You are on page 1of 37

CHAPTER

90
Disorders of the Body Mass
Patricia A Donohoue and Omar Ali
Medical College of Wisconsin, Milwaukee, WI, USA

90.1 OVERVIEW >30 kg/m2 is accepted as defining obesity in adult Ameri-


There are numerous human conditions that produce cans. For growing children, age- and sex-specific BMI
both elevated and reduced body mass, and extremes at standards are used (Figure 90-1).
both ends of the body-size spectrum are associated with In the past, the CDC recommended that the term obe-
increased relative risk of mortality (1). This chapter sity be avoided, and that the following terms be used:
will be devoted to increased body mass associated with At risk for overweight—BMI between the 85th and 95th
excess fat stores (obesity). Obesity-associated comorbidi- percentile for age and gender; overweight—BMI higher
ties include significantly increased risks for diabetes, car- than 95th percentile for age and sex. But in 2010, based
diovascular disease, the “metabolic syndrome,’’ cancer on the recommendations of an AMA expert panel, it
(2), respiratory disease (asthma, sleep apnea), infertil- was decided that the terminology for children would be
ity, degenerative joint disease, depression, anxiety, and the same as that used in adults, i.e. children whose BMI
discrimination both in social life and in the workplace. ranges from the 85th to 95th percentile for age and sex
It is likely that diabetes-related morbidities will contrib- would be classified as overweight and those who exceed
ute most to the disabilities related to obesity (3). The the 95th percentile will be classified as obese (8).
metabolic syndrome, defined as the combined presence The regulation of body fat stores is complex, and is
of obesity, hyperinsulinemia, hypertension, and hyper- controlled by the interaction of environment and genetic
lipidemia, is increasing in prevalence in parallel with background. The great importance of environmental fac-
obesity in adults (4) and in youth (5,6). For the first tors on body size is underscored by the marked increase
time in known history, the life expectancy in the United in obesity prevalence over the past 30 years, a time period
States may potentially decline because of, in large part, whose brevity most certainly did not allow for a signifi-
the obesity epidemic (7). The etiology of human obesity cant change in the gene pool. The prevalence of obesity
is undoubtedly multifactorial, reflecting complex interac- is increasing at a dramatic rate. This is illustrated in a
tions between genetic background, environmental condi- dramatic animation of US obesity prevalence by states
tions, and developmental processes. compiled by the CDC (http://www.cdc.gov/obesity/data/
trends.html). The prevalence is significantly higher, and
the trend is even more pronounced, in American minor-
90.1.1 Definition ity groups, particularly in females. This is not only occur-
Centers for Disease Control and Prevention (CDC) ring in adults (9) (Figure 90-2), but in youth as well (5)
defines individuals as obese whose body mass index (Table 90-1). There is some indication that the trend may
(BMI) (weight in kilograms divided by the square of the be flattening in some groups, but in others, the preva-
height in meters; kg/m2) exceeds the age- and sex-specific lence of overweight and obesity continues to rise. This
95th percentile. Those who have a BMI between the 85th pattern has been observed throughout the United States
and 95th percentiles are overweight and are at increased (10–12), has accelerated over the past 15  years, and is
risk for obesity-related comorbidities. High BMI corre- mirrored by a rise in the prevalence of type 2 diabetes
lates well with excess body fat in all age groups and in mellitus in adults and youth. International trends paral-
both genders, with the exception of persons with very lel those of the United States (13,14). The prevalence in
high muscle mass (e.g. “body builders”). A cutoff of a given population, such as China, must be determined
© 2013, Elsevier Ltd. All rights reserved. 1
2 CHAPTER 90  Disorders of the Body Mass

FIGURE 90-1  Childhood BMI curves, age 2–20 years. A. BMI for age, boys. http://www.cdc.gov/growthcharts/data/set1clinical/cj41c021.pdf.

using population-specific standards in order to avoid While this pronounced secular trend is almost cer-
underestimating the scale of the problem (15). tainly environmental, it is also clear that there is a sig-
The impact of environment on the epidemic of obesity nificant heritable component in obesity risk, and that this
includes unfavorable trends in food intake and physical genetic component explains up to 80% of the variation
activity, as well as barriers to reversing these trends (16). in BMI within a given environment. This genetic risk
The impact of environment on body size is also under- of obesity may have been acquired during the stage of
scored by the development of obesity in patients who human evolution when acquisition of food was achieved
have survived leukemia (17,18), or who have suffered through strenuous activity (hunting, digging, etc.), and
hypothalamic damage (19), especially patients treated periods of prolonged fasting and famine were constant
for craniopharyngioma (20). In addition, a human virus threats, that genotypes were enriched to favor energy
has been implicated in the etiology of animal obesity storage. In the current environment of plentiful and eas-
(21,22) and human obesity (23). ily accessible high-calorie food, these so-called “thrifty
CHAPTER 90  Disorders of the Body Mass 3

FIGURE 90-1  cont’d. B. BMI for age, girls. http://www.cdc.gov/growthcharts/data/set1clinical/cj41c022.pdf.

genotypes” are highly prevalent and, unfortunately, fat distribution also includes a genetic component (26).
are now detrimental. This evolutionary pressure to pre- Body fat may be preferentially located in the abdomen
vent weight loss has led to the “absence of protection” (android or central obesity pattern) or surrounding the
(against weight gain) model of genetic control of obe- hips and thighs (gynoid obesity pattern). The android
sity. Another model, termed the “Central Resistance” obesity pattern is more strongly associated with dyslip-
model, suggests that human genetic background would idemia, hypertension, and glucose intolerance (27), and
provide equal defenses against weight gain or loss, but is associated with increased intra-abdominal or visceral
that additional genetic or acquired defects impair the fat stores. This is the complex of features known as the
homeostatic control (24). In addition to absolute differ- metabolic syndrome. Thus, even at the same level of
ence in BMI, body composition and the distribution of overweight, an individual with a greater amount of vis-
body fat also play an important role in the development ceral fat is more likely to have or develop serious health
of obesity-related morbidities (25) and variation in body consequences associated with obesity.
4 CHAPTER 90  Disorders of the Body Mass

(A) 40

Overweight

30

Obese
Percentage

20

10

Extremely obese

0
1988–1994 1999–2000 2003–2004 2007–2008
2001–2002 2005–2006
Years

(B) 40

Overweight

30
Percentage

20

Obese

10

Extremely obese
0
1960–1962 1971–1974 1976–1980 1988–1994 1999–2000 2007–2008
2003–2004
Years
FIGURE 90-2  A. Trends in overweight, obesity, and extreme obesity among adults aged 20 years and over: United States, 1988–2008.
B. Trends in overweight, obesity, and extreme obesity among adults aged 20–74 years: United States, 1960–2008.

90.1.2 Heritability of BMI a significant correlation of BMI in biological sibs across


The important influence of heredity on human body size the entire distribution of body sizes. This correlation was
has been demonstrated in multiple studies of dizygotic and much stronger for the full sibs (30). In a study of pediat-
monozygotic twins, and of adopted individuals and their ric twins, there was a genetic contribution to percentage
biological siblings (28). Studies of twin pairs have con- body fat (PBF) measured by bioelectric impedance that
sistently demonstrated higher concordance for body size was distinct from the genetic contribution to BMI. This
among monozygotic than dizygotic twins. In a study of contribution accounted for 75–80% of the variation in
1974 monozygotic and 2097 dizygotic pairs, concordance PBF, and 62.5% of the total genetic variation in PBF was
for body size at six different degrees of overweight (15, due to genes that influenced PBF but not BMI (31).
20, 25, 30, 35, and 40% overweight) at approximately In phenotypic analyses of large unrelated populations,
20 years of age were 1.9–3.6-fold higher for monozygotic there is statistical evidence for recessive gene effects on
than dizygotic pairs (29). In a study of adult adoptees and body size variables, though the specific genes were not
their biological siblings, both full and half sibs, there was identified. Some examples include BMI in Muscatine,
CHAPTER 90  Disorders of the Body Mass 5

(C) 50
1988–1994 2007–2008

40

30
Percentage

20

10

0
Non-Hispanic white Non-Hispanic black Mexican American
Race/ethnicity

(D) 50
1988–1994 2007–2008

40

30
Percentage

20

10

0
Non-Hispanic white Non-Hispanic black Mexican American
Race/ethnicity
FIGURE 90-2  cont’d. C. Prevalence of obesity among men aged 20 years and older, by race and ethnicity: United States, 1988–1994 and
2007–2008. D. Prevalence of obesity among women aged 20 years and over, by race/ethnicity: United States, 1988–1994 and 2007–2008.
All figures available in Reference (9).

Iowa (32), BMI in Caucasian and African-American has been studied extensively with longitudinal follow-up
families (33), abdominal visceral fat in Québec (34), of body size and relative fatness, cardiovascular risk fac-
relative fat pattern in Utah pedigrees (35), and obesity in tors, blood lipid levels, blood pressure, and other phe-
American Pima Indians (36). notypic features for over 40  years. The data obtained
Studies to identify factors contributing to the genetic from the children in this community established normal
epidemiology of childhood obesity have been undertaken ranges for tracking of height, weight, skin fold thickness,
using the population of Muscatine, Iowa. This population blood pressure, cholesterol and triglyceride levels (37).
6 CHAPTER 90  Disorders of the Body Mass

factors controlling this set point (described in Section 90.2).


TA B L E 9 0 - 1    Prevalence of Overweight (BMI
>95th percentile) for American Sustained weight gain is also associated with an increase in
Children 6–19 years of Age urinary norepinephrine excretion and in serum triiodothy-
ronine concentrations, with the reverse patterns seen with
Years Age 6–11 years Age 12–19 years sustained weight loss. The percentage of changes in these
1963–1970a 4.2 4.6 parameters correlated with the percentage of changes in
1971–1974 4.0 6.1 energy expenditure (53). Changes in carbohydrate metab-
1976–1980 6.5 5.0 olism were also noted. In subjects with sustained weight
1988–1994 11.3 10.5
gain, trends toward insulin resistance were more apparent
1999–2000 15.1 14.8
2001–2002 16.3 16.7
in those who were obese than in never obese subjects, sug-
2003–2004 18.8 17.4 gesting to the investigators that there is a threshold effect
2005–2006 15.1 17.8 of total body fat on insulin sensitivity (53). Ethnic variabil-
2007–2008 19.6 18.1 ity in resting energy expenditure has been demonstrated,
a1963–1965
being higher in white than in black prepubertal girls (54),
data are for 6–11-year-old children, and 1966–1970 data are
for 12–17 (not 12–19)-year-old children.
and higher in white than in black prepubertal children,
Data obtained from References (412) and (5). independent of PBF and sex (55). There is ethnic variabil-
ity in insulin sensitivity and atherogenic risk among ado-
lescents of comparable BMI. Black-obese adolescents have
The relationship between ponderosity and increased cor- a more diabetogenic insulin secretion and sensitivity pro-
onary risk factors was established for school children and file than white-obese adolescents. On the other hand, the
their family members (38). As an extension of this study, white-obese youth had more visceral adiposity and athero-
it was demonstrated that there was increased familial car- genic risk factors than their black peers with similar BMI
diovascular mortality among the obese school children (56). Genetic factors are likely to have major influence on
(39,40). A longitudinal study of trends in BMI in this the lower physical activity and resting energy expendi-
population indicated that both genetic and environmen- ture observed in infants who later become obese children
tal factors were involved in the variability of BMI (32). (57). A somewhat genetically isolated population, the Old
The data indicated strong support for a single autosomal- Order Amish, that is known to have high levels of physi-
recessive locus with a major effect that accounted for cal activity has a lower rate of obesity despite high caloric
almost 35% of the variation in adjusted BMI. Polygenic consumption (58).
loci accounted for an additional 42% of the variation.
Therefore, only 23% of the variation was not explained
90.2 GENETIC ARCHITECTURE
by genetic factors and was presumed to represent the
OF OBESITY
environmental influence. The most recent report from the
Muscatine Studies show that childhood BMI is the most The detailed genetic architecture of obesity risk has not
predictive phenotype for adult obesity (41). yet been precisely defined. Even the true magnitude of
In other communities, longitudinal studies have also the heritability of obesity is not yet settled and estimates
demonstrated familial aggregation of obesity and cardio- range from as low as 20% to as much as 80% (59). A
vascular risk. These include the Bogalusa Heart Study relatively small percentage (5% or less) of obesity cases
(42,43), the San Antonio Family Heart Study (44), the is due to monogenic or syndromic obesity (60), but most
Heritage Study (45), the Québec Family Study (46), and cases are due to complex interaction between multiple
studies of American Pima Indians (47). genes and environmental factors. Whether the genetic
Human obesity in rare instances may be associated component of this risk is due to multiple common genetic
with defects at a single genetic locus. These include the variants of small effect (common disease common vari-
Prader–Willi (48) and Bardet–Biedl/McKusick–Kaufman ant hypothesis) or whether the effect is due to multiple
Syndromes (49), Alström Syndrome (50), and intersti- rare variants or even a few rare variants of large effect
tial deletion of chromosome 18 (q12.2q21.1) (51). The has not yet been clarified. But whatever the nature of this
mechanisms by which these genetic defects produce the genetic influence, it does appear to become more promi-
obesity phenotype are not completely known. Several nent as the prevalence and severity of obesity increases
rare single gene defects produce obesity through better in a given population, indicating that genes influencing
understood mechanisms (see Section 90.3). obesity may be more aggressively expressed in an obeso-
An extremely important factor in maintenance of body genic environment (61).
weight is the relationship between body weight and total It is also becoming apparent that individual genomes
energy expenditure (52). The trend toward returning to a differ from each other much more than previously
specific set point for body weight is powerful, and results assumed; for example, it has been estimated that as com-
from not only a reduction in total energy expenditure pared to the reference haploid genome, each individual
in response to weight loss but also an increase in energy human genome on average contains some three and a
expenditure with weight gain. There are no doubt genetic half million single nucleotide variants (SNV) and about
CHAPTER 90  Disorders of the Body Mass 7

1000 copy number variants (CNV) of >450 bp, many of strain, they cause the identical phenotypes of severe
which are rare in the population from which the indi- hyperphagia, obesity, diabetes, defective thermogenesis,
vidual was sampled and are unique to the individual’s and infertility due to hypogonadotropic hypogonad-
family or clan. Each individual genome is truly unique, ism. Parabiosis experiments showed that ob/ob animals
not just in terms of sequence variants in individual genes, were unable to produce a circulating factor regulating
but in terms of the complex interaction between multiple food consumption, and that db/db mice were unable
genes and gene networks. Our models of genetic risk, to respond to this factor despite producing it in exces-
therefore, need to be modified to take the interplay of sive quantities (63). The phenotype of the ob/ob mouse
multiple variants within each unique individual genome is partially obliterated by adrenalectomy (except for the
into consideration (62). thermogenic defect) and these adrenalectomized obese
In the following pages, we will outline the known mice have significantly increased sensitivity to corticoste-
monogenic and syndromic forms of obesity, their animal rone (64). Mice with electrolytic lesioning of the ventro-
models and human correlates, and the results of recent medial hypothalamus (VMH) were found to be resistant
genome-wide scans and linkage studies of common to the circulating factor thought to be absent in ob/ob
(polygenic) obesity, keeping in mind that the boundaries mice, but lacked the other phenotypic features of ob/ob
between these categories are not sharply defined. Genes and db/db such as diabetes, defective thermoregulation,
and chromosomal regions involved in monogenic forms and infertility (65). This led to the theory that the VMH
of obesity are also involved in polygenic common obesity was only one of the several sites of action of this circulat-
and all forms of obesity are ultimately the product of ing “satiety” factor. The phenotypic expression of the
complex interactions between multiple genes and envi- ob/ob and db/db genotypes is highly dependent upon the
ronmental factors. Thus, the same mutation that causes background strain; for example, the BL/Ks background
morbid obesity in one individual may have a significantly is associated with more severe diabetes than the BL/6
attenuated effect in another individual with a different background (65).
individual genetic background, environmental exposures The mutant gene responsible for the phenotype in Lepob
and family and clan genomic structure. mice encodes the protein leptin (66,67), which is deficient
in these animals. The main site of expression of ob (Lep)
mRNA is white adipose tissue (67). The Lep sequence
90.2.1 Animal Models and Human is highly conserved among vertebrates, and the human
Correlates (Spontaneous Mutations) gene maps to chromosome 7p31. Leptin is a cytokine-like
In rodents, multiple examples of spontaneous single hormone secreted mainly by white (not brown) adipose
gene mutations producing obesity are known, and form tissue (67,68), with myriad effects including modulation
the basis for a candidate gene approach to identify the of satiety and basal energy expenditure, and sexual matu-
genes responsible for human obesity (Table 90-2). Several ration. Treatment of ob/ob mice with recombinant leptin
human counterparts of these rodent obesity syndromes or through leptin gene therapy (69,70) corrects the obe-
have been identified, and will be described in Section 90.3. sity/diabetes phenotype.
90.2.1.1 Leptin and Its Receptor.  The prototypic Recombinant leptin treatment causes some weight
obese mice with single gene defects are the obese (ob/ob, reduction in normal mice and rats, but has no effect in
Lepob) and diabetes (db/db, Leprdb) autosomal-recessive db/db mice (71,72). Recombinant leptin also corrects
mutations. If present on the same genetic background the infertility in ob/ob mice (73), and stimulates early

TA B L E 9 0 - 2    Rodent Obesity Mutations, Human Regions of Synteny, and Human Homologs


Mode of Inheritance Rodent Human Syntenic Human Mutation
Mutation Gene (Autosomal) Chromosome Region Described Mutant Protein
Mouse
Agouti Ay Dominant 2 20q11 Noa ASP
Diabetes db Recessive 4 1p31 Yes Lepr
Fat fat Recessive 8 4q21 Nob Carboxypeptidase E
Obese ob Recessive 6 7q31 Yes Leptin
Tubby tub Recessive 7 11p15 Noc Phosphodiesterase
Rat
Fatty fa Recessive 5 1p31 Yes Lepr
Corpulent faK Recessive 5 1p31 Yes Lepr
ASP, Agouti signaling protein; Lepr, leptin receptor; CPE, carboxypeptidase E.
aASP prevents binding of αMSH to its receptor MC4R. Several dominant MC4R mutations are associated with human obesity.
bCPE is required for normal prohormone processing by prohormone convertase 1 (PC1). PC1 mutations are associated with human obesity.
cPhenotype is similar to human Bardet–Biedl and Usher Syndromes.
8 CHAPTER 90  Disorders of the Body Mass

reproductive function despite slower growth in normal (101,102), to obesity-related traits and insulin precur-
mice (74). Leptin acts as a “metabolic gate” allowing sors in Mexican-American families with a type II dia-
for pubertal maturation in the rat (75), similar to the betic proband (103), and to lower leptin levels in obese
effect seen in normal mice. In normal rats, the effects of subjects (104). A leptin gene polymorphism is associated
recombinant leptin are enhanced by VMH injection rela- with hypertension, independent of obesity, in a Japanese
tive to peripheral infusion (76), and leptin induces rapid population (105).
modulation of synaptic transmission in isolated arcuate The cloning of the leptin receptor gene Lepr (106)
nucleus slices from rat hypothalami (77). In rats with from mouse choroid plexus led to characterization of the
streptozocin-induced diabetes, the iatrogenic leptin defi- mutations causing the mouse db/db phenotype, and its
ciency is reversed, and even overcompensated, by leptin rat homologs, the Zucker fatty (fa/fa) and obese Koletsky
treatment. This effect is independent of the amount of phenotypes (107–111). Thus, mutation of the same gene,
weight regained or the resulting blood glucose concen- Lepr, occurred spontaneously in three different strains
trations (78). and produced strikingly similar phenotypes. Multiple
Leptin exists in protein-bound and free states in the splice variants of Lepr exist (112–114), and are expressed
plasma, the latter being the predominant form in obese in various tissues within and outside of the central ner-
subjects (79). The demonstration of high leptin levels in vous system (CNS), including lung, liver, skeletal mus-
the plasma of obese individuals correlating with BMI cle and kidney (106), the major site of leptin clearance
and %BF, along with elevated leptin mRNA in adipose (115). One of these encodes a circulating (short) form
tissue, lead to the hypothesis that obese humans are, to of the leptin receptor, which transports leptin across
some degree, leptin resistant (80). Leptin levels in plasma the blood–brain barrier producing a saturable system of
are normally higher in women than in men (81–83), and transport into the CNS (79). If excess leptin is secreted,
they correlate with %BF rather than genetic background such as in obese individuals with a large fat mass, plasma
in identical twins discordant for obesity (84). Circulat- levels of free leptin will increase significantly because of
ing leptin concentration, when normalized for total fat the saturable transport of the system. The production of
mass, decreases in males as they progress to late puber- this short or soluble form of the receptor is regulated
tal stages, whereas the reverse trend is seen in females independently by many different physiological condi-
(85). Plasma levels decrease significantly after 60  years tions, and serves to modulate the amount of free leptin
of age in both sexes (86). Fasting and sustained weight available to the transmembrane signaling long form of
loss result in a reduction in plasma leptin, however, high- the receptor (116).
fat diet and energy expenditure per se have no effect The cascade of effects downstream of leptin binding to
(83,87). As expected, leptin levels are low in women with its receptors is not completely understood, but involves
anorexia nervosa (88), high in patients with the Prader– activation of members of the STAT family of transcrip-
Willi Syndrome (PWS) (89), and relatively high in new- tional activators (117,118). Both insulin and leptin work
borns (90), but correlate with %BF in all groups. There in parallel in many of these pathways. Treatment of fa/
is a normal oscillation and diurnal rhythm of leptin levels fa (leptin receptor defective) rats with a thermogenic
(91), which shows a blunted pattern in women athletes mixture of sympathomimetic amines (ephedrine and
with amenorrhea relative to athletes with normal menses methylxanthines) can reverse or prevent their obesity
(92). This perhaps provides a correlation to the defect (119,120). Such findings suggest that the downstream
in sexual maturation seen in ob/ob mice. Exposure of effects of leptin include actions on the sympathetic
cultured human hepatocytes to high concentrations of nervous system. This has been confirmed in studies of
leptin results in attenuation of insulin-induced activities rodents as well as humans (121,122). Mice that are het-
including downregulation of gluconeogenesis (93), lead- erozygous for either the ob or the db mutation have body
ing to the possibility that leptin is involved in the patho- composition and leptin homeostasis phenotypes that are
genesis of the diabetes associated with human obesity. intermediate between homozygous wild-type and mutant
The gene encoding human leptin has been studied animals (123).
extensively, but with the exception of leptin deficiency Other downstream effects of leptin include inhibition
caused by rare LEP gene mutations (94), its impor- of the neurons that produce neuropeptide Y (NPY), a
tance in altered human satiety and abnormal body hypothalamic neurotransmitter that can cause obesity
size determination has not been clearly demonstrated. through appetite stimulation (described in more detail
Studies of this gene in large panels of obese and/or dia- below), and which is found at high levels in the hypo-
betic individuals have failed to demonstrate mutations thalami of ob/ob mice (124). Leptin also stimulates
(95–97). Linkage of obesity to this genetic region was neurons that increase production of POMC, which is
not demonstrated in Pima Indians (98), nor in Mexican- known to inhibit feeding. The NPY and POMC neurons
Americans with NIDDM (99), and Mexican-American in the arcuate nuclei of mice are rapidly altered by leptin
obese sib pairs from Starr County, Texas (100). How- administration. Genetically engineered wild-type and
ever, some investigators have shown a suggestion of link- leptin-deficient (ob/ob) mice, who express two differ-
age of polymorphic markers near LEP to extreme obesity ent green fluorescent proteins in the two neuronal types,
CHAPTER 90  Disorders of the Body Mass 9

were administered systemic leptin treatment. In the In population genetic studies, the LEPR region has
ob/ob mice, who normally had increased NPY excitatory shown no relation to body size variables in the Pima
synapses and decreased inhibitory POMC synapses, Indians (98), but a possible correlation was seen with
leptin treatment caused a rapid change in this pattern. acute insulin response in the same group (129). In the
Within 6 h, before any effect on food intake or weight Québec Family Study, results of sib-pair linkage analy-
was observed, the numbers of excitatory and inhibitory sis with 137 adult sibships suggested linkage of body
synapses became indistinguishable from those of the fat and insulin levels to polymorphic markers on chro-
wild-type, untreated mice (Figure 90-3) (125). mosome 1p32–p22, the cytogenetic location of LEPR
In addition to the effects on feeding, leptin receptor- (130). These markers spanned the regions syntenic with
mediated effects alter bone metabolism, the immune mouse db as well as one of the 10 mouse quantitative
system, angiogenesis, and the cardiovascular system trait loci (QTLs) related to susceptibility to diet-induced
(126,127). Thus, it is clear that animals or humans with obesity, termed Do1 (131). Polymorphisms within the
defective leptin receptors would require therapeutic LEPR introns (130) and exons (132) have been tested
intervention aimed at a number of organ systems in order for their relationship to obesity in humans. Among these,
to correct the defect. Multiple factors exist that result the sequence variations at coding exons 2 (Lys109Arg),
in leptin resistance at the tissue level in obese subjects, 4 (Lys204Arg and Gln223Arg), and 12 (Lys656Asn)
including interactions with transcription factors that are are the most likely to have effects on receptor function.
associated with obesity-induced endoplasmic reticulum These amino acid substitutions are at positions that are
(ER) stress. These factors attenuate leptin signaling, and conserved in rat, mouse, and humans. In addition, the
may, in fact, promote more weight gain and adiposity in codon 223 and 656 variants cause changes in charge
obese individuals (128). (neutral to positive, and positive to neutral, respectively).
Of 20 nondiabetic American Pima Indians chosen for
extremes in body size, seven LEPR sequence variations
were identified, including Lys109Arg and Gln223Arg.
Three of these variants were within noncoding regions
of the gene and these were found exclusively in obese
Pima Indians (133). Lys109Arg and Gln223Arg were
associated with BMI and fat mass in Caucasians, but not
in African-Americans in the HERITAGE Family Study
(134). The Lys656Asn variant was significantly linked to
BMI in the Muscatine (135), and Gln223Arg was linked
to fat mass in the Québec Family Study (136). On the
other hand, in adult subjects from Baltimore, Gln223Arg
and Lys656Asn were not associated with obesity traits
(137), and the codon 109, 204, 223, and 656 changes
were not positively associated with juvenile-onset obesity
in 56 Danish men (138). However, a meta-analysis of
LEPR polymorphisms failed to confirm this as an impor-
tant population-wide locus (139).
Severe early-onset obesity caused by a mutant leptin
receptor was originally identified in three female sib-
lings (140). These homozygotes had failure of pubertal
development, and reduced growth hormone and thyroid
stimulating hormone secretion. Despite absence of leptin
signaling, and massively elevated plasma leptin concen-
trations, leptin mRNA levels in adipose tissue, assessed
by quantitative reverse transcription polymerase chain
reaction, were as expected for their BMI (141). This
argues against a direct negative feedback loop in regula-
tion of leptin gene expression in humans.
FIGURE 90-3  Changes in hypothalamic synapses of ob/ob mice 90.2.1.2 Tubby.  The tubby (tub) mutation in mice is an
after leptin treatment. This diagram demonstrates the difference in autosomal-recessive trait, which is associated with less-
the number of excitatory (red) and inhibitory (blue) synaptic inputs severe obesity and insulin resistance than ob or db, is not
onto NPY (coexpressing AgRP) (yellow) and POMC (green) neurons associated with significant hyperphagia, causes variable
in wild-type and ob/ob mice. Leptin treatment to ob/ob mice rapidly
reversed the number of inputs onto NPY and POMC to wild-type degrees of hyperlipidemia depending on background
levels. NPY, neuropeptide Y; AgRP, Agouti-related protein; POMC, strain, and is more severe in males than females. The pheno-
proopiomelanocortin. Pinto et al. (125), Figure 94-4C, page 114. type also includes retinal degeneration and sensorineural
10 CHAPTER 90  Disorders of the Body Mass

