ArticlePDF AvailableLiterature Review

What the Genetics of Lipodystrophy Can Teach Us About Insulin Resistance and Diabetes

Authors:

Abstract and Figures

Genetic lipodystrophic syndromes are rare diseases characterized by generalized or partial fat atrophy (lipoatrophy) associated with severe metabolic complications such as insulin resistance (IR), diabetes, dyslipidemia, nonalcoholic fatty liver disease, and ovarian hyperandrogenism. During the last 15 years, mutations in several genes have been shown to be responsible for monogenic forms of lipodystrophic syndromes, of autosomal dominant or recessive transmission. Although the molecular basis of lipodystrophies is heterogeneous, most mutated genes lead to impaired adipogenesis, adipocyte lipid storage, and/or formation or maintenance of the adipocyte lipid droplet (LD), showing that primary alterations of adipose tissue (AT) can result in severe systemic metabolic and endocrine consequences. The reduced expandability of AT alters its ability to buffer excess caloric intake, leading to ectopic lipid storage that impairs insulin signaling and other cellular functions ("lipotoxicity"). Genetic studies have also pointed out the close relationships between ageing, inflammatory processes, lipodystrophy, and IR.
Content may be subject to copyright.
GENETICS (TM FRAYLING, SECTION EDITOR)
What the Genetics of Lipodystrophy Can Teach Us About
Insulin Resistance and Diabetes
Camille Vatier & Guillaume Bidault & Nolwenn Briand &
Anne-Claire Guénantin & Laurence Teyssières &
Olivier Lascols & Jacqueline Capeau & Corinne Vigouroux
#
Springer Science+Business Media New York 2013
Abstract Genetic lipodystrophic syndromes are rare diseases
characterized by generalized or partial fa t atrophy (lipoatrophy)
associated with severe metabolic complications such as insulin
resistance (IR), diabetes, dyslipidemia, nonalcoholic fatty liver
disease, a nd ovarian hy perandrog enism. During the last
15 years, mutations in several genes have been shown to be
responsible for monogenic forms of lipodystrophic syndromes,
of autosomal dominant or recessive transmission. Although the
molecular basis of lipodystrophies is heterogeneous, most mu-
tated genes lead to impaired adipogenesis, adipocyte lipid
storage, and/or formation or maintenance of the adipocyte lipid
droplet (LD), showing that primary alterations of adipose tissue
(AT) can result in severe systemic metabolic and endocrine
consequences. The reduced expandability of AT alters its ability
to buffer excess caloric intake, leading to ectopic lipid storage
that impairs insulin signaling and other cellular functions
(lipotoxicity). Genetic studies have also pointed out the close
relationships between ageing, inflammatory processes,
lipodystrophy, and IR.
Keywords Adipose tissue
.
Lipodystrophy
.
Lipid droplets
.
Adipogenesis
.
Insulin resistance
.
Diabetes
.
Dyslipidemia
.
Liver steatosis
.
Polycystic ovary syndrome
.
Ageing
.
Genetics
.
Seipin
.
AGPAT2
.
A-type lamins
.
PPARγ
.
Perilipin
.
Caveolin 1
.
Cavin 1
.
Caveolae
.
CIDEC
.
Akt2
.
PSMB8
.
Progeria
.
ZMPSTE24
.
Genetics
Introduction
Insulin resistance (IR), frequently associated with obesity, is a
highly prevalent risk factor for cardiovascular diseases, type 2
diabetes, and fatty liver disease. A better understanding of its
pathophysiology is a prerequisite for an efficient prevention
and treatment in the general population. In this setting, the
studies of genetic lipodystrophic syndromes, rare monogenic
models of insulin resistance, have provided important clues.
Lipodystrophic syndromes are rare diseases with congeni-
tal or acquired loss of body fat. Paradoxically, the metabolic
consequences of having «too little» fat (lipoatrophy) are re-
markably similar to those of having «too much» fat (obesity).
Excess energy, stored as lipids in ectopic sites, leads to IR,
dyslipidemia, diabetes, with cardiovascular, reproductive, and
liver complications.
Recent advances in molecular genetics of lipodystrophic
syndromes have revealed that primary defects in biogenesis
and metabolism of adipose lipid droplet (LD) are important
pathophysiological issues [1, 2], highlighting the central role
for AT in systemic metabolism.
C. Vatier
:
G. Bidault
:
N. Briand
:
A.<C. Guénantin
:
L. Teyssières
:
O. Lascols
:
J. Capeau
:
C. Vigouroux
INSERM UMR_S938, Centre de Recherche Saint-Antoine,
75012 Paris, France
C. Vatier
:
G. Bidault
:
N. Briand
:
A.<C. Guénantin
:
L. Teyssières
:
O. Lascols
:
J. Capeau
:
C. Vigouroux
UPMC Univ Paris 6, UMR_S938, 75012 Paris, France
C. Vatier
:
G. Bidault
:
N. Briand
:
A.<C. Guénantin
:
L. Teyssières
:
O. Lascols
:
J. Capeau
:
C. Vigouroux
ICAN, Institute of Cardiometabolism and Nutrition, Paris, France
C. Vatier
AP-HP, Hôpital Pitié-Salpêtrière, Service de Nutrition, 75013 Paris,
France
O. Lascols
AP-HP, Hôpital Saint-Antoine, Laboratoire Commun de Biologie et
Génétique Moléculaires, 75012 Paris, France
J. Capeau
:
C. Vigouroux
AP-HP, Hôpital Tenon, Service de Biochimie et Hormonologie,
75020 Paris, France
C. Vigouroux (*)
Faculté de Médecine Pierre et Marie Curie, site Saint-Antoine,
UMR_S938, 27 rue Chaligny, 75012 Paris, France
e-mail: corinne.vigouroux@inserm.fr
Curr Diab Rep
DOI 10.1007/s11892-013-0431-7
Since 1999, more than 10 genes involved in lipodystro-
phies have been identified [3, 4, 5, 6, 7, 8, 9, 10, 11, 12••]
(Table 1). In addition, complex syndromes associating
lipodystrophy and premature aging or auto-inflammatory fea-
tures have revealed that AT can be damaged by senescence
and/or immuno-inflammatory processes [1317, 18••, 19].
Due to the growing availability of powerful genetic tools,
new genes will probably be linked to still undeciphered
lipodystrophic syndromes.
AT is now well-known not only as a metabolic tissue, but
also as an important paracrine and endocrine organ [20, 21].
Among ma ny adipose secretory products, leptin, w hich
plasma levels generally reflect the AT mass, regulates food
intake and energy consumption, but also distribution of
cellular storage. Adiponectin also m odulates i nsulin sensi-
tivity. Several proteins of the renin-angiotensin system,
produced by AT, influence its development and functions
and could link both obesity and lipodystrophy to hyperten-
sion [22]. Moreover, adipokines secreted by AT contribute
to immune and i nflammatory processes [23]andtheirrole
as important cancer microenvironment factors has recently
emerged [24].
In this review, we report how genetic studies of monogenic
forms of lipodystrophic syndromes have offered the unique
opportunity to reveal the importance of AT for the whole body
metabolism.
Murine Models of Lipodystrophies
Several mouse models of generalized lipodystrophy have been
generated, using knock-out or overexpression of key gene s.
Models of generalized lipodystrophy recapitulated most of the
features of human diseases, but partial lipodystrophy pheno-
types were more difficult to generate, probably because of
important differences in AT distribution between mice and
humans. Nevertheless, murine models have been important
tools showing that loss of AT initiates both lipodystrophy and
metabolic consequences [25, 26]. In accordance, transplanta-
tion of healthy AT [27] but not leptin-deficient ob/ob AT [ 28]
reversed insulin resistance in lipodystrophic mice, showing
the major role of both fat-storing capacities and leptin for
metabolic homeostasis. Metabolic improvements obtained in
lipodystrophic mice with leptin infusion or overexpression
[29] have allowed initiation of successful therapeutic trials
of leptin treatment in human lipodystrophies.
