genetics of obesity: gene3diet interaction.
Int J Vitam Nutr Res 2006;76:184–93.
[25] Ochoa MC, Marti A, Azcona C, Chueca M, Oyarzabal M, et al
Grupo de Estudio Navarro de Obesidad Infantil (GENOI). Gene–
gene interaction between PPARG2 and ADR beta 3 increases
obesity risk in children. Int J Obes Relat Metab Disord
2004;28(Suppl. 3):S37–41.
[26] Ochoa Mdel C, Marti A, Martinez JA. [Obesity studies in
candidate genes]. Med Clin (Barcelona) 2004;122:542–51.
[27] Park HS, Kim Y, Lee C. Single nucleotide variants in the beta2-
adrenergic and beta3-adrenergic receptor genes explained 18.3%
of adolescent obesity variation. J Hum Genet 2005;50:365–9.
16 R.M. Shawky, D.I. Sadik
[28] Li S, Chen W, Srinivasan SR, Boerwinkle E, Berenson GS.
Influence of lipoprotein lipase gene Ser447Stop and beta(1)-
adrenergic receptor gene Arg389Gly polymorphisms and their
interaction on obesity from childhood to adulthood: the Bogalusa
Heart Study. Int J Obes (London) 2006;30(8):1183–8.
[29] Linne Y, Dahlman I, Hoffstedt J. beta1-Adrenoceptor gene
polymorphism predicts long-term changes in body weight. Int J
Obes (London) 2005;29:458–62.
[30] Zurbano R, Ochoa MC, Moreno-Aliaga MJ, Martinez JA, Marti
AGrupo de Estudio Navarro de la obesidad infantil. [Influence of
the 2866G/A polymorphism of the UCP2 gene on an obese
pediatric population]. Nutr Hosp 2006;21:52–6.
[31] Marti A, Corbalan MS, Forga L, Martinez-Gonzalez MA,
Martinez JA. Higher obesity risk associated with the exon-8
insertion of the UCP2 gene in a Spanish case-control study.
Nutrition 2004;20:498–501.
[32] Alonso A, Marti A, Corbalan MS, Martinez-Gonzalez MA,
Forga L, Martinez JA. Association of UCP3 gene 255C.T
polymorphism and obesity in a Spanish population. Ann Nutr
Metab 2005;49:183–8.
[33] Marti A, Corbalan MS, Martinez-Gonzalez MA, Forga L,
Martinez JA. CHO intake alters obesity risk associated with
Pro12Ala polymorphism of PPARgamma Gene. J Physiol Bio-
chem 2002;58:219–20.
[34] Bell CG, Walley AJ, Froguel P. The genetics of human obesity.
Nat Rev Genet 2005;6:221–34.
[35] Chiang AP, Beck JS, Yen HJ, Tayeh MK, Scheetz TE, Swiderski
RE, et al. Homozygosity mapping with SNP arrays identifies
TRIM32, an E3 ubiquitin ligase, as a Bardet-Biedl syndrome gene
(BBS11). Proc Natl Acad Sci USA 2006;103:6287–92.
[36] Lower KM, Turner G, Kerr BA, et al. Mutations in PHF6 are
associated with Borjeson–Forssman–Lehmann syndrome. Nat
Genet 2002;32:661–5.
[37] Cohen Jr MM, Hall BD, Smith DW, et al. A new syndrome with
hypotonia, obesity, mental deficiency, and facial, oral, ocular, and
limb anomalies. J Pediatr 1973;83:280–4.
[38] Chandler KE, Kidd A, Al-Gazali L, et al. Diagnostic criteria,
clinical characteristics, and natural history of Cohen syndrome. J
Med Genet 2003;40:233–41.
[39] Russell-Eggitt IM, Clayton PT, Coffey R, et al. Alstrom syn-
drome. Report of 22 cases and literature review. Ophthalmology
1998;105:1274–80.
[40] Michaud JL, Heon E, Guilbert F, et al. Natural history of
Alstrom syndrome in early childhood: onset with dilated cardio-
myopathy. J Pediatr 1996;128:225–9.