hearing loss similar to the human Usher, Alström, and been identified with compound heterozygous POMC
Bardet–Biedl syndromes (BBS) (50,142,143). The map- gene mutations. They had early-onset severe obesity,
ping (144) and cloning (145,146) of tub has led to its congenital ACTH deficiency, and red hair pigmenta-
characterization as a gene which is expressed mainly tion (154). Both mutations alter the cleavage of POMC,
in the hypothalamus and encodes a protein with phos- and blood levels of POMC cleavage products (ACTH,
phodiesterase-like sequences, possibly affecting cellular αMSH) were subnormal. The heterozygous parents were
apoptosis. Further studies have identified and character- unaffected. In genome-wide searches, the region contain-
ized several Tubby-like proteins (TULPs), mutations of ing the POMC locus is linked to serum leptin levels and
which in mammals can cause retinal degeneration, and/ fat mass in Mexican-Americans (155,156), and to serum
or hearing loss (147), as well as alterations in neural tube leptin levels in African-Americans (157) and French
formation (148). The exact biological functions of these Caucasians (158). However, POMC coding region and
proteins remain unknown, however, their roles in com- promoter region sequence variants were not related to
plex cellular functions are being characterized. They are obesity in Caucasians with juvenile-onset obesity (159).
membrane-bound transcriptional activators that translo- A similar negative association with either extreme of
cate to the nucleus, and interact in the G-protein system body size was seen when the POMC coding region was
of intracellular signaling (149). It appears that TUB is screened in patients who were obese, underweight, or
the only member of this protein family that is implicated with anorexia nervosa (160).
in obesity. It is highly concentrated in the nuclei of the 90.2.1.5 Agouti.  The yellow mutation of Agouti mice is
hypothalamus that are involved in energy regulation a dominant trait which causes yellow coat color (rather
(148). Human tub maps to chromosome 11p15. Linkage than the wild-type hairs which are banded black and
of human obesity to this cytogenetic region has not been yellow), obesity and diabetes. This was the first rodent
demonstrated, nor have tub mutations. obesity gene to be cloned, and subsequently over 30
90.2.1.3 Fatty.  In the fatty (fat/fat) mouse, the reces- Agouti yellow alleles have been identified, which vary in
sively inherited mutation causes hyperinsulinemia with- their phenotypic expression. The molecular defect in the
out hyperglycemia, and postpubertal obesity that is most dominant form Ay is a deletion in the 5′ regulatory
less severe than that seen in ob/ob or db/db mice. The region, which brings a different promoter into contact
“hyperinsulinemia” is actually due to hyperproinsu- with the coding region. This results in excessive produc-
linemia. The molecular defect is in the gene encoding tion of Agouti signaling protein (ASP) through ectopic
carboxypeptidase E (CPE), an endoprotease required for overexpression of normal Agouti mRNA in many tissues,
normal processing of prohormones to active hormones, rather than its normal site of expression which is limited
including proinsulin to insulin (150), and for transport to the skin (161). Other Agouti alleles also result from
and processing of propeptides in the granules of the regu- mutations that cause abnormal promoters to be utilized,
lated secretory pathways of the CNS. The primary exam- such as Aiapy which has an insertion of retroviral DNA
ple of such a peptide is proopiomelanocortin (POMC), between exons C and D, directing use of an aberrant
which is the precursor for adrenocorticotropic hormone promoter. This mutation is paternally imprinted. The
(ACTH), melanocyte-stimulating hormone (MSH), beta- Aiy and Asy mutations have insertions upstream of the
endorphin, and beta-lipoprotein. POMC has a “sorting first coding exon acting as ubiquitous promoters. When
signal” consisting of two acidic N-terminal residues, transgenic mice overexpress ASP only in the hair follicles
which bind to the two basic residues of CPE. Proinsulin of skin, obesity and diabetes are not observed, leading
and proenkephalin also attach to this CPE-binding site to the hypothesis that these effects are due to a para-
(151). POMC and proinsulin are then cleaved by prohor- crine rather than an endocrine effect of ASP (161). The
mone convertase 1 (PC1). large number of these dominant Agouti alleles makes it
The role of CPE mutations (fa) in the expression of likely that this locus has a propensity toward spontane-
the human obesity phenotype is unknown; however, a ous mutations, making it an interesting candidate gene
correlate has been described in which there are muta- for studies of the genetics of obesity.
tions of the PC1 gene (152). This patient has obesity ASP and another similar peptide, Agouti-related pep-
along with impaired processing of insulin leading to dia- tide (AgRP) act as antagonists of native ligand binding
betes and hyperproinsulinemia, and ACTH deficiency to melanocortin receptors 1–4 (MC1R through MC4R),
due to impaired processing of POMC. Polymorphic each of which are expressed differentially in specific tis-
genotypes of the PC1 gene and neighboring anonymous sues. AgRP binds specifically to the MC4R in the CNS,
DNA markers, however, do not confer susceptibility to and ASP binds to both CNS MC4R and to skin MC1R
obesity, NIDDM, or gestational diabetes (153), but may (162). Transgenic mice with a knockout (KO) mutation
play a role as one of the important polygenes in these of the CNS-expressed MC4R (MC4R-KO) have features
syndromes. of the yellow Agouti phenotype. Both Ay and MC4R-KO
90.2.1.4 POMC.  Since POMC processing defects lead to mice have high levels of NPY expression in an abnormal
obesity, it is not surprising that the POMC locus itself is CNS site, the dorsal medial hypothalamus, which may
important in body size determination. Two siblings have be an etiologic factor for the obesity phenotype in this
CHAPTER 90  Disorders of the Body Mass 11

syndrome (163). Mice with MC4R haploinsufficiency UCP-mediated thermogenesis in BAT. The β3-AR is
have an obese phenotype that is intermediate between thought to be adipose specific, is the predominant
wild-type and null mice (164). subtype in rodent BAT, and can be chronically
90.2.1.6 Melanocortin Receptors.  Although ASP stimulated by its specific agonists, as opposed to the
mutations have not yet been described in humans, sev- β1- and β2-ARs which become refractory to chronic
eral MC4R mutations are now known to be associated stimulation (178). In transgenic mice deficient in BAT
with obesity. Two frameshifts, a 4-bp deletion at codon through targeted disruption of the UCP-1 gene, obesity
211 and a 4-bp insertion at nucleotide 732 of the cod- develops in the absence of hyperphagia (179). The
ing sequence, are associated with dominantly inherited BAT-deficient mice have enhanced susceptibility to diet-
obesity (165,166). A novel in-frame deletion of codons induced obesity and its comorbidities (insulin resistance,
88–92 has been described in one obese female (167). Sev- hyperglycemia, and hyperlipidemia) (180), which are
eral other MC4R mutations have been identified among not reversed by leptin treatment (181). Alternatively,
obese cohorts (168,169). Up to 10% of obese subjects in constitutive transgenic overexpression of UCP-1 in both
Germany have MC4R mutations (168). The prevalence BAT and WAT of mice cause significant reduction in
rate appears to be lower in the United States (170). Func- subcutaneous fat, both in normal and genetically obese
tional analyses of several mutations have confirmed their (Avy) mice (182). Consumption of a high-fat diet can
deleterious effects on receptor function (169–171). The alter UCP gene expression in some mouse strains but
codon 88–92 deletion prevents ligand binding (167). The not others, indicating variable polygenic control of body
dominant nature of these mutations is presumed to be composition and thermogenesis (183). In normal rats,
due to haploinsufficiency as it is in the transgenic mouse UCP protein and mRNA are regulated in a tissue-specific
model. In one child, homozygous for a 2-bp deletion and subtype-specific manner by fasting and by leptin
causing premature termination of MC4R, the obesity treatment. In some circumstances, the effects on specific
phenotype was more severe and of earlier onset than UCP subtype protein and mRNA are disparate (184).
in his heterozygous siblings and parents (172). Interest- In normal mice, there is strain-specific variation in the
ingly, this child did not have any significant differences prevention of high-fat-diet-induced obesity by β3-AR
in insulin or glucose levels during an oral glucose toler- agonists, which is related to the ability to recruit BAT
ance test, when compared with 39 BMI/age/sex-matched in WAT anatomic sites (185). Targeted disruption of
control children. the β3-AR gene results in a much milder phenotype than
In the Québec Family Study, linkage and association seen in the BAT-deficient mice. They have only modestly
analyses showed that both MC4R and MC5R genotypes increased fat stores (females greater than males), but also
were related to obesity phenotypes (173). The melano- have upregulated levels of β1 but not β2-AR mRNA in
cortin receptor MC3R mutation I183N, identified in a adipose tissue (in BAT more so than in WAT) suggesting
father and daughter with high PBF, alters agonist-induced crosstalk among the receptor subtypes (186). In WAT
G-protein activation (174). It does not exert a dominant of ob/ob mice, lipolysis and adenylyl cyclase activation
negative effect on the wild-type receptor, thus is likely to by the βARs is deficient. Whereas the β3-AR is the main
cause its phenotypic effect through haploinsufficiency. In activator of cyclase activity in WAT of lean mice, it is
mice with MC3R KO, there is a decrease in food intake only partially responsible for adenylyl cyclase activity in
and normal energy expenditure, but they have twice the ob/ob mouse WAT (187,188). In normal rats, the inter-
fat mass of their wild-type littermates (174). action between the sympathetic nervous system, leptin,
UCPs, and regulation of fat cell energy utilization is com-
plex. The in vivo modulation of leptin and UCPs appears
90.2.2 Energy Expenditure to depend on sympathetic nerve activity, plasma insulin
90.2.2.1 Hormonal and Genetic Control.  The genetic and glucose, and hemodynamic factors (189).
control of energy expenditure and heat production is an Variation of the human β3-AR (ADRB3) gene sequence
area of intense investigation as many of these metabolic has been studied extensively, again with disparate results.
and neuronal pathways are involved in expression of the The human gene has been cloned and its product exhibits
genetic defects described above. Defects specific to this properties pharmacologically identical to rodent β3-ARs
system are also important as independent contributors (190). In omental fat tissue from obese individuals, there
to obesity. In rodents, the generation of heat by brown is increased catecholamine-induced lipolysis relative to
adipose tissue (BAT) is related to the pathways controlling cells from nonobese subjects, and this effect is mainly
body fat content and distribution. BAT generates heat due to increased β3-AR function (191). An ADRB3 mis-
through its capacity for uncoupling of mitochondrial sense mutation (Trp64Arg) has been associated with ear-
respiration from oxidative phosphorylation via several lier onset of NIDDM and lower resting metabolic rate
mitochondrial uncoupling proteins (UCPs) (175–177). in Pima Indians (192), abdominal obesity, insulin resis-
BAT as well as white adipose tissue (WAT) contains the tance, and BMR in Finns (193,194), and an increased
beta adrenergic receptors (βARs). The βARs mediate capacity to gain weight in a French cohort (195). There is
lipolysis in BAT and WAT, as well as mitochondrial no association between this polymorphism and NIDDM
12 CHAPTER 90  Disorders of the Body Mass

susceptibility in at risk family members (196), obesity no association between UCP-2 genotypes and obesity in
phenotypes in the Québec Family Study or in the Swed- French Caucasians (217). Allele frequencies of a UCP-
ish Obese Subjects Cohorts (197), obesity and NIDDM 2 gene polymorphism at codon 55 (A55V) were similar
susceptibility in Nauruans (198), obesity in Japanese in African-Americans and Caucasian Americans (218),
men (199), BMI gained or response to a hypocaloric diet but its association with body size was not character-
in morbidly obese Finns (200), or obesity in a population ized. A 45-bp insertion polymorphism of the 3′ flanking
of healthy British men (201). region of the UCP-2 gene is correlated with high BMI
A polymorphism of UCP-1 (202) is reportedly associ- and %BF in children (219). This insertion/deletion poly-
ated with resistance to weight loss with a low-calorie diet morphism results in mRNA species of different lengths,
in the French (203), and this polymorphism may have an and the insertion variant is less stable. The insertion vari-
additive effect with the ADRB3 polymorphism on rate of ant is in linkage disequilibrium with a pair of linked pro-
weight gain in morbid obesity (204). From a physiologic moter region sequence variants (T/A at −2723 and G/A
standpoint, there is no evidence quantifying the amount at −866). The minor allele −866A is associated with a
of BAT in humans, and studies of brown adipocytes from decreased risk of obesity in middle-aged subjects from
nonhuman primates indicate a lack of functional β3-ARs Austria (220). Regarding UCP3, an exon 5 C–T sub-
(205). In addition, pharmacological characterization stitution that does not change the codon 210 Tyrosine
of the Trp64Arg-mutant β3-AR shows no difference in residue was associated with significantly lower resting
its response to agonists when compared with wild-type energy expenditure in African-American but not White
receptors in CHO cells (206). In 204 Japanese subjects, women (221). There were no effects of genotypes of three
the Trp64Arg variant allele was not associated with other UCP-3 variants, nor of two UCP-1 and two UCP-
differences in BMI, plasma glucose or insulin, or fam- 2 variants. In a review of over 40 studies of UCP gene
ily history of obesity or diabetes. However, those with variants in humans, UCP-1 was not felt to contribute
the variant had lower resting autonomic nervous system significantly to variability in BMI. However, UCP-2 and
activity than those without it (207). This conflicting evi- UCP-3 effects on BMI are accepted. As they are adjacent
dence, combined with the mild phenotype in β3-AR KO genes, it is likely that their genetic variants are in linkage
mice, would rule against this locus having a major gene disequilibrium, and the effects of one gene on BMI may
effect on obesity. However, a minor effect, when com- reflect variants in the other (222).
bined with other defects affecting body fat deposition,
may contribute to the obesity phenotype.
90.2.3 Control of Feeding
UCP activity is stimulated by a number of physi-
ologic factors, including cold, βARs, and fatty acids Energy intake and expenditure is under the control of
(208–211). When activated, the protein allows hydrogen complex interactions between peripheral signaling and
ions to “leak” through the respiratory chain of the inner effector systems (e.g. βARs and UCPs discussed above)
mitochondrial membrane, which abolishes the gradi- and neuroendocrine systems (162,223). There is great
ent of hydrogen ions required for ATP synthesis from complexity in these interactions (224), and the details of
stored nutrients (209). Mammalian UCP-1 is expressed these pathways are under widespread investigation. Some
mainly in BAT, which is present to a significant degree in of the pathways are illustrated in Figure 90-4. The con-
humans only in the newborn period. The human UCP-1 tribution of signals from the gastrointestinal tract repre-
gene maps to chromosome 4q31 (210). Human UCP-2 sents the more acute responses to feeding, rather than the
and -3 are also expressed in WAT and skeletal muscle chronic responses to energy stores (225). Among these
(78,208,209), and the genes map to a duplicated locus as are ghrelin from the stomach, insulin and glucagon from
adjacent genes on chromosome 11q13 (212,213). UCP-3 the pancreas, and PYY and glucagon-like peptides from
is preferentially expressed in skeletal muscle (212), and the intestinal tract. The factors important in determin-
has both long and short transcripts (214). In American ing an individual’s set point for food intake may change
Pima Indians, BMI is negatively correlated with both the as individuals become obese because of attenuation of
UCP-3 long and short forms, but not with UCP-2. Meta- leptin signaling induced by increasing adiposity (128).
bolic rate during sleep correlated positively with the long 90.2.3.1 Ghrelin.  Ghrelin was first discovered in the
form of UCP-3 (214). In the skeletal muscle of UCP-3 1970s as a growth hormone secretagog. It’s major role,
KO mice, there was reduced mitochondrial uncoupling however, is not modulation of growth hormone secretion
activity. However, there was no effect seen on body from the anterior pituitary, but rather its role in increasing
weight regulation, exercise tolerance, fatty acid oxida- energy stores is felt to be more important, physiologically
tion, or cold-induced thermogenesis (215). (226). It is secreted mainly by the stomach, with minor
A UCP-1-associated Bcl-I RFLP may be associated contributions from the brain, kidney, and placenta. These
with increased body size (202). Seven sequence variants other sources may compensate for gastric secretion as
of the UCP-1 gene were identified in Danish subjects, but patients having gastrectomies only have 65% reductions
genetic variation in the coding region of the gene did not in blood levels. Serum levels are inversely correlated with
contribute to obesity in this population (216). There is BMI but are paradoxically elevated in children with the
CHAPTER 90  Disorders of the Body Mass 13

FIGURE 90-4  Interactions that regulate food intake and body fat mass. Dashed lines: inhibitory effects. Solid lines: stimulatory effects. Y1R
and Y2R, neuropeptide Y (NPY) receptor subtypes; MC4R, melanocortin 4 receptor; GHsR, growth hormone secretagog receptor (ghrelin
receptor); AgRP, Agouti-related protein; POMC, proopiomelanocortin; α-MSH, α-melanocyte-stimulating hormone; LEPR, leptin receptor; INSR,
insulin receptor. (With permission from Kevin G. Murphy and Stephen R. Bloom. Gut Hormones and the Regulation of Energy Homeostasis.
Nature 444, 854–859.)

PWS (227). Ghrelin levels are high in anorexia nervosa, releasing hormone (CRH), among others (232). The
and levels drop after food intake in lean subjects. How- list of neuropeptides which are known to alter energy
ever, in obese subjects, the levels do not drop after eating balance is growing rapidly. Some of these are listed in
(228). Ghrelin binds to specific receptors in the hypothal- Table 90-3.
amus on NPY/AgRP neurons, and acts to some degree as 90.2.3.3 Neuropeptide Y.  NPY is a neurotransmit-
a stimulator of hunger (225). Despite evidence that sug- ter that is abundant in the hypothalamus, and may be
gests a potentially strong influence of ghrelin on body size, important in mediating obesity. Hypothalamic levels of
ghrelin-deficient mice created by gene KO are the same as NPY and its message are elevated in ob/ob mice, db/db
their wild-type littermates in terms of size, growth rate, mice, and fa/fa rats. Central administration of NPY to
food intake, response to fasting, body composition, and lean rats causes hyperphagia, obesity, insulin resistance
reproduction (229). In humans, variation in the ghrelin and increased triglyceride deposition in WAT. These
gene was associated with higher BMI and lower insulin effects are prevented by adrenalectomy before NPY
response to glucose in a group of 70 tall obese children treatment (233). In a special strain of ob/ob mice that
(230). Other evidence points to a genetic contribution of are deficient in NPY, the obesity and hyperphagia are
ghrelin to body size, including a study showing that ghre- less severe, infertility is less marked, and diabetes is seen
lin levels at baseline and in response to overfeeding, are later and is milder than in ob/ob mice with intact NPY
more similar in monozygotic than dizygotic twins (231). (234). Otherwise, normal mice homozygous for NPY
90.2.3.2 Central Control of Feeding and Energy KO mutations have normal fed and fasting plasma lev-
Expenditure.  The hormone leptin is made almost els of leptin, thyroxine, corticosterone, and sex steroids,
exclusively in peripheral (mainly adipose) tissues, and indicating NPY is not vital for normal pituitary func-
acts centrally in the hypothalamus by modifying two tion (235). They also have lower seizure thresholds and
major effector systems. Low plasma concentrations of higher ethanol consumption and tolerance. However,
leptin and insulin (e.g. during weight loss) increase food after a fasted state, NPY KO mice have less food intake
intake and decrease energy expenditure by stimulating than WT mice, have anxiogenic-like behavior, and hypo-
NPY synthesis, and perhaps by inhibiting sympathetic algesia (236). NPY receptor 1 KO mice have marked
activity and other catabolic pathways. High leptin and obesity (159), but NPY receptor 5 KO mice have a less
insulin concentrations (e.g. during feeding and weight severe obesity phenotype (237).
gain) decrease food intake and increase energy expendi- In human studies, iv NPY administration increases
ture through release of melanocortin and corticotropin sleep and decreases ACTH release in males (238).
14 CHAPTER 90  Disorders of the Body Mass

90.2.3.5 The Hypothalamic–Pituitary–Adrenal (HPA)


TA B L E 9 0 - 3    Central Nervous System Proteins
(Neuropeptides) Involved in
Axis in Feeding and Body Size Determination.  It
Energy Homeostasis has been demonstrated in multiple animal models that
adrenalectomy obliterates or attenuates the obesity syn-
Regulation by Leptin dromes expressed in genetically obese rats and mice,
Neuropeptide or Insulin
in diet-induced obesity, and in hypothalamic obesity
Orexigenic (stimulates feeding) (233). Replacement of only trace quantities of gluco-
Neuropeptide Ya Decreased corticoid to these adrenalectomized animals causes
Agouti-related peptidea Decreased prompt return of the phenotypes indicating heightened
Melanin concentrating hormone Decreased
sensitivity to the steroid. In humans and animals with
Orexin A and B (Hypocretin 1 and 2) Decreased
Galanin Decreased hypercortisolemia, obesity is prominent. However, in
Norepinephrine adrenalectomized animals, the effects seen could result
β-endorphinb from a combination of glucocorticoid deficiency alone
Ghrelinc or in combination with the secondary elevation of hypo-
Anorexigenic (inhibits feeding) thalamic CRH and pituitary ACTH production. Obese
Leptin Zucker (fa/fa) rats have no difference in hypothalamic
α-Melanocyte-stimulating hormonea Increased levels of CRH or in magnitude of CRH response to
Corticotropin releasing hormonea Increased
stressors when compared to lean littermates. However,
Thyrotropin releasing hormonea Increased
Cocaine- and amphetamine-regu- Increased
basal secretion of CRH is less in the obese rats, and
lated transcript (CART)a CRH response to adrenalectomy is more pronounced in
Interleukin-1βa Increased the obese rats. Corticosterone infusion is more effective
Urocortina in suppressing CRH levels in adrenalectomized obese
Glucagon-like peptide 1 rats than in adrenalectomized controls (247), leading to
Oxytocin the hypothesis that fa/fa rats have an abnormality of the
Neurotensin HPA axis at a site proximal to that which mediates glu-
Serotonin cocorticoid responsiveness. Central administration of
Somatostatin
CRH as well as a related hormone, urocortin (248,249),
aEffects
both energy intake and expenditure that change energy stores. mimics the events seen in the “stress response” includ-
bβ-endorphin treatment stimulates feeding in many animal models, but ing anorexia. This treatment prevents excessive weight
β-endorphin knockout male mice have hyperphagia and obesity. gain in fa/fa rats when compared with pair fed fa/fa
cMost ghrelin is secreted from the stomach, but some is produced in the

hypothalamus.
­controls (250,251).
Information obtained from Schwartz et al. (232), and Sahu (413). Leptin treatment of normal mice and rats causes
a blunted HPA axis response to stress, perhaps via
decreased release of CRH (252). In Ay/a obese mice,
Genetic studies of the human genes encoding NPY have the effects of POMC and leptin pathway activation are
identified no deleterious mutations. Linkage analysis of additive, yet independent (253). Full-length LEPR mes-
the NPY gene showed evidence of linkage to body size sage has been identified in human adrenal cortex, and to
in Mexican-American obese sib pairs (239), but not in a lesser degree in adrenal medulla. In cultured adrenal
French Caucasian obese families (240). cells, leptin inhibited ACTH-stimulated cortisol, aldo-
The NPY Y1 and Y5 receptors are transcribed from sterone, and dehydroepiandrosterone secretion (254). In
a common promoter region in opposite directions, sug- addition, some individuals may have heightened sensitiv-
gesting they evolved from a gene duplication event (241). ity to glucocorticoid because of a sequence variant of the
Linkage and association studies with these genes have glucocorticoid receptor gene, which has been associated
failed to identify an effect on body size in several popula- with higher BMI and lower bone density in the Rotterdam
tions (239,240,242). Study (255). These data suggest that therapeutic manipu-
90.2.3.4 Orexins.  Orexins A and B are stimulators lations of the HPA axis could alter the phenotype of cer-
of feeding that are secreted as preprohormones by the tain genetically obese individuals.
hypothalamus. They act through binding to orexin One of the most interesting developments in this area
receptors, which are of the classical G-protein-coupled is the role of adipose tissue in the synthesis and degrada-
seven transmembrane domain class (243). Orexin KO tion of cortisol through the action of 11β-hydroxysteroid
mice have narcolepsy (244), a finding that may correlate dehydrogenase (11β-HSD). 11β-HSD2 converts cortisol
with NPY-induced sleep in humans, described above. to its inactive metabolite cortisone. In the kidney, this
Human orexin receptors are localized in the pituitary prevents activation of the mineralocorticoid receptor by
gland (245) and in pheochromocytomas (246). Muta- cortisol, which is present in plasma in much higher con-
tional analyses and linkage studies of the genes encod- centrations than its native ligand, aldosterone. Cortisol
ing these peptides have not yet been reported in humans has equal affinity for the receptor, and aldosterone is
with altered body size. not a substrate for the enzyme. If 11β-HSD2 is deficient,
CHAPTER 90  Disorders of the Body Mass 15

the syndrome of apparent mineralocorticoid excess preadipocytes is accompanied by a marked increase in the
occurs (256). Glycyrrhetinic acid in licorice causes salt expression of adipose-related transcripts such as leptin
retention and hypertension because it inhibits 11β-HSD2 and UCPs. The terminal differentiation of adipocytes is
activity (257). Topical administration of glycyrrhetinic controlled by several transcription factors, one of the
acid in females caused reduction in the underlying femo- more important being peroxisome proliferator-activated
ral subcutaneous fat (258), presumably by causing a local receptor-γ2 (PPAR-γ2). After differentiation, the fat cells
reduction in adipose tissue cortisol. Chronic ingestion of produce many products other than metabolic fuels that
licorice was associated with a reduction in fat mass in 15 are secreted into the circulation. Prominent examples
normal-weight young adults, without a change in BMI or of these include the cytokines leptin (discussed in Sec-
in caloric intake (259). tion 90.2.1) and TNFα. In addition, adipocyte macro-
11β-HSD1 (cortisone reductase) converts cortisone phage colony-­stimulating factor has been shown to be
back into cortisol in the liver. Defects in 11β-HSD1 important in stimulating adipocyte hyperplasia (270).
activity have been implicated in several clinical settings Hormone-­sensitive lipase (HSL) stimulates breakdown
(260,261). There is mounting evidence that increased of triglycerides into fatty acids, which have been impli-
11β-HSD1 activity in visceral adipose tissue contrib- cated in the development of insulin resistance. The gene
utes to obesity and the metabolic syndrome. Trans- encoding human HSL maps to chromosome 19q13, and
genic mice that overexpress 11β-HSD1 in adipose tissue contains a dinucleotide repeat polymorphism within one
develop the typical metabolic syndrome, with visceral of its introns. This polymorphism was associated with
obesity exaggerated by a high-fat diet, insulin-resistant obesity and type 2 diabetes in a French population (271).
diabetes, hyperphagia, hyperleptinemia, and hyperlip- 90.2.4.1 PPARs.  Members of the PPAR family of nuclear
idemia (262). Patients with hypothalamic obesity may receptors regulate adipocyte differentiation. PPARs are
have increased 11β-HSD1 activity (263), and adipose divided into subtypes α, β, γ, and δ, each of which is
tissue of individuals with idiopathic obesity has been expressed in a tissue-specific manner. The PPAR-γ class
shown to have higher than expected 11β-HSD1 activity is expressed in adipose tissue, and has two isoforms, γ1
(264,265). and γ2, which are differentially spliced products of the
The human gene encoding 11β-HSD1 contains two same gene. The PPAR-γ2 isoform is the longer protein,
polymorphic intronic microsatellites, and one intronic and is adipose specific (272). When PPAR-γ2 is binds its
polymorphism useful for linkage and association stud- ligands (prostaglandin derivatives, fatty acids, and thia-
ies. In a study of 439 normal adults from Glasgow, and zolidinediones), it forms a heterodimer with the retinoid
of 557 Danish military draftees (234 with ­juvenile-onset X receptor that activates a cascade of fatty acid metabo-
obesity and the rest normal controls), there was no lism and preadipocyte differentiation (272–274). This
significant association of 11β-HSD1 genotypes with effect is adipose-depot specific (273), and can even occur
BMI in either group, but there were weak associations in fibroblasts (274). Homozygous PPARγ KO mice have
between 11β-HSD1 activity and waist-to-hip ratio a prenatal lethal phenotype due to placental dysfunc-
(266). In American children, the intronic ins4436A tion. Heterozygous mice seem to be protected from the
polymorphism was associated with BMI and insulin development of insulin resistance, and have reduced fat
resistance (267). mass, smaller adipocyte size, and hypersecretion of leptin
(275). Mice that have a muscle-specific KO of PPARγ
had normal responsiveness to thiazoliadinediones, as
90.2.4 Adipose Tissue Development well as normal glucose and insulin levels, despite excess
and Function adiposity with reduced caloric intake. However, they
Adipose tissue is a biologically active organ, involved not have whole-body insulin resistance when tested with
only with energy storage and release but also with auto- a hyperinsulinemic euglycemic clamp, due to localized
crine and paracrine effects that are widespread (268). insulin resistance in nonmuscle tissues (liver, and per-
These effects are manifested in endocrine/hormonal haps, fat) (276).
function, immune function including its role in cardio- The human gene encoding PPAR-γ2 has been cloned,
vascular risk, and the CNS communication through and maps to chromosome 3p25 (277). Several sequence
leptin, tumor necrosis factor alpha (TNFα), interleukin 6 variants have been described, and as with other candi-
(IL6), adiponectin, resistin, and other adipokines. Leptin date genes for obesity, studies in human populations
and adiponectin are thought to be insulin sparing in their have revealed differing results. A Pro115Gln variant was
effects, whereas TNFα, IL6, and resistin are thought to shown to accelerate differentiation of murine fibroblasts
contribute to the insulin resistance phenotype associated into adipocytes when it was over expressed (278). This
with obesity (268,269). variant was identified more frequently in German obese
Adipocytes play a pivotal role in the maintenance of subjects than nonobese subjects (278). However, this
energy balance. The roles of BAT, WAT, sympathetic ner- variant was not found in a different study of ­German
vous system signals, and UCPs were discussed in ­Section subjects (279), nor in Danish men (280). Another more
90.2.2. The development of mature adipose tissue from widely studied variant, Pro12Ala, has also shown both
16 CHAPTER 90  Disorders of the Body Mass