Human Inherited Lipodystrophies
Epidemiology
The worldwide prevalence of genetic lipodystrophies is not
precisely known. It was evaluated at about 1 per 10 million for
Table 1 Human genetic lipodystrophic syndromes and main affected genes
Main role of affected protein Disease Gene/protein AD or AR transmission
Partial lipodystrophy
Protein of the nuclear envelope FPLD2 LMNA / lamin A/C AD
Protein of the nuclear envelope Metabolic laminopathies LMNA / lamin A/C AD
Protein of the nuclear envelope MAD-A LMNA / lamin A/C AR
Key transcription factor for adipocyte differentiation FPLD3 PPARG /PPARγ AD
Lipolysis regulation of the adipocyte lipid droplet FPLD4 PLIN1 / perilipin AD
Structure of the adipocyte lipid droplet FPLD CIDEC / CIDEC AR
Serine threonine kinase involved in insulin signaling FPLD AKT2 / Akt2 AD
Protein of the nuclear envelope APL LMNB2 / lamin B2 risk factor
Generalized lipodystrophy
Enzyme of triglyceride and glycerophospholipid synthesis BSCL 1 AGPAT2 /AGPAT2 AR
Formation of lipid droplet BSCL 2 BSCL2 /seipin AR
Structural protein of caveolae and adipocyte lipid droplet BSCL3 CAV1 /caveolin1 AR
Structural protein of caveolae and adipocyte lipid droplet BSCL4 PTRF / cavin 1 AR
Protein of the nuclear envelope Hutchinson-Gilford progeria LMNA / lamin A/C Heterozygous,
de novo
mutations
Endoprotease involved in lamin A maturation MAD-B ZMPSTE24 / Zmpste24 AR
Immunoproteasome subunit JMP PSMB8 / PSMB8 AR
AD autosomal dominant, AGPA T2 acylglycerol-3-phosphate O-Acyltransferase, APL acquired partial lipodystrophy, AR autosomal recessive, BSCL
Berardinelli-Seip congenital lipodystrophy, FPLD familial partial lipodystrophy, JMP joint contractures, muscle atrophy, microcytic anemia, and
panniculitis-induced lipodystrophy MAD mandibuloacral dysplasia, PSMB8 proteasome subunit, beta-type, 8
Curr Diab Rep
congenital generalized lipodystrophies (CGL) in the US [30];
in France our records suggest that it could be of 1 to 5 per
million. The prevalence of familial partial lipodystrophies
(FPLD) is also probably under-estimated. Patients, and more
specifically men, in whom the morphotype is frequently mod-
erate, are frequently diagnosed with a metabolic syndrome
without any search for molecular alterations, an android dis-
tribution of fat being frequent in metabolic syndrome and type
2diabetes.
Clinical Characteristics
In inherited lipodystrophies, lipoatrophy could be present at
birth or later in life, the extent of fat loss varying from partial
to complete. In addition to lipoatrophy, clinical signs of IR,
thought to be linked to effects of severe hyperinsulinemia on
IGF1 receptors, can help diagnosis: skin lesions of acanthosis
nigricans, a brownish lesion of axillae, neck and other body
folds; acromegaloid features, striking at the level of face and
extremities; muscular hypertrophy, even more visible due
to the lack of subcutaneous adipose tissue (SAT); ovar-
ian hyperandrogenism with hirsutism or virilization and
oligomenorrhea in the context of polycystic ovary syndrome
or hyperthecosis.
Patients with CGL are characterized by total absence of AT
evident from birth [31], marked muscular appearance with
prominent veins, severe acromegaloid features, acanthosis
nigricans, hepatomegaly, and umbilical prominence. They
often have a voracious appetite and children frequently show
an accelerated linear growth.
In FPLD, lipoatrophy mostly involves the extremities with
variable fat loss from the trunk. The body fat distribution is
unremarkable at birth and during childhood, progressive loss
of fat occurring during late childhood or after puberty. In
lamin A-linked forms, peripheral lipoatrophy contrasts with
accumulation of subcutaneous fat (SAT) over the face, chin,
supraclavicular and dorsocervical re gions, and frequently
intra-abdominal fat stores [32, 33]. Similar to patients with
CGL, acanthosis nigricans and hepatomegaly can be promi-
nent, and female patients frequently show features of polycys-
tic ovarian syndrome [34]. CGL and FPLD are of autosomal
recessive or dominant inheritance, depending on the gene
involved (Table 1).
Both partial and generalized lipodystrophy have also been
reported in patients with premature ageing syndromes, as
presented below.
Biological Signs
Metabolic alterations can be mild or absent during childhood,
and increase with age. Lipid alterations associate increased
triglyceride (TG) level and decreased HDL-cholesterol, with a
high risk of acute pancreatitis. Striking hyperinsulinemia is
usually able to control glycemia in childhood, but glucose
intolerance and then diabetes frequently occur around puberty.
Serum adiponectin and leptin concentrations are reduced in
proportion to the extent of fat loss [35]. In CGL2, a surpris-
ingly detectable adiponectin level has been reported [36].
Imagery
Whole-body dual-energy X-ray absorptiometry (DEXA) eval-
uates the total body fat amount, and the distribution of AT. It
can help for phenotypic characterization of lipodystrophies, in
particular in precocious identification of partial forms [37].
Computerized tomography (CT) or magnetic resonance im-
aging (MRI) at the lumbar L4 level, assessing abdominal fat,
distinguishes between subcutaneous and visceral fat depots
[38].
Hepatomegaly and nonalcoholic fatty liver disease are eval-
uated using ultrasound, CT, and MRI. Hepatic elastometry
helps to diagnose liver fibrotic changes.
As patients with CGL could present with bone lesions [39]
(see below), conventional bone radiographies and CT are need-
ed. Cardiovascular investigatio ns are also useful for the pheno-
type characterization and the medical care in lipodystrophic
patients; cardiac rhythm and conduction disturbances have to
be checked carefully in case of lamin A-linked lipodystrophies
[40, 61].
Complications
Chronic complications are related to diabetes and dyslipidemia
(microangiopathy, recurrent acute pancreatitis, cardiovascular
diseases), to nonalcoholic fatty liver disease (steatohepatitis
and cirrhosis), and to reproductive abnormalities due to ovarian
hyperandrogenism (infertility).
Early-onset and severe hypertension is frequent, particular-
ly in peroxisome prolifer ator-activat ed receptor gam ma
(PPAR-γ) -linked forms.
Differential Diagnosis
Genetic lipodystrophies can be difficult to differentiate first
from primary syndromes of IR and second from acquired
lipodystrophic syndromes.
Indeed, syndromes of severe IR due to mutations of the
insulin receptor are associated with a generalized paucity of
body fat when both alleles are affected (leprechaunism or
Donohue syndrome and Rabson-Me ndenhall syndrome).
These syndromes of extreme insulin resistance expressed in
newborns or children are developmental disorders, with dys-
morphic features and growth delay [41]. Notable differences
with lipodystrophic syndromes are the usual absence of
dyslipidemia and liver steatosis, and the high adiponectin
levels, consistent with the generalized impairment of insulin-
Curr Diab Rep
stimulated pathways. In contrast, in lipodystrophies, primary
adipose tissue defects induce insulin resistance at a post-
receptor level: the ability of insulin to suppress hepatic gluco-
neogenesis is impaired, leading to hyperglycemia and
hyperinsulinemia, but the insulin-stimulated lipogenesis path-
way is preserved, with increased production of TG by the liver
[42]. Through a collaborative study, we have recently shown
that the SHORT syndrome (short stature, hyperextensibility of
joints and/or inguinal hernia, ocular depression, Reiger anom-
aly and teething delay), another developmental disorder asso-
ciated with insulin resistance and generalized paucity of fat, is
a syndrome of prima ry insulin resistance due to PIK3R1
mutations affecting a proximal intermediate of the insulin
signaling pathways [43].
Other anomalies of fat distribution must be distinguished
from genetic lipodystrophic syndromes. Among them, the
Launois-Bensaude lipomatosis is characterized by multiple
localized fat tumors of the proximal limbs and neck, of un-
known origin, often associated with peripheral neuropathy
and increased alcohol intake, and variable metabolic alter-
ations. In rare cases, lipomatosis is associated with other
diseases linked to altered mitochondrial DNA. Acquired
lipodystrophic syndromes occurring in late infancy or young
adulthood, are frequently associated with autoimmune fea-
tures. Some HIV antiretrovirals or endogenous or exogenous
hypercortisolism also lead to lipodystrophic syndromes.
Finally, the aging process leads to fat redistribution with
relative peripheral lipoatrophy and visceral fat accumulation
and increased risk of metabolic disturbances [2].
Human Genetic Lipodystrophic Syndromes
Are Heterogeneous Monogenic Diseases
(Table 1 and Fig. 1)
In the last 14 years, mutations in more than 10 genes have
been found to be responsible for lipodystrophic syndromes,
bearing new light on the relationships between AT and
metabolism.
Congenital Generalized Lipodystrophies (CGL)
The genetic defect underlying CGL (or BSCL, Berardinelli-
Seip congenital lipodystrophy), of recessive inheritance, has
been identified in more than 90 % of cases, with 1 of 2 main
genes involved in most cases (encoding seipin (BSCL2)[6], or
AGPAT2 (1-acylglycerol-3-phosphate-O-acyltransferase 2)
(BSCL1 )[7]. Far less often caveolin-1 (BSCL3 )[9]or
cavin-1/PTRF (polymerase I and transcript release factor)
(BSCL4) are involved [11]. All the mutated proteins act on
the pathways of TG synthesis and/or storage in the adipocyte
LD (Fig. 1).