[41] Collin GB, Marshall JD, Ikeda A, et al. Mutations in ALMS1
cause obesity, type 2 diabetes and neurosensory degeneration in
Alstrom syndrome. Nat Genet 2002;31:74–8.
[42] Hagerman PJ, Hagerman RJ. The fragile-X premutation: a
maturing perspective. Am J Hum Genet 2004;74:805–16.
[43] Jin P, Warren ST. New insights into fragile X syndrome: from
molecules to neurobehaviors. Trends Biochem Sci 2003;28:152–8.
[44] Bamshad M, Lin RC, Law DJ, Watkins WC, Krakowiak PA,
Moore ME, Franceschini P, Lala R, Holmes LB, Gebuhr TC,
Bruneau BG, Schinzel A, Seidman JG, Seidman CE, Jorde LB.
Mutations in human TBX3 alter limb, apocrine and genital
development in ulnar-mammary syndrome. Nat Genet
1997;16:311–5.
[45] Brzustowicz LMFS, Khan MB, Weksberg R. Mapping of a new
SGBS locus to chromosome Xp22 in a family with a severe form
of Simpson-Golabi-Behmel syndrome. Am J Hum Genet
1999;65:779–83.
[46] Wilson M, Mulley J, Gedeon A, Robinson H, Turner G. New X-
linked syndrome of mental retardation, gynecomastia, and obesity
is linked to DXS255. Am J Med Genet 1991;40:406–13.
[47] Leshinsky-Silver E, Zinger A, Bibi CN, Barash V, Sadeh M, Lev
D, Sagie TL. MEHMO (mental retardation, epileptic seizures,
hypogenitalism, microcephaly, obesity): a new X-linked mito-
chondrial disorder. Eur J Hum Genet 2002;10:226–30.
[48] Goldstone AP. Prader–Willi syndrome: advances in genetics,
pathophysiology and treatment. Trends Endocrinol Metab
2004;15:12–20.
[49] Michaud JL, Boucher F, Melnyk A, et al. Sim1 haploinsufficiency
causes hyperphagia, obesity and reduction of the paraventricular
nucleus of the hypothalamus. Hum Mol Genet 2001;10:1465–73.
[50] Faivre L, Cormier-Daire V, Lapierre JM, et al. Deletion of the
SIM1 gene [6q16.2] in a patient with a Prader–Willi-like pheno-
type. J Med Genet 2002;39:594–6.
[51] Rose EA, Glaser T, Jones C, et al. Complete physical map of the
WAGR region of 11p13 localizes a candidate Wilms’ tumor gene.
Cell 1990;60:495–508.
[52] Gul D, Ogur G, Tunca Y, Ozcan O. Third case of WAGR
syndrome with severe obesity and constitutional deletion of
chromosome [11][p12p14]. Am J Med Genet 2002;107:70–1.
Genetics of obesity 17
... Indeed, one molecular genetic study showed that more than 20 obesity susceptible loci have been detected (Herrera and Lindgren 2010). Mutations in those genes encoding food-intake-regulating factors, such as leptin and its pro-opiomelanocortin and melanocortin-4 receptors, are associated with hyperphagia (Shawky and Sadik 2012). Although there is a genetic predisposition to obesity, the risk of obesity is only manifested in an "obesogenic environment," following the interaction between susceptible genes and unfavorable conditions, which include unhealthy lifestyles, foodrich surroundings and influence from family and friends (Pigeyre et al. 2016;Higgs and Thomas 2016). ...