positive and negative correlations with obesity. It was histocompatibility locus and the steroid 21-hydroxylase
initially studied and ruled out as a candidate gene genes (295). Polymorphic dinucleotide repeat markers
for lipoatrophic diabetes (277). PPAR-γ2 Pro12Ala that map near TNFα were genotyped in American Pima
occurred with the same gene frequency in obese and Indian families. The marker closest to TNFα was linked
lean German subjects (279). Association studies with to %BF in a sib-pair analysis, and was associated with
two different Caucasian populations revealed a positive BMI by analysis of variance (296). However, screening
relationship between Pro12Ala and high BMI (281). In of the TNFα coding region and promoter identified only
French subjects, studies of this variant suggested a role a single polymorphism, and this was not associated with
for it in weight control and lipid homeostasis, but not obesity in this population. A G–A substitution at position
in the etiology of type 2 diabetes (282). Among patients −308 in the TNFα promoter, which causes a restriction
with type 1 and type 2 diabetes, the variant had no effect fragment length polymorphism (RFLP) with the enzyme
on lipids or BMI (283). In Mexican-Americans from NcoI, was associated with a higher rate of transcription
the San Antonio Heart Study, presence of the Pro12Ala of TNFα in vitro. In Spanish overweight individuals, this
allele was positively associated with BMI and serum RFLP was associated with %BF, insulin sensitivity, and
leptin levels (284). A large study of Danish men under- leptin levels, despite no differences in BMI and plasma
scores the conflicting nature of these study results. Obese TNFα levels (297).
Danish men who were homozygous for Pro12Ala had 90.2.4.3 Adiponectin.  Adiponectin is a peptide secreted
higher BMI and higher weight gain than wild-type carri- by adipose tissue, and is preferentially secreted by mature
ers. However, homozygotes for Pro12Ala in the control adipocytes over preadipocytes (298). It was originally
group had a lower BMI and less weight gain than the termed Acrp30 (adipocyte complement-related protein
wild-type carriers (280). of 30 kDa), because of its homology to complement fac-
90.2.4.2 TNFα.  TNFα is one of the several cytokines, tor C1q. It was also known as adipocyte most abundant
including leptin, interleukin 1 and 6, interferon, and gene transcript 1 (apM1), having been isolated as the
TNFβ, known to have profound effects on lipid metabo- most abundant mRNA in human adipose tissue (299).
lism. It not only is secreted by adipose tissue but also Plasma levels are strongly positively correlated with
has direct effects on adipocyte metabolism (285). These insulin sensitivity, and it has putative antiatherogenic
effects are mostly catabolic, including suppression of properties (298). Plasma levels are the reverse of those
many lipogenic enzymes, and development of insu- of leptin—adiponectin concentrations are lower in obese
lin resistance (285). TNFα inhibits human adipose cell than lean subjects (300). Its gene expression in and secre-
insulin signaling (286) and causes apoptosis in human tion from 3T3-L1 adipocytes is reversibly inhibited by
adipose cells (287), and in rat brown adipocytes (288). interleukin 6, an adipocytokine that is elevated in states
TNFα stimulates leptin release from cultured human of insulin resistance (301). In mice, intravenous injection
adipocytes (289), and levels of the soluble TNFα recep- of adiponectin was followed by a rise in CSF levels, con-
tor (sTNFα-R55) are positively correlated with plasma sistent with transport to the CNS. Intracerebroventricu-
insulin and leptin levels in patients with type 2 diabetes lar administration of adiponectin in these animals caused
and in controls (290). TNFα infusion in humans induces a decrease in body weight, due to an increase in energy
an increase in serum leptin levels in humans (291). KO expenditure (302).
mice that are deficient in the adipocyte fatty-acid-binding The adiponectin gene spans a 17-kb region on human
protein aP2 are equally susceptible to diet-induced obe- chromosome 3q27, and contains three exons and two
sity as are control mice. However, the aP2 null mice fail introns (303). Several polymorphisms of the gene have
to express TNFα, and also failed to develop the insulin been detected and analyzed in human subjects (303,304).
resistance seen in the obese control mice (292). A conservative G/T substitution in exon 2 did not corre-
TNFα KO mice did not have a significant change in late with plasma adiponectin levels or with the presence
body size by 28 weeks of age, but there was an increase of obesity in Japanese subjects (304), but was associated
in insulin levels, and a decrease in triglyceride, leptin, with serum cholesterol and waist circumference in obese
and glucose levels (293). In mice with gold thioglucose- Swedish subjects (305). In the latter study, the frequen-
induced obesity, TNFα deficient animals had lower lev- cies of the G or T allele did not differ between obese
els of glucose and insulin, and lower insulin and glucose and lean subjects. In obese subjects undergoing gastric
responses to a glucose tolerance test than the obese wild- banding surgery, genotype haplotypes of two linked
type animals (293). In KO mice simultaneously deficient polymorphisms 11,377 C/G and 11,391 G/A correlated
for both the p.55 and p.75 types of the TNFα receptor, with adiponectin levels at baseline (306).
obesity and diabetes were present. However, KO of 90.2.4.4 Visfatin.  Visfatin is one of the members of the
either receptor alone did not produce this phenotype, adipokine group. It was formerly known as pre-B-cell
and db/db mice lacking p.55 were still diabetic and insu- colony-enhancing factor (PBEF), and also small-molecule
lin resistant (294). insulin mimetic compound 2 (307,308). This nonpep-
The human TNFα gene maps to chromosome tidal activator of insulin receptor action is secreted by
6p21, within the region containing the class 3 major visceral fat as well as lymphocytes. Plasma levels in
CHAPTER 90  Disorders of the Body Mass 17

humans and in mice correlate positively with the quan-


TABLE 90-4     Transgenic Models for Altered
tity of visceral fat but not subcutaneous fat (309). Treat- Body Size or Body Fat
ment of mice with this compound reduces food intake
and prevents high-fat-diet-induced obesity (310). When Increased
mice with diabetes are treated with recombinant visfa- MC4R-KO (164)
tin, their diabetes improves whether they have type 2 or 5-HT2c Receptor KO (414)
streptozocin-induced diabetes. Homozygous visfatin KO CRH overexpression (415)
is a prenatal lethal mutation, and heterozygous mutants BAT ablation (416)
β3-AR KO (+/−) (186)
have a diabetic tendency but no change in body fat (309).
GLUT4 overexpressed in fat (417)
Studies of variation in the human gene for visfatin may NPY receptor 1 KO (159)
provide insight into the link between visceral adiposity Nhlh2 gene KO (418)
and type 2 diabetes. 11β-HSD1 KO (262)
MCH overexpression (413)
β-endorphin KO (413)
90.2.5 Gene Targeting and Effects MC3R KO (174)
on Body Fat/Feeding Muscle PPARγ-KO (276)
The widespread use of gene targeting to study a myriad Decreased
of genes has been applied extensively to the study of Dopamine D1Receptor KO (419)
obesity. Many examples of these animal models have UCP-1 overexpression (182)
been described above. These, along with other relevant Tyrosine phosphatase 1B KO (420)
GLUT4 KO (421)
animal models are summarized in Table 90-4, and they
MCH KO (422)
demonstrate both monogenic and polygenic effects on LPL overexpression in muscle (417)
body size variables. PKA RIIβ KO (423)
PPARγ KO heterozygotes (275)
MCH or MCH-R ablation (413)
90.2.6 Polygenic Models
No change on normal diet
The polygenic mouse models of obesity have allowed CRH KO (424)
identification of multiple autosomal and nonautosomal NPY KO (235)
QTLs within individual strains, which modify obesity, TNFα KO (293)
plasma cholesterol levels, specific deposition of body UCP-3 KO (215)
fat depots (311–314) and propensity toward develop- Ghrelin KO (229)
ment of high leptin levels and obesity on a high-fat diet Visfatin partial KO (309)
(315,316). To underscore the complexity of the contri- MC4R, melanocortin receptor 4; KO, knockout; 5-HT2c, 5-hyroxy-tryptophan
bution of genetic background to autosomal-recessive 2c; CRH, corticotropin releasing hormone; BAT, brown adipose tissue; β3-AR,
­obesity, obese Zucker (fa)/NKY13H intercross F2 rats beta 3-adrenergic receptor; GLUT4, glucose transporter 4; NPY, neuropeptide
have at least three QTLs unrelated to Lepr that control Y; Nhlh2, one of two helix–loop–helix transcription factors expressed in the
developing mouse nervous system; UCP, uncoupling protein; MCH, melanin
BMI and glucose homeostasis (317). In addition, het- concentrating hormone; LPL, lipoprotein lipase; PKA RIIβ, protein kinase A
erozygosity for Lepob and Leprdb in mice is known to regulatory subunit II beta; PPAR, peroxisome proliferator-activated receptor;
affect body composition and leptin homeostasis (123). TNF, tumor necrosis factor; 11β-HSD1, 11beta-hydroxysteroid dehydroge-
Polygenic models may more closely resemble the human nase type 1; MCH, melanin concentrating hormone; MCH-R, MCH receptor;
obesity phenotypes, however, the single gene defects pro- MC3R,melanocortin 3 receptor; PPARγ, peroxisome proliferator-activated
receptor gamma.
ducing recessive traits, dominant traits, promoter altera-
tions, and those subject to parental imprinting must
continue to be considered candidates for genetic effects
in human obesity (224). humans, physical activity may counteract the obesity
related to specific genotypes of the beta 2 adrenergic
receptor β2AR gene (318). The β3AR (319) and lipo-
90.2.7 Gene–Environment Interactions
protein lipase (LPL) genes (320) may be involved in
There are a number of examples of environmental fac- similar interactions. Both β2AR- and β3AR-mediated
tors influencing genotype expression. Consumption of effects involve interactions with UCPs in their produc-
a high-fat diet can alter UCP gene expression in some tion of thermogenesis and response to high-fat diets
mouse strains, indicating variable polygenic control of (321). Other loci that may impact individual responses
body composition and thermogenesis (183). In normal to increased energy intake include ApoE, ApoA-II, and
mice, there is strain-specific variation in the preven- the LDL receptor (322). These and other as-yet unchar-
tion of high-fat-diet-induced obesity by β3AR agonists, acterized interactions would be important in predicting
which is related to the ability to recruit brown adipose the responses to prevention strategies or treatments for
tissue in white adipose tissue anatomic sites (185). In obesity.
18 CHAPTER 90  Disorders of the Body Mass

we will briefly review known human genetic defects


90.2.8 Nonmammalian Models and their clinical features.
While the above discussion has focused on rodent mod-
els, nonmammalian animal models are also playing an
90.3.1 Leptin Deficiency
increasing role in the investigation of obesity and related
traits. These nonmammalian animal models have sev- Missense and nonsense mutations in the leptin gene are
eral advantages over rodents, including shorter genera- an extremely rare cause of morbid obesity. Leptin defi-
tion times, ease of breeding, large population size and ciency is inherited in an autosomal-recessive manner
the existence of tools for the creation and screening of and leads to early-onset extreme obesity characterized
mutants and phenocopies on a large scale. Some high- by intense hyperphagia. Resting and free-living energy
lights of genetic findings in nonmammalian models are expenditure is normal. Patients also have hypogonado-
listed below. tropic hypogonadism with delayed spontaneous puberty,
90.2.8.1 Drosophila.  Mutations in the Laminin A immune deficiency and may have other neuroendocrine
(LanA) gene, a fruit fly gene that is closely related with the defects (e.g. hypothyroidism) (330). They respond dra-
human LAMA5 gene, lead to a decrease in ­triacylglycerol matically to treatment with recombinant human leptin,
storage, body weight and total protein content (323). with resolution of hyperphagia and associated pheno-
The human LAMA5 gene maps to a well-replicated types (331,332). Recently, a patient with a leptin muta-
­obesity-linkage region on chromosome 20q13.2–q13.3 tion and relatively mild obesity and hypogonadotropic
and variants may play a role in body composition and hypogonadism has been described, raising the possibility
lipid phenotypes (324). Other studies indicate that mem- that milder clinical forms of leptin deficiency may also
bers of the syndecan gene family may play a role in energy exist (333).
metabolism and regulation of body weight in Drosophila 90.3.1.1 Leptin Receptor Mutations.  Human leptin
and that variants in homologous human genes may influ- receptor deficiency was initially reported in a single
ence the same phenotypes in humans (325). Further stud- family with very severe early-onset obesity and growth
ies are needed to confirm if these genes play a role in retardation secondary to impaired growth hormone
human obesity and to elucidate the mechanisms involved secretion. Since then, it has been found that mutations
in any such role. in the leptin receptor may be present in up to 3% of the
90.2.8.2 Zebrafish.  Zebrafish are being used to study cases of morbid obesity (334). These subjects presented
the effects of KO or knockdown of various genes with early-onset hyperphagia and morbid obesity with-
involved in obesity and metabolic regulation (326) and out significant elevation of leptin levels or developmental
may provide a means of interrogating known obesity- abnormalities or dysmorphism.
related genes as well as the discovery of novel genes in 90.3.1.2 MC4R.  Defects in the melanocortin 4 receptor
the future. may be the most common form of monogenic obesity
90.2.8.3 Caenorhabditis elegans.  C. elegans provides in humans. These mutations appear to be codominant,
an excellent model for the study of the basic biology of with more severe obesity seen in homozygotes. Preva-
obesity and energy regulation (327). Genes identified in lence of MC4R mutations ranges from 1% to 6% in
human genetic studies can be knocked down or mutated morbidly obese patients (335), but has not been con-
in C. elegans to study their physiology, and conversely, firmed in all populations (336,337). Functional studies
targeted mutagenesis and screening of mutants can reveal of these mutants have confirmed the deleterious effects
novel genes involved in energy metabolism and the regu- in many cases (167,171). Nonpathogenic mutations and
lation of adiposity (328). For example, the Indy (I am mutations that exert only a mild effect on BMI are more
not dead yet) gene was shown to regulate lifespan and common than mutations associated with morbid obesity,
adiposity phenotypes in Drosophila and C. elegans and making genetic diagnosis relatively complicated in these
this finding was later replicated in rodents using a mouse cases (335).
KO (329). 90.3.1.3 POMC Mutation.  POMC is processed to
produce the hormones ACTH, MSH alpha, beta and
gamma, and beta-endorphin. Defects in POMC are an
90.3 MENDELIAN DISORDERS extremely rare cause of obesity, ACTH deficiency and
ASSOCIATED WITH INCREASED BMI red hair (154) though mutations that were not associated
IN HUMANS with red hair have also been reported (338,339).
Increased risk of obesity can, in relatively rare cases, 90.3.1.4 PC1 Mutation.  PC1 cleaves prohormones
be secondary to single gene disorders of large effect. before processing by carboxypeptidase. A mutation in
While these disorders are uncommon, they do shed PC1 has been described a patient with obesity, elevated
light on some of the complex hormonal and neural net- proinsulin, hypocortisolism, hypoglycemia, hypogonad-
works that regulate adiposity. Their murine models, otropic hypogonadism and elevated POMC (152). In at
physiological role and known counterparts in humans least one case, a mutation in PC1 was also associated
have been discussed in detail in Section 90.2.1. Here with neonatal diarrhea and malabsorption (340).
CHAPTER 90  Disorders of the Body Mass 19

90.3.1.5 Cohen Syndrome.  Mutations in the VPS13B Some of the variations in clinical phenotypes between
gene (vacoular protein sorting 13, yeast, homolog B), different PWS patients are due to variation in the num-
located on chromosome 8q22, are associated with ber of deleted or silenced genes in this region. But while
Cohen syndrome. Cohen syndrome is characterized by several genes contribute to the phenotype, deletion of
truncal obesity, thin extremities and short stature and is genes involved in the production and processing of small
inherited in an autosomal-recessive manner (341). nucleolar RNAs, particularly snoRNA SNORD116 clus-
90.3.1.6 Alstrom Syndrome.  Alstrom syndrome ter (HBII-85) appear to be sufficient to cause the core
(ALMS) is an autosomal-recessive ciliopathy that is phenotypes of PWS (349).
characterized by truncal obesity, short stature, reti- 90.3.2.2 Bardet–Biedl Syndrome.  BBS is a rare syn-
nal cone dystrophy, progressive hearing loss, dilated drome characterized by early-onset obesity, rod–cone
­cardiomyopathy, type 2 diabetes and progressive dystrophy, dyslexia, learning disabilities, postaxial poly-
­pulmonary, hepatic, and renal dysfunction. The involved dactyly, hypogonadism and progressive renal disease.
gene, ALMS1, is located on chromosome 2p13 and The McKusick–Kaufman syndrome is an allelic form of
encodes a large protein that is involved in ciliary func- BBS. It was initially thought to be a classic autosomal-
tion (342). How and why a ciliopathy causes ­obesity is recessive disorder, but the genetics of BBS turn out to
the focus of intense research and may shed new light on be much more complex, with at least 15 different genes
the regulation of adiposity and energy metabolism in now identified as being associated with this syndrome
humans (343). (350,351). Some behave in an autosomal-recessive man-
90.3.1.7 Others.  Individual cases of mutations in sev- ner, but others are “triallelic,” requiring a homozygous
eral other genes involved in the CNS appetite regula- recessive mutation in one gene and an additional muta-
tion and the development of neuronal circuits have been tion at a second locus. Most of these genes are involved
described in patients with morbid obesity, e.g. in SIM1 in ciliary function and the syndrome is now considered
(single-minded, Drosophila, homolog of, 1) gene (344) ciliopathy that overlaps clinically with ALMS (352). Its
and NTRK2 (neurotropic tyrosine kinase receptor 2) recurrence risk depends on the specific locus containing
gene (345). genetic mutation of each pedigree (353).
90.3.2.3 WAGR Syndrome.  Wilms' Tumor, aniridia,
genitourinary abnormalities and mental retardation
90.3.2 Syndromic Obesity (WAGR) syndrome is associated with heterozygous
Syndromic obesity describes obesity that is associated interstitial deletions of chromosome 11p-13. This syn-
with well-defined combinations of other phenotypes drome included obesity in some but not all patients and
such as mental retardation, dysmorphic features and comparison of the deleted regions between obese and
other developmental abnormalities. Examples of such nonobese patients indicated that obesity is associated
syndromes with their known genetic cognates include with deletion of the brain-derived neurotrophic factor
the following. (BNDF) gene (354). Polymorphisms in the same gene
90.3.2.1 Prader–Willi Syndrome.  PWS is the most have since been found to be associated with obesity in
common obesity syndrome with an incidence of GWAS cohorts. This gene is expressed in the CNS and
1/15,000–1/25,000 live births. It is caused by loss of appears to be involved in the regulation of appetite.
expression of paternally expressed genes in the imprinted 90.3.2.4 16p11.2 Deletions.  Deletions in this region are
(differentially methylated) 15q11–13 region. This loss associated with obesity and in some cases with autism.
may be secondary to deletions of the paternal 15q11– This region includes the gene SH2B1, which codes for
13 region (70–75% of cases), maternal uniparental an adapter protein involved in tyrosine kinase signaling
disomy of chromosome 15 (20–25%), microdeletions including leptin and insulin signaling (355).
in the imprinting center at the SNURF–SNRPN gene
locus (<3%) and paternal translocations (<1%) (346).
90.3.3 Polygenic Obesity
Diagnostic testing is best performed by analyzing the
methylation status of the PWS region using methylation- In most cases, the inherited risk of obesity appears to be
specific PCR. The clinical features of PWS include low complex and is very likely polygenic in origin. It remains
birth weight, severe hypotonia and feeding difficulties a matter of dispute if this risk is due to rare variants of
in early infancy, followed by hyperphagia and obesity large effect or common variants of small effect. At the
starting in early childhood. Other characteristic features dawn of the genomic era, it was generally assumed that
include hypogonadism, short stature, small hands and common variants, each of small effect, are responsible
feet, facial dysmorphology (narrow bifrontal diameter, for the main genetic contribution to risk of obesity. But
almond-shaped eyes, triangular mouth) and a distinctive very large genome-wide association studies (GWAS) have
behavioral phenotype (347). The 15q11–13 region in explained a relatively small proportion of the genetic risk
an imprinted region in which at least 10 genes are only of obesity and some researchers have proposed that his
expressed from the paternal chromosome and these were may be due to the fact that rare variants of large effect
therefore considered candidate genes for PWS (348). that arise relatively recently in extended families or clans
20 CHAPTER 90  Disorders of the Body Mass

are responsible for a large proportion of the hereditary intake after correction for lean body mass. Heterozy-
risk of obesity (62). gotes, interestingly, are resistant to high-fat-diet-induced
90.3.3.1 Linkage and Candidate Gene Studies of obesity (359). A different mutation that causes partial
the Genetics of Human Obesity.  Starting in the 90s, loss-of-function of the FTO gene is associated with
several research groups attempted to identify genes asso- decreased fat mass and increased energy expenditure,
ciated with obesity using candidate gene and linkage but no hyperphagia or growth retardation is seen (360).
studies. Genes thought to be involved in appetite regu- The FTO gene is widely expressed in the body, with high
lation, partitioning of body fat, energy storage, energy levels of expression in the CNS and particularly in the
expenditure and other physiologic processes that could hypothalamus. Overexpression of FTO in the arcuate
play a role in obesity were investigated to see if variants nucleus in mice leads to a reduction in daily food intake,
in their sequences were linked to risk of obesity. Link- while knocking down its expression leads to increased
age studies using minisatellites and other markers were food intake (361).
used to identify genomic regions that appeared linked to The effects of an FTO mutation in humans have been
BMI variation. Among the regions and genes found to be studied in one extended family and all affected homozy-
possibly linked to obesity were a region on chromosome gotes suffered from a polymalformation syndrome that
3 that included the adiponectin gene as well as other includes postnatal growth retardation, microcephaly,
genes that were previously not known to be involved in severe brain deficits, dysmorphic facies, cardiac abnor-
the control of body composition or energy metabolism malities and cleft palate and all affected members died
(e.g. PSARL and VPS8) (356). Overall, the results of within the first 30 months of life. Heterozygotes in the
these studies were somewhat disappointing and did not extended family have not been studied in detail, but no
explain most of the genetic risk of obesity. overt body weight phenotype has been reported. Thus,
90.3.3.1.1 GWAS in Humans.  With advancements FTO appears to be essential for the normal development
in technology, it has become feasible to genotype hun- of a number of major organs and a complete lack of FTO
dreds of thousands to several million SNPs in individual activity is incompatible with life beyond early childhood.
microarrays and to test their association with obesity The physiological role of FTO in body-weight regula-
and related traits. The first large GWAS in humans tion remains unknown at this time. The FTO protein may
led to the identification of FTO (Fat Mass and Obe- play a role in appetite regulation as well as resting energy
sity associated gene) as a gene that is associated with expenditure. As it is capable of demethylating both DNA
variation in BMI in almost every population studied to and RNA (362), it may be involved in epigenetic modula-
date (357). Since then, large GWAS and meta-analyses tion of DNA as well as in the regulation of various RNAs.
have identified dozens of new genes and intergenic loci 90.3.3.1.3 Peroxisome Proliferator-Activated
associated with BMI, while also replicating many of the Receptor Gamma Gene (PPARγ).  A polymorphism
genes identified earlier by candidate and linkage studies (Pro115Gln) in this gene was found to be associated
(Table 90-5) shows many of the genes found to be asso- with adipocyte differentiation and greater cellular accu-
ciated with obesity in GWAS. We will discuss a few of mulation of triglyceride (278). Another polymorphism
these in greater detail: (Ala allele of Pro12Ala polymorphism) was associated
90.3.3.1.2 FTO.  FTO (Fat mass and obesity asso- with lower BMI, increased insulin sensitivity and higher
ciated) is a large gene, over 400 kb in size, and several plasma HDL levels (363). Associations of these and
different SNPs in the first intron have been robustly asso- other (PPARγ) polymorphisms with obesity and other
ciated with obesity in at least 22 different population metabolic syndrome phenotypes were replicated in sev-
groups (358). The effect size is modest (3 kg increase in eral populations (364,365), but explained only a tiny
weight in individuals homozygous for the risk allele) but fraction of the inherited risk of obesity.
the risk allele is prevalent at a level of almost 50% in the 90.3.3.1.4 Beta 2- and Beta 3-Adrenergic Recep-
Caucasian population, so the gene may still contribute tor Genes (ADRB2, ADRB3).  As described in Section
to obesity in a very large number of individuals. When 90.2.2.1, polymorphisms in this adrenergic receptor gene
it was initially discovered, nothing was known about its were associated with BMI and fasting fatty acid levels
function, let alone its role in the regulation of BMI. Since in some populations in linkage and candidate gene stud-
then, it has been discovered that the FTO gene product ies (366,367). The beta 2- and beta3-adrenergic receptor
is an oxoglutarate-dependent demethylase that is able to genes were also implicated in onset of obesity as well as
bind and demethylate single-stranded DNA and RNA. blood pressure elevation in other studies (368) but these
How this relates to regulation of body weight remains associations have not replicated in all studies (369,370).
unknown at this time. Some evidence suggests an additive effect of the Beta 3
The first complete Fto null mouse (Fto−/−) was reported adrenergic gene and UCP-1 gene variants on body-size
in 2009. The homozygote displayed a phenotype that phenotypes (371,372).
includes growth retardation, decreased fat mass and 90.3.3.1.5 MC4R.  The melanocortin 4 receptor
lean mass, increased metabolic rate and increased food is an integral component of the leptin–melanocortin
CHAPTER 90  Disorders of the Body Mass 21

TA B L E 9 0 - 5    
Chromosomal Location Suspected Gene(s) Function Reference
1p31 NEGR1(Neuronal growth regulator 1) Axonal growth promoter (425)
1q25 SEC16B, RASAL2 Endoplasmic reticulum/protein export, GTPase (425)
activator
1q41 LYPLAL1, ZC3H11B Lipase, zinc finger pseudogene (26)
1q43-44 SDCCAG8 Centrosomal protein (426)
2p25 TMEM18 Cell migration modulator (427)
3q27 Near ETV5 Transcription factor (425)
4p13 GNPDA2 Glucosamine-6-phosphate isomerase (427)
6p12 TFAP2B Transcription factor (26)
6p22.2–p21.3 PRL Prolactin (428)
8p23.1 MSRA Reduction of methionine sulfoxide/repair (26,426)
oxidative damage
10p12 PTER Phosphodiesterase related (428)
11p11.2 MTCH2 Mitochondrial carrier protein (427)
12q13 FAIM2, BCDIN3D Neuronal membrane protein/apoptosis (425)
­inhibition. Methyltransferase
14q31 NRXN3 Neurexin; CNS cell adhesion (428,429)
16p11.2 SH2B1 Kinase signaling pathways (425,427)
16q22–q23 MAF Transcription factor; pancreas development (428)
and insulin gene transcription
16q22.2 FTO Nucleic acid demethylase; other functions? (357) Multiple others
18q11.2 NPC1 Niemann–Pick disease gene; intracellular (428)
cholesterol trafficking
18q22 MC4R Melanocortin 4 receptor, appetite regulation. (430) Multiple others
Associated with monogenic obesity
19q13.11 KCTD15 Potassium channel (425,427)
Human genetic loci associated with GWAS or epigenetics studies. Based on Reference (375), Figure 90-1.