If clinical presentation is very similar whatever the geno-
type, patients with BSCL2 have the most severe phenotype
(lipoatrophy implicating all fat depots with also paucity of
mechanical fat, hypertrophic cardiomyopathy) [44, 45], with
frequent mild mental retardation whereas AGPAT2 mutations
are frequently associated with skeletal alterations (osteoblastic
nodules, diffuse bone sclerosis, lytic lesions [39]) and cavin-1/
PTRF mutations with muscular dystrophy.
AGPAT2
Bi-allelic mutations affecting AGPAT2 [7]onchromosome
9q34, are expected to entirely abrogate protein function or
expression.
AGPAT2 is an enzyme that catalyses the synthesis of
phosphatidic acid from acyl-CoAs in the endoplasmic reticu-
lum (ER) of adipocytes, the site of LD formation [46].
AGPAT2 deficiency might thus prevent TG synthesis and
mature lipid adipocytes droplet formation. In addition, it could
also impair the synthesis of other lipids involved in adipocyte
differentiation [47].
Seipin
Seipin is encoded by BSCL2 on chromosome 11q13 [6
]. CGL
is linked to BSCL2 bi-allelic inactivating mutations.
Seipin is an integral protein of the ER involved in the
adipogenesis process [48]. At the ER level, it mediates the
lipidation of nascent LDs and their maintenance [49, 50, 51]
and regulates fatty acid monoinsaturation [48, 50]. To note,
gain-of-toxic-function heterozygous BSCL2 mutations are re-
sponsible for motor neuron diseases [52], with decreased
neuronal TG content and endoplasmic reticulum stress [53].
Caveolin-1
Mutations in CAV1, encoding caveolin-1 were identified in a
patient with CGL, short stature, and resistance to vitamin D
[9] (homozygous nonsense mutation) and thereafter in cases
of atypical partial lipodystrophy (heterozygous mutations)
[54].
Caveolin-1 is the major coating protein of caveolae, plasma
membrane invaginations particularly abundant in adipocytes,
but can also be found at the LD surface [55, 56]. Caveolin-1
regulates several signaling pathways in adipocytes, including
insulin signaling and lipolysis. Defects in insulin and lipolytic
responses in caveolin-1 deficient adipocytes may contribute to
a nutrient shortage, leading to autophagy [57]. In addition,
caveolin-1 deficient adipocytes show a global alteration of
phospholipid composition of the LD surface, suggesting a
regulatory role of caveolin-1 on LD expandability [58].
Curr Diab Rep
PTRF/Cavin-1
Mutations affecting both alleles of cavin-1/PTRF lead to a
mixed phenotype of generalized lipodystrophy and muscular
dystrophy [11]. Pyloric stenosis and cardiac arrhythmia have
also been described [59].
Cavin-1 is colocalized with caveolin-1 on the adipocyte LD
[58] and is involved in the stabilization and last phase of
biogenesis of caveolae [60]. Loss of cavin-1 causes loss of
caveolae and a reduced expression and mislocalization of the
caveolins [10].
Causes of Genetic Partial Lipodystrophies
Partial forms of genetic lipodystrophies, collectively named
FPLD, are more common than generalized forms. FPLD are
generally dominantly inherited, except some forms, due to
mutations in Cell death-inducing DFF45-like effector C
(CIDEC) [10]orLMNA in rare cases [61, 62], which are
codominantly or recessively transmitted.
Lamin A/C
The most typical form of FPLD is the Dunnigan type (or
FPLD2), linked to heterozygous LMNA mutations. Mutations
in this gene cause a group of rare disorders (laminopathies), of
wide clinical heterogeneity ranging from cardiac and muscular
phenotypes to lipodystrophic syndromes and premature aging
syndromes [63].
The 2 major A-type lamins, prelamin A and lamin C, arise
from alternative splicing of the LMNA gene. Prelamin A is
posttranslati onally processed with a step of farnesylation follow-
ed by ZMPSTE24-mediated cleavage, resulting in mature non-
farnesylated lamin A release. A-type lamins, expressed in most
differentiated cells, form with B-type lamins the lamina network
beneath the inner nuclear membrane, which maintains the
Fig. 1 Proteins involved in genetic lipodystrophic syndromes. Pluripo-
tent mesenchymal stem cells can differentiate into preadipocytes
depending upon the signals from hormones such as insulin, steroids and
other adipogenic transcription factors, mainly CCAAT (cytidine-cytidine-
adenosine-adenosine-thymidine)-enhancer-binding proteins (C/EBP)
β/δ,PPARγ,C/EBPα, and sterol regulatory element-binding protein
(SREBP) 1c. Seipin, AGPAT2 and AKT2 may also be involved in
adipocyte differentiation. Other proteins involved in lipodystrophies are
critical for maturation of preadipocytes and/or maintenance of mature
adipocyte phenotype
Curr Diab Rep
nucleus shape and interact with the cytoskeleton. Also found in
the nucleoplasm, they interact with chromatin, DNA, and tran-
scription factors, and regulate several functions including gene
transcription [64].
FPLD2 is mainly due to p.R482 heterozygous substitutions
affecting the C-terminal domain of A-type lamins, involved
DNA and the adipogenic transcription factor SREBP-1c
binding [4, 5]. The phenotype of the disease is more severe
in women than in men [65, 66]. In addition to lipoatrophy,
FPLD2 is characterized by an expansion of dystrophic facio-
cervical fat [67].
Other LMNA mutations result in atypical lipodystrophies
[40, 68] and/or in mixed phenotypes with muscular or cardiac
dystrophies or progeria-like features [69, 70],andtowell-
characterized accelerated ageing syndromes [1315](seebelow).
The pathophysiology of the different laminopathies, which
affect highly specialized tissues but are due to alterations in
ubiquitously expressed proteins, remains to be clarified. Both
cellular mechanical stress and alterations of gene expression
could be involved. We and others described the toxicity of
farnesylated R482-mutated prelamin-A, leading to premature
cellular senescence [17, 71]. However, we observed that hu-
man homozygous LMNA p.T655fsX49 mutation, resulting in
the expression of non-farnesylated prelamin A without mature
lamin A, led to a lipodystrophic syndrome [61], showing that
permanent farnesylation of mutated lamin A is not the unique
factor leading to LMNA -linked lipodystrophies.
Interestingly, studies using induced pluripotent stem cells
(iPSC), which express wild-type or mutated A-type lamins
during differentiation, have shown that mutated A-type lamins
lead to defects in the mesenchymal and smooth muscle line-
ages [72••, 73••]. In addition, LMNA mutations perturb the
balance between proliferation and differentiation in adult stem
cells [7476]. Defects in specific tissular differentiation in
response to different A-type lamins alterations could underlie
the heterogeneous laminopathic phenotypes.
PPARγ
The transcription factor PPARγ plays a leading role in
adipogenesis and also exerts anti-inflammatory and antioxi-
dant effects [77].
Rare dominant-negative and loss-of-function heterozygous
mutations affecting the ligand- or the DNA-binding domains of
PPARγ have been identified in patients with FPLD (FPLD3)
[3, 78]. Severe hypertension linked to FPLD3 may result from
the ability of mutated PPARγ to activate the cel lular renin -
angiotensin system in cells from the vascular wall [79
].
Akt2
AKT2 encodes the protein kinase B, an important intermediate
of insulin signaling, has been involved in one FPLD family
[8]. Post-receptor insulin resistance sparing the lipogenesis
pathway has been reported in this family [42••].
CIDEC
CIDEC participates in adipose LD formation and contributes
to the preadipocyte differentiation process [80]. A homozy-
gous truncating non-sense mutation of CIDEC (p.E186X) has
been identified in a case of partial lipoatrophic syndrome,
leading to decreased fat mass and multiloculated adipose LD
[10].
Perilipin
Perilipin (now called perilipin-1) is localized at the surface of
LD in adipocytes and steroidogenic cells. Perilipin-1 reduces
basal lipolysis, but also activa tes lipolysis in response to
catecholamines.
Heterozygous inactivating mutations of the perilipin gene
(PLIN1), inducing a sustained constitutive lipolysis [12••, 81],
are responsible for FPLD4 [12••], with inflammation and
fibrosis of fat tissue, and whole body metabolic consequences
(ie, insulin resistance and dyslipidemia).
Acquired Partial Lipodystrophy and Lamin B2
Barraquer-Simons syndrome, also called acquired generalized
lipodystrophy (APL), with lipoatrophy affecting progressively
the face and upper body, was classically considered as an
immune disease, since it is associated with auto-immune
disorders and/or membrano-proliferative glomerulonephritis.