Article
Full-text available
Aim To investigate the association among genetics, lifestyle and environmental factors in obese and non-obese university students in Malaysia. Subjects and methods A cross-sectional study was conducted on students who were studying at universities in Selangor and Kuala Lumpur, Malaysia. Face-to-face interviews were conducted using a pre-tested, self-administered questionnaire, consisting of three major parts: demographics, factors influencing body weight and actions to alter or maintain the present body weight. Results Most of the parents and siblings of young obese participants were also obese compared with non-obese participants (p < 0.001). Family and friends had an influence on the eating habits of both obese and non-obese participants. About 40% of obese participants had a smoking habit. Non-obese participants did not eat much differently, whilst obese participants increased their food intake when they had fewer sleeping hours (p = 0.002). Only 29% of obese participants preferred fruit or vegetables, while 71% preferred salty, baked, fatty and sugary food. More than 50% of non-obese participants always had breakfast. None of the obese participants exercised daily. Stress and poor time management were the major factors preventing obese participants from losing body weight, while encouragement from family members, peers and social influencers were the major motivators for non-obese participants to maintain their body weight. Conclusion Genetics, family and friends, sleep, eating habits, smoking, physical activity and stress have influences on both obese and non-obese cohorts. Obese populations should adopt good discipline, have adequate sleep, observe a healthy diet and maintain an active lifestyle to reduce body weight.
... Peroxisome proliferator-activated receptor gamma (PPARG) and CCAAT/enhancer binding protein-a (C/EBPα) are considered the master regulators of adipogenesis (Rosen & MacDougald, 2006). The storage of excess energy as fat in the adipose tissue results due to an imbalance in the physiological energy homeostasis (Shawky & Sadik, 2012). The energy balance (intake/expenditure) under normal physiological conditions is controlled by environmental factors like diet and physical activity. ...
Article
Full-text available
Obesity is a metabolic disorder that manifests into various forms. Recent studies have indicated that the pomegranate (Punica granatum) seed oil (PSO) has many biologically active components that help in controlling diet‐induced obesity and insulin resistance. However, its impact on the adipogenic differentiation of human adipose‐derived mesenchymal stem cells (HADMSC) remains unclear. Here we have attempted to study the anti‐obesity potential of SHAMstat3pg, a fatty acid composite extracted from PSO. It is composed of three dietary fatty acids: punicic acid [(9Z,11E,13Z)‐9,11,13‐Octadecatrienoic acid], oleic acid [Cis‐9‐Octadecenoic acid], and linoleic acid [(9Z,12Z)‐octadeca‐9,12‐dienoic acid]. In this study, we discuss the impact of the fatty acids on adipogenesis, inflammation, glucose uptake, and mitochondrial ATP production. The impact of SHAMstat3pg on the expression of various obesity‐associated protein and mRNA transcripts in HADMSC was also analyzed. The results indicate that exposure to 10 µg/ml of SHAMstat3pg (24 hr) inhibited adipogenesis of HADMSC, ameliorated inflammation, attenuated ATP production, and glucose uptake. Also, the extract favorably regulated the mRNA expression of the studied obesity‐associated gene transcripts. Practical applications SHAMstat3pg has the potential to serve as a multi‐targeted therapy for the management of obesity. This study demonstrated that the dietary fatty acids inhibited the differentiation of preadipocytes to adipocytes. SHAMstat3pg has also shown to have a favorable impact on the expression of the obesity‐linked proteins and genes in HADMSC that are associated with adipogenesis, inflammation, satiety, energy intake/expenditure (central and peripheral signaling molecules). The study gives an overview of the vast number of genes impacted by the treatment with SHAMstat3pg paving the way for future studies to demonstrate the exact mode of action of how dietary fatty acids can help manage obesity, insulin resistance, and type 2 diabetes.
... The effect of obesity and its consequences affect the annual direct cost of the National Health Service (NHS) which was estimated to be approximately £5.1 billion in 2006-07 [3]. Childhood obesity is also a major public health problem [4], with obesity prevalence increasing from 11.1% and 12.2% to 16.8% and 15.2% in boys and girls respectively in the UK from 1995 to 2009 [5]. Obese children are more likely to become obese adults, with associated health problems and consequential costs [6]. ...