regulatory pathway and disruptions in this receptor are imprinting or epigenetics studies as correlated with body
the most common monogenic cause of severe obesity. size variables are listed in Table 90-5.
GWAS scans reveal that polymorphisms in and around 90.3.3.2 Addressing the Human Obesity Problem:
this gene are also associated with BMI in diverse pop- Predictive Factors, Prevention Strategies, and
ulations (373,374). It is likely that variations in the Treatments.
functionality of this receptor lead to alterations in sati- 90.3.3.2.1 Prediction of Adult Obesity during
ety and a change in body weight because of increased Childhood.  It is well known that blood pressure, blood
appetite. lipid levels, and obesity in childhood tracks into adulthood
90.3.3.1.6 UCP Genes.  UCPs are mitochondrial (377). Thus childhood obesity itself is a predictor of adult
proteins that help to dissipate the proton gradient at the obesity (378), and of higher-than-expected adult morbid-
inner mitochondrial membrane, decreasing the genera- ity and mortality (379). These late effects may be unre-
tion of ATP. It has been postulated that polymorphisms lated to the presence of overweight in adulthood (380).
in the various uncoupling proteins (UCP1, 2 and 3) can The presence of a specific growth pattern termed early
lead to alteration in energy expenditure and thus in the adiposity rebound may predict adult obesity in young
accumulation of fat. Members of this gene family were children (381). Early adiposity rebound is associated with
initially implicated in obesity using candidate gene parental obesity but not with socioeconomic status or
approaches (177) and these associations have been repli- dietary variables (382). Whereas most of the comorbidi-
cated in several populations. ties of obesity occur in adults, they are definitely present
90.3.3.1.7 Others.  Other genes including TNFα in youth in their presymptomatic or early symptomatic
gene (TNFA), angiotensin-converting enzyme gene forms (38,42,383,384), and when present in children,
(ACE), G-protein beta3 subunit gene (GNB3), leptin they are associated with increased cardiovascular mor-
gene (LEP), leptin receptor (LEPR), SLC6A14,GAD2, bidity and mortality in their relatives (38–40). The preva-
TMEM18, INSIG2 and ENPP1 have been linked to the lence of clinically significant obesity-related morbidities
risk of obesity in different studies (375,376). Their exact in youth is definitely on the rise (385), and predicts earlier
role remains unclear in many cases and their combined onset of more severe problems in adolescents, especially
contribution to the obesity epidemic is relatively small. among African-Americans (386), and in young adults
Many examples of human loci identified in GWAS and (387). The alarming increase in type 2 ­diabetes among
22 CHAPTER 90  Disorders of the Body Mass

children and adolescents is directly related to the obesity resulting in obesity, novel efficacious therapeutic inter-
epidemic (388). ventions have not been forthcoming.
Many investigators have established the importance 90.3.3.2.4 Medications.  Current medical therapeu-
of parental obesity as a predictive factor for childhood tic options for treatment of obesity are not very promis-
obesity. In a study of the influence of different aspects ing, and the principals of therapy are flawed. Therapy
of the home environment on obesity in nearly 3000 chil- must be long-term and ongoing, as is the case with treat-
dren <8 years of age, maternal obesity was the most sig- ment of hypertension and diabetes. Current obesity drugs
nificant predictor of childhood obesity (389). High birth are not approved for prolonged use, or for use in youth,
weight is a significant predictor of later obesity, and the have not been extensively tested for safety over long
most important factor contributing to high birth weight periods of time, may only benefit a minority of patients
is maternal diabetes and, to a lesser degree, maternal (12,397), and are associated with serious cardiovascu-
obesity (390). The relative risk of developing obesity lar side effects (398). The latter has resulted in two such
in young adulthood is higher for young children if they agents being taken off the market. The major classes of
have obese parents, and higher for older children if they drugs are those that reduce food intake (monoamine
themselves are obese (391) (Table 90-6). oxidase inhibitors, sympathomimetic drugs), those that
The influence of lack of physical activity on the devel- increase energy expenditure (ephedrine, caffeine), and
opment of obesity is reflected in the NHANES data, those that inhibit fat absorption. The use of agents to
especially from the most recent survey. Lack of physi- induce dietary fat malabsorption has been only mini-
cal activity is directly related to television viewing, and mally successful, and is associated with significant intes-
hours of television are significantly correlated to weight tinal discomfort if not accompanied by a low-fat diet. Of
gain during the growing years (392). Reduction in tele- course, the diet itself may be nearly equally effective over
vision viewing significantly reduced the rate of weight the long-term.
gain in a study of third-grade children, when compared The use of recombinant leptin in humans resulted in
to third graders with no intervention (393). a modest and highly variable loss of weight (loss of fat
90.3.3.2.2 Prevention Strategies.  Obviously, mass), which was dose related, and occurred in both lean
community-wide efforts need to be directed toward and obese subjects (399). Leptin treatment in the rare
increasing physical activity, and changing dietary hab- leptin-deficient patients produced a rapid reduction in
its. Reduction of dietary calories and fat, and increas- weight and increase in energy expenditure, and antibod-
ing dietary fiber are recommended (394). The CDC has ies to leptin developed after 2 months (400).
developed a resource guide for prevention of obesity and In an animal study, mice that were treated with fatty
other chronic diseases that cover many aspects of nutri- acid synthase inhibitors had a reduction in food intake
tion, physical activity, and other lifestyle interventions and in body weight (401). This treatment was effective
(395). Counseling obese parents about the risk of child- when given either systemically or intracerebroventricu-
hood obesity in their children must be practiced by health larly. It inhibited NPY expression in the hypothalamus,
care providers. Enhancement of physical education pro- as well as leptin expression in WAT.
grams must be instituted by the school systems. It has 90.3.3.2.5 Surgery.  Bariatric surgery perhaps
been established that breast-fed infants are less likely to provides the best weight loss results (402), but both
develop adult obesity than bottle-fed infants (396), add- short- and long-term risks are present. Bariatric surgi-
ing obesity prevention to the list of benefits of breast cal procedures fall into two categories—those reducing
feeding, which health care providers should convey. gastric volume, such as vertical banded gastroplasty or
90.3.3.2.3 Treatments.  Despite the identification of adjustable gastric band, and those that also result in
specific single gene defects and metabolic abnormalities moderate selective malabsorption such as gastric bypass.
Bypass results in more rapid and more significant weight
loss. As a result, there is a recent trend toward gastric
TA B L E 9 0 - 6    Odds Ratios for Obesity in Young bypass as the preferred procedure. However, there is an
Adulthood According to Obesity altered gut hormone response to feeding, potential for
Status in Childhood and Parents’ deficiencies of vitamin B12, folate, and iron leading to
Obesity Status anemia, as well as secondary hyperparathyroidism, pre-
sumably due to reduction of calcium absorption due
Obese as Child Number of Obese Parents
to loss of gastric acidity. Vertical banded gastroplasty
Age (yr) Yes versus No 1 versus 0 2 versus 0 has been associated with loss of bone density without
1–2 1.3 3.2 13.6 hyperparathyroidism (403). Several studies confirmed
3–5 4.7 3.0 15.3 that gastric bypass results in reduction in diabetes and
6–9 8.8 2.6 5.0 hypertension (404,405), but the risk for metabolic bone
10–14 22.3 2.2 2.0
disease, vitamin deficiencies, and intestinal complications
15–17 17.5 2.2 5.6
is high (406–409). Surgery to remove fat (liposuction), if
Data were obtained with permission from Reference (391). used alone, is not a long-term solution. However, it may
CHAPTER 90  Disorders of the Body Mass 23

be a useful cosmetic adjunct in patients who are success- 11. Mokdad, A. H.; Ford, E. S.; Bowman, B. A.; Dietz, W. H.;
ful with diet and exercise, or surgical gastroplasty. Vinicor, F.; Bales, V. S.; Marks, J. S. Prevalence of Obesity,
90.3.3.2.6 Future Directions in Treatment.  Diabetes, and Obesity-related Health Risk Factors, 2001. J.
Of course, the most significant impact on the obesity Am. Med. Assoc. 2003, 289 (1), 76–79.
12. National Task Force on the Treatment of Obesity ­Long-Term
epidemic will have to be achieved through patient edu- Pharmacotherapy in the Management of Obesity. J. Am.
cation and prevention strategies. Many resources offer- Med. Assoc. 1996, 276, 1907–1915.
ing guidance in these areas are available (410) (see also 13. Wang, M. Trends of Obesity and Underweight in Older
www.cdc.gov and www.surgeongeneral.gov). New Children and Adolescents in the United States, Brazil, China,
medical treatments, including potent agonists for the and Russia. Am. J. Clin. Nutr. 2002, 75 (6), 971–977.
14. Friedrich, M. J. Epidemic of Obesity Expands its Spread to
β3-AR and the MC4R, are under development. Pres- Developing Countries. J. Am. Med. Assoc. 2002, 287 (11),
ymptomatic genotyping using microarray technologies 1382–1386.
may be able to identify those individuals genetically pre- 15. Fu, W. P.C.; Lee, H. C.; Ng, C. J.; Tay, Y. -K.D.; Kau, C.
disposed to obesity, allowing prevention strategies to be Y.; Seow, C. J.; Siak, J.; Hong, C. Y. Screening for Child-
initiated early. Gene or hormone replacement therapy hood Obesity: International vs Population-Specific Defini-
tions. Which Is More Appropriate? Int. J. Obes. 2003, 27 (9),
may become available for those rare patients with single 1121–1126.
gene defects. Direct electrostimulation of specific hypo- 16. Hill, J.; Peters, J. Environmental Contributions to the Obe-
thalamic nuclei has been successful in increasing met- sity Epidemic. Science 1998, 280, 1371–1374.
abolic rate and fat utilization, and decreasing feeding 17. Didi, M.; Didcock, E.; Davies, H. A.; Oglivy-Stuart, A. L.;
in rats (411). Similar techniques are used in humans to Wales, J. K. H.; Shalet, S. M. High Incidence of Obesity in
Young Adults After Treatment of Acute Lymphoblastic Leu-
treat tremors, and perhaps could be applied to obesity kemia in Childhood. J. Pediatr. 1995, 127, 63–67.
treatment. 18. Van Dongen-Melman, J. E.W.M.; Hokken-Koelega, A.
C. S.; Hählen, K.; De Groot, A.; Tromp, C. G.; Egeler, R.
M. Obesity After Successful Treatment of Acute Lympho-
REFERENCES blastic Leukemia in Childhood. Pediatr. Res. 1995, 38,
86–90.
1. Strawbridge, W. J.; Wallhagen, M. I.; Shema, S. J. New 19. Choi, S.; Dallman, M. F. Hypothalamic Obesity: Multiple
NHLBI Clinical Guidelines for Obesity and Overweight: Routes Mediated by Loss of Function in Medial Cell Groups.
Will They Promote Health? Am. J. Public. Health. 2000, 90 Endocrinology 1999, 140, 4081–4088.
(3), 340–343. 20. De Vile, C. J.; Grant, D. B.; Hayward, R. D.; Kendall, B. E.;
2. Calle, E. E.; Rodriguez, C.; Walker-Thurmond, K.; Thun, M. J. Neville, B. G.R.; Stanhope, R. Obesity in Childhood Cra-
Overweight, Obesity, and Mortality from Cancer in a Prospec- niopharyngioma: Relation to Post-Operative Hypothalamic
tively Studies Cohort of U.S. Adults. N. Engl. J. Med. 2003, Damage Shown by Magnetic Resonance Imaging. J. Clin.
348, 1625–1638. Endocrinol. Metab. 1996, 81, 2734–2737.
3. Visscher, T. L.S.; Seidell, J. C. The Public Health Impact of 21. Dhurandhar, N.; Israel, B.; Kolesar, J.; Mayhew, G.; Cook, M.;
Obesity. Annu. Rev. Public. Health. 2001, 22, 355–375. Atkinson, R. Increased Adiposity in Animals Due to a Human
4. Ford, E. S.; Giles, W. H.; Mokdad, A. H. Increasing Preva- Virus. Int. J. Obes. 2000, 24, 989–996.
lence of the Metabolic Syndrome among U.S. Adults. Diabe- 22. Dhurandhar, N.; Kulkarni, P.; Ajinkya, S. M.; Sherikar, A.;
tes Care 2004, 27 (10), 2444–2449. Atkinson, R. Association of Adenovirus Infection with Human
5. Hedley, A. A.; Ogden, C. L.; Johnson, C. L.; Carroll, M. D.; Obesity. Obes. Res. 1997, 5, 464–469.
Curtin, L. R.; Flegal, K. M. Prevalence of Overweight and 23. Dhurandhar, N. V.; Atkinson, R.; Ahmed, A. Obesity of
Obesity among US Children, Adolescents, and Adults, 1999- Infectious Origin—A Review. Growth Genet. Horm. 2004,
2002. J. Am. Med. Assoc. 2004, 291, 2847–2850. 20, 33–39.
6. Weiss, R.; Dziura, J.; Burgert, T. S.; Tamborlane, W. V.; 24. Schwartz, M. W.; Niswender, K. D. Adiposity Signaling and
Taksali, S. E.; Yeckel, C. W.; Allen, K.; Lopes, M.; Savoye, Biological Defense Against Weight Gain: Absence of Protec-
M.; Morrison, J., et al. Obesity and the Metabolic Syndrome tion or Central Hormone Resistance? J. Clin. Endocrinol.
in Children and Adolscents. N. Engl. J. Med. 2004, 350, Metab. 2004, 89 (12), 5889–5897.
2362–2374. 25. Dulloo, A. G.; Jacquet, J.; Solinas, G.; Montani, J. P.;
7. Olshansky, S. J.; Passaro, D. J.; Hershow, R. C.; Layden, J.; Schultz, Y. Body Composition Phenotypes in Pathways to
Carnes, B. A.; Brody, J.; Hayflick, L.; Butler, R. N.; Allison, Obesity and the Metabolic Syndrome. Int. J. Obes. 2010, 34
D. B.; Ludwig, D. S. A Potential Decline in Life Expectancy (Suppl 2), S4–S17.
in the United States in the 21st Century. N. Engl. J. Med. 26. Lindgren, C. M.; Heid, I. M.; Randall, J. C., et  al. Giant
2005, 352 (11), 1138–1145. Consortium Genome-Wide Association Scan Meta-Analysis
8. Ogden, C. L.; Flegal, K. M. Changes in Terminology for Identifies Three Loci Influencing Adiposity and Fat Distribu-
Childhood Overweight and Obesity. Natl. Health Stat. Rep. tion. PLoS 2009, 5 (6), e1000508.
2010, 25 (25), 1–5. 27. Kissebah, A. H.; Krakower, G. R. Regional Adiposity and
9. Ogden, C. L.; Carroll, M. D. Prevalence of Overweight, Morbidity. Physiol. Rev. 1994, 74, 761–811.
Obesity, and Extreme Obesity among Adults: United States, 28. Maes, H. H.; Neale, M. C.; Eaves, L. J. Genetic and Environ-
Trends 1960–1962 through 2007–2008; National Center for mental Factors in Relative Body Weight and Human Obe-
Health Statistics, Centers for Disease Control and Preven- sity. Behav. Genet. 1997, 27 (4), 325–351.
tion, 2010. 29. Stunkard, A. J.; Foch, T. T.; Hrubec, Z. A Twin Study of
10. Mokdad, A. H.; Serdula, M. K.; Dietz, W. H.; Bowman, B. A.; Human Obesity. J. Am. Med. Assoc. 1986, 256, 51–54.
Marks, J. S.; Koplan, J. P. The Spread of the Obesity Epidemic 30. Sorensen, T.; Price, R. A.; Stunkard, A. J.; Schulsinger, F.
in the United States, 1991–1998. J. Am. Med. Assoc. 1999, Genetics of Obesity in Adult Adoptees and their Biological
282 (16), 1519–1522. Siblings. Br. Med. J. 1989, 298, 87–90, (January 14).
24 CHAPTER 90  Disorders of the Body Mass

31. Faith, M. S.; Pietrobelli, A.; Nunez, C.; Heo, M.; Lalande, M. FISH Ordering of References Markers and of
Heymsfield, S. B.; Allison, D. B. Evidence for Independent the Gene for the α5 Subunit of the Gamma-Aminobutyric
Genetic Influences on Fat Mass and Body Mass Index in a Acid Receptor (GABRA5) within the Angelman and
Pediatric Twin Sample. Pediatrics 1999, 104, 61–67. Prader–Willi Syndrome Chromosomal Regions. Hum.
32. Moll, P. P.; Burns, T. L.; Lauer, R. M. The Genetic and Mol. Genet. 1993, 2, 183–189.
Environmental Sources of Body Mass Index Variability: The 49. Sheffield, V. C. Use of Isolated Populations in the Study of
Muscatine Ponderosity Family Study. Am. J. Hum. Genet. a Human Obesity Syndrome, the Bardet–Biedl Syndrome.
1991, 49, 1243–1255. Pediatr. Res. 2004, 55 (6), 908–911.
33. Price, R. A. Within Birth Cohort Segregation Analyses 50. Michaud, J. L.; Heon, E.; Guilbert, F.; Weill, J.; Puech, B.;
­Support Recessive Inheritance of Body Mass Index in White Benson, L.; Smallhorn, J. F.; Shuman, C. T.; Buncic, J. R.;
and African-American Families. Int. J. Obes. 1996, 20, Levin, A. V., et  al. Natural History of Alstrom Syndrome
1044–1047. in Early Childhood: Onset with Dilated Cardiomyopathy. J.
34. Bouchard, C.; Rice, T.; Lemieux, S.; Després, J.-P.; Pérusse, L.; Pediatr. 1996, 128, 225–229.
Rao, D. C. Major Gene for Abdominal Visceral Fat Area in the 51. Wilson, G. N.; Al Saadi, A. A. Obesity and Abnormal Behav-
Québec Family Study. Int. J. Obes. 1996, 20, 420–427. iour Associated with Interstitial Deletion of Chromosome 18
35. Hasstedt, S. J.; Ramirez, M. E.; Kuida, H.; Williams, R. R. (q12.2q21.1). J. Med. Genet. 1988, 26, 62–63.
Recessive Inheritance of a Relative Fat Pattern. Am. J. Hum. 52. Leibel, R. L.; Rosenbaum, M.; Hirsch, J. Changes in Energy
Genet. 1988, 45, 917–925. Expenditure Resulting from Altered Body Weight. N. Engl.
36. Knowler, W. C.; Pettitt, D. J.; Saad, M. F.; Charles, M. A.; J. Med. 1995, 332, 621–628.
Nelson, R. G.; Howard, B. V.; Bogardus, C.; Bennett, P. H. 53. Rosenbaum, H. Effects of Changes in Body Weight on
Obesity in the Pima Indians: Its Magnitude and Relationship Carbohydrate Metabolism, Catecholamine Excretion,
with Diabetes. Am. J. Clin. Nutr. 1991, 53, 1543s–1551s. and Thyroid Function. Am. J. Clin. Nutr. 2000, 71 (6),
37. Coleman, D. L.; Hummel, K. P. The Influence of Genetic 1421–1432.
Background in the Expression of the Obese (ob) Gene in the 54. Morrison, J. A.; Alfaro, M. P.; Khoury, P.; Thornton, B. B.;
Mouse. Diabetologia 1973, 9, 287–293. Daniels, S. R. Determinants of Resting Energy Expenditure
38. Burns, T. L.; Moll, P. P.; Lauer, R. M. The Relation Between in Young Black Girls and Young White Girls. J. Pediatr.
Ponderosity and Coronary Risk Factors in Children and 1996, 129, 637–642.
Their Relatives. Am. J. Epidemiol. 1989, 129, 973–987. 55. Kaplan, A. S.; Zemel, B. S.; Stallings, V. A. Differences in
39. Burns, T. L.; Moll, P. P.; Lauer, R. M. Increased Familial Resting Energy Expenditure in Prepubertal Black Children
Cardiovascular Mortality in Obese School Children: The and White Children. J. Pediatr. 1996, 129, 643–647.
Muscatine Ponderosity Family Study. Pediatrics 1992, 89 56. Bacha, F.; Saad, R.; Gungor, N.; Janosky, J.; Arslanian, S. A.
(2), 262–268. Obesity, Regional Fat Distribution, and Syndrome X in Obese
40. Schrott, H. G.; Clarke, W. R.; Wiebe, D. A.; Connor, W. E.; Black Versus White Adolescents: Race Differential in Diabe-
Lauer, R. M. Increased Coronary Mortality in Relatives of togenic and Atherogenic Risk Factors. J. Clin. Endocrinol.
Hypercholesterolemic School Children: The Muscatine Study. Metab. 2003, 88 (6), 2534–2540.
Circulation 1979, 59 (2), 320–326. 57. Roberts, S. B. Energy Expenditure and the Development of
41. Burns, T. L.; Letuchy, E. M.; Paulos, R.; Witt, J. Child- Early Obesity. Ann. NY Acad. Sci. 1993, 699, 18–25.
hood Predictors of the Metabolic Syndrome in Middle-Aged 58. Bassett, D. R., Jr.; Schneider, P. L.; Huntington, G. E. Physi-
Adults: The Muscatine Study. J. Pediatr. 2009, 155 (3), S5. cal Activity in an Old Order Amish Community. Med. Sci.
42. Freedman, D. S.; Dietz, W. H.; Srinivasan, S. R.; Berenson, Sports Excerc. 2004, 36, 79–85.
G. S. The Relation of Overweight to Cardiovascular Risk 59. Bouchard, C. Defining the Genetic Architecture of the Pre-
Factors among Children and Adolescents: The Bogalusa disposition to Obesity: A Challenging but not Insurmount-
Heart Study. Pediatrics 1999, 103, 1175–1182. able Task. Am. J. Clin. Nutr. 2010, 91 (1), 5–6.
43. Srinivasan, S. R.; Myers, L.; Berenson, G. S. Temporal Asso- 60. Chung, W. K. An Overview of Monogenic and Syndromic
ciation Between Obesity and Hyperinsulinemia in Children, Obesities in Humans. Pediatr. Blood Cancer 201110.1002/
Adolescents, and Young Adults: The Bogalusa Heart Study. pbc.23372.
Metab. Clin. Exp. 1999, 48, 928–934. 61. Rokholm, B.; Silventoinen, K.; Skytthe, A.; Kyvik, K. O.;
44. Mahaney, M. C.; Blangero, J.; Comuzzie, A. G.; VandeBerg, Sørensen, T. I. Increased Genetic Variance of BMI with
J. L.; Stern, M. P.; MacCluer, J. W. Plasma HDL Choles- a Higher Prevalence of Obesity. PLoS One 2011, 6 (6),
terol, Triglycerides, and Adiposity: A Quantitative Genetic e20816.
Test of the Conjoint Trait Hypothesis in the San Antonio 62. Lupski, J. R.; Belmont, J. W.; Boerwinkle, E.; Gibbs, R. A.
Family Heart Study. Circulation 1995, 92 (11), 3240–3248. Clan Genomics and the Complex Architecture of Human
45. Hong, Y.; Rice, T.; Gagnon, J.; Despres, J.-P.; Nadeau, Disease. Cell 2011, 147 (1), 32–43.
A.; Perusse, L.; Bouchard, C.; Leon, A. S.; Skinner, J. S.; 63. Coleman, D. L. Effects of Parabiosis of Obese with Diabetes
Wilmore, J. H., et  al. Familial Clustering of Insulin and and Normal Mice. Diabetologia 1973, 9, 294–298.
Abdominal Visceral Fat: The Heritage Family Study. J. Clin. 64. Tokuyama, K.; Himms-Hagen, J. Increased Sensitivity of the
Endocrinol. Metab. 1998, 83, 4239–4245. Genetically Obese Mouse to Corticosterone. Am. J. Phys.
46. Rice, T.; Nadeau, A.; Perusse, L.; Bouchard, C.; Rao, D. C. 1987, 252, E202–E208.
Familial Correlations in the Québec Family Study: Cross- 65. Coleman, D. L. Obese and Diabetes: Two Mutant Genes
trait Familial Resemblance for Body Fat with Plasma Glu- Causing Diabetes-Obesity Syndromes in Mice. Diabetologia
cose and Insulin. Diabetologia 1996, 39, 1357–1364. 1978, 14, 141–148.
47. Norman, R. A.; Thompsom, D. B.; Foroud, T.; Garvey, W. 66. Friedman, J. M.; Leibel, R. L.; Siegel, D. S.; Walsh, J.;
T.; Bennett, P. H.; Bogardus, C.; Ravussin, E. Genome Wide Bahary, N. Molecular Mapping of the Mouse ob Mutation.
Search for Genes Influencing Percent Body Fat in Pima Indi- Genomics 1991, 11, 1054–1062.
ans: Suggestive Linkage at Chromosome 11q21-q22. Am. J. 67. Zhang, Y.; Proenca, R.; Maffel, M.; Barone, M.; Leopold, L.;
Hum. Genet. 1997, 60, 166–173. Friedman, J. M. Positional Cloning of the Mouse Obese Gene
48. Knoll, J. H.; Sinnett, D.; Wagstaff, J.; Glatt, K.; Schanz and its Human Homologue. Nature 1994, 372, 425–432,
Wilcox, A.; Whiting, P. M.; Wingrove, P.; Sikela, J. M.; (December 1).
CHAPTER 90  Disorders of the Body Mass 25