However, 5 patients with AGL have been reported with het-
erozygous mutations in LMNB2, encoding lamin B2, a partner
of A-type lamins at the nuclear envelope. These genetic alter-
ations could be risk factors for the disease [82, 83].
Lipodystrophies Associated With Premature Ageing
Generalized or partial forms of lipoatrophy are observed in
several premature ageing syndromes. First, heterozygous mu-
tations of LMNA are responsible for the typical form of
Hutchinson-Gilford progeria, due to the synthesis of a perma-
nently farnesylated mutated prelamin A, [14, 15] and to other
progeroid syndromes [69, 70], characterized by post-natal
growth retardation, craniofacial dysmorphy, skeletal and skin
abnormalities, and generalized lipoatrophy. Other rare,
biallelic, LMNA mutations lead to A-type mandibuloacral
dysplasia (MAD), with alterations of the same tissues and
FPLD-like partial lipodystrophy [13]. Homozygous mutations
of the zinc metalloproteinase ZMPSTE24, which cleaves
farnesylated prelamin A into mature lamin A, can lead to
MAD type B, with generalized lipoatrophy [16]. These dis-
eases point to the involvement of farnesylated prelamin A,
Curr Diab Rep
which, by altering its binding properties, especially for the
membranes, leads to cellular senescence. In accordance, the
inhibition of ZMPSTE24 by some HIV protease inhibitors
could also contribute to the antiretroviral-linked lipodystrophic
syndromes [17, 84]. However, the therapeutical benefits of
farnesyl-transferase inhibitors in children with Hutchinson-
Gilford progeria were only partial, improving predominantly
vascular stiffness [85••].
Other premature ageing syndromes, linked to DNA repair
defects, can also associate with IR and altered fat repartition.
Our group recently showed that partial lipodystrophy and
severe IR can reveal Werner syndrome, due to inactivating
biallelic mutations of WRN, a DNA helicase. Increased lamin
B1 expression could play a role in premature senescence of
WRN-mutated cells [86]. In addition, Weedon et al recently
observed that heterozygous mutations in POLD1 ,encoding
DNA polymerase δ, which cooperates with WRN to maintain
genome stability, also lead to a multisystem disorder with
lipodystrophy [87].
Lipodystrophy Associated With Autoinflammation
Inflammatory or immune processes can lead to inflammation
then fibrosis of AT, as observed in localized lipoatrophies due
to iatrogenic aggression of AT (for example injections of
corticosteroid, insulin, or antibiotics) [88].
Recently, Garg et al deciphered the molecular basis of the
JMP syndrome (joint contractures, muscle atrophy, microcytic
anemia, and panniculitis-induced lipodystrophy) associated
with autoinflammation (recurrent fever with osteo-articular
and muscular symptoms). Partial lipoatrophy, developed dur-
ing infancy, affects mainly the upper part of the body.
Homozygous mutations affected PSMB8 , encoding a catalytic
subunit of the immunoproteasome [18••]. Several groups con-
firmed the alterations of this gene in similar phenotypes
denominated CANDLE or JASP (Japanese autoinflammatory
syndrome) [19].
Metabolic Consequences of Lipodystrophies (Fig. 1)
Patients with lipodystrophies present metabolic consequences
that are remarkably similar to those observed in obesity, but
generally more severe, showing that maintaining a healthy fat
amount is an essential requirement for metabolic homeostasis.
In addition, these diseases also illustrate the fact that subcuta-
neous fat of the lower part of the body is mostly metabolically
favorable, being able to buffer fat excess and protect against
lipotoxicity [89]. Conversely, in the upper part of the body,
limited fat expansion leads to overwhelming of fat and ectopic
lipid deposition in other tissues, as liver, muscles, heart and
vessels, and pancreas [90]. However, Barraquer-Simons syn-
drome with the reverse distribution of fat compared with
FPLD2, can be associated, although less frequently, with
severe insulin resistance.
AT has emerged as an integrator of a wide array of homeo-
static processes, including blood pressure control, insulin
sensitivity, and regulation of inflammation and immune pro-
cesses through the secretion of numerous adipokines [2024].
Most of the proteins mutated in human lipodystrophies are
directly involved in adipogenesis (PPARγ,probablylaminA/
C, seipin, AGPAT2), the synthesis and maintenance of the LD
(seipin, perilipin, CIDEC, caveolin 1 and cavin-1) or are acting
in the pathways leading to lipid synthesis or storage, at the level
of the nucleus, ER or caveolae (lamins A/C through their
binding to SREBP-1c, AGPAT2, caveolin 1, and cavin-1).
It is considered that AT expandability is finite up to a
particular set point that varies on an individual basis. Beyond
this set point, additional energy excess results in AT failure with
oxidative stre ss, re cruitment of mac rophages, release of
proinflammatory cytokines and of free fatty acid (FFA) and
decreased adiponectin and leptin, leading t o lipotoxicity.
Genetically limited expansion of fat in lipodystrophies lowers
the se t point for fat storage leading to ectopic deposition of
lipids and severe metabolic deregulation.
Therapeutic Options for Lipodystrophic Syndromes
Diet and Exercise
A well-balanced diet is particularly important for lipodystrophic
patients, who are not capable to correctly store excess energy as
fat. Reduction of energy intake, particularly dietary fat, is very
useful to avoid ectopic lipid deposition, and thus metabolic
disturbances, but excess consumption of carbohydrates,
which enhances de novo lipogenesis, should also be avoided.
However, reduction of energy intake is frequently difficult for
lipodystrophic patients, due to a voracious appetite resulting
from very low levels of leptin. In addition, diet should be
carefully monitored in children to allow adequate growth and
development.
Similarly, increasing energy expenditure with daily physi-
cal activity, particularly aerobic exercise, can notably improve
metabolic complications in lipodystrophy.
Medications
Due to insulin resistance, diabetes is generally difficult to
control in lipodystrophies. Insulin sensitizers are the first line
of therapy. However metformin therapy has not been studied
specifically in patients with lipodystrophies. A treatment with
thiazolidinediones was beneficial in several patients, even
those with mutations in PPARγ, but it is no longer available
in France and its use is restricted in several countries. Very
high doses of insulin are frequently required. Medium chain
Curr Diab Rep
triglycerides supplementation and /or omega-3 polyunsaturat-
ed fatty acids from fish oils could contribute to lower TG [91].
Otherwise, hypolipidemic drugs such as fibrates are required
to avoid major hypertriglyceridemia [92].
In women with lipodystrophies, ethinylestradiol should be
used with caution because of the risks of hypertriglyceridemia
and acute pancreatitis.
Substitution with leptin, administered as human recombi-
nant metreleptin, has resulted in markedly improved metabol-
ic homeostasis and regression of liver steatosis, particularly in
severe lipoatrophic and hypoleptinemic patients [9396].
Leptin therapy reduces appetite and results in weight loss,
which contributes to metabolic improvement. In addition,
leptin can enhance insulin sensitivity by activating muscular
adenosine monophosphate kinase (AMPK), thus decreasing
lipotoxicity. Our recent results showed an improvement in
insulin secretion in leptin-treated lipodystrophic patients
(Vatier et al in preparation). Metreleptin is available through
compassionate programs for the moment.
Other therapeutic options have been proposed, but have not
been evaluated in humans. Among them, AMPK activators
[97], adiponectin replacement, which has been reported to
lower plasma glucose and FFA levels in mouse models of
lipodystrophy or diabetes [98], inhibitors of 11β-HSD1 or
glucocorticoid receptor inhibitors, which could reduce IR,
and inhibitors of pancreatic endoplasmic reticulum kinase,
which may reduce endoplasmic reticulum stress.
Plastic Surgery
The altered body fat repartition can benefit from plastic sur-
gery, in particular for patients with partial lipodystrophy [99].
Conclusions
Lipodystrophies represent a heterogeneous group of severe
diseases leading to early complications. Genetic studies of
these diseases, showing the involvement of several proteins
involved in adipocyte differentiation and/or in adipocyte LD
biogenesis and metabolism, enlightened the leading role of
adipose tissue for global metabolic homeostasis. In addition,
metabolic studies have shown that primary defects of adipose
tissue resulted in secondary insulin resistance, affecting het-
erogeneously the different insulin signaling pathways.
Therefore, studies on human lipodystrophies help to un-
derstand the complex physiology and pathophysiology of fat.
They point to new genes and new targets, which could lead to
the discovery of new and innovative therapeutic clues that
could also offer new perspectives for metabolic syndrome and
type 2 diabetes.