Article
Full-text available
In this paper, association results from genome-wide association studies (GWAS) are combined with a deep learning framework to test the predictive capacity of statistically significant single nucleotide polymorphism (SNPs) associated with obesity phenotype. Our approach demonstrates the potential of deep learning as a powerful framework for GWAS analysis that can capture information about SNPs and the important interactions between them. Basic statistical methods and techniques for the analysis of genetic SNP data from population-based genome-wide studies have been considered. Statistical association testing between individual SNPs and obesity was conducted under an additive model using logistic regression. Four subsets of loci after quality-control (QC) and association analysis were selected: P-values lower than 1x10-5 (5 SNPs), 1x10-4 (32 SNPs), 1x10-3 (248 SNPs) and 1x10-2 (2465 SNPs). A deep learning classifier is initialised using these sets of SNPs and fine-tuned to classify obese and non-obese observations. Using a deep learning classifier model and genetic variants with P-value < 1x10-2 (2465 SNPs) it was possible to obtain results (SE=0.9604, SP=0.9712, Gini=0.9817, LogLoss=0.1150, AUC=0.9908 and MSE=0.0300). As the P-value increased, an evident deterioration in performance was observed. Results demonstrate that single SNP analysis fails to capture the cumulative effect of less significant variants and their overall contribution to the outcome in disease prediction, which is captured using a deep learning framework.
... Genetic factors are the one, which can affects the total cholesterol and fat levels. Mutations in genes encoding leptin and its receptors (LEP, LEPR), situated at hypothalamus, leads to deficiency in leptin sensitivity [19]. POMC deficiency is due to the lack of activation of the melanocortin-4 receptor leads to early-onset obesity. ...
Article
Full-text available
As per WHO, Obesity is abnormal or excessive fat accumulation that may impair health. Women are more obese than the men in Gujarat State. Between the age of 15-49 years, 23.7% Women and 19.7% Men are overweight or obese (BMI ≥ 25.0 kg/m2) as per National Family Health Survey - 4, 2016 -17. Obesity leads to Diabetes mellitus, hypertension, and dyslipidemia. There are two major factors (i) Environmental (ii) Genetic, enhance the blood lipid levels. Basic Pathophysiology of obesity indicates insufficiency of Leptin, Ghrelin Hormones or insufficiency of Leptin, Ghrelin the receptors. Current Drug Treatment includes statins, bile acid sequestrants, ezetimibe that have side effects like rhabdomyolysis, muscle complaints, gastrointestinal disturbances, myalgia. Recent targets of the obesity are Amylin analogues, leptin analogues, GLP-1 analogues, Neuropeptide Y antagonists etc., which affects the hormonal balance into the body. Herbal Drugs are same effective as Current treatment, with no significant side effects. They are easily available from the natural sources, bio-compatible, Eco-friendly in nature. Certain edible mulberries, algaes, are the recent herbal sources for the treatment of the obesity. Key words: Obesity, BMI, Rhabdomyolysis, Myalgia, Eco-Friendly, Edible mulberries, Algae.
... Genetics influence the normal physiology, embryonic development, adaptation, and obesity. Still the research in "genetic environment interactions" is in the early stage [1,2]. ...
Article
Full-text available
p>Obesity is a state of “positive” energy balance when energy intake in is much greater than energy expenditure. The familial aggregation of body size was first published by Sir Francis Galton in 1889, which become a well-established risk factor for childhood obesity. Obesity is often allied with insulin resistance, dyslipidemia and cardiovascular disease. Monogenic obesity includes the involvements of Leptin (LEP) gene, leptin receptor(LEPR), Proopiomelanocortin (POMC), Melanocortin-4 receptor (MC4R), single-minded gene 1 (SIM 1), Proprotein convertase subtilisin/ kexin type 1 (PCSK1), Neuropeptide Y (NPY). Polygenic obesity is due to Adrenoceptor beta 1 (ADRB1), ADRB2, ADRB3, Uncoupling protein 1(UCP1), UCP2, UCP3. Monogenic and polygenic obesity; pleiotropic syndromes, chromosomal rearrangement are different types of obesity with genetic cause. Surprisingly all “obesity genes” carriers do not become overweight, and exercise, maintaining healthy routine is a significant contributing factor to nullify genetic prediposition. Journal of Biomedical Sciences , Vol. 3, No.2, 2016, Page: 24-28</p
Article
Full-text available
Introduction: Endocrine-disruptors are exogenous compounds that interfere with the human biological system. Bisphenol-A and toxic ele-mental mixtures (e.g. As, Pb, Hg, Cd, and U) are major endocrine-disruptive chemicals documented by the USEPA. Globally obesi-ty is a major health problem with increasing fast-food intake among children. The use of food packaging material is rising global-ly, becoming a primary source of chemical migration from food contact materials. Material and methods: This protocol is a cross-sectional study, and the primary outcome is to assess the various dietary and non-dietary exposure sources of endocrine-disruptive chemicals (bisphenol A and heavy metals) through a questionnaire, and quantification of urinary bisphenol A and heavy metals using LC-MS/MS and ICP-MS, respectively, among children. In this study, anthropometric assess-ment, socio-demographic characteristics, and laboratory investigations will be performed. Exposure pathway assessment will be done using questions about household characteristics, surroundings, food and water sources, physical/dietary habits, and nutri-tional assessment. Results: An exposure pathway model will be developed based on the questions on source, pathway/exposure, and receptor (child), of those exposed to or potentially exposed to the endocrine-disruptive chemicals. Conclusions: The children who are exposed or potentially exposed to the chemical migration sources need intervention through local bodies, school curriculum, and training programs. Also, methodological points of view implication of regression models and the LASSO approach will be assessed to identify the emerging risk factors of childhood obesity and even reverse causality through multi-pathway exposure sources. The feasibility of the current study outcome can be implicated in developing countries.