68. Cinti, S.; Frederich, R. C.; Zingaretti, M. C.; De Matteis, R.; of Leptin in Humans: Gender-based Differences in Rela-
Flier, J. S.; Lowell, B. B. Immunohistochemical Localization tionship to Adiposity, Insulin Sensitivity, and Energy
of Leptin and Uncoupling Protein in White and Brown Adi- Expenditure. J. Clin. Endocrinol. Metab. 1997, 82,
pose Tissue. Endocrinology 1997, 138, 797–804. 1293–1300.
69. Pelleymounter, M. A.; Cullen, M. J.; Baker, M. B.; Hecht, R.; 84. Ronnemaa, R.; Karonen, S. L.; Rissanen, A.; Koskenvuo, M.;
Winters, D.; Boone, T.; Collins, F. Effects of the Obese Gene Koivisto, V. A. Relation Between Plasma Leptin Levels and
Product on Body Weight Regulation in ob/ob Mice. Science Measures of Body Fat in Identical Twins Discordant for Obe-
1995, 269, 540–543, (July 28). sity. Ann. Intern. Med. 1997, 126, 26–31.
70. Muzzin, P.; Eisensmith, R. C.; Copeland, K. C.; Woo, S. L.C. 85. Horlick, M. B.; Rosenbaum, M.; Nicolson, M.; Levine, L. S.;
Correction of Obesity and Diabetes in Genetically Obese Fedun, B.; Wang, J.; Pierson, R. N., Jr.; Leibel, R. L. Effect of
Mice by Leptin Gene Therapy. Proc. Natl. Acad. Sci. 1996, Puberty on the Relationship Between Circulating Leptin and
93, 14804–14808. Body Composition. J. Clin. Endocrinol. Metab. 2000, 85 (7),
71. Halaas, J. L.; Gajiwala, K. S.; Maffei, M.; Cohen, S. L.; 2509–2518.
Chait, B. T.; Rabinowitz, D.; Lallone, R. L.; Burley, S. K.; 86. Ostlund, R. E.; Yang, J. W.; Klein, S.; Gingerich, R. Rela-
Friedman, J. M. Weight-Reducing Effects of the Plasma tion Between Plasma Leptin Concentration and Body Fat,
Protein Encoded by the Obese Gene. Science 1995, 269, Gender, Diet, Age, and Metabolic Covariates. J. Clin. Endo-
543–546, (July 28). crinol. Metab. 1996, 81, 3909–3913.
72. Chen, G.; Koyama, K.; Yuan, X.; Lee, Y.; Zhou, Y. T.; 87. Havel, P. J.; Kasim-Karakas, S.; Mueller, W.; Johnson, P. R.;
O’Doherty, R.; Newgard, C. B.; Unger, R. H. Disappear- Gingerich, R. L.; Stern, J. S. Relationship of Plasma Leptin
ance of Body Fat in Normal Rats Induced by Adenovirus-­ to Plasma Insulin and Adiposity in Normal Weight and
mediated Leptin Gene Therapy. Proc. Natl. Acad. Sci. 1996, ­Overweight Women: Effects of Dietary Fat Content and Sus-
93, 14795–14799, (December). tained Weight Loss. J. Clin. Endocrinol. Metab. 1996, 81,
73. Chehab, F. F.; Lim, M. E.; Lu, R. Correction of the Steril- 4406–4413.
ity Defect in Homozygous Obese Female Mice by Treatment 88. Grinspoon, S.; Gulick, T.; Askari, H.; Landt, M.; Lee,
with the Human Recombinant Leptin. Nat. Genet. 1996, 12, K.; Anderson, E.; Ma, Z.; Vignati, L.; Bowsher, R.;
318–320, (March). Herzog, D., et  al. Serum Leptin Levels in Women with
74. Chehab, F. F.; Mounzih, K.; Lu, R.; Lim, M. E. Early Onset Anorexia ­Nervosa. J. Clin. Endocrinol. Metab. 1996, 81,
of Reproductive Function in Normal Female Mice Treated 3861–3863.
with Leptin. Science 1997, 275, 88–90, (3 January). 89. Weigle, D. S.; Ganter, S. L.; Kuijper, J. L.; Leonetti, D. L.;
75. Cheung, C. C.; Thornton, J. E.; Kuijper, J.; Weigle, D. S.; Boyko, E. J.; Fujimoto, W. Y. Effect of Regional Fat Distri-
Clifton, D. K.; Steiner, R. A. Leptin Is a Metabolic Gate for bution and Prader–Willi Syndrome on Plasma Leptin Levels.
the Onset of Puberty in The Female Rat. Endocrinology J. Clin. Endocrinol. Metab. 1997, 82, 566–570.
1997, 138 (2), 855–858. 90. Hassink, S. G.; Delancey, E.; Sheslow, D. V.; Smithkirwin,
76. Jacob, R. J.; Dziura, J.; Medwick, M. B.; Leone, P.; Caprio, S.; S. M.; O’Connor, D. M.; Considine, R. V.; Opentanova, I.;
During, M.; Shulman, G. I.; Sherwin, R. S. The Effect of Leptin Dostal, K.; Spear, M. L.; Leef, K., et al. Placental Leptin—An
is Enhanced by Microinjection into the Ventromedial Hypo- Important New Growth Factor in Intrauterine and Neonatal
thalamus. Diabetes 1997, 46, 150–152. Development. Pediatrics 1997, 100, E11–E16.
77. Glaum, S. R.; Hara, M.; Bindokas, V. P.; Lee, C. C.; 91. Sinha, M. K.; Sturis, J.; Ohannesian, J.; Magosin, S.;
­Polonsky, K. S.; Bell, G. I.; Miller, R. J. Leptin, the Obese ­Stephens, T.; Heiman, M. L.; Polonsky, K. S.; Caro, J. F.
Gene Product, Rapidly Modulates Synaptic Transmission in Ultradian Oscillations of Leptin Secretion in Humans. Bio-
the Hypothalamus. Mol. Pharmacol. 1996, 50, 230–235. chem. Biophys. Res. Commun. 1996, 228 (1724), 733–738.
78. Sivitz, W. I.; Walsh, S.; Morgan, D.; Donohoue, P.; 92. Laughlin, G. A.; Yen, S. S. C. Hypoleptinemia in Women
Haynes, W.; Leibel, R. L. Plasma Leptin in Diabetic and Athletes: Absence of a Diurnal Rhythm with Amenorrhea. J.
Insulin-Treated Diabetic and Normal Rats. Metabolism Clin. Endocrinol. Metab. 1997, 82 (1), 318–321.
1998, 8, 584–591. 93. Cohen, B.; Novick, D.; Rubinstein, M. Modulation of
79. Sinha, M. K.; Opentanova, I.; Ohannesian, J. P.; Kolaczynski, ­Insulin Activities by Leptin. Science 1996, 274, 1185–1188,
J. W.; Heiman, M. L.; Hale, J.; Becker, G. W.; Bowsher, R. (15 November).
R.; Stephens, T. W.; Caro, J. F. Evidence of Free and Bound 94. Montague, C. T.; Farooqi, S.; Whitehead, J. P.; Soos, M.
Leptin in Human Circulation. J. Clin. Invest. 1996, 98, A.; Rau, H.; Wareham, N. J.; Sewter, C. P.; Digby, J. E.;
1277–1282. ­Mohammed, S. N.; Hurst, J. A., et  al. Congenital Leptin
80. Considine, R. V.; Sinha, M. K.; Heiman, M. L.; Deficiency Is Associated with Severe Early-Onset Obesity in
Kriauciunas, A.; Stephens, T. W.; Nyce, M. R.; Ohan- Humans. Nature 1997, 387, 903–908.
nesian, J. P.; Marco, C. C.; McKee, L. J.; Bauer, T. L., 95. Considine, R. V.; Considine, E. L.; Williams, C. J.;
et  al. Serum Immunoreactive-leptin Concentrations in Nyce, M. R.; Magosin, S. A.; Bauer, T.; Rosato, E. L.;
Normal-Weight and Obese Humans. N. Engl. J. Med. ­Colberg, J.; Caro, J. F. Evidence Against Either a Premature
1996, 334, 292–295. Stop Codon or the Absence of Obese Gene mRNA in Human
81. Kichey, M. S.; Israel, R. G.; Gardiner, S. N.; Obesity. J. Clin. Invest. 1995, 95, 2986–2988.
Considine, R. V.; McCammon, M. R.; Tyndall, G. L.; 96. Maffei, M.; Stoffel, M.; Barone, M.; Moon, B.;
Houmard, J. A.; Marks, R. H. L.; Caro, J. F. Gender Dif- Dammerman, M.; Ravussin, E.; Bogardus, C.; Ludwig, D.
ferences in Serum Leptin Levels in Humans. Biochem. Soc. S.; Flier, J.; Talley, M., et  al. Absence of Mutations in the
Trans. 1996, 59, 1–6. Human ob Gene in Obese/Diabetic Subjects. Diabetes 1996,
82. Rosenbaum, M.; Nicolson, M.; Hirsch, J.; Heymsfield, S. 45, 679–682.
B.; Gallagher, D.; Chu, F.; Leibel, R. L. Effects of Gender, 97. Considine, R. V.; Concsidine, E. L.; Williams, C. J.;
Body Composition, and Menopause on Plasma Concen- Nyce, M. R.; Zhang, P.; Opentanova, I.; Ohannesian, J. P.;
trations of Leptin. J. Clin. Endocrinol. Metab. 1996, 81, Kolaczynski, J. W.; Bauer, T. L.; Moore, J. H., et al. Muta-
3424–3427. tion Screening and Identification of a Sequence Variation
83. Kennedy, A.; Gettys, T. W.; Watson, P.; Wallace, P.; Gan- in the Human ob Coding Region. Biochem. Biophys. Res.
away, E.; Pan, Q.; Garvey, T. The Metabolic Significance Commun. 1996, 220, 735–739.
26 CHAPTER 90  Disorders of the Body Mass

98. Norman, R. A.; Leibel, R. L.; Chung, W. K.; Power-Kehoe, L.; 113. Chen, H.; Charlat, O.; Tartaglia, L. A.; Woolf, E. A.; Weng, X.;
Chua, S. C., Jr.; Knowler, W. C.; Thompson, D. B.; Bogardus, Ellis, S. J.; Lakey, N. D.; Culpepper, J.; Moore, K. J.; Breitbart,
C.; Ravussin, E. Absence of Linkage of Obesity and Energy R. E., et al. Evidence that the Diabetes Gene Encodes the Leptin
Metabolism to Markers Flanking Homologues of Rodent Obe- Receptor: Identification of a Mutation in the Leptin Receptor
sity Genes in Pima Indians. Diabetes 1996, 45, 1229–1232. in db/db Mice. Cell 1996, 84, 491–495.
99. Stirling, B.; Cox, N. J.; Bell, G. I.; Hanis, C. L.; Speilman, R. S.; 114. Cioffi, J. A.; Shafer, A. W.; Zupancic, T. J.; Smith-Gbur, J.;
Concannon, P. Identification of Microsatellite Markers near Mikhail, A.; Platika, D.; Snodgrass, H. R. Novel B219/OB
the Human ob Gene and Linkage Studies in NIDDM-Affected Receptor Isoforms: Possible Role of Leptin in Hematopoi-
Sib Pairs. Diabetes 1995, 44, 999–1001. esis and Reproduction. Nat. Med. 1996, 2 (5), 585–589,
100. Bray, M.; Boerwinkle, E.; Hanis, C. L. OB Gene not (May).
Linked to Human Obesity in Mexican American Affected 115. Cumin, F.; Baum, H. P.; Levens, N. Leptin Is Cleared from
Sib Pairs from Starr County, Texas. Hum. Genet. 1996, the Circulation Primarily by the Kidney. Int. J. Obes. 1996,
98, 590–595. 20, 1120–1126.
101. Clement, K.; Garner, C.; Hager, J.; Philippi, A.; LeDuc, C.; 116. Yang, G.; Ge, H.; Boucher, A.; Yu, X.; Li, C. Modulation
Carey, A.; Harris, T. J. R.; Jury, C.; Cardon, L. R.; Bas- of Direct Leptin Signaling by Soluble Leptin Receptor. Mol.
devant, A., et  al. Indication for Linkage of the Human ob Endocrinol. 2004, 18 (6), 1354–1362.
Gene Region with Extreme Obesity. Diabetes 1996, 45, 117. Ghilardi, N.; Ziegler, S.; Wiestner, A.; Stoffel, R.; Heim, M.
687–690. H.; Skoda, R. C. Defective STAT Signaling by the Leptin
102. Reed, D. R.; Ding, Y.; Xu, W.; Cather, C.; Green, E. D.; Receptor in Diabetic Mice. Proc. Natl. Acad. Sci. U.S.A.
Price, R. A. Extreme Obesity May Be Linked to Markers 1996, 93, 6231–6235.
Flanking the Human ob Gene. Diabetes 1996, 45, 691–694. 118. Vaisse, C.; Halaas, J. L.; Horvath, C. M.; Darnell, J. E.;
103. Duggirala, R.; Stern, M. P.; Mitchell, B. D.; Reinhart, L. J.; ­Stoffel, M.; Friedman, J. M. Leptin Activation of STAT3 in
Shipman, P. A.; Uresandi, O. C.; Chung, W. K.; Leibel, R. L.; the Hypothalamus of Wildtype and ob/ob Mice but not in
Hales, C. N.; O’Connell, P., et al. Quantitative Variation in db/db Mice. Nat. Genet. 1996, 14, 95–97.
Obesity-Related Traits and Insulin Presursors Linked to the 119. Dulloo, A. G.; Miller, D. S. Prevention of Genetic fa/fa Obe-
ob Gene Region on Human Chromosome 7. Am. J. Hum. sity with an Ephedrine-Methylxanthines Thermogenic Mix-
Genet. 1996, 59, 694–703. ture. Am. J. Phys. 1987, 252, R507–R513.
104. Hager, J.; Clement, K.; Francke, S.; Dina, C.; Raison, J.; 120. Dulloo, A. G.; Miller, D. S. Reversal of Obesity in the Genet-
Lahlou, N.; Rich, N.; Pelloux, V.; Basdevant, A.; Guy-Grand, ically Obese fa/fa Zucker Rat with an Ephedrine/Methylxan-
B., et al. A Polymorphism in the 5’ Untranslated Region of thines Thermogenic Mixture. J. Nutr. 1987, 117, 383–389.
Human ob Gene Is Associated with Low Leptin Levels. Int. 121. Eikelis, S. A.K.E. Interactions Between Leptin and the
J. Obes. 1998, 22, 200–205. Human Sympathetic Nervous System. Hypertension 2003,
105. Shintani, M.; Ikegami, H.; Fujisawa, T.; Kawaguchi, Y.; 41 (5), 1072–1079.
Ohishi, M.; Katsuya, T.; Higaki, J.; Shimamoto, K.; Ogi- 122. Hausberg, M.; Morgan, D. A.; Chapleau, M. A.; Sivitz, W.
hara, T. Leptin Gene Polymorphism Is Associated with I.; Mark, A. L.; Haynes, W. G. Differential Modulation of
Hypertension Independent of Obesity. J. Clin. Endocrinol. Leptin-induced Sympathoexcitation by Baroreflex Activa-
Metab. 2002, 87 (6), 2909–2912. tion. J. Hypertens. 2002, 20 (8), 1633–1641.
106. Tartaglia, L. A.; Dembski, M.; Weng, X.; Deng, N.; 123. Chung, W. K.; Belfi, K.; Chua, M.; Wiley, J.; Mackintosh, R.;
­Culpepper, J.; Devos, R.; Richards, G. J.; Campfield, L. Nicolson, M.; Boozer, C.; Leibel, R. L. Heterozygosity for Lepob
A.; Clark, F. T.; Deeds, J., et al. Identification and Expres- and Leprdb Affects Body Composition and Leptin Homeostasis
sion Cloning of a Leptin Receptor, OB-R. Cell 1996, 83, in Adult Mice. Am. J. Phys. 1998, 274, R985–R990.
1263–1271. 124. Stephens, T. W.; Basinski, M.; Bristow, P. K.; Bue-Valleskey,
107. Truett, G. E.; Bahary, N.; Friedman, J. M.; Leibel, R. L. Rat J. M.; Burgett, S. G.; Craft, L.; Hale, J.; Hoffmann, J.; Hsi-
Obesity Gene Fatty (fa) Maps to Chromosome 5: Evidence ung, H. M.; Kriauciunas, A. The Role of Neuropeptide Y in
for Homology with the Mouse Gene Diabetes (db). Proc. the Antiobesity Action of the Obese Gene Product. Nature
Natl. Acad. Sci. U.S.A. 1991, 88, 7806–7809. 1995, 377 (6549), 530–532, (October 12).
108. Chua, S. C.; White, D. W.; Wu-Peng, X. S.; Liu, S. M.; 125. Pinto, S.; Roseberry, A. G.; Liu, H.; Diano, S.; Shanabrough,
Okada, N.; Kershaw, E. E.; Chung, W. K.; Power-Kehoe, L.; M.; Cai, X.; Friedman, J. M.; Horvath, T. L. Rapid Rewir-
Chua, M.; Tartaglia, L. A., et al. Phenotype of Fatty due to ing of Arcuate Nucleus Feeding Circuits by Leptin. Science
Gln269Pro Mutation in the Leptin Receptor (lepr). Diabetes 2004, 304, 110–115.
1996, 45, 1141–1143. 126. Huang, L. Leptin: A Multifunctional Hormone. Cell
109. Iida, M.; Murakami, T.; Ishida, K.; Mizuno, A.; Kuwajima, M.; Research 2000, 10 (2), 81–92.
Shima, K. Phenotype-Linked Amino Acid Alteration in Leptin 127. Shirasaka, T.; Takasaki, M.; Kannan, H. Cardiovascular
Receptor cDNA from Zucker Fatty (fa/fa) Rat. Biochem. Bio- effects of Leptin and Orexins. Am. J. Physiol. Regul. Integr.
phys. Res. Commun. 1996, 222, 19–26. Comp. Physiol. 2003, 284 (3), R639–R651.
110. Chua, S. C.; Chung, W. K.; Wu-Peng, S.; Zhang, Y.; 128. Myers, M. G.; Leibel, R. L.; Seeley, R. J.; Schwartz, M. W.
Liu, S. -M.; Tartaglia, L.; Leibel, R. L. Phenotypes of Mouse Obesity and Leptin Resistance: Distiguishing Cause from
Diabetes and Rat Fatty Due to Mutations in the OB (Leptin) Effect. Trends. Endocrinol. Metab. 2010, 21 (11), 643–651.
Receptor. Science 1996, 271, 994–996. 129. Thompson, D. B.; Janssen, R. C.; Ossowski, V. M. P. M.;
111. Wupeng, X. S.; Chua, S. C.; Okada, N.; Liu, S. M.; Knowler, W. C.; Bogardus, C. Evidence for Linkage Between
Nicolson, M.; Leibel, R. L. Phenotype of the Obese Koletsky a Region on Chromosome 1 p and the Acute Insulin Response
(F) Rat Due to Tyr763Stop Mutation in the Extracellular in Pima Indians. Diabetes 1995, 44, 478–481.
Domain of the Leptin Receptor (Lepr)- Evidence for Defi- 130. Chung, W. K.; Power-Kehoe, L.; Chua, M.; Lee, R.;
cient Plasma-to-CSF Transport of Leptin in both Zucker and Leibel, R. L. Genomic Structure of the Human OB Recep-
Koletsky Obese Rat. Diabetes 1997, 46, 513–518. tor and Identification of Two Novel Intronic Microsatellites.
112. Lee, G. -H.; Proenca, R.; Montez, J. M.; Carroll, K. M.; Genome. Res. 1996, 6, 1192–1199.
­Darvishzadeh, J. G.; Lee, J. I.; Friedman, J. M. Abnormal 131. Chagnon, Y. C.; Perusse, L.; Lamothe, M.; Chagnon, M.;
Splicing of the Leptin Receptor in Diabetic Mice. Nature Nadeau, A.; Dionne, F. T.; Gagnon, J.; Chung, W. K.; Leibel,
1996, 379, 632–635. R. L.; Bouchard, C. Suggestive Linkages Between Markers
CHAPTER 90  Disorders of the Body Mass 27

on Human 1p.32-p22 and Body Fat and Insulin Levels in 146. Kleyn, P. W.; Fan, W.; Kovats, S. G.; Lee, J. J.; Pulido, J. C.;
the Québec Family Study. Obes. Res. 1997, 5 (2), 115–121. Wu, Y.; Berkemeier, L. R.; Misumi, D. J.; Holmgren, L.; Char-
132. Chung, W. K.; Power-Kehoe, L.; Chua, M.; Chu, F.; lat, O., et al. Identification and Characterization of the Mouse
Aronne, L.; Huma, Z.; Sothern, M.; Udall, J. N.; Kahle, B.; Obesity Gene Tubby: A Member of a Novel Gene Family. Cell
Leibel, R. L. Exonic and Intronic Sequence Variation in the 1996, 85, 281–290.
Human Leptin Receptor Gene (LEPR). Diabetes 1997, 46, 147. Boggon, T. J.; Shan, W.-S.; Santagata, S.; Myers, S. C.;
1509–1511. ­Shapiro, L. Implication of Tubby Proteins as Transcription
133. Thompson, D. B.; Ravussin, E.; Bennett, P. H.; Bogardus, C. Factors by Structure-Based Functional Analysis. Science
Structure and Sequence Variation at the Leptin Receptor Gene 1999, 286, 2119–2125.
in Lean and Obese Pima Indians. Hum. Mol. Genet. 1997, 6, 148. Carroll, G. Tubby Proteins: The Plot Thickens. Nature
675–679. Reviews. Molecular Cell Biology 2004, 5 (1), 55–63.
134. Chagnon, Y. C.; Wilmore, J. H.; Borecki, I. B.; Gagnon, J.; 149. Santagata, S.; Boggon, T. J.; Baird, C. L.; Gomez, C. L.; Zhao,
Perusse, L.; Chagnon, M.; Collier, G. R.; Leon, A. S.; Skin- J.; Shan, W. S.; Myszka, D. G.; Shapiro, L. G-Protein Signal-
ner, J. S.; Rao, D. C., et al. Associations Between the Leptin ing through Tubby Proteins. Science 2001, 292, 2041–2050.
Receptor Gene and Adiposity in Middle-Age Caucasian 150. Naggert, J. K.; Fricker, L. D.; Varlamov, O.; Nishina, P. M.;
Males from the HERITAGE Family Study. J. Clin. Endocri- Rouille, Y.; Steiner, D. F.; Carroll, R. J.; Paigen, B. J.; Leiter,
nol. Metab. 2000, 85, 29–34. E. H. Hyperproinsulinaemia in Obese Fat/Fat Mice Associ-
135. Donohoue, P. A.; Burns, T. L.; Mendoza, M. C.B.; ated with a Carboxypeptidase E Mutation which Reduces
Chung, W. K.; Leibel, R. L. Lys656Asn Variant of the Leptin Enzyme Activity. Nat. Genet. 1995, 10, 135–142.
Receptor Gene (LEPR) and the β-3 Adrenergic Receptor 151. Zhang, C. -F.; Snell, C. R.; Loh, Y. P. Identification of a
(B3AR) Gene Linked to Body Mass Index in Humans: The Novel Prohormone Sorting Signal-Binding Site on Carboxy-
Muscatine Study. Pediatr. Res. 2000, 47, 127A. peptidase E, a Regulated Secretory Pathway-Sorting Recep-
136. Chagnon, Y. C.; Chung, W. K.; Perusse, L.; Chagnon, M.; tor. Mol. Endocrinol. 1999, 13, 527–536.
­Leibel, R. L.; Bouchard, C. Linkages and Associations 152. Jackson, R. S.; Creemers, J. W.M.; Ohagi, S.; Raffin-Sansom,
Between the Leptin Receptor (LEPR) Gene and Human Body M. -L.; Sanders, L.; Montague, C. T.; Hutton, J. C.; O’Rahilly,
Composition in the Québec Family Study. Int. J. Obes. 1999, S. Obesity and Impaired Prohormone Processing Associated
23, 278–286, Ref Type: Abstract. with Mutations in the Prohormone Convertase 1 Gene. Nat.
137. Silver, K.; Walston, J.; Chung, W. K.; Yao, F.; Parikh, V. V.; Genet. 1997, 16, 303–306.
Andersen, R.; Cheskin, L. J.; Elahi, D.; Muller, D.; Leibel, 153. Kalidas, K.; Dow, E.; Saker, P. J.; Wareham, N.; Halsall, D.;
R. L., et al. The Gln223Arg and Lys656Asn Polymorphisms in Jackson, R. S.; Chan, S.-P.; Gelding, S.; Walker, M.;
the Human Leptin Receptor Do Not Associate with Traits Kousta, E., et al. Prohormone Convertase 1 in Obesity, Ges-
Related to Obesity. Diabetes 1997, 46, 1898–1900. tational Diabetes Mellitus, and NIDDM. Diabetes 1998, 47,
138. Echwald, S. M.; Sorenson, T. D.; Sorenson, T. I. A.; 287–289.
­Tybjaerg-Hansen, A.; Andersen, T.; Chung, W. K.; Leibel, 154. Krude, H.; Beibermann, H.; Luck, W.; Horn, R.; Brabant, G.;
R. L.; Pedersen, O. Amino Acid Variants on the Human Grüters, A. Severe Early-Onset Obesity, Adrenal Insufficiency
Leptin Receptor: Lack of Association to Juvenile Onset and Red Hair Pigmentation Caused by POMC Mutations in
Obesity. Biochem. Biophys. Res. Commun. 1997, 233, Humans. Nat. Genet. 1998, 19 (2), 155–157.
248–252. 155. Hixson, J. E.; Almasy, L.; Cole, S.; Birnbaum, S.;
139. Heo, M.; Leibel, R. L.; Fintaine, K. R.; Boyer, B. B.; Chung, Mitchell, B. D.; Mahaney, M. C.; MacCluer, J. W.; Blangero,
W. K.; Koulu, M.; Karvonene, M. K.; Pesonen, U.; Rissanen, J.; Comuzzie, A. G. Normal Variation in Leptin Levels Is
A.; Laasko, M., et al. A Meta-Analytic Investigation of Link- Associated with Polymorphisms in the Proopiomelanocor-
age and Association of Common Leptin Receptor (Lepr) tin Gene. POMC. J. Clin. Endocrinol. Metab. 1999, 84,
Polymorphisms with Body Mass Index and Waist Circum- 3187–3191.
ference. Int J Obesity 2002, 26, 640–646. 156. Comuzzie, A. G.; Hixson, J. E.; Almasy, L.; Mitchell, B. D.;
140. Clement, K.; Vaisse, C.; Lahlous, N.; Cabrol, S.; Pelloux, V.; Mahaney, M. C.; Dyer, T. D.; Stern, M. P.; MacCluer, J. W.;
Cassuto, D.; Gourmelen, M.; Dina, C.; Chambaz, J.; Lacortw, Blangero, J. A Major Quantitative Trait Locus Determining
J. -M., et al. A Mutation in the Human Leptin Receptor Gene Serum Leptin Levels and Fat Mass Is Located on Human
Causes Obesity and Pituitary Dysfunction. Nature 1998, Chromosome 2. Nat. Genet. 1997, 15, 273–276.
392, 398–401. 157. Rotimi, C.; Comuzzie, A. G.; Lowe, W.; Luke, A.; Blangero,
141. Clement, K.; Vega, N.; Laville, M.; Pelloux, V.; Guy-Grand, J.; Cooper, R. The Quantitative Trait Locus on Chromo-
B.; Basdevant, A.; Vidal, H. Adipose Tissue Gene Expression some 2 for Serum Leptin Levels Is Confirmed in African-
in Patients with a Loss of Function Mutation in the Leptin Americans. Diabetes 1999, 48, 643–644.
Receptor. Int. J. Obes. 2002, 26 (12), 1533–1538. 158. Hager, J.; Dina, C.; Francke, S.; Dubois, S.; Houari, M.;
142. Smith, R. J.; Berlin, C. I.; Hejtmancil, J. F.; Keats, B. J.; Vatin, V.; Vaillant, E.; Lorentz, N.; Basdevant, A.; Clement,
­Kimberling, W. J.; Lewis, R. A.; Moller, C. G.; Pelias, M. Z.; K., et  al. A Genome-Wide Scan for Human Obesity Genes
Tranebjaerg, L. Clinical Diagnosis of the Usher Syndromes. Reveals a Major Susceptibility Locus on Chromosome 10.
Usher Syndrome Consortium. Am. J. Med. Genet. 1994, 50, Nat. Genet. 1998, 20, 304–308.
32–38. 159. Kushi, A.; Sasai, H.; Koizumi, H.; Takeda, N.; Yokoyama, M.;
143. Carmi, R.; Rokhlina, T.; Kwitek-Black, A. E.; Elbedour, K.; Nakamura, M. Obesity and Mild Hyperinsulinemia Found
Nishimura, D.; Stone, E. M.; Sheffield, V. C. Use of a DNA in Neuropeptide Y-Y1 Receptor-Deficient Mice. Proc. Natl.
Pooling Strategy to Identify a Human Obesity Syndrome Acad. Sci. U.S.A. 1998, 95, 15659–15664.
Locus on Chromosome 15. Hum. Mol. Genet. 1995, 4, 9–13. 160. Hinney, A.; Becker, I.; Heibült, O.; Nottebom, K.;
144. Chung, W. K.; Goldberg-Berman, J.; Power-Kehoe, L.; Schmidt, A.; Ziegler, A.; Mayer, H.; Siegfried, W.; Blum,
Leibel, R. L. Molecular Mapping of the Tubby (tub) W. F.; ­Remschmidt, H., et  al. Systematic Screening of the
­Mutation on Mouse Chromosome 7. Genomics 1996, 32, ­Pro-opiomelanocortin Gene: Identification of Several Genetic
210–217. Variants Including Three Different Insertions, One Nonsense
145. Noben-Trauth, K.; Naggert, J. K.; North, M. A.; Nishina, and Two Missense Point Mutations in Probands of Differ-
P. M. A Candidate Gene for the Mouse Mutation Tubby. ent Weight Extremes. J. Clin. Endocrinol. Metab. 1998, 83,
Nature 1996, 380, 534–538. 3737–3741.
28 CHAPTER 90  Disorders of the Body Mass