Acknowledgments The researches of the authors are supported by Institut
de la Santé et de la Recherche Médicale (INSERM), Université Pierre et
Marie Curie - Paris 6 (UPMC), and Agence Nationale de la Recherche
(program Investments for the Future, Insti tute of Cardiometabol ism and
Nutrition [ICAN]; grant no. ANR-10-IAHU).
C. Vatier is the recipient of a PhD grant from the Conseil Régional
dIle de France (Cardiovasculaire-Obésité-Diabète Domaine dIntérêt
Majeur), G. Bidault. of a PhD grant from the Fondation pour la Recherche
Médicale, N. Briand of post-doctoral grant from Région Ile-de-France
(DIM Biotherapies), A-C. Guénantin of a post-doctoral grant from Insti-
tute of Cardiometabolism and Nutrition (Innovative projects 2012) and L.
Teyssières of a master grant from Agence Régionale de Santé Limousin.
Compliance with Ethics Guidelines
Conflict of Interest Camille Vatier has been on the Advisory board on
lipodystrophy and leptin for Astra-Zeneca; has received the SFE 2012
Oral communication award from Novartis; has received honoraria from
Sanofi; has received payment for manuscript preparation from Elsevier
Masson; and has received travel/accommodations expenses covered or
reimbursed for meetings from Novo-Nordisk, Servier, and Lilly.
Guillaume Bidault declares that he has n o conflict of interest.
Nolwenn Briand declares that she has no conflict of interest. Anne-
Claire Guénantin declares that she has no conflict of interest. Laurence
Teyssières declares that she has no conflict of interest. Olivier Lascols
declares that he has no conflict of interest. Jacqueline Capeau declares
that she has no conflict of interest. Corinne Vigouroux has been on the
Advisory board on lipodystrophy and leptin for Astra-Zeneca; and has
received travel/accommodations expenses covered or reimbursed for
meetings from Boehringer-Ingelheim, Novo-Nordisk, Edimark Santé,
and Vitalaire.
Human and Animal Rights and Informed Consent This article does
not contain any studies with human or animal subjects performed by any
of the authors.
References
Papers of particular interest, published recently, have been
highlighted as:
Of importance
•• Of major importance
1. Garg A. Lipodystrophies: genetic and acquired body fat disorders. J
Clin Endocrinol Metab. 2011;96:331325.
2. Vigouroux C, Caron-Debarle M, Le Dour C, Magré J, Capeau J.
Molecular mechanisms of human lipodystrophies: from adipocyte
lipid droplet to oxidative stress and lipotoxicity. Int J Biochem Cell
Biol. 2011;43:86276.
3. Barroso I, Gurnell M, Crowley VE, et al. Dominant negative muta-
tions in human PPARgamma associated with severe insulin resis-
tance, diabetes mellitus and hypertension. Nature. 1999;402:8803.
4. Cao H, Hegele RA. Nuclear lamin A/C R482Q mutation in Canadian
kindreds with Dunnigan-type familial partial lipodystrophy. Hum Mol
Genet. 2000;9:10912.
5. Shackleton S, Lloyd DJ, Jackson SN, et al. LMNA, encoding lamin
A/C, is mutated in partial lipodystrophy. Nat Genet. 2000;24:1536.
6. Mag J, Delépine M, Khallouf E, et al. Identification of the gene
altered in Berardinelli-Seip congenital lipodystrophy on chromosome
11q13. Nature Genet. 2001;28:36570.
Curr Diab Rep
7. Agarwal AK, Arioglu E, De Almeida S, et al. AGPAT2 is mutated in
congenital generalized lipodystrophy linked to chromosome 9q34.
Nat Genet. 2002;31:213.
8. George S, Rochford JJ, Wolfrum C, et al. A family with severe
insulin resistance and diabetes due to a mutation in AKT2. Science.
2004;304:13258.
9. Kim C, Delépine M, Boutet E, et al. Association of a homozygous
nonsense Caveolin-1 mutation with Berardinelli-Seip Congenital
Lipodystrophy. J Clin Endocrinol Metab. 2008;93:112934.
10. Rubio-Cabezas O, Puri V, Murano I, et al. Partial lipodystrophy and
insulin resistant diabetes in a patient with a homozygous nonsense
mutation in CIDEC. EMBO Mol Med. 2009;1:2807.
11. Hayashi YK, Matsuda C, Ogawa M, et al. Human PTRF mutations
cause secondary deficiency of caveolins resulting in muscular dystro-
phy with generalized lipodystrophy. J Clin Invest. 2009;119:262333.
12. •• Gandotra S, Le Dour C, Bottomley W, et al. Perilipin deficiency
and autosomal dominant partial lipodystrophy. N Engl J Med.
2011;364:7408. Each of these studies (references 312) identified
genes involved in lipodystrophic syndromes, underlying the impor-
tance of a primary defect in adipose tissue for metabolism at the
systemic level.
13. Novelli G, Muchir A, Sangiuolo F, et al. Mandibuloacral dysplasia is
caused by a mutation in LMNA-encoding lamin A/C. Am J Hum
Genet. 2002;71:42631.
14. De Sandre-Giovannoli A, Bernard R, Cau P, et al. Lamin A truncation
in Hutchinson-Gilford progeria. Science. 2003;300:2055.
15. Eriksson M, Brown WT, Gordon LB, et al. Recurrent de novo point
mutations in lamin A cause Hutchinson-Gilford progeria syndrome.
Nature. 2003;423:2938.
16. Agarwal AK, Fryns JP, Auchus RJ, Garg A. Zinc metalloproteinase,
ZMPSTE24, is mutated in mandibuloacral dysplasia. Hum Mol
Genet. 2003;12:19952001.
17. Caron M, Auclair M, Donadille B, et al. Human lipodystrophies linked
to mutations in A-type lamins and to HIV protease inhibitor therapy are
both associated with prelamin A accumulation, oxidative stress, and
premature cellular senescence. Cell Death Differ. 2007;14:175967.
18. •• Agarwal AK, Xing C, DeMartino GN, et al. PSMB8 encoding the
beta5i proteasome subunit is mutated in joint contractures, muscle
atrophy, microcytic anemia, and panniculitis-induced lipodystrophy
syndrome. Am J Hum Genet. 2010;87:86672. This work reported
the identification of PSMB8 as the gene involved in JMP syndrome,
showing that autoinflammation can lead to lipodystrophy.
19. Kitamura A, Maekawa Y, Uehara H, et al. A mutation in the
immunoproteasome subunit PSMB8 causes autoinflammation and
lipodystrophy in humans. J Clin Invest. 2011;121:415060.
20. Ailhaud G. Adipose tissue as an endocrine organ. Int J Obes Relat
Metab Disord. 2000;24 Suppl 2:S13.
21. Bastard JP, Fève B. The secretory face of the adipose cell: a tribute to
two queens, leptin and adiponectin. Biochimie. 2012;94:20634.
22. Goossens GH, Blaak EE, van Baak MA. Possible involvement of the
adipose tissue renin-angiotensin system in the pathophysiology of
obesity and obesity-related disorders. Obes Rev. 2003;4:4355.
23. Caspar-Bauguil S, Cousin B, Bour S, et al. Adipose tissue lympho-
cytes: types and roles. J Physiol Biochem. 2009;65:42336.
24. Dirat B, Bochet L, Dabek M, et al. Cancer-associated adipocytes
exhibit an activated phenotype and contribute to breast cancer inva-
sion. Cancer Res. 2011;71:245565.
25. Shimomura I, Hammer RE, Richardson JA, et al. Insulin resistance
and diabetes mellitus in transgenic mice expressing nuclear SREBP-
1c in adipose tissue: model for congenital generalized lipodystrophy.
Genes Dev . 1998;12:318294.
26. Moitra J, Mason MM, Olive M, et al. Life without white fat: a
transgenic mouse. Genes Dev. 1998;12:3168
81.
27. Gavrilova O, Marcus-Samuels B, Graham D, et al. Surgical implan-
tation of adipose tissue reverses diabetes in lipoatrophic mice. J Clin
Invest. 2000;105:2718.
28. Colombo C, Cutson JJ, Yamauchi T, et al. Transplantation of adipose
tissue lacking leptin is unable to reverse the metabolic abnormalities
associated with lipoatrophy. Diabetes. 2002;51:272733.
29. Shimomura I, Hammer RE, Ikemoto S, Brown MS, Goldstein JL.
Leptin reverses insulin resistance and diabetes mellitus in mice with
congenital lipodystrophy. Nature. 1999;401:736.
30. Garg A. Acquired and inherited lipodystrophies. N Engl J Med.
2004;350:122034.
31. Seip M, Trygstad O. Generalized lipodystrophy, congenital and ac-
quired (lipoatrophy). Acta Paediatr Suppl. 1996;413:228.