Article
Full-text available
This study investigates the effect of environmental risk factors among North Indian children aged between 6 and 16 years associated with body mass index by comparing fasting and non-fasting urine Bisphenol-A, urine creatinine, and serum thyroid levels with other potential confounding factors. A total of 301 children were enrolled, and term fasting and non-fasting were used based on the sample collection, either first morning or random. Children were grouped into obese, non-obese, and underweight categories based on age- and gender-specific Centre for Disease Control (CDC) 2000 growth chart and grouped according to their body mass index. The overall mean ± SD (standard deviation) of age (years) was 10.45±2.39 (boys age 10.38±2.38 and girls age 10.45±2.40). Urinary Bisphenol-A concentration (ng/mL ± SD) was higher among obese (2.40±4.70) than non-obese (1.30±1.67) and underweight (1.84±3.46) category children. Higher levels of fasting urinary Bisphenol-A (≥2 ng/mL) and thyroid-stimulating hormone (TSH) (≥12.7 μg/dL) were associated with obesity than those of non-fasting children. Being non-fasting among girls at quartile 4 (obese, >2.1800 ng/mL; underweight, >2.1325 ng/mL) had higher odds (obese adjusted odds ratio (aOR) 7.72 (95% CI 0.08–1368.52), underweight adjusted odds ratio (aOR) 12.45 (95% CI 1.55–171.24)] than fasting children. We recommend awareness programs and policy guidelines by engaging teachers and parents for the physical assessment of the presence of Bisphenol-A in food packaging materials to reduce the potential exposure to other chemical migrations sources. Graphical abstract
Article
Full-text available
Obesity is a common serious health problem leading to many serious health disorders. This phenomenon is defined as the over-storage of lipids in adipose tissue that occurs when there is an imbalance between the energy intake and energy used. During obesity, many metabolic alterations occur that can damage several organs, such as vascular or skeletal muscle resulting in the dysfunction of these tissues. In this review, we will discuss molecular genetics and causes of obesity, some of the disorders related to human obesity as well as anti-obesity tool. An interesting solution to the obesity problem is natural substances, revealing anti-obesity activity, as well as functional food enriched with aforementioned substances. Functional foods are products exhibiting a potentially positive effect on health beyond basic nutrition. They contain well-known biologically active natural compounds, which promote optimal health and reduce the risk of many diseases, including obesity.
Article
Full-text available
The regulation of body weight is a very good example of homeostasis, that it follows the feedback regulatory loops. The adipocyte hormone, leptin (OB protein), is proposed to be an "adiposity signal" that acts in the brain to lower food intake, increase the energy expenditure and thus finally the adiposity. As plasma leptin levels are elevated in most overweight individuals, obesity may be associated with leptin resistance. The saturation capacity of leptin transport to reach its site of action i.e. hypothalamus might be one of the probable mechanism for leptin resistance. This review throws light on the mechanism of genesis of obesity with special interest on the role of leptin and its resistance as an important mechanism.