161. Kucera, G. T.; Bortner, D. M.; Rosenberg, M. P. Overex- 177. Fleury, C.; Neverova, M.; Collins, S.; Raimbault, S.;
pression of an Agouti cDNA in the Skin of Transgenic Mice ­Champigny, O.; Levi-Meyrueis, C.; Bouillaud, F.; Seldin, M.
Recapitulates Dominant Coat Color Phenotypes of Sponta- F.; Surwit, R. S.; Ricquier, D., et al. Uncoupling Protein-2: A
neous Mutants. Dev. Biol. 1996, 173, 162–173. Novel Gene Linked to Obesity and Hyperinsulinemia. Nat.
162. Seeley, R. J.; Schwartz, M. W. Neuroendocrine Regulation Genet. 1997, 15, 269–272, (March).
of Food Intake. Acta. Paediatr. Suppl. 1999, 88 (Suppl 428), 178. Giacobino, J. P. β3-Adrenoceptor: An Update. Eur. J. Endo-
58–61. crinol. 1995, 132, 377–385.
163. Kesterson, R. A.; Huszar, D.; Lynch, C. A.; Simerly, R. B.; 179. Lowell, B. B.; S-susulic, V.; Hamann, A.; Lawitts, J. A.;
Cone, R. D. Induction of Neuropeptide Y Gene Expression Himms-Hagen, J.; Boyers, B. B.; Kozak, L. P.; Flier, J. S.
in the Dorsal Medial Hypothalamic Nucleus in Two Models Development of Obesity in Transgenic Mice After Genetic
of the Agouti Obesity Syndrome. Mol. Endocrinol. 1997, 11, Ablation of Brown Adipose Tissue. Nature 1993, 366 (23),
630–637. 740–742.
164. Huszar, D.; Lynch, C. A.; Fairchild-Huntress, V.; Dunmore, J.; 180. Hamann, A.; Flier, J. S.; Lowell, B. B. Decreased Brown Fat
Fang, Q.; Berkemeier, L. R.; Gu, W.; Kesterson, R. A.; Boston, B. Markedly Enhances Susceptibility to Diet-Induced Obesity,
A.; Cone, R. D., et al. Targeted Disruption of the Melanocortin-4 Diabetes, and Hyperlipidemia. Endocrinology 1996, 137
Receptor Results in Obesity in Mice. Cell 1997, 88, 131–141. (1), 21–29, (January).
165. Yeo, G. S.H.; Farooqi, S.; Aminian, S.; Halsall, D.; 181. Hamann, A.; Busing, B.; Kausch, C.; Ertl, J.; Preibisch, G.;
Stanhope, R.; O’Rahilly, S. A Frameshift Mutation in MC4R Greten, H.; Matthei, S. Chronic Leptin Treatment Does Not
Associated with Dominantly Inherited Human Obesity. Nat. Prevent the Development of Obesity in Transgenic Mice with
Genet. 1998, 20, 111–112. Brown Fat Deficiency. Diabetologia 1997, 40, 810–815.
166. Vaisse, C.; Clement, K.; Guy-Grand, B.; Froguel, P. A 182. Kopecky, J.; Clarke, G.; Enerback, S.; Spiegelman, B.;
Frameshift Mutation in Human MC4R Is Associated with a Kozak, L. P. Expression of the Mitochondrial Uncoupling
Dominant Form of Obesity. Nat. Genet. 1998, 20, 113–114. Protein Gene from the aP2 Gene Promoter Prevents Genetic
167. Donohoue, P. A.; Tao, Y. X.; Collins, M.; Yeo, G. S.H.; Obesity. J. Clin. Invest. 1995, 96, 2914–2923.
O’Rahilly, S.; Segaloff, D. L. Deletion of Codons 88-92 of 183. Surwit, R. S.; Wang, S.; Petro, A. E.; Sanchis, D.;
the Melanocortin-4 Receptor Gene: A Novel Deleterious Raimbault, S.; Ricquier, D.; Collins, S. Diet-Induced
Mutation in an Obese Female. J. Clin. Endocrinol. Metab. Changes in Uncoupling Proteins in Obesity-Prone and
2003, 88 (12), 5841–5845. ­Obesity-Resistant Strains of Mice. Proc. Natl. Acad. Sci.
168. Hinney, A.; Schmidt, A.; Nottebaum, K.; Heibült, O.; U.S.A. 1998, 95, 4061–4065.
Becker, I.; Ziegler, A.; Gerber, G.; Sina, M.; Görg, T.; Mayer, 184. Sivitz, W. I.; Fink, B. D.; Donohoue, P. A. Fasting and Leptin
H., et  al. Several Mutations in the Melanocortin-4 Recep- Modulate Adipose and Muscle Uncoupling Protien: Diver-
tor Gene Including a Nonsense and a Frameshift Mutation gent Effects Between mRNA and Protein Expression. Endo-
Associated with Dominantly Inherited Obestiy in Humans. J. crinology 1998, 140, 1511–1519.
Clin. Endocrinol. Metab. 1999, 84, 1483–1486. 185. Collins, S.; Daniel, K. W.; Petro, A. E.; Surwit, R. S. Strain-
169. Donohoue, P. A.; Tao, Y.-X.; Collins, M.; Yeo, G. S. H.; Specific Response to β3-Adrenergic Receptor Agonist Treat-
O'Rahilly, S.; Segaloff, D. L. Deletion of Codons 88–92 of ment of Diet-Induced Obesity in Mice. Endocrinology 1997,
the Melanocortin-4 Receptor Gene: A Novel Deleterious 138 (1), 405–413.
Mutation in an Obese Female. J. Clin. Endocrin. Metab. 186. Susulic, V. S.; Frederich, R. C.; Lawitts, J.; Tozzo, E.;
2003, 88, 5841–5845. Kahn, B. B.; Harper, M. E.; Himms-Hagen, J.; Flier, J. S.;
170. Gu, W.; Tu, Z.; Kleyn, P. W.; Kissebah, A.; Duprat, L.; Lee, Lowell, B. B. Targeted Disruption of the β3-Adrenergic
J.; Chin, W.; Maruti, S.; Deng, N.; Fisher, S. L., et al. Iden- Receptor Gene. J. Biol. Chem. 1995, 270 (49), 29483–
tification and Functional Analysis of Novel Human Mela- 29492, (December).
nocortin-4 Receptor Variants. Diabetes 1999, 48, 635–639. 187. Begin-Heick, N. β3-Adrenergic Activation of ­Adenylyl
171. Tao, Y. X.; Segaloff, D. L. Functional Characterization of Cyclase in Mouse White Adipocytes: Modulation by
Melanocortin-4 Receptor Mutations Associated with Child- Gtp and Effect of Obesity. J. Cell. Biochem. 1995, 58,
hood Obesity. Endocrinology 2003, 144 (10), 4544–4551. ­464–473.
172. Lubrano-Berthelier, C.; Le Stunff, C.; Bougneres, P.; Vaisse, 188. Trayhurn, P.; Duncan, J. S.; Rayner, D. V.; Hardie, L. Rapid
C. A Homozygous Null Mutation Delineates the Role of the Inhibition of ob Gene Expression and Circulating Leptin
Melanocortin-4 Receptor in Humans. J. Clin. Endocrinol. Levels in Lean Mice by the β3-Adrenoceptor Agonists BRL
Metab. 2004, 89 (5), 2028–2032. 35135A and ZD2079. Biochem. Biophys. Res. Commun.
173. Chagnon, Y. C.; Chen, W.; Perusse, L.; Chagnon, M.; 1996, 228 (1704), 605–610.
Nadeau, A.; Wilkison, W.; Bouchard, C. Linkage and Asso- 189. Sivitz, W. I.; Fink, B. D.; Morgan, D.; Fox, J.; Donohoue, P.;
ciation Studies Between the Melanocortin Receptors 4 and 5 Haynes, W. Sympathetic Inhibition, Leptin, and Uncoupling
Genes and Obesity-Related Phenotypes in the Québec Fam- Protein Subtype Expression in Normal Fasting Rats. Am. J.
ily Study. Mol. Med. 1997, 3, 663–673. Phys. 1999, 277, E668–E677, (Endocrinol Metab 40).
174. Tao, Y. X.; Segaloff, D. L. Functional Characterization 190. Emorine, L. J.; Marullo, S.; Briend-Sutren, M. M.; Patey, G.;
of Melanocortin-3 Receptor Variants Identify a Loss-of- Tate, K.; Delavier-Klutchko, C.; Strosberg, A. D. Molecular
Function Mutation Involving an Amino Acid Critical for G Characterization of the Human β3-Adrenergic Receptor. Sci-
Protein-Coupled Receptor Activation. J. Clin. Endocrinol. ence 1989, 245, 1118–1121, (September).
Metab. 2004, 89 (8), 3936–3942. 191. Lonnqvist, F.; Thorne, A.; Nilsell, K.; Hoffstedt, J.; Arner, P.
175. Vidal-Puig, A.; Solanes, G.; Grujic, D.; Flier, J. S.; Lowell, B. B. A Pathogenic Role of Visceral Fat β3-Adrenoceptors in Obe-
UCP3: An Uncoupling Protein Homologue Expressed Preferen- sity. J. Clin. Invest. 1995, 95, 1109–1116, March.
tially and Abundantly in Skeletal Muscle and Brown Adipose 192. Walston, J.; Silver, K.; Bogardus, C.; Knowler, W. C.;
Tissue. Biochem. Biophys. Res. Commun. 1997, 235, 79–82. Celi, F. S.; Austin, S.; Manning, B.; Strosberg, A. D.; Stern,
176. Gimeno, R. E.; Dembski, M.; Weng, X.; Deng, N.; Shyjan, M. P.; Raben, N., et  al. Time of Onset of Non-Insulin-
A. W.; Gimeno, C. J.; Iris, F.; Ellis, S. J.; Woolf, E. A.; Tar- Dependent Diabetes Mellitus and Genetic Variation in the
taglia, L. A. Cloning and Characterization of an Uncoupling β3-Adrenergic-Receptor Gene. N. Engl. J. Med. 1995, 333,
Protein Homolog. Diabetes 1997, 46, 900–906. 343–347.
CHAPTER 90  Disorders of the Body Mass 29

193. Widen, E.; Lehto, M.; Kanninen, T.; Walston, J.; Shuldiner, 206. Candelore, M. R.; Deng, L.; Tota, L. J.; Cascieri, M. A.;
A. R.; Groop, L. C. Association of a Polymorphism in the Strader, C. D. Pharmacological Characterization of a
β3-Adrenergic-receptor Gene with Features of the Insulin Recently Described Human β3-Adrenergic Receptor Mutant.
Resistance Syndrome in Finns. N. Engl. J. Med. 1995, 333, Endocrinology 1996, 137, 2638–2641.
348–351. 207. Shihara, N.; Yasuda, K.; Moritani, Y.; Ue, H.; Adachi, T.;
194. Sipilainen, R.; Uusitupa, M.; Heikkinen, S.; Rissanen, A.; Tanaka, K.; Seino, Y. The Association Between Trp64Arg
Laakso, M. Polymorphism of the β3-Adrenergic Receptor Polymorphism of the β3-Adrenergic Receptor and Auto-
Gene Affects Basal Metabolic Rate in Obese Finns. Diabetes nomic Nervous System Activity. J. Clin. Endocrinol. Metab.
1997, 46, 77–80. 1999, 84, 1623–1627.
195. Clement, K.; Vaisse, C.; Manning, B. S.J.; Basdevant, A.; 208. Boivin, M.; Camirand, A.; Carli, F.; Hoffer, L.; Silva, J.
Guy-Grand, B.; Ruiz, J.; Silver, K. D.; Shuldiner, A. R.; Uncoupling Protein-2 and -3 Messenger Ribonucleic Acids in
Froguel, P.; Strosberg, A. D. Genetic Variation in the β3- Adipose Tissue and Skeletal Muscle of Healthy Males: Vari-
Adrenergic Receptor and Increased Capacity to Gain Weight ability, Factors Affecting Expression, and Relation to Mea-
in Patients with Morbid Obesity. N. Engl. J. Med. 1995, sures of Metabolic Rate. J. Clin. Endocrinol. Metab. 2000,
333, 352–354. 85, 1975–1983.
196. Elbein, S. C.; Hoffman, M.; Barrett, K.; Wegner, K.; Miles, C.; 209. Boss, O.; Hagen, T.; Lowell, B. B. Uncoupling Proteins 2 and
Bachman, K.; Berkowitz, D.; Shuldiner, A. R.; Leppert, M. F.; 3: Potential Regulators of Mitochondrial Energy Metabo-
Hasstedt, S. Role of the β3-Adrenergic Receptor Locus in Obe- lism. Diabetes 2000, 49, 143–156.
sity and Noninsulin-Dependent Diabetes among Members of 210. Palou, A.; Picó, C.; Bonet, M.; Oliver, P. The Uncoupling Pro-
Caucasian Families with a Diabetic Sibling Pair. J. Clin. Endo- tein, Thermogenin. Int. J. Biochem. Cell. Biol. 1998, 30, 7–11.
crinol. Metab. 1996, 81, 4422–4427. 211. Samec, S.; Seydoux, J.; Dulloo, A. G. Interorgan Signalling
197. Gagnon, J.; Mauriege, P.; Roy, S.; Sjostrom, D.; Chagnon, Between Adipose Tissue Metabolism and Skeletal Muscle
Y. C.; Dionne, F. T.; Oppert, J. M.; Perusse, L.; Sjostrom, L.; Uncoupling Protein Homologs: Is There a Role for Circulat-
Bouchard, C. The Trp64Arg Mutation of the β3 Adrenergic ing Free Fatty Acids? Diabetes 1998, 47, 1693–1698.
Receptor Gene has no Effect on Obesity Phenotypes in the 212. Solanes, G.; Vidal-Puig, A.; Grujic, D.; Flier, J.; Lowell, B. B.
Québec Family Study and Swedish Obese Subjects Cohorts. The Human Uncouplin Protein-3 Gene. J. Biol. Chem. 1997,
J. Clin. Invest. 1996, 98 (9), 2086–2093. 272, 25433–25436.
198. Silver, K.; Walston, J.; Wang, Y.; Dowse, G.; Zimmet, P.; 213. Pecqueur, C.; Cassard-Doulcier, A. M.; Raimbault, S.;
Shuldiner, A. R. Molecular Scanning for Mutations in the ­Miroux, B.; Fleury, C.; Gelly, C.; Bouillaud, F.; Ricquier, D.
β3-Adrenergic Receptor Gene in Nauruans with Obesity and Functional Organization of the Human Uncoupling ­Protein-2
Noninsulin-Dependent Diabetes Mellitus. J. Clin. Endocri- Gene, and Juxtaposition to the Uncoupling ­Protein-3 Gene.
nol. Metab. 1996, 81, 4155–4158. Biochem. Biophys. Res. Commun. 1999, 255, 40–46.
199. Nagase, T.; Aoki, A.; Yamamoto, M.; Yasuda, H.; Kado, S.; 214. Schrauwen, P.; Xia, J.; Bogardus, C.; Pratley, R. E.; ­Ravussin,
Nishikawa, M.; Kugai, N.; Akatsu, T.; Nagata, N. Lack of Asso- E. Skeletal Muscle Uncoupling Protein 3 Expression Is a
ciation Between the Trp64Arg Mutation in the β3-Adrenergic Determinant of Energy Expenditure in Pima Indians. Diabe-
Receptor Gene and Obesity in Japanese Men: A Longitudinal tes 1999, 48, 146–149.
Analysis. J. Clin. Endocrinol. Metab. 1997, 82, 1284–1287. 215. Vidal-Puig, A.; Grujic, D.; Zhang, C. Y.; Hagen, T.; Boss, O.;
200. Oksanen, L.; Mustajoki, P.; Kaprio, J.; Kainulainen, K.; Ido, Y.; Szczepanik, A.; Wade, J.; Mootha, V.; Cortright, R.;
Janne, O.; Peoltonen, L.; Kontula, K. Polymorphism of the Muoio, D.; Lowell, B. B. Energy Metabolism in Uncou-
β3-Adrenergic Receptor Gene in Morbid Obesity. Int. J. pling Protein 3 Knockout Mice. J. Biol. Chem. 2000, 275,
Obes. 1996, 20, 1055–1061. 16258–16266.
201. O’Dell, S. D.; Bolla, M. K.; Miller, G. J.; Cooper, J. A.; 216. Urhammer, S.; Fridberg, M.; Sorensen, T.; Echwald, S. M.;
Humphries, S. E.; Day, I. N.M. W64R Mutation in β-3- Andersen, T.; Tybjaerg-Hansen, A.; Clausen, J. O.; Pedersen,
Adrenergic Receptor Gene and Weight in a Large Population O. Studies of Genetic Variability of the Uncoupling Protein 1
Sample. Int. J. Obes. 1997, 22, 377–379. Gene in Caucasian Subjects with Juvenile-Onset Obesity. J.
202. Cassard-Doulcier, A. M.; Bouillaud, F.; Chagnon, M.; Clin. Endocrinol. Metab. 1997, 82, 4069–4074.
Gelly, C.; Dionne, F. T.; Oppert, J. M.; Bouchard, C.; 217. Otabe, S.; Clement, K.; Rich, N.; Warden, C.; Pecqueur, C.;
Chagnon, Y.; Ricquier, D. The Bcl I Polymorphism of the Neverova, M.; Raimbault, S.; Guy-Grand, B.; Basdevant, A.;
Human Uncoupling Protein (UCP) Gene Is Due to a Point Ricquier, D., et al. Mutation Screening of the Human UCP-2
Mutation in the 5′-Flanking Region. Int. J. Obes. 1994, 18, Gene in Normoglycemic and NIDDM Morbidly Obese
526–531. Patients: Lack of Association Between New UCP-2 Polymor-
203. Fumeron, F.; Durack-Brown, I.; Betoulle, D.; Cassard- phisms and Obesity in French Caucasians. Diabetes 1998,
Doulcier, A. M.; Tuzet, S.; Bouillaud, F.; Melchior, J. C.; 47, 840–842.
Ricquier, D.; Apfelbaum, M. Polymorphisms of Uncoupling 218. Argyropoulos, G.; Brown, A.; Peterson, R.; Likes, C.;
Protein (UCP) and β3 Adrenoreceptor Genes in Obese People ­Watson, D.; Garvey, W. Structure and Organization of
Submitted to a Low Calorie Diet. Int. J. Obes. 1996, 20, the Human Uncoupling Protein 2 Gene and Identification
1051–1054. of a Common Biallelic Variant in Caucasian and African-­
204. Clement, K.; Ruiz, J.; Cassard-Doulcier, A. M.; Bouillaud, F.; American Subjects. Diabetes 1998, 47, 685–687.
Ricquier, D.; Basdevant, A.; Guy-Grand, B.; Froguel, P. Addi- 219. Yanovski, J. A.; Diament, A.; Sovik, K.; Nguyen, T.; Li, H.;
tive Effect of A to G (-3826) Variant of the Uncoupling Pro- Sebring, N.; Warden, C. Associations Between Uncoupling
tein Gene and the Trp64Arg Mutation of the β3-Adrenergic Protein 2, Body Composition, and Resting Energy Expendi-
Receptor Gene on Weight Gain in Morbid Obesity. Int. J. ture in Lean and Obese African American, White, and Asian
Obes. 1996, 20, 1062–1066. Children. Am. J. Clin. Nutr. 2000, 71, 1405–1412.
205. Viguerie-Bascands, N.; Bousquet-Melou, A.; Galitzky, J.; 220. Esterbauer, H.; Schneitler, C.; Oberkofler, H.; Ebenbichler, C.;
Larrouy, D.; Ricquier, D.; Berlan, M.; Casteilla, L. Evi- Paulweber, B.; Sandhofer, F.; Ladurner, G.; Hell, E.; Strosberg,
dence for Numerous Brown Adipocytes Lacking Functional A. D.; Patsch, J. R., et  al. A Common Polymorphism in the
β3-Adrenoceptors in Fat Pads from Nonhuman Primates. J. Promoter of UCP2 Is Associated with Decreased Risk of Obe-
Clin. Endocrinol. Metab. 1996, 81, 368–375. sity in Middle-Aged Humans. Nat. Genet. 2001, 28, 178–183.
30 CHAPTER 90  Disorders of the Body Mass

221. Kimm, S. Y.; Glynn, N. W.; Aston, C. E.; Damcott, C. M.; 239. Bray, M.; Boerwinkle, E.; Hanis, C. L. Linkage Analysis of
Poehlman, E. T.; Daniels, S. R.; Ferrell, R. E. Racial Differ- Candidate Obesity Genes Among the Mexican-American
ences in the Relation Between Uncoupling Protein Genes and Population of Starr County, Texas. Genet. Epidemiol. 2000,
Resting Energy Expenditure. Am. J. Clin. Nutr. 2002, 75 (4), 16, 397–411.
714–719. 240. Roche, F.; Boutin, P.; Dina, C.; Gyapay, G.; Basdevant, A.;
222. Schonfeld-Warden, N. A. Warden CH Physiological Effects Hager, J.; Guy-Grand, B.; Clement, K. Frogu Genetic Stud-
of Variants in Human Uncoupling Proteins: UCP2 Influ- ies of Neuropeptide Y and Neuropeptide Y Receptors Y1
ences Body-Mass Index. Biochem. Soc. Trans. 2001, 29, and Y5 Regions in Morbid Obesity. Diabetologia 1997, 40,
777–784. 671–675.
223. Uehara, Y.; Shimizu, H.; Ohtani, K.; Sato, N.; Mori, M. 241. Herzog, H.; Darby, K.; Ball, H.; Hort, Y.; Beck-Sickinger, A.;
Hypothalamic Corticotropin-releasing Hormone Is a Media- Shine, J. Overlapping Gene Structure of the Human Neuro-
tor of the Anorexigenic Effect of Leptin. Diabetes 1998, 47, peptide Y Receptor Subtypes Y1 and Y5 Suggests Coordinate
890–893. Transcriptional Regulation. Genomics 1997, 41, 315–319.
224. Bell, C. G.; Walley, A. J.; Froguel, P. The Genetics of Human 242. Rosenkranz, K.; Hinney, A.; Ziegler, A.; von Prittwitz, S.;
Obesity. Nat. Rev. Genet. 2005, 6, 221–234. Barth, N.; Roth, H.; Mayer, H.; Siegfried, W.; Lehmkuhl, G.;
225. Korner, J.; Leibel, R. L. To Eat Ornot to Eat—How the Gut Poutska, F., et al. Screening for Mutations in the Neuropep-
Talks to the Brain. N. Engl. J. Med. 2003, 349, 926–928. tide Y Y5 Receptor Gene in Cohorts Belonging to Different
226. Horvath, T. L.; Diano, S.; Sotonyi, P.; Heiman, M.; Tschop, Weight Extremes. Int. J. Obes. 1998, 22, 157–163.
M. Minireview: Ghrelin and the Regulation of Energy 243. Sakuri, T.; Ameniya, A.; Ishii, M.; Matsuzaki, I.;
­Balance—A Hypothalamic Perspective. Endocrinology Chemelli, R. M.; Tanaka, H.; Williams, S. C.; Richardson, J.
2001, 142 (10), 4163–4169. A.; Kozlowski, G. P.; Wilson, S., et al. Orexins and Orexin
227. Haqq, A. M.; Farooqi, I. S.; O’Rahilly, S.; Stadler, D. D.; Receptors: A Family of Hypothalamic Neuropeptides and G
Rosenfeld, R. G.; Pratt, K. L.; LaFranchi, S. H.; Purnell, J. Protein-Coupled Receptors that Regulate Feeding Behavior.
Q. Serum Ghrelin Levels Are Inversely Correlated with Body Cell 1998, 92, 573–585.
Mass Index, Age, and Insulin Concentrations in Normal 244. Chemelli, R. M.; Willie, J.; Sinton, C.; Elmquist, J.;
Children and are Markedly Increased in Prader–Willi Syn- Scammell, T.; Lee, C.; Richardson, J. A.; Williams, S.; Xiong,
drome. J. Clin. Endocrinol. Metab. 2003, 88 (1), 174–178. Y.; Kisanuki, Y., et  al. Narcolepsy in Orexin Knock-Out
228. English, P. J.; Ghatei, M. A.; Malik, I. A.; Bloom, S. R.; Mice: Molecular Genetics of Sleep Regulation. Cell 1999,
­Wilding, J. P. H. Food Fails to Suppress Ghrelin Levels in 98, 437–451.
Obese Humans. J. Clin. Endocrinol. Metab. 2002, 87 (6), 245. Blanco, M.; Lopez, M.; GarcIa-Caballero, T.; Gallego, R.;
2984. Vazquez-Boquete, A.; Morel, G.; SenarIs, R.; Casanueva,
229. Sun, A. Deletion of Ghrelin Impairs neither Growth nor F.; Dieguez, C.; Beiras, A. Cellular Localization of Orexin
Appetite. Mol. Cell. Biol. 2003, 23 (22), 7973–7981. Receptors in Human Pituitary. J. Clin. Endocrinol. Metab.
230. Korbonits, M.; Gueorguiev, M.; O’Grady, E.; Lecoeur, C.; 2001, 86 (4), 1616–1619.
Swan, D. C.; Mein, C. A.; Weill, J.; Grossman, A. B.; Froguel, 246. Mazzocchi, G.; Malendowicz, L. K.; Aragona, F.; Rebuffat, P.;
P. A Variation in the Ghrelin Gene Increases Weight and Gottardo, L.; Nussdorfer, G. G. Human Pheochromocytomas
Decreases Insulin Secretion in Tall, Obese Children. J. Clin. Express Orexin Receptor Type 2 Gene and Display an in Vitro
Endocrinol. Metab. 2002, 87 (8), 4005–4008. Secretory Response to Orexins A and B. J. Clin. Endocrinol.
231. Wu, J. T.; Kral, J. G. Ghrelin: Integrative Neuroendocrine Metab. 2001, 86 (10), 4818–4821.
Paptide in Health and Disease. Ann. Surg. 2004, 239, 247. Plotsky, P. M.; Thrivikraman, K. V.; Watts, A. G.; Hauger,
464–474. R. L. Hypothalamic–Pituitary–Adrenal Axis Function in the
232. Schwartz, M. W.; Woods, S. C.; Porte, D., Jr.; Seeley, R. J.; Zucker Obese Rat. Endocrinology 1992, 130, 1931–1941.
Baskin, D. Central Nervous System Control of Food Intake. 248. Moreau, J.; Kilpatrick, G.; Jenck, F. Urocortin, a Novel Neu-
Nature 2000, 404, 661–671. ropeptide with Anxiogenic-Like Properties. NeuroReport
233. Sainsbury, A.; Cusin, I.; Rohner-Jeanrenaud, F.; Jeanrenaud, 1997, 8, 1697–1701.
B. Adrenalectomy Prevents the Obesity Syndrome Produced 249. Spina, M.; Merlo-Pich, E.; Chan, R.; Basso, A. M.; Rivier,
by Chronic Central Neuropeptide Y Infusion in Normal J.; Vale, W.; Koob, G. F. Appetite-Suppressing Effects of
Rats. Diabetes 1997, 46, 209–214. Urocortin, a CRF-related Neuropeptide. Science 1996, 273,
234. Erickson, J. C.; Hollopeter, G.; Palmiter, R. D. Attenuation 1561–1564.
of the Obesity Syndrome of ob/ob Mice by the Loss of Neu- 250. Rohner-Jeanrenaud, F.; Walker, C. D.; Greco-Perotto, R.;
ropeptide Y. Science 1996, 274, 1704–1707, (6 December). Jeanrenaud, B. Central Corticotropin-Releasing Factor Admin-
235. Erickson, J. C.; Ahima, R. S.; Hollopeter, G.; Flier, J.; istration Prevents the Excessive Body Weight Gain of Geneti-
­Palmiter, R. D. Endocrine Function of Neuropeptide Y cally Obese (fa/fa) Rats. Endocrinology 1989, 124, 733–739.
Knockout Mice. Regul. Pept. 1997, 70, 199–202. 251. Spina, M.; Merlo-Pich, E.; Chan, R. K. W.; Basso, A. M.;
236. Bannon, A.; Seda, J.; Carmouche, M.; Francis, J.; Norman, Rivier, J.; Vale, W.; Koob, G. F. Appetite-Suppressing Effects
M.; Karbon, B.; McCaleb, M. Behavioral Characterization of Urocortin, a CRF-related Neuropeptide. Science 1996,
of Neuropeptide Y Knockout Mice. Brain. Res. 2000, 868, 273, 1561–1564, (13 September).
79–87. 252. Heiman, M. L.; Ahima, R. S.; Craft, L. S.; Schoner, B.;
237. Kanatani, A.; Mashiko, S.; Murai, N.; Sugimoto, N.; Ito, J.; Stephens, T. W.; Flier, J. Leptin Inhibition of the
Fukuroda, T.; Fukami, T.; Morin, N.; MacNeil, D.; Van der ­Hypothalamic–Pituitary–Adrenal Axis in Response to
Ploeg, L., et  al. Role of the Y1 Receptor in the Reguation Stress. Endocrinology 1997, 138, 3859–3863.
of Neuropeptide Y-Mediated Feeding: Comparison of Wild- 253. Boston, B.; Blaydon, K. M.; Varnerin, J.; Cone, R. D.
type, Y1 Receptor-Deficient, and Y5 Receptor-Deficient ­Independent and Additive Effects of Central POMC and
Mice. Endocrinology 2000, 141, 1011–1016. Leptin Pathways on Murine Obesity. Science 1997, 278,
238. Antonijevic, I.; Murck, H.; Bohlhalter, S.; Frieboes, R.; Hols- 1641–1644.
boer, F.; Steiger, A. Neuropeptide Y Promotes Sleep and 254. Glasow, A.; Haidan, A.; Hilbers, U.; Breidert, M.; ­Gillespie, J.;
Inhibits ACTH and Cortisol Release in Young Men. Neuro- Scherbaum, W. A.; Bornstein, S. R.; Chrousos, G. P.
pharmacology 2000, 39, 1474–1481. Expression of Ob Receptor in Normal Human Adrenals:
CHAPTER 90  Disorders of the Body Mass 31