32. Dunnigan MG, Cochrane MA, Kelly A, Scott JW. Familial
lipoatrophic diabetes with dominant transmission. A new syndrome.
Q J Med. 1974;43:3348.
33. Garg A, Peshock RM, Fleckenstein JL. Adipose tissue distribution
pattern in patients with familial partial lipodystrophy (Dunnigan
variety). J Clin Endocrinol Metab. 1999;84:1704.
34. Vantyghem MC, Vincent-Desplanques D, Defrance-Faivre F, et al.
Fertility and obstetrical complications in women with LMNA-related
familial partial lipodystrophy. J Clin Endocrinol Metab. 2008;96:
22239.
35. Haque WA, Shimomura I, Matsuzawa Y, Garg A. Serum adiponectin
and leptin levels in patients with lipodystrophies. J Clin Endocrinol
Metab. 2002;87:23958.
36. Antuna-Puente B, Boutet E, V ig ouroux C, et al. Higher adiponectin
levels in pati ents with Berardinel li-Seip congenital lipodystrophy due to
seipin compared with 1-acylglycerol-3-phosphate-o-acyltransferase-2
deficiency . J Clin Endocrinol Metab. 2010;95:14638.
37. Valerio CM, Godoy-Matos A, Moreira RO, et al. Dual-energy X-ray
absorptiometry study of body composition in patients with
lipodystrophy. Diabetes Care. 2007;30:18579.
38. Abate N, Burns D, Peshock RM, Garg A, Grundy SM. Estimation of
adipose tissue mass by magnetic resonance imaging: validation
against dissection in human cadavers. J Lipid Res. 1994;35:14906.
39. Brunzell JD, Shankle SW, Bethune JE. Congenital generalized
lipodystrophy accompanied by cystic angiomatosis. Ann Intern
Med. 1968;69:50116.
40. Decaudain A, Vantyghem MC, Guerci B, et al. New metabolic
phenotypes in laminopathies: LMNA mutations in patients with severe
metabolic syndrome. J Clin Endocrinol Metab. 2007;92:483544.
41. Semple RK, Savage DB, Cochran EK, Gorden P, ORahilly S.
Genetic syndromes of severe insulin resistance. Endocr Rev.
2011;32:498514.
42. Semple RK, Sleigh A, Murgatroyd PR, et al. Postr eceptor insulin
resistance contributes to human dyslipidemia and hepatic steatosis. J
Clin Invest. 2009;1 19:31522.
43. Thauvin-Robinet C, Auclair M, Duplomb L, et al. Mutations in
PIK3R1 cause syndromic insulin resistance with lipoatrophy. Am J
Hum Genet. 2013;93:1419.
44. Van Maldergem L, Magré J, Gedde-Dahl Jr T, et al. Genotype-
phenotype relationships in berardinelli-seip congenital lipodystrophy.
J Med Genet. 2002;39:72233.
45. Simha V, Garg A. Phenotypic heterogeneity in body fat distribution
in patients with congenital generalized lipodystrophy caused by
mutations in the AGPAT2 or Seipin genes. J Clin Endocrinol
Metab. 2003;88:54337.
46. Agarwal AK. Lysophospholipid acyltransferases: 1-acylglycerol-3-
phosphate O-acyltransferases. From discovery to disease. Curr Opin
Lipidol. 2012;23:290302. A comprehensive review about the role of
AGPAT isoforms.
47. Subauste AR, Das AK, Li X, et al. Alterations in lipid signaling
underlie lipodystrophy secondary to AGPAT2 mutations. Diabetes.
2012;61:292231.
48. Cartwright BR, Goodman JM. Seipin: from human disease to
molecular mechanism. J Lipid Res. 2012;53:104255. A recent com-
plete review on the role of seipin, in particular in lipid droplet
assembly and maintenance and in adipocyte differentiation.
Curr Diab Rep
49. Szymanski KM, Binns D, Bartz R, et al. The lipodystrophy protein
seipin is found at endoplasmic reticulum lipid droplet junctions and is
important for droplet morphology. Proc Natl Acad Sci U S A.
2007;104:208905.
50. Boutet E, El Mourabit H, Prot M, et al. Seipin deficiency alters fatty
acid Delta9 desaturation and lipid droplet formation in Berardinelli-
Seip congenital lipodystrophy. Biochimie. 2009;91:796803.
51. Yang H, Galea A, Sytnyk V, Crossley M. Controlling the size of lipid
droplets: lipid and protein factors. Curr Opin Cell Biol. 2012;24:509
16. A comprehensive r eview about the structur e of lipid dr oplets and
the mechanisms involved in their assembly and growth.
52. Ito D, Suzuki N. Seipinopathy: a novel endoplasmic reticulum stress-
associated disease. Brain. 2009;132:815.
53. Holtta-Vuori M, Salo VT, Ohsaki Y, Suster ML, Ikonen E.
Alleviation of seipinopathy-related ER stress by triglyceride storage.
Hum Mol Genet. 2013;22:115766.
54. Cao H, Alston L, Ruschman J, Hegele RA. Heterozygous CAV1
frameshift mutations (MIM 601047) in patients with atypical partial
lipodys troph y and hypertriglyce ride mia. Lipids Health Di s. 2008;7:3 .
55. Ostermeyer AG, Paci JM, Zeng Y, et al. Accumulation of caveolin in
the endoplasmic reticulum redirects the protein to lipid storage drop-
lets. J Cell Biol. 2001;152:10718.
56. Pol A, Luetterforst R, Lindsay M, et al. A caveolin dominant negative
mutant associates with lipid bodies and induces intracellular choles-
terol imbalance. J Cell Biol. 2001;152:105770.
57. Le Lay S, Briand N, Blouin CM, et al. The lipoatrophic caveolin-1
deficient mouse model reveals autophagy in mature adipocytes.
Autophagy. 2010;6:75463. Autophagy, a new pathway involved in
lipodystrophy in caveolin-1 deficient mice.
58. Blouin CM, Le Lay S, Eberl A, et al. Lipid droplet analysis in
caveolin-deficient adipocytes: alterations in surface phospholipid
composition and maturation defects. J Lipid Res. 2010;51:94556.
59. Rajab A, Straub V, McCann LJ, et al. Fatal cardiac arrhythmia and
long-QT syndrome in a new form of congenital generalized
lipodystrophy with muscle rippling (CGL4) due to PTRF-CAVIN
mutations. PLoS Genet. 2010;6:e1000874.
60. Hill MM, Bastiani M, Luetterforst R, et al. PTRF-Cavin, a conserved
cytoplasmic protein required for caveola formation and function.
Cell. 2008;132:11324.
61. Le Dour C, Schneebeli S, Bakiri F, et al. A homozygous mutation of
prelamin-A preventing its farnesylation and maturation leads to a
severe lipodystrophic phenotype: new insights into the pathogenicity
of nonfarnesylated prelamin-A. J Clin Endocrinol Metab. 2011;96:
E85662.
62. Wiltshire KM, Hegele RA, Innes AM, Brownell AK. Homozygous
lamin A/C familial lipodystrophy R482Q mutation in autosomal
recessive Emery Dreifuss muscular dystrophy. Neuromuscul
Disord. 2013;23:2658.
63. Worman HJ. Nuclear lamins and laminopathies. J Pathol. 2012;226:
31625.
64. Burke B, Stewart CL. The nuclear lamins: flexibility in function. Nat
Rev Mol Cell Biol. 2013;14:1324.
65. Vigouroux C, Magré J, Vantyghem MC, et al. Lamin A/C gene: sex-
determined expression of mutations in Dunnigan-type familial partial
lipodystrophy and absence of coding mutations in congenital and
acquired generalized lipoatrophy. Diabetes. 2000;49:195862.
66. Araújo-Vilar D, Loidi L, Domínguez F, Cabezas-Cerrato J.
Phenotypic gender differences in subjects with familial partial
lipodystrophy (Dunnigan variety) due to a nuclear lamin A/C
R482W mutation. Horm Metab Res. 2003;35:2935.
67. réziat V, Cervera P, Le Dour C, et al. LMNA mutations induce a
non-inflammatory fibrosis and a brown fat-like dystrophy of enlarged
cervical adipose tissue. Am J Pathol. 2011;179:244353. This work
reported that accumulated adipose tissue in familial partial
lipodystrophy is dystrophic, showing fibrosis and an altered differ-
entiation pattern with brown fat-like adipocytes.
68. Mory PB, Crispim F, Freire MB, et al. Phenotypic diversity in
patients with lipodystrophy associated with LMNA mutations. Eur
J Endocrinol. 2012;167:42331.