Article
Full-text available
We review the literature on the familial resemblance of body mass index (BMI) and other adiposity measures and find strikingly convergent results for a variety of relationships. Results from twin studies suggest that genetic factors explain 50 to 90% of the variance in BMI. Family studies generally report estimates of parent-offspring and sibling correlations in agreement with heritabilities of 20 to 80%. Data from adoption studies are consistent with genetic factors accounting for 20 to 60% of the variation in BMI. Based on data from more than 25,000 twin pairs and 50,000 biological and adoptive family members, the weighted mean correlations are .74 for MZ twins, .32 for DZ twins, .25 for siblings, .19 for parent-offspring pairs, .06 for adoptive relatives, and .12 for spouses. Advantages and disadvantages of twin, family, and adoption studies are reviewed. Data from the Virginia 30,000, including twins and their parents, siblings, spouses, and children, were analyzed using a structural equation model (Stealth) which estimates additive and dominance genetic variance, cultural transmission, assortative mating, nonparental shared environment, and special twin and MZ twin environmental variance. Genetic factors explained 67% of the variance in males and females, of which half is due to dominance. A small proportion of the genetic variance was attributed to the consequences of assortative mating. The remainder of the variance is accounted for by unique environmental factors, of which 7% is correlated across twins. No evidence was found for a special MZ twin environment, thereby supporting the equal environment assumption. These results are consistent with other studies in suggesting that genetic factors play a significant role in the causes of individual differences in relative body weight and human adiposity.
Article
The bHLH-PAS transcription factor SIM1 is required for the development of the paraventricular nucleus (PVN) of the hypothalamus. Mice homozygous for a null allele of Sim1 (Sim1 -/- ) lack a PVN and die perinatally. In contrast, we show here that Sim1 heterozygous mice are viable but develop early-onset obesity, with increased linear growth, hyperinsulinemia and hyperleptinemia. Sim1 +/- mice are hyperphagic but their energy expenditure is not decreased, distinguishing them from other mouse models of early-onset obesity such as deficiencies in leptin and melanocortin receptor 4. Quantitative histological comparison with normal littermates showed that the PVN of Sim1 +/- mice contains on average 24% fewer cells without a selective loss of any identifiable major cell type. Since acquired lesions in the PVN also induce increased appetite without a decrease in energy expenditure, we propose that abnormalities of PVN development cause the obesity of Sim1 +/- mice. Severe obesity was described recently in a patient with a balanced translocation disrupting SIM1. Pathways controlling the development of the PVN thus have the potential to cause obesity in both mice and humans.
Article
Cohen syndrome is a rare, recessively inherited condition associated with facial dysmorphism, developmental delay, and visual disability. A delay in making the diagnosis commonly occurs, contributed to by the lack of a definitive molecular test and the clinical variability of published case reports. A specific clinical phenotype has been delineated in a homogeneous cohort of Finnish Cohen syndrome patients, but the applicability of their diagnostic criteria to non-Finnish patients has been debated. Detailed delineation of Cohen syndrome in patients from outside Finland is therefore warranted. We report on the clinical features of 33 non-Finnish Cohen syndrome patients. Variability within the clinical spectrum is identified and the natural history of Cohen syndrome described. Diagnostic guidelines for facilitating accurate and early diagnosis are discussed. Results from molecular genetic analysis using markers located within the previously mapped COH1 critical region support allelic but not genetic heterogeneity in this UK cohort.
Article
Height, weight, and body mass index (BMI) were assessed in a sample of 1974 monozygotic and 2097 dizygotic male twin pairs. Concordance rates for different degrees of overweight were twice as high for monozygotic twins as for dizygotic twins. Classic twin methods estimated a high heritability for height, weight, and BMI, both at age 20 years (.80,.78, and.77, respectively) and at a 25-year follow-up (.80,.81, and.84, respectively). Height, weight, and BMI were highly correlated across time, and a path analysis suggested that the major part of that covariation was genetic. These results are similar to those of other twin studies of these measures and suggest that human fatness is under substantial genetic control.(JAMA 1986;256:51-54)