Differential Regulation of Adrenocortical and Adrenomedul- Obesity and the Metabolic Syndrome. J. Clin. Endocrinol.
lary Function by Leptin. J. Clin. Endocrinol. Metab. 1998, 83, Metab. 2001, 86 (9), 4321–4325.
4459–4466. 270. Levine, J. A.; Jensen, M.; Eberhardt, N. L.; O’Brien, T.
255. Huizenga, N.; Koper, J.; De Lange, P.; Pols, H.; Stolk, R.; ­Adipocyte Macrophage Colony-Stimulating Factor Is a
Burger, H.; Grobbee, D.; Brinkmann, A.; De Jong, F.; Lam- Mediator of Adipose Tissue Growth. J. Clin. Invest. 1998,
berts, S. A Polymorphism in the Glucocorticoid Receptor 101, 1557–1564.
Gene May Be Associated with an Increased Sensitivity to 271. Magre, J.; Laurell, H.; Fizames, C.; Antoinne, P. J.; Dib, C.;
Glucocorticoids In Vivo. J. Clin. Endocrinol. Metab. 1998, Vigouroux, C.; Bourot, C.; Capeau, J.; Weissenbach, J.;
83, 144–151. ­Langin, D. Human Hormone-Sensitive Lipase: Genetic
256. White, P. C.; Tomoatsu, M.; Agarwal, A. K. Mapping, Identification of a New Dinucleotide Repeat, and
11β-Hydroxysteroid Dehydrogenase and the Syndrome of Association with Obesity and NIDDM. Diabetes 1998, 47,
Apparent Mineralocorticoid Excess. Endocr. Rev. 1997, 18, 284–286.
135–156. 272. Spiegelman, B. M. PPAR-γ: Adipogenic Regulator and Thia-
257. Farese, R. V., Jr.; Biglieri, E. G.; Shackleton, C. H. L.; zolidinedione Receptor. Diabetes 1998, 47, 507–514.
Irony, I.; Gomez-Fontes, R. Licorice-Induced Hyperminer- 273. Adams, M.; Montague, C. T.; Prins, J. B.; Holder, J.;
alocorticoidism. N. Engl. J. Med. 1991, 325, 1223. Smith, S.; Sanders, L.; Digby, J. E.; Sewter, C. P.; Lazar,
258. Armanini D; Francini-Pesenti F; Battagin G; Fiore C; M.; Chatterjee, K., et al. Activators of Peroxisome Prolif-
Mariniello B; Sartorato P; Nacamulli D. Effect of a Cream erator–Activated Receptor γ have Depot-Specific Effects on
with Glycyrrhetinic Acid on the Reduction of Subcutaneous Human Preadipocyte Differentiation. J. Clin. Invest. 1997,
Fat Layer in Women. The Endocrine Society 85th Annual 100, 3149–3153.
Meeting, 583, 2003. Ref Type: Abstract. 274. Tontonoa, P.; Singer, S.; Forman, B.; Sarraf, P.; Fletcher, J.;
259. Armanini, D.; DePalo, C. B.; Mattarello, M. J.; Spinella, P.; Fletcher, C.; Brun, R.; Mueller, E.; Altiok, S.; Oppenheim, H.,
Zaccaria, M.; Ermolao, A.; Palermo, M.; Fiore, C.; Sartor- et al. Terminal Differentiation of Human Liposarcoma Cells
ato, P.; Francini-Pesenti, F., et  al. Effect of Licorice in the Induced by Ligands for Peroxisome ­Proliferator-Activated
Reduction of Body Fat Mass in Healthy Subjects. J. Endocri- Receptor γ and the Retionoid X Receptor. Proc. Natl. Acad.
nol. Invest. 2003, 26, 646–650. Sci. U.S.A. 1997, 94, 237–241.
260. Nordström, A.; Marcus, C.; Axelson, M.; Wedell, A.; 275. Kubota, N.; Terauchi, Y.; Miki, H.; Tamemoto, H.;
Ritzén, M. Failure of Conrtisone Acetate Treatment of ­Yamauchi, T.; Komeda, K.; Satoh, S.; Nakano, R.; Ishii, C.,
Congeital Adrenal Hyperplasia Because of Defective et al. PPAR Gamma Mediates High-fat Diet-Induced Adipo-
11β-Hydroxysteroid Dehydrogenase Reductase Activity. J. cyte Hypertrophy and Insulin Resistance. Mol. Cell. 1999, 4,
Clin. Endocrinol. Metab. 1999, 84, 1210–1213. 597–609.
261. Jamieson, A.; Wallace, A. M.; Andrew, R.; Nunez, B. S.; 276. Norris, A. W.; Chen, L.; Fisher, S. J.; Szanto, I.; Ristow, M.;
Walker, B. R.; Fraser, R.; White, P. C.; Connell, J. M. C. Jozsi, A. C.; Hirshman, M. F.; Rosen, E. D.; Goodyear, L.
Apparent Cortisone Reductase Deficiency: A Functional J.; Gonzalez, F. J., et  al. Muscle-Specific PPAR­{Gamma}-
Defect in 11β-Hydroxysteroid Dehydrogenase Type 1. J. Deficient Mice Develop Increased Adiposity and Insulin
Clin. Endocrinol. Metab. 1999, 84, 3570–3574. Resistance But Respond to Thiazolidinediones. J. Clin.
262. Masuzaki, H.; Paterson, J.; Shinyama, H.; Morton, N. M.; Invest. 2003, 112 (4), 608–618.
Mullins, J. J.; Seckl, J. R.; Flier, J. S. A Transgenic Model of 277. Vigouroux, C.; Fajas, L.; Khallouf, E.; Meier, M.; Gyapay, G.;
Visceral Obesity and the Metabolic Syndrome. Science 2001, Lascols, O.; Auwerx, J.; Weissenbach, J.; Capeau, J.; Magre,
294, 2166–2170. J. Human Peroxisome Proliferator-Activated Receptor-γ2:
263. Tiosano, D.; Einsentein, I.; Militianu, D.; Hrousos, G. P.; Genetic Mapping, Identification of a Variant in the Cod-
Hochberg, Z. 11β-Hydroxysteroid Dehydrogenase Activity ing Sequence, and Exclusion as the Gene Responsible for
in Hypothalamic Obesity. J. Clin. Endocrinol. Metab. 2003, Lipoatrophic Diabetes. Diabetes 1998, 47, 490–492.
88, 379–384. 278. Ristow, M.; Müller-Wieland, D.; Pfeiffer, A.; Krone, W.;
264. Rask, E.; Olsson, T.; Söderberg; Andrew, R.; Livingstone, D. Kahn, C. Obesity Associated with a Mutation in a Genetic
E.W.; Johnson, O.; Walker, B. R. Tissue-Specific Dysregu- Regulator of Adipocyte Differentiation. N. Engl. J. Med.
lation of Cortisol Metabolism in Human Obesity. J. Clin. 1998, 339, 953–959.
Endocrinol. Metab. 2003, 86, 1418–1421. 279. Hamann, A.; Munzberg, H.; Buttron, P.; Busing, B.;
265. Bujalska, I. J.; Kumar, S. Does Central Obesity Reflect Hinney, A.; Mayer, H.; Siegfried, W.; Hebebrand, J.;
“Cushing’s Disease of the Omentum?” Lancet 1997, 349, ­Greten, H. Missense Variants in the Human Peroxisome
1210–1215. ­Proliferator-Activated Receptor Gamma 2 Gene in Lean and
266. Draper, N.; Echwald, S. M.; Lavery, G. G.; Walker, E. A.; Obese Subjects. Eur. J. Endocrinol. 1999, 141, 90–92.
Fraser, R.; Davies, E.; Sørensen, T. I. A.; Astrup, A.; Adam- 280. Ek, J.; Urhammer, S.; Sorensen, T.; Andersen, T.;
ski, J.; Hewison, M., et  al. Association Studies Between Auwerx, J.; Pedersen, O. Homozygosity of the Pro12Ala
Microsatellite Markers within the Gene Encoding Human Variant of the Peroxisome Proliferative-Activated Receptor
11β-Hydroxysteroid Dehydrogenase Type 1 and Body Mass Gamma 2 (PPAR-γ2): Divergent Modulating Effects on Body
Index, Waist to Hip Ratio, and Glucocorticoid Metabolism. Mass Index in Obese and Lean Caucasian men. Diabetologia
J. Clin. Endocrinol. Metab. 2002, 87, 4984–4990. 1999, 42, 892–895.
267. Gelernter-Yaniv, L.; Feng, N.; Sebring, N. G.; Hochberg, Z.; 281. Beamer, B. A.; Yen, C. J.; Andersen, R. E.; Muller, D.;
Yanovski, J. A. Associations Between a Ploymorphism in the Elahi, D.; Cheskin, L. J.; Andres, R.; Roth, J.; Shuldiner, A.
11 Beta Hydorxysteroid Dehydrogenase Type 1 Gene and R. Association of the Pro12Ala Variant in the Peroxisome
Body Composition. Int. J. Obes. 2003, 27, 983–986. ­Proliferator-Activated Receptor-γ2 Gene with Obesity in
268. Diamond, F. The Endocrine Function of Adipose Tissue. Two Caucasian Populations. Diabetes 1998, 47, 1806–1808.
Growth Genet. Horm. 2002, 18, 17–22. 282. Meirhaeghe, A.; Fajas, L.; Helbecque, N.; Cottel, D.;
269. Comuzzie, A. G.; Funahashi, T.; Sonnenberg, G.; Martin, L. ­Auwerx, J.; Deeb, S.; Amouyel, P. Impact of the Peroxisome
J.; Jacob, H. J.; Black, A. E. K.; Maas, D.; Takahashi, M.; Proliferator-Activated Receptor Gamma 2 Pro12Ala Poly-
Kihara, S.; Tanaka, S., et  al. The Genetic Basis of Plasma morphism on Adiposity, Lipids, and Non-Insulin-Dependent
Variation in Adiponectin, a Global Endophenotype for Diabetes Mellitus. Int. J. Obes. 2000, 24, 195–199.
32 CHAPTER 90  Disorders of the Body Mass

283. Ringel, J.; Engeli, S.; Distler, A.; Sharma, A. Pro12Ala Mis- Expression of a Novel Adipose Specific Collagen-Like Fac-
sense Mutation of the Peroxisome Proliferator-Activated tor, apM1(Adipose Most Abundant Gene Transcript 1). Bio-
Receptor Gamma and Diabetes Mellitus. Biochem. Biophys. chem. Biophys. Res. Commun. 1996, 221, 286–289.
Res. Commun. 1999, 254, 450–453. 300. Silha, J. F.; Krsek, M.; Skrha, J. V.; Sucharda, P.; Nyomba,
284. Cole, S.; Mitchell, B. D.; Hsueh, W.; Pineda, P.; Beamer, B. B. L. G.; Murphy, L. J. Plasma Resistin, Adiponectin and
A.; Shuldiner, A. R. The Pro12Ala Variant of Peroxisome Leptin Levels in Lean and Obese Subjects: Correlations
­Proliferator-Activated Receptor-gamma 2 (PPAR-γ2) Is Asso- with Insulin Resistance. Eur. J. Endocrinol. 2003, 149 (4),
ciated with Measures of Obesity in Mexican Americans. Int. 331–335.
J. Obes. 2000, 24, 522–524. 301. Fasshauer, M.; Kralisch, S.; Klier, M.; Lossner, U.;
285. Hotamisligil, G. S.; Shargill, N. S.; Spiegelman, B. M. Adi- Bluher, M.; Klein, J.; Paschke, R. Adiponectin Gene Expres-
pose Expression of Tumor Necrosis Factor-α: Direct Role sion and Secretion Is Inhibited by Interleukin-6 in 3T3-L1
in Obesity-Linked Insulin Resistance. Science 1993, 259, Adipocytes. Biochem. Biophy. Res. Commun. 2003, 301
87–91, (1 January). (4), 1045–1050.
286. Liu, L.; Spelleken, M.; Röhrig, K.; Hauner, H.; Eckel, J. 302. Qi, Y.; Takahashi, N.; Hileman, S. M.; Patel, H. R.; Berg, A.
Tumor Necrosis Factor-α Inhibits Insulin Signalling in H.; Pajvani, U. B.; Scherer, P. E.; Ahima, R. S. Adiponectin
Human Adipocytes. Diabetes 1998, 47, 515–522. Acts in the Brain to Decrease Body Weight. Nat. Med. 2004,
287. Prins, J. B.; Niesler, C.; Winterford, C.; Bright, N.; 10 (5), 524–529.
Siddle, K.; O’Rahilly, S.; Walker, N.; Cameron, D. Tumor 303. Takahashi, M.; Arita, Y.; Yamagata, K.; Matsukawa, Y.;
Necrosis Factor-α Induces Apoptosis of Human Adipose Okutomi, K.; Horie, M.; Shimomura, I.; Hotta, K.; Kuri-
Cells. Diabetes 1997, 46, 1939–1944. yama, H.; Kihara, S., et  al. Genomic Structure and Muta-
288. Nisoli, E.; Briscini, L.; Tonello, C.; Degiulimorghen, C.; tions in Adipose-Specific Gene, Adiponectin. Int. J. Obes.
Carruba, M. Tumor Necrosis Factor-α Induces Apoptosis 2000, 24, 861–868.
in Rat Brown Adipocytes. Cell. Death. Differ. 1997, 4, 304. Schaffler, A.; Barth, N.; Palitzsch, K. D.; Drobnik, W.;
771–778. S­cholmerich, J.; Schmitz, G. Mutation Analysis of the
289. Kirchgessner, T.; Uysal, K.; Wiesbrock, S.; Marino, M.; Human Adipocyte-Specific apM-1 Gene. Eur. J. Clin. Invest.
Hotamisligil, G. S. Tumor Necrosis Factor-α Contributes 2000, 30 (10), 879–887.
to Obesity-Related Hyperleptinemia by Regulating Leptin 305. Ukkola, O.; Ravussin, E.; Jacobson, P.; Sjöström, L.;
Release from Adipocytes. J. Clin. Invest. 1997, 100, Bouchard, C. Mutations in the Adiponectin Gene in Lean
2777–2782. and Obese Subjects from the Swedish Obese Subjects Cohort.
290. Mantzoros, C. S.; Moschos, S.; Avramopoulos, I.; ­Kaklamani, Metab. Clin. Exp. 2003, 52 (7), 881–884.
V.; Liolios, A.; Dougerakis, D.; Griveas, I.; Katsilambros, 306. Poitou, C.; Lacorte, J.-M.; Coupaye, M.; Bertrais, S.; Bedel,
N.; Flier, J. Leptin Concentrations in Relation to Body Mass J.-F.; Lafon, N.; Bouillot, J.-L.; Galan, P.; Borson-Chazot, F.;
Index and the Tumor Necrosis Factor-α System in Humans. Basdevant, A., et al. Relationship Between Single Nucleotide
J. Clin. Endocrinol. Metab. 1997, 82, 3408–3413. Polymorphisms in Leptin, IL6 and Adiponectin Genes and
291. Zumbach, M.; Boehme, M.; Wahl, P.; Stremmel, W.; their Circulating Product in Morbidly Obese Subjects Before
Ziegler, R.; Nawroth, P. Tumor Necrosis Factor Increases and After Gastric Banding Surgery. Obes. Surg. 2005, 15,
Serum Leptin Levels in Humans. J. Endocrinol. Invest. 1997, 11–23.
82, 4080–4082. 307. Samal, B.; Sun, Y.; Stearns, G.; Xie, C.; Suggs, S.; McNiece, I.
292. Hotamisligil, G. S.; Johnson, R. S.; Distel, R. J.; Ellis, R.; Cloning and Characterization of the cDNA Incoding a Novel
Papaioannou, V. E.; Spiegelman, B. M. Uncoupling of Obe- Pre-B-Cell Colony-Enhancing Factor. Mol. Cell. Biol. 1994,
sity from Insulin Resistance through a Targeted Mutation in 14, 1431–1437.
aP2, the Adipocyte Fatty Acid Binding Protein. Science 1996, 308. Strowski, M. Z.; Li, Z.; Szalkowski, D.; Shen, X.; Guan, X.
274, 1377–1379, (22 November). M.; Juttner, S.; Moller, D. E.; Zhang, B. B. Small-Molecule
293. Ventre, J.; Doebbler, T.; Wu, M.; MacNaul, K.; Stevens, K.; Insulin Mimetic Reduces Hyperglycemia and Obesity in a
Pasparakis, M.; Kollias, G.; Moller, D. Targeted Disruption Nongenetic Mouse Model of Type 2 Diabetes. Endocrinol-
of the Tumor Necrosis Factor-α Gene. Diabetes 1997, 46, ogy 2004, 145 (11), 5259–5268.
1526–1531. 309. Fukuhara, A.; Matsuda, M.; Nishizawa, M.; Segawa,
294. Scheyer, S.; Chua, S.; Leboeuf, R. Obesity and Diabetes in K.; Tanaka, M.; Kishimoto, K.; Matsuki, Y.; Murakami,
TNF-Alpha Receptor-Deficient Mice. J. Clin. Invest. 1998, M.; Ichisaka, T.; Murakami, H., et  al. Visfatin: A Protein
102, 402–411. Secreted by Visceral Fat That Mimics the Effects of Insulin.
295. Carroll, M. C.; Katzman, P.; Alicot, E.; Koller, B.; Geraghty, Science 2005, 307 (5708), 426–430.
D.; Orr, H.; Strominger, J. L.; Spies, T. Linkage Map of the 310. Air, E. L.; Strowski, M. Z.; Benoit, S. C.; Conarello, S. L.;
Major Histocompatibility Complex Including the Tumor Salitro, G. M.; Guan, X. -M.; Liu, K.; Woods, S. C.; Zhang,
Necrosis Factor Genes. Proc. Natl. Acad. Sci. U.S.A. 1987, 84, B. B. Small Molecule Insulin Mimetics Reduce Food Intake
8535–8539. and Body Weight and Prevent Development of Obesity. Nat.
296. Norman, R. A.; Bogardus, C.; Ravussin, E. Linkage Between Med. 2002, 8, 179–183.
Obesity and a Marker Near the Tumor Necrosis Factor-α 311. Warden, C. H.; Fisler, J. S.; Pace, M. J.; Svenson, K. L.;
Locus in Pima Indians. J. Clin. Invest. 1995, 96, 158–162. Lusis, A. J. Coincidence of Genetic Loci for Plasma Choles-
297. Fernandez-Real, J. M.; Gutierrez, C.; Ricart, W.; ­Casamitjana, terol Levels and Obesity in a Multifactorial Mouse Model. J.
R.; Fernandez-Castaner, M.; Vendrell, J.; Richart, C.; Clin. Invest. 1993, 92, 773–779.
Soler, J. The TNF-α Gene Nco I Polymorphism Influences 312. Warden, C. H.; Fisler, J. S.; Shoemaker, S. M.; Wen, P.-Z.;
the Relationship Among Insulin Resistance, Percent Body Svenson, K. L. Identification of Four Chromosomal Loci
Fat, and Increased Serum Leptin Levels. Diabetes 1998, 46, Determining Obesity in a Multifactorial Mouse Model. J.
1468–1472. Clin. Invest. 1995, 95, 1545–1552.
298. Berg, A. H.; Combs, T. P.; Scherer, P. E. ACRP30/Adiponec- 313. Taylor, B. A.; Phillips, S. J. Obesity QTLs on Mouse Chro-
tin: An Adipokine Regulating Glucose and Lipid Metabo- mosomes 2 and 17. Genomics 1997, 43, 249–257.
lism. Trends. Endocrinol. Metab. 2002, 13, 84–89. 314. York, B.; Kailian, L.; West, D. B. Inherited Non-Autosomal
299. Maeda, K.; Okubo, K.; Shimomura, I.; Funahashi, T.; Effects on Body Fat in F2 Mice Derived from an AKR/J ×
­Matsuzawa, Y.; Matsubara, K. cDNA Cloning and SWR/J Cross. Mamm. Genome. 1997, 8, 726–730.
CHAPTER 90  Disorders of the Body Mass 33

315. West, D. B.; Goudey-Lefevre, J.; York, B.; Truett, G. E. 331. Farooqi, I. S.; Matarese, G.; Lord, G. M.; Keogh, J. M.;
Dietary Obesity Linked to Genetic Loci on Chromosomes 9 Lawrence, E.; Agwu, C.; Sanna, V., et al. Beneficial Effects
and 15 in a Polygenic Mouse Model. J Clin Invest 1994, 94, of Letpin on Obesity, T Cell Hyporesponsiveness, and Neu-
1410–1416. roendocrine/Metabolic Dysfunction of Human Congenital
316. Frederich, R. C.; Hamann, A.; Anderson, S.; Lollmann, B.; Leptin Deficiency. J. Clin. Invest. 2002, 110, 1093–1103.
Lowell, B. B.; Flier, J. S. Leptin Levels Reflect Body Lipid 332. Licinio, J.; Caglayan, S.; Ozata, M.; Yildiz, B. O.;
Content in Mice: Evidence for Diet-Induced Resistance to de Miranda, P. B.; O’Kirwan, F., et al. Phenotypic Effects of
Leptin Action. Nat. Med. 1995, 1, 1311–1314. Leptin Replacement on Morbid Obesity, Diabetes Mellitus,
317. Chung, W. K.; Zheng, M.; Chua, M.; Kershaw, E.; Hypogonadism, and Behavior in Leptin-Deficient Adults.
­Power-Kehoe, E.; Tsuji, M.; Wupeng, X. S.; Williams, J.; Proc. Natl. Acad. Sci. U.S.A. 2004, 101, 4531–4536.
Chua, S. C.; Leibel, R. L. Genetic Modifiers of Lepr (fa) 333. Fischer-Posovoszky, P.; von Schnurbein, J.; Moepps, B.;
Associated with Variability in Insulin Production and Lahr, G.; Strauss, G.; Barth, T. F., et  al. A New Missense
­Susceptibility to NIDDM. Genomics 1997, 41, 332–344. Mutation in the Leptin Gene Causes Mild Obesity and
318. Meirhaeghe, A.; Heibecque, N.; Cottel, D.; Amouyel, P. Hypogonadism without Affecting T Cell Responsiveness. J.
β2-Adrenoreceptor Gene Polymorphism, Body Weight, and Clin. Endocrinol. Metab. 2010, 95 (6), 2836–2840.
Physical Activity. Lancet 1999, 353, 896. 334. Farooqi, I. S.; Wangensteen, T.; Collins, S.; Kimber, W.;
319. Donohoue, P. A.; Mendoza, M. B.; Collins, M. M.; Matarese, G.; Keogh, J. M.; Lank, E.; Bottomley, B.;
Hamilton, W. L.; Burns, T. L. The Association of Geno- Lopez-Fernandez, J., et  al. Clinical and Molecular Genetic
type, Physical Activity, and their Interaction on Age/Gender Spectrum of Congenital Deficiency of the Leptin Receptor.
Adjusted Body Mass Index (BMI): The Muscatine Study. N. Engl. J. Med. 2007, 96 (5), 237–247.
Pediatr. Res. 2001, 49. 335. Santini, F.; Maffei, M.; Pelosini, C.; Salvetti, G.; Scartabelli, G.;
320. Garenc, C.; Perusse, L.; Bergeron, J.; Gagnon, J.; Chagnon, Pinchera, A. Melanocortin-4 Receptor Mutations in Obesity.
Y. C.; Borecki, I. B.; Leon, A. S.; Skinner, J. S.; Wilmore, J. Am. J. Clin. Nutr. 2009, 49, 95–109.
H.; Rao, D. C., et al. Evidence of LPL Gene-Exercise Interac- 336. Miraglia Del Giudice, E.; Cirillo, G.; Nigro, V.; Santoro, N.;
tion for Body Fat and LPL Activity: The HERITAGE Family D’Urso, L.; Raimondo, P.; Cozzolino, D.; Scafato, D. Per-
Study. J. Appl. Phys. 2001, 91 (3), 1334–1340. rone L Low Frequency of Melanocortin-4 Receptor (MC4R)
321. Lowell, BB.; Bachman, E. S. β-Adrenergic Receptors, Diet- Mutations in a Mediterranean Population with Early Onset
Induced Thermogenesis, and Obesity. J. Biol. Chem. 2003, Obesity. Int. J. Obes. Relat. Metab. Disord. 2002, 26 (5),
278, 29385–29388. 647–651.
322. Perusse, B. Gene–Diet Interactions in Obesity. Am. J. Clin. 337. Nowacka-Woszuk, J.; Cieslak, J.; Skowronska, B.;
Nutr. 2000, 72 (5), 1285S–1290S. ­Majewska, K. A.; Stankiewicz, W.; Fichna, P.; Switonski, M.
323. De Luca, M.; Chambers, M. M.; Casazza, K.; Lok, K. H.; Missense Mutations and Polymorhisms of the MC4R Gene
Hunter, G. R.; Gower, B. A.; Fernández, J. R. Genetic Varia- in Polish Obese Children and Adolescents in Relation to the
tion in a Member of the Laminin Gene Family Affects Varia- Relative Body Mass Index. J. Appl. Genet. 2011, 52 (3),
tion in Body Composition in Drosophila and Humans. BMC. 319–323.
Genet. 2008, 11 (9), 52. 338. Mendiratta, M. S.; Yang, Y.; Balazs, A. E.; Willis, A. S.;
324. De Luca, M.; Crocco, P.; Wiener, H.; Tiwari, H. K.; Eng, C. M.; Karaviti, L. P.; Potocki, L. C. Early Onset Obe-
­Passarino, G.; Rose, G. Association of a Common LAMA5 sity and Adrenal Insufficiency Associated with a Homozygous
Variant with Anthropometric and Metabolic Traits in an POMC Mutation. Int. J. Pediatr. Endocrinol. 2011, 5 (1).
Italian Cohort of Healthy Elderly Subjects. Exp. Gerentol. 339. Clément, K.; Dubern, B.; Mencarelli, M.; Czernichow, P.;
2011, 46 (1), 60–64. Ito, S.; Wakamatsu, K.; Barsh, G. S.; Vaisse, C.; Leger, J.
325. De Luca, M.; Klimentidis, Y. C.; Casazza, K.; Chambers, M. Unexpected Endocrine Features and Normal Pigmentation
M.; Cho, R.; Harbison, S. T.; Jumbo-Lucioni, P.; Zhang, S.; in a Young Adult Patient Carrying a Novel Homozygous
Leips, J.; Fernández, J. R. A Conserved Role for Syndecan Mutation in the POMC Gene. J. Clin. Endocrinol. Metab.
Family Members in the Regulation of Whole-Body Energy 2008, 93 (12), 4955–4962.
Metabolism. PLoS One 2010, 5 (6), e11286. 340. Jackson, R. S.; Creemers, J. W.; Farooqi, I. S.; Raffin-Sanson,
326. Blechman, J.; Amir-Zilberstein, L.; Gutnick, A.; Ben-Dor, S.; M. L.; Varro, A.; Dockray, G. J.; Holst, J. J.; Brubaker, P. L.,
Levkowitz, G. The Metabolic Regulator PGC-1 Alpha et  al. Small-Intestinal Dysfunction Accompanies the Com-
Directly Controls the Expression of the Hypothalamic plex Endocrinopathy of Human Proprotein Convertase 1
­Neuropeptide Oxytocin. J. Neurosci. 2011, 31 (42), Deficiency. J. Clin. Invest. 2003, 112 (10), 1550–1560.
14835–14840. 341. Douzgou, S.; Petersen, M. B. Clinical Variability of Genetic Iso-
327. Jones, K. T.; Ashrafi, K. Caenorhabditis Elegans as an lates of Cohen Syndrome. Clin. Genet. 2011, 79 (6), 501–506.
Emerging Model for Studying the Basic Biology of Obesity. 342. Marshall, J. D.; Maffei, P.; Collin, G. B.; Naggert, J. K.
Dis. Model Mech. 2011, 2 (5–6), 224–229. Alström Syndrome: Genetics and Clinical Overview. Curr.
328. Ashrafi, K.; Chang, F. Y.; Watts, J. L.; Fraser, A. G.; Kamath, Genomics. 2011, 12 (3), 225–235.
R. S.; Ahringer, J.; Ruvkun, G. Genome-Wide RNAi Analy- 343. Girard, D.; Petrovsky, N. Alström Syndrome: Insights into
sis of Caenorhabditis Elegans Fat Regulatory Genes. Nature the Pathogenesis of Metabolic Disorders. Nat. Rev. Endocri-
2003, 421 (6920), 268–272. nol. 2011, 7 (2), 77–88.
329. Birkenfeld, A. L.; Lee, H. Y.; Guebre-Egziabher, F.; Alves, 344. Wang, J. C.; Turner, L.; Lomax, B.; Eydoux, P. A 5-Mb
T. C.; Jurczak, M. J.; Momauvaz, F. R., et  al. Deletion of Microdeletion at 6q16.1-q16.3 with SIM Gene Deletion and
the Mammalian INDY Homolog Mimics Aspects of Dietary Obesity. Am. J. Med. Genet. 2008, 146A (22), 2975–2978.
Restriction and Protects Against Adiposity and Insulin Resis- 345. Gray, J.; Yeo, G.; Hung, C.; Keogh, J.; Clayton, P.; Baner-
tance in Mice. Cell. Metab. 2011, 14 (2), 184–195. jee, K.; McAulay, A.; O’Rahilly, S.; Farooqi, I. S. Functional
330. Gibson, T.; Farooqi, I. S.; Moreau, M.; DePoli, A. M.; Charaterization of Human NTRK2 Mutations Identified in
Lwrence, E.; O’Rahilly, S.; Trussell, R. A. Congenital Patients with Severe Early-Onset Obesity. Int. J. Obes. 2007,
Leptin Deficiency Due to Homozygosity for the DELTA 31 (2), 359–364.
133 G Mutation: Report of Another Case and Evaluation of 346. Buiting, K. Prader–Willi Syndrome and Angelman Syndrome.
Response to Four Years of Leptin Therapy. J. Clin. Endocri- Am. J. Med. Genet. C Semin. Med. Genet. 2010, 154C (3),
nol. Metab. 2004, 89 (10), 4821–4826. 365–376.
34 CHAPTER 90  Disorders of the Body Mass