69. Caux F , Dubosclard E, Lascols O, et al. A new clinical condition linked
to a novel mutation in lamins A and C with generalized lipoatrophy,
insulin-resistant diabetes, disseminated leukomelanodermic papules, liv-
er steatosis, and cardiomyopathy. J Clin Endocrinol Metab. 2003;88:
100613.
70. Garg A, Subramanyam L, Agarwal AK, et al. Atypical progeroid
syndrome due to heterozygous missense LMNA mutations. J Clin
Endocrinol Metab. 2009;94:497183.
71. Capanni C, Mattioli E, Columbaro M, et al. Altered pre-lamin A
processing is a common mechanism leading to lipodystrophy. Hum
Mol Genet. 2005;14:1489502.
72. •• Liu GH, Barkho BZ, Ruiz S, et al. Recapitulation of premature
ageing with iPSCs from Hutchinson-Gilford progeria syndrome.
Nature. 2011;472:2215. These 2 studies highlighted the role of
progerin (prelamin A mutated in Hutchinson-Gilford progeria) in
ageing-associated cell phenotypes during differentiation and showed
that iPSC are a model for studying the pathogenesis of laminopathies.
73. •• Zhang J, Lian Q, Zhu G, et al. A human iPSC model of Hutchinson
Gilford Progeria reveals vascular smooth muscle and mesenchymal
stem cell defects. Cell Stem Cell. 2011;8:3145. These 2 studies
highlighted the r ole of progerin (prelamin A mutated in Hutchinson-
Gilford progeria) in ageing-associated cell phenotypes during differ-
entiation and showed that iPSC ar e a model for studying the patho-
genesis of laminopathies.
74. Scaffidi P, Misteli T. Lamin A-dependent misregulation of adult stem
cells associated with accelerated ageing. Nat Cell Biol. 2008;10:4529.
75. Pekovic V, Hutchison CJ. Adult stem cell maintenance and tissue
regeneration in the ageing context: the role for A-type lamins as
intrinsic modulators of ageing in adult stem cells and their niches. J
Anat. 2008;213:525.
76. Naetar N, Foisner R. Lamin complexes in the nuclear interior control
progenitor cell proliferation and tissue homeostasis. Cell Cycle.
2009;8:148893.
77. Tontonoz P, Spiegelman BM. Fat and beyond: the diverse biology of
PPARgamma. Annu Rev Biochem. 2008;77:289312.
78. Jeninga EH, Gurnell M, Kalkhoven E. Functional implications of
genetic variation in human PPARgamma. Trends Endocrinol Metab.
2009;20:3807.
79. Auclair M, Vigouroux C, Boccara F, et al. Peroxisome proliferator-
activated receptor-gamma mutations responsible for lipodystrophy
with severe hypertension activate the cellular renin-angiotensin sys-
tem. Arterioscler Thrombos Vasc Biol. 2013;33:82938. Atransla-
tional study of peroxisome proliferator-activated receptor γ transla-
tional study of peroxisome proliferator-activated rsupporting a role
for PPARγ as a regulator of blood pressure through its ability to
modulate the cellular renin-angiotensin system.
80. Li F, Gu Y, Dong W, et al. Cell death-inducing DFF45-like effector, a
lipid droplet-associated protein, might be involved in the differenti-
ation of human adipocytes. FEBS J. 2010;277:417383.
81. Gandotra S, Lim K, Girousse A, et al. Human frame shift mutations
affecting the carboxyl terminus of perilipin increase lipolysis by
failing to sequester the adipose triglyceride lipase (ATGL) coactivator
AB-hydrolase-containing 5 (ABHD5). J Biol Chem. 2011;286:
349985006.
82. Hegele RA, Cao H, Liu DM, et al. Sequencing of the reannotated
LMNB2 gene reveals novel mutations in patients with acquired
partial lipodystrophy. Am J Hum Genet. 2006;79:3839.
83. Gao J, Li Y, Fu X, Luo X. A Chinese patient with acquired partial
lipodystrophy caused by a novel mutation with LMNB2 gene. J
Pediatr Endocrinol Metab. 2012;25:3757.
84. Coffinier C, Hudon SE, Farber EA, et al. HIV protease inhibitors block
the zinc metalloproteinase ZMPSTE24 and lead to an accumulation of
prelamin A in cells. Proc Natl Acad Sci U S A. 2007;104:134327.
Curr Diab Rep
85. •• Gordon LB, Kleinman ME, Miller DT, et al. Clinical trial of a
farnesyltransferase inhibitor in children with Hutchinson-Gilford
progeria syndrome. Proc Natl Acad Sci U S A. 2012;109:16666
71. Results from the first clinical trial with farnesyl-transferase
inhibitor in Hutchinson-Gilford progeria syndrome, showing the
improvement of vascular stiffness and bone structure.
86. Donadille B, DAnella P, Auclair M, et al. Partial lipodystrophy with
severe insulin resistance and adu lt progeria Werner syn drome.
Orphanet J Rare Dis. 2013;8:106.
87. Weedon MN, Ellard S, Prindle MJ, et al. An in-frame deletion at the
polymerase active site of POLD1 causes a multisystem disorder with
lipodystrophy. Nat Genet. 2013; [In press].
88. Dahl PR, Zalla MJ, Winkelman n RK. Localized involutional
lipoatrophy: a clinicopathologic study of 16 patients. J Am Acad
Dermatol. 1996;35:5238.
89. Manolopoulos KN, Karpe F, Frayn KN. Gluteofemoral body fat as a
determinant of metabolic health. Int J Obes. 2010;34:94959.
90. Virtue S, Vidal-Puig A. Adipose tissue expandability, lipotoxicity and
the Metabolic Syndromean allostatic perspective. Biochim Biophys
Acta. 1801;2010:33849.
91. Wilson DE, Chan IF , Stevenson KB, Horton SC, Schipke C. Eucaloric
substitution of medium chain triglycerides for dietary long chain fatty
acids in acquired total lipodystrophy: effects on hyperlipop roteinemia and
endogenous insulin resistance. J Clin Endocrinol Metab. 1983;57:51723.
92. Agarwal AK, Garg A. Genetic basis of lipodystrophies and manage-
ment of metabolic complications. Annu Rev Med. 2006;57:29731 1.
93. Oral EA, Simha V, Ruiz E, et al. Leptin-replacement therapy for
lipodystrophy. N Engl J Med. 2002;346:5708.
94. Oral EA, Ruiz E, Andewelt A, et al. Effect of leptin replacement on
pituitary hormone regulation in patients with severe lipodystrophy. J
Clin Endocrinol Metab. 2002;87:31107.
95. Javor ED, Cochran EK, Musso C, et al. Long-term efficacy of leptin
replacement in patients with generalized lipodystrophy. Diabetes.
2005;54:19942002.
96. Ebihara K, Kusakabe T, Hirata M, et al. Efficacy and safety of leptin-
replacement therapy and possible mechanisms of leptin actions in
patients with generalized lipodystrophy. J Clin Endocrinol Metab.
2007;92:53241.
97. Yu X, McCorkle S, Wang M, et al. Leptinomimetic effects of the AMP
kinase activator AICAR in leptin-resistant rats: prevention of diabetes
and ectopic lipid deposition. Diabetologia. 2004;47:201221.
98. Yamauchi T, Kamon J, Waki H, et al. The fat-derived hormone
adiponectin reverses insulin resi stance associated with both
lipoatrophy and obesity. Nat Med. 2001;7:9416.
99. Calderoni DR, Ramos TM, de Castro JR, Kharmandayan P. Surgical
management of phenotypic alterations related to the Dunnigan vari-
ety of familial partial lipodystrophy. J Plast Reconstr Aesthet Surg.
2011;64:124850.
Curr Diab Rep
... Homozygous pathogenic variants that eliminate enzyme activity have been determined in the majority of CGL1 cases. Compound heterozygous or homozygous pathogenic variants with low levels of in vitro enzyme activity have also been reported (7,13). Although patients with CGL1 lack metabolically active adipose tissue, the preservation of residual mechanical adipose tissue in the palms, soles, scalp, orbital and periarticular regions, and the perineum is clinically apparent (14,15). ...
... The majority of identified variants have been classified as null pathogenic variants based on functional investigations which lead to severe disruption of the protein. Missense pathogenic variants have also been reported (7,13,18). ...