347. Cassidy, S. B.; Driscoll, D. J. Prader–Willi Syndrome. Eur. J. 363. Deeb, S. S.; Fajas, L.; Nemoto, M.; Pihlajamäki, J.;
Hum. Genet. 2009, 17, 3–13. ­Mykkänen, L.; Kuusisto, J.; Laakso, M.; Fujimoto, W.;
348. Butler, M. G. Prader–Willi Syndrome: Obesity Due to Auwerx, J. A. Pro12Ala Substitution in PPARgamma2 Asso-
Genomic Imprinting. Curr. Genomics. 2011, 12 (3), 204–215. ciated with Decreased Receptor Activity, Lower Body Mass
349. Duker, A. L.; Balllif, B. C.; Bawle, E. V.; Person, R. E.; Index and Improved Insulin Sensitivity. Nat. Genet. 1998,
Mahadevan, S.; Alliman, S.; Thompson, R.; Traylor, 20 (3), 284–287.
R.; Bejjani, B. A.; Shaffer, L. G., et  al. Paternally Inher- 364. Lei, H. H.; Chen, M. H.; Yang, W. S.; Chiu, M. C.;
ited Microdeletion at 15q11.2 Confirms a Significant Chen, M. C.; Tai, T. Y.; Chuang, L. M. Peroxisome
Role for the SNORD116 C/D box snoRNA Cluster in ­Proliferator-Activated Receptor Gamma 2 Pro12Ala Gene
Prader–Willi Syndrome. Eur. J. Hum. Genet. 2010, 18 (11), Variant Is Strongly Associated with Larger Body Mass in the
1196–1201. Taiwanese. Metabolism 2000, 49 (10), 1267–1270.
350. Janssen, S.; Ramaswami, G.; Davis, E. E.; Hurd, T.; 365. Kawasaki, I.; Tahara, H.; Emoto, M.; Shoji, T.; Okuno,
Airik, R.; Kasanuki, J. M.; Van Der Kraak, L.; Allen, S. Y.; Inaba, M.; Nishizawa, Y. Impact of Pro12Ala Variant
J.; Beales, B. A.; Katsanis, N., et al. Mutation Analysis in in the Peroxisome Proliferative-Activated Receptor (PPAR)
Bardet–Biedl Syndrome by DNA Pooling and Massively Gamma2 on Obesity and Insulin Resistance in Japanese Type
Parallel Resequencing in 105 Individuals. J. Appl. Genet. 2 Diabetic and Healthy Subjects. Osaka City Med. 2002, 48
2011, 129 (1), 79–90. (1), 23–28.
351. Muller, J.; Stoetzel, C.; Vincent, M. C.; Leitch, C. C.; 366. Ishiyama-Shigemoto, S.; Yamada, K.; Yuan, X.; Ichikawa,
Laurier, V.; Danse, J. M.; Hellé, S.; Marion, V.; Bennouna- F.; Nonaka, K. Association of Polymorphisms in the Beta2-
Greene, V., et al. Identification of 28 Novel Mutations in the Adrenergic Receptor Gene with Obesity, Hypertriglyceri-
Bardet–Biedl Syndrome Genes: The Burden of Private Muta- demia, and Diabetes Mellitus. Diabetologia 1999, 42 (1),
tions in an Extensively Heterogeneous Disease. Hum. Genet. 98–101.
2010, 127 (5), 583–593. 367. Masuo, K.; Katsuya, T.; Kawaguchi, H.; Fu, Y.;
352. Aliferis, K.; Hellé, S.; Gyapay, G.; Duchatelet, S.; Stoetzel, Rakugi, H.; Ogihara, T.; Tuck, M. L. Beta2-Adrenoceptor
C.; Mandel, J. L.; Dollfus, H. Differentiating Alström from Polymorphisms Relate to Obesity through Blunted Leptin-
Bardet–Biedl Syndrome (BBS) Using Systematic Ciliopathy Medicated Sympathetic Activation. Am. J. Hypertens. 2006,
Genes Sequencing. Ophthalmic. Genet. Oct 17, 2011, e pub 19 (10), 1084–1091.
ahead of print. 368. Masuo, K.; Katsuya, T.; Fu, Y.; Rakugi, H.; Ogihara, T.;
353. Sapp, J. C.; Nishimura, D.; Johnston, J. J.; Stone, E. M.; Tuck, M. L. Beta2- and Beta3-Adrenergic Receptor Polymor-
Heon, E.; Sheffield, V. C.; Biesecker, L. G. Recurrence Risks phisms Are Related to the Onset of Weight Gain and Blood
for Bardet–Biedl Syndrome: Implications of Locus Heteroge- Pressure Elevation Over 5 Years. Circulation 2005, 111 (25),
neity. Genet. Med. 2010, 12 (10), 623–627. 3429–3434.
354. Han, J. C.; Liu, Q. R.; Jones, M.; Levinn, R. L.; 369. Santos, J. L.; Pérez-Bravo, F.; Martinez, J. A.; Montalvo,
Menzie, C. M.; Jefferson-George, K. S.; Adler-Wailles, D. D.; Albala, C.; Carrasco, E. No Evidence for an Association
C., et  al. Brain-Derived Neurotrophic Factor and Obesity Between Genetic Polymorphisms of Beta(2)- and Beta(3)-
in the WAGR Syndrome. N. Engl. J. Med. 2008, 359 (9), Adrenergic Receptor Genes with Body Mass Index in Aymara
918–927. Natives from Chile. Nutrition 2002, 18 (3), 255–258.
355. Bochukova, E. G.; Huang, N.; Keogh, J.; Henning, E.; 370. Lowe, W. L., Jr.; Rotimi, C.; Luke, A.; Guo, X.; Zhu, X.;
­Purmann, C.; Blaszczyk, K.; Saeed, S.; Hamilton-Shield, J.; Comuzzie, A. G.; Schuh, T. S.; Halback, S.; Kotlar, T. J.;
Clayton-Smith, J.; O’Rahilly, S., et al. Large, Rare Chromo- Cooper, R. S. The Beta 3-Adrenergic Receptor Gene and
somal Deletions Associated with Severe Early-Onset Obesity. Opbesity in a Population Sample of African Americans. Int.
Nature 2010, 463 (7281), 666–670. J. Obes. Relat. Metab. Disord. 2001, 25 (1), 54–60.
356. Kissebah, A. H.; Sonnenberg, G. E.; Myklebust, J.; ­Goldstein, 371. Fogelholm, M.; Valve, R.; Kukkonen-Harjula, K.; Nenonen, A.;
M.; Broman, K., et  al. Quantitative Trait Loci on Chro- Hakkarainen, V.; Laakso, M.; Uusitupa, M. Additive Effects of
mosomes 3 and 17 Influence Phenotypes of the Metabolic the Mutations in the {beta}3-Adrenergic Receptor and Uncou-
Syndrome. Proc. Natl. Acad. Sci. U.S.A. 2000, 97 (26), pling Protein-1 Genes on Weight Loss and Weight Maintenance
14478–14483. in Finnish Women. J. Clin. Endocrinol. Metab. 1998, 83 (12),
357. Frayling, T. M.; Timpson, N. J.; Weedon, M. N., et  al. A 4246–4250.
Common Variant in the FTO Gene Is Associated with Body 372. Sivenius, V. L. N. L. U. Synergistic Effect of Polymorphisms
Mass Index and Predisposes to Childhood and Adult Obe- in Uncoupling Protein 1 and Beta3-Adrenergic Receptor
sity. Science 2007, 316, 889–894. Genes on Long-Term Body Weight Change in Finnish Type
358. Tung, Y. C.; Yeo, G. S. From GWAS to Biology: Lessons 2 Diabetic and Non-Diabetic Control Subjects. Int. J. Obes.
from FTO. Ann. NY Acad. Sci. 2011, 1220, 162–171. 2000, 24 (4), 514–519.
359. Van d Hoeven, F.; Schimmang, T.; Volkmann, A., et al. Pro- 373. Kring, S. I.; Holst, C.; Toubro, S.; Astrup, A.; Hansen, T.;
grammed Cell Death is Affected in the Novel Mouse Mutant Pedersen, O.; Sørensen, T. I. Common Variants Near MC4R
Fused Toes (Ft). Development 1994, 120, 2601–2607. in Relation to Body Fat, Body Fat Distribution, Metabolic
360. Church, C.; Lee, S.; Bagg, E. A., et al. A Mouse Model for Traits and Energy Expenditure. Int. J. Obes. 2010, 34 (1),
the Metabolic Effects of the Human Fat Mass and Obesity 182–189.
Associated FTO Gene. PLoS Genet. 2009, 5, e1000599. 374. Loos, R. J.; Lindgren, C. M.; Li, S., et al. Common Variants
361. Tung, Y. C.; Ayuso, E.; Shan, X., et  al. Hypothalamic- Near MC4R Are Associated with Fat Mass, Weigth and Risk
Specific Manipulation of Fto, the Ortholog of the Human of Obesity. Nat. Genet. 2008, 40 (6), 768–775.
Obesity Gene FTO, Affects Food Intake in Rats. PLoS One 375. Herrera, B. M.; Keildson, S.; Lindgren, C. M. Genetics and
2010, 5, e8771. Epigenetics of Obesity. Maturitas 2011, 69 (1), 41–49.
362. Jia, G.; Yang, G. G.; Yang, S., et al. Oxidative Demthylation 376. Ichihara, S.; Yamada, Y. Genetic Factors for Human Obe-
of 3-Methylthymine and 3-Methyluracil in Single-Stranded sity. Cell. Mol. Life. Sci. 2008, 65 (7–8), 1086–1098.
DNA and RNA by Mouse and Human FTO. FEBS. Lett. 377. Clarke, W. R.; Schrott, H. G.; Leaverton, P. E.; Connor, W. E.;
2008, 582, 3313–3319. Lauer, R. M. Tracking of Blood Lipids and Blood Pressures in
CHAPTER 90  Disorders of the Body Mass 35

School Age Children: The Muscatine Study. Circulation 1978, and Obesity: Cross Sectional Study. Br. Med. J. 1999, 319,
58, 626–634, (October). 147–150.
378. He, Q.; Karlberg, J. Prediction of Adult Overweight During 397. Denke, M. A. Weight Loss as an Endpoint: Measuring the
the Pediatric Years. Pediatr. Res. 1999, 46, 697–703. Effectiveness of Treatment in Obesity. Endocrinologist
379. Mossberg, H.-O. 40-Year Follow-Up of Overweight Chil- 1996, 6, 375–383.
dren. Lancet 1989, 491–493, August 26, 1989. 398. Abenhaim, L.; Moride, Y.; Brenot, F.; Rich, S.; Benichou, J.;
380. Must, A.; Jacques, P. F.; Dallal, G. E.; Bajema, C. J.; Kurz, X.; Higenbottam, T.; Oakley, C.; Wouters, E.; Aubier,
Dietz, W. H. Long-Term Morbidity and Mortality of M., et  al. Appetite-Suppressant Drugs and the Risk of Pri-
Overweight Adolescents. N. Engl. J. Med. 1992, 327, mary Pulmonary Hypertension. N. Engl. J. Med. 1996, 335,
1350–1355. 609–616.
381. Whitaker, R. C.; Pepe, M. S.; Wright, J. A.; Seidel, K. D.; 399. Heymsfield, S. B.; Greenberg, A. S.; Fujioka, K.; Dixon, R.
Dietz, W. H. Early Adiposity Rebound and the Risk of Adult M.; Kushner, R.; Hunt, T.; Lubina, J. A.; Patane, J.; Self,
Obesity. Pediatrics 1998, 101 (3), e5. B.; Hunt, P., et al. Recombinant Leptin for Weight Loss in
382. Dorosty, A. R.; Emmett, P. M.; Reilly, J. J. The ALSPAC Obese and Lean Adults. J. Am. Med. Assoc. 1999, 282 (16),
Study Team Factors Associated with Early Adiposity 1568–1575.
Rebound. Pediatrics 2000, 105 (5), 1115–1118. 400. Farooqi, S.; Jebb, S. A.; Langmack, G.; Lawrence, E.;
383. Morrison, J. A.; Sprecher, D. L.; Barton, B. A.; Waclawiw, Cheetham, C. H.; Prentice, A. M.; Hughes, I. A.; McCamish,
M. A.; Daniels, S. R. Overweight, Fat Patterning, and Car- M.; O’Rahilly, S. Effects of Recombinant Leptin Therapy in
diovascular Disease Risk Factors in Black and White Girls: a Child with Congenital Leptin Deficiency. N. Engl. J. Med.
The National Heart, Lung, and Blood Institute Growth and 1999, 341, 879–884.
Health Study. J. Pediatr. 1999, 135, 458–464. 401. Loftus, T.; Jaworski, D.; Frehywot, G.; Townsend, C.;
384. Lauer, R. M.; Connor, W. E.; Leaverton, P. E.; ­Ronnett, G.; Lane, M. D.; Kuhajga, F. Reduced Food Intake
Reiter, M. A.; Clarke, W. R. Coronary Heart Disease Risk and Body Weight in Mice Treated with Fatty Acid Synthase
Factors in School Children: The Muscatine Study. J. Pediatr. Inhibitors. Science 2000, 288, 2379–2381.
1975, 86 (5), 697–706. 402. Buchwald, H.; Avidor, Y.; Braunwald, E.; Jensen, M. D.;
385. Figueroa-Colon, R.; Franklin, F.; Lee, J.; Aldridge, R.; Pories, W.; Fahrbach, K.; Schoelles, K. Bariatric Surgery: A
­Alexander, L. Prevalence of Obesity with Increased Blood Systematic Review and Meta-Analysis. J. Am. Med. Assoc.
Pressure in Elementary School-Aged Children. South. Med. 2004, 292 (14), 1724–1737.
J. 1997, 90, 806–813. 403. Pugnale, N.; Giusti, V.; Suter, M.; Zysset, E.; Héraief, E.;
386. Young-Hyman, D.; Schlundt, D. G.; Herman, L.; De Luca, F.; Gaillard, R. C.; Burckhardt, P. Bone Metabolism and Risk
Counts, D. Evaluation of the Insulin Resistance Syndrome in of Secondary Hyperparathyroidism 12 Months After Gas-
5- to 10-Year-Old Overweight/Obese African-American Chil- tric Banding in Obese Pre-Menopausal Women. Int. J. Obes.
dren. Diabetes Care 2001, 24 (8), 1359–1364. 2002, 27, 110–116.
387. Dietz, W. H. Health Consequences of Obesity in Youth: 404. Sjostrom, C. D.; Peltonen, M.; Wedel, H.; Sjostrom, L. Dif-
Childhood Predictors of Adult Disease. Pediatrics 1997, 101 ferentiated Long-Term Effects of Intentional Weight Loss
(supplement), 518–525. on Diabetes and Hypertension. Hypertension 2000, 36,
388. Fagot-Campagna, A.; Pettitt, D. J.; Engelgau, M. M.; 20–25.
­Burrows, N. R.; Geiss, L. S.; Valdez, R.; Beckles, G. L.A.; 405. MacDonald, K. G.; Long, S. D.; Swanson, M. S.; Brown,
Saadine, J.; Gregg, E. W.; Williamson, D. F., et  al. Type 2 B. M.; Morris, P.; Dohm, G. L.; Pories, W. J. The Gastric
Diabetes Among North American Children and Adolescents: Bypass Operation Reduces the Progression and Mortality of
An Epidemiological Review and a Public Health Perspective. Non-Insulin-Dependent Diabetes Mellitus. J. Gastrointest.
J. Pediatr. 2000, 136, 664–672. Surg. 1997, 1, 213–220.
389. Strauss, R. S.; Knight, J. Influence of the Home Environment 406. Goldner, W.; O’Dorisio, T. M.; Dillon, J. S.; Mason, E. E.
on the Development of Obesity in Children. Pediatrics 1999, Severe Metabolic Bone Disease as a Long-Term Complica-
103 (6), e85. tion of Obesity Surgery. Obes. Surg. 2002, 12, 685–692.
390. Whitaker, R. C.; Dietz, W. H. Role of the Prenatal Environment 407. Mason, E. E. Development and Future of Gastroplasties.
in the Development of Obesity. J. Pediatr. 1998, 132, 768–776. Arch. Surg. 2003, 138, 361–366.
391. Whitaker, R. C.; Wright, J. A.; Pepe, M. S.; Seidel, K. D.; 408. Mason, E. E. Bone Disease from Duodenal Exclusion. Obes.
Dietz, W. H. Predicting Obesity in Young Adulthood from Surg. 2000, 10, 585–586.
Childhood and Parental Obesity. N. Engl. J. Med. 1997, 337 409. Newbury, L.; Dolan, K.; Hatzifotis, M.; Low, N.; Fielding,
(13), 869–873. G. Calcium and Vitamin D Depletion and Elevated Parathy-
392. Andersen, R. E.; Crespo, C. J.; Bartlett, S. J.; Cheskin, L. J.; roid Hormone Following Biliopancreatic Diversion. Obes.
Pratt, M. Relationship of Physical Activity and Television Surg. 2003, 13, 893–895.
Watching with Body Weight and Level of Fatness Among 410. Committee on Nutrition American Academy of Pediatrics
Children. J. Am. Med. Assoc. 1998, 279, 938–942. Policy Statement: Prevention of Pediatric Overweight and
393. Robinson, T. N. Ruducing Children’s Television Viewing to Obesity. Pediatrics 2003, 112, 424–430.
Prevent Obesity. J. Am. Med. Assoc. 1999, 282, 1561–1567. 411. Ruffin, M. P.; Nicolaidis, S. Electrical Stimulation of the
394. Ludwig, D. S.; Pereira, M. A.; Kroenke, C. H.; Hilner, J. E.; Ventromedial Hypothalamus Enhances Both Fat Utili-
Horn, L. V.; Slattery, M. L.; Jacobs, D. R. Dietary Fiber, zation and Metabolic Rate that Precede and Parallel the
Weight Gain, and Cardiovascular Disease Risk Factors in Inhibition of Feeding Behavior. Brain Res. 1999, 846 (1),
Young Adults. J. Am. Med. Assoc. 1999, 282 (16), 1539–1546. 23–29.
395. National Center for Chronic Disease Prevention and Health 412. Advisory Board for The Endocrine Society and The Hormone
Promotion Resource Guide for Nutrition and Physical Activ- Foundation. The Endocrine Society Weighs. In: A Handbook
ity Interventions to Prevent Obesity and Other Chronic Dis- on Obesity in America; 2005.
eases. CDC 2003. 413. Sahu, A. Minireview: A Hypothalamic Role in Energy Bal-
396. Von Kries, R.; Koletzko, B.; Sauerwald, T.; von Mutius, ance with Special Emphasis on Leptin. Endocrinology 2004,
E.; Barnert, D.; Grunert, V.; von Voss, H. Breast Feeding 145 (6), 2613–2620.
36 CHAPTER 90  Disorders of the Body Mass

414. Tecott, L.; Sun, L.; Akana, S.; Strack, A.; Lowenstein, D.; 423. Cummings, D.; Brandon, E.; Planas, J.; Motamed, K.;
Dallman, M.; Julius, D. Eating Disorder and Epilepsy in ­Idzerda, R.; McKnight, G. Genetically Lean Mice Result
Mice Lacking 5-HT2c Serotonin Receptors. Nature 1995, from Targeted Disruption of the RII Beta Subunit of Protein
374, 542–546. Kinase A. Nature 1996, 382, 622–626.
415. Stenzel-Poore, M.; Cameron, V.; Vaughan, J.; Sawchenko, 424. Muglia, L.; Jacobson, L.; Dikkes, P.; Majzoub, J.
P.; Vale, W. Development of Cushing’s Syndrome in ­Corticotropin-Releasing Hormone Deficiency Reveals Major
­Corticotropin-Releasing Factor Transgenic Mice. Fetal But Not Adult Glucocorticoid Need. Nature 1995,
­Endocrinology 1992, 130, 3378–3386. 373, 427–432.
416. Melynk, A.; Himms-Hagen, J. Temperature-Dependent 425. Thorliefsson, G.; Walters, G. B.; Gudbjartsson, D. F., et al.
Feeding: Lack of a Role for Leptin and Defects in Brown Genome-Wide Association Yields New Sequence Variants at
Adipose Tissue-Ablated Obese Mice. Am. J. Phys. 1998, Seven Loci That Associate with Measures of Obesity. Nat.
274, R1131–R1135. Genet. 2009, 41, 18–24.
417. Morin, C.; Eckel, R. Transgenic and Knockout Rodents— 426. Scherag, A.; Dina, C.; Hinney, A., et al. Two New Loci for
Novel Insights into Mechanisms of Body Weight Regulation. Body-Weight Regulation Identified in a Joint Analysis of
J. Nutr. Biochem. 2000, 8, 702–706. Genome-Wide Association Studies for Early-Onset Extreme
418. Good, D. J.; Porter, F. D.; Mahon, K. A.; Parlow, A. F.; Obesity in French and German Study Groups. PLoS Genet.
Westphal, H.; Kirsch, I. R. Hypogonadism and Obesity in 2010, 6, e1000916.
Mice with a Targeted Deletion of the Nhlh2 Gene. Nat. 427. Willer, C. J.; Speliotes, E. K.; Loos, R. J., et  al. Six New
Genet. 1997, 15, 397–401. Loci Associated with Body Mass Index Highlight a Neuronal
419. Drago, J.; Padungchaichot, P.; Accili, D.; Fuchs, S. Dopa- Influence on Body Weight Regulation. Nat. Genet. 2009, 41,
mine Receptors and Dopamine Transporter in Brain Func- 25–34.
tion and Addictive Behaviors: Insights from Targeted Mouse 428. Meyre, D.; Delplanque, J.; Chevre, J. C., et al. Genome-Wide
Mutants. Dev. Neurosci. 1998, 20, 188–203. Association Study for Early-Onset and Morbid Adult Obe-
420. Elchebly, M.; Payette, P.; Machaliszyn, E.; Cromlish, W.; sity Identifies Three New Risk Loci in European Populations.
Collins, S.; Loy, A.; Normandin, D.; Cheng, A.; Himms- Nat. Genet. 2009, 41, 157–159.
Hagen, J.; Chan, C.-C., et  al. Increased Insulin Sensitivity 429. Gratacos, M.; Gonzalez, J. R.; Mercader, J. M., et  al.
and Obesity Resistance in Mice Lacking the Protein Tyrosine Brain-Derived Neurotrophic Factor Val66Met and Psy-
Phophatase-1B Gene. Science 2000, 283, 1544–1548. chiatric Disorders: Meta-analysis of Case-Control Studies
421. Charron, M. J.; Katz, E. Metabolic and Therapeutic Lessons Confirm ­Association to Substance-Related Disorders, Eat-
from Genetic Manipulation of GLUT4. Mol. Cell. Biochem. ing Disorders, and Schizophrenia. Biol. Psychiatry. 2007,
1998, 182, 143–152. 61, 911–922.
422. Shimada, M.; Tritos, N. A.; Lowell, B. B.; Flier, J. S.; 430. Chambers, J. C.; Elliott, P.; Zabaneh, D., et  al. Common
Maratos-Flier, E. Mice Lacking Melanin-Concentrating Genetic Variation Near MC4R Is Associated with Waist
Hormone Are Hypophagic and Lean. Nature 1998, 396, ­Circumference and Insulin Resistance. Nat. Genet. 2008, 40,
670–674. 716–718.
CHAPTER 90  Disorders of the Body Mass 37

Biographies

 atricia A Donohoue attended medical school at The Ohio State University, and completed her
P
internship in Pediatrics there at what is now known as Nationwide Children’s Hospital. She
completed her second and third years of pediatric residency at Rainbow Babies and Children’s
Hospital, Case Western Reserve University. Her Pediatric Endocrinology Fellowship was at
Johns Hopkins under the mentorship of Professor Claude Migeon and the late Professor Neil
Van Dop. She stayed there as a junior faculty member for 4 years before moving to the Univer-
sity of Iowa. At Iowa, she continued her NIH-funded laboratory research on the genetics of ste-
roid 21-hydroxylase deficiency, and then focused her interest on other inherited defects of the
hypothalamic–pituitary–adrenal axis, and eventually on the genetics of obesity. After 18 years
at the University of Iowa, she moved to the Medical College of Wisconsin in Milwaukee in
2008, where she is a Professor of Pediatrics and Section Chief of Pediatric Endocrinology and
Diabetes.

 mar Ali, MD attended medical school at King Edward Medical College in Lahore, Pakistan,
O
and completed his residency in pediatrics at Jersey City Medical Center. He then did a 1-year
fellowship in community pediatrics at the Children’s Hospital of Pittsburgh. He worked in
general pediatrics for the next 12 years, but then decided to do a fellowship in Pediatric Endo-
crinology at UCLA-Mattel Children’s Hospital under Dr Hassy Cohen. Subsequently, he joined
the Medical College of Wisconsin in 2006. He is an Assistant Professor of Pediatrics in the
Section of Pediatric Endocrinology and Diabetes, and Director of the Pediatric Endocrinology
Fellowship Program. His research interest is the genetics and epigenetics of obesity and the
metabolic syndrome, and he is currently working on grants funded by the NIH, the National
Children’s study and the Children’s Research Institute.

You might also like