Article
Full-text available
Lipodystrophy is a heterogeneous group of disorders characterized by lack of body fat, which can be genetic or acquired. Lipodystrophy is associated with insulin resistance that can develop in childhood and youth, and usually leads to severe metabolic complications. Diabetes mellitus, hypertriglyceridemia, and hepatic steatosis ordinarily develop in these patients, and most girls suffer from menstrual abnormalities. Severe complications develop at a relatively young age, which include episodes of acute pancreatitis, renal failure, cirrhosis, and complex cardiovascular diseases, and they all are associated with serious morbidity. Treatment of lipodystrophy consists of medical nutrition therapy, exercise, and the use of anti-hyperglycemic and lipid-lowering agents. New treatment modalities such as metreleptin replacement offer a great benefit in the treatment of metabolic abnormalities secondary to lipodystrophy. Current challenges in lipodystrophy management in children and youth include, but not limited to, (1) establishing specialized centers with experience in providing care for lipodystrophy presenting in childhood and adolescence; (2) optimizing algorithms that can provide some guidance for the use of standard and novel therapies to ensure adequate metabolic control and to prevent complications; (3) educating patients and their parents about lipodystrophy management; (4) improving patient adherence to chronic therapies; (5) reducing barriers to access novel treatments; and (5) improving the quality of life of these patients and their families.
... Lipodystrophies are a group of genetic or acquired diseases characterized by abnormal adipose tissue deposition, including peripheral fat loss (lipoatrophy) and central fat accumulation (lipohypertrophy or abdominal prominence), which could be present separately or combined in the same individual [1][2][3]. These syndromes are usually, but not invariably, linked with severe metabolic complications, such as insulin resistance, diabetes, lipid abnormalities, hypertension, and hepatic steatosis [4]. Many complications of lipodystrophy are secondary to deficient adipose mass, resulting in ectopic lipid storage in the liver, muscles, and other organs, with associated insulin resistance [5]. ...
... Lipodystrophies are a group of genetic or acquired diseases characterized by abnormal adipose tissue deposition, including peripheral fat loss (lipoatrophy) and central fat accumulation (lipohypertrophy or abdominal prominence), which could be present separately or combined in the same individual [1][2][3]. These syndromes are usually, but not invariably, linked with severe metabolic complications, such as insulin resistance, diabetes, lipid abnormalities, hypertension, and hepatic steatosis [4]. Many complications of lipodystrophy are secondary to deficient adipose mass, resulting in ectopic lipid storage in the liver, muscles, and other organs, with associated insulin resistance [5]. ...
Article
Full-text available
Background: Lipodystrophies are a group of diseases which are characterized by abnormal adipose tissue deposition and are frequently associated with metabolic changes. Congenital generalized lipodystrophy is an autosomal recessive syndrome, with a prevalence < 1:10 million. Acromegaly is a rare disease, secondary to the chronic hypersecretion of growth hormone and insulin-like growth factor-1, with characteristic metabolic and somatic effects. "Acromegaloidism" is a term used for patients who manifest clinical features of acromegaly, but do not present a demonstrable hormone growth hypersecretion. The extreme shortage of subcutaneous adipose tissues and muscle hypertrophy confer an acromegaloid-like appearance in these patients. Case presentation: We describe a case of a patient with the rare combination of Berardinelli-Seip congenital lipodystrophy and acromegaly; our patient is a 63-year-old white man, who was referred to an endocrinology consultation for suspected lipodystrophy. He had lipoatrophy of upper and lower limbs, trunk, and buttocks, with muscular prominence, acromegaloid facial appearance, large extremities, and soft tissue tumescence. In addition, he had dyslipidemia and prediabetes. His fat mass ratio (% trunk fat mass/% lower limbs fat mass) was 1.02 by densitometry and he also had hepatomegaly, with mild steatosis (from an abdominal ultrasound), and left ventricular hypertrophy (from an electrocardiogram). His first oral glucose tolerance test had growth hormone nadir of 0.92 ng/mL, and the second test, 10 months afterwards, registered growth hormone nadir of 0.64 ng/mL (growth hormone nadir < 0.3 ng/mL excludes acromegaly). Pituitary magnetic resonance imaging identified an area of hypocaptation of contrast product in relation to a pituitary adenoma and he was subsequently submitted to transsphenoidal surgical resection of the mass. A pathological evaluation showed pituitary adenoma with extensive expression of growth hormone and adrenocorticotropic hormone, as well as a rare expression of follicle-stimulating hormone and prolactin. A genetic study revealed an exon 3/exon 4 deletion of the AGPAT2 gene in homozygosity. Conclusions: Congenital generalized lipodystrophy is a rare disease which occurs with acromegaloid features. As far as we know, we have described the first case of genetic lipodystrophy associated with true acromegaly. Although this is a rare association, the presence of congenital generalized lipodystrophy should not exclude the possibility of simultaneous acromegaly.
... This condition predisposes to diabetes type 2, cardiovascular diseases and hepatic or pancreatic comorbidities. FPLD type 2 (FPLD2), commonly named Dunnigan syndrome, is the most frequent FPLD and results from LMNA mutations transmitted in a dominant pattern [2][3][4]. LMNA encodes for lamin A and C proteins as the result of an alternative splicing. These proteins are type V intermediate filaments located both in the nucleoplasm and at the nuclear lamina, a meshwork underlying the inner nuclear envelope. ...
Article
Full-text available
This study details the clinical and cellular phenotypes associated with two missense heterozygous mutations in LMNA, c.1745G > T p.(Arg582Leu), and c.1892G> A p.(Gly631Asp), in two patients with early onset of diabetes mellitus, hypertriglyceridemia and non-alcoholic fatty liver disease. In these two patients, subcutaneous adipose tissue was persistent, at least on the abdomen, and the serum leptin level remained in the normal range. Cellular studies showed elevated nuclear anomalies, an accelerated senescence rate and a decrease of replication capacity in patient cells. In cellular models, the overexpression of mutated prelamin A phenocopied misshapen nuclei, while the partial reduction of lamin A expression in patient cells significantly improved nuclear morphology. Altogether, these results suggest a link between lamin A mutant expression and senescence associated phenotypes. Transcriptome analysis of the whole subcutaneous adipose tissue from the two patients and three controls, paired for age and sex using RNA sequencing, showed the up regulation of genes implicated in immunity and the down regulation of genes involved in development and cell differentiation in patient adipose tissue. Therefore, our results suggest that some mutations in LMNA are associated with severe metabolic phenotypes without subcutaneous lipoatrophy, and are associated with nuclear misshaping.
... In cases of partial fat loss, hypertrophy may occur in other fat depots 2 . Lipodystrophic syndromes are associated with metabolic dysfunction including severe insulin resistance, dyslipidemia, fatty liver and abnormal adipocytokine secretion, all of which lead to early disease complications 2,3 . Several lipodystrophic syndromes are monogenic disorders. ...
Article
Background: Lipodystrophic syndromes are rare diseases of genetic or acquired origin characterized by partial or generalized lack of body fat. Early detection and diagnosis are crucial to prevent and manage associated metabolic dysfunctions, i.e. insulin resistance, dyslipidemia, fatty liver, and diabetes, and to provide appropriate genetic counseling. By means of several representative case studies, this article illustrates the diagnostic and management challenges of lipodystrophic syndromes. Review: Berardinelli-Seip congenital lipodystrophy (BSCL) is typically diagnosed at birth, or soon thereafter, with generalized lipoatrophy and hepatomegaly secondary to hepatic steatosis. Physicians must also consider this diagnosis in adults with atypical non-autoimmune diabetes, hypertriglyceridemia, and a lean and muscular phenotype. The BSCL1 subtype due to mutations in the AGPAT2 gene can have an unusual presentation, especially in neonates and infants. Particular attention should be paid to infants presenting failure to thrive who also have hepatomegaly and metabolic derangements. The BSCL2 sub-type due to mutations in the BSCL gene tends to be more severe than BSCL1, and is characterized by greater fat loss, mild intellectual disability, earlier onset of diabetes, and higher incidence of premature death. Effective management from an earlier age may moderate the natural disease course. Partial lipodystrophies may easily be confused with common central obesity and/or metabolic syndrome. In patients with unexplained pancreatitis and hypertriglyceridemia, lipodystrophies such as familial partial lipodystrophy type 2 (FPLD2; Dunnigan type, due to LMNA mutations) should be considered. Oral combined contraceptives, which can reveal the disease by inducing severe hypertriglyceridemia, are contraindicated. Endogenous estrogens may also lead to "unmasking" of the FPLD2 phenotype, which often appears at puberty, and is more severe in females than males. Conclusions: Diet and exercise, adapted to age and potential comorbidities, are essential prerequisites for therapeutic management of lipodystrophic syndromes. Metreleptin therapy can be useful to manage lipodystrophy-related metabolic complications.
... [1][2][3][4][5][6][7][8][9][10][11][12] In HIV-infected patients, insulin resistance (IR) and DM have been related to lipodystrophy and exposure to the first generation of protease inhibitors (PI) or to D-thymidine nucleoside analogues and didanosine. 1,13 In HCV-infected patients, apart from cirrhosis from any cause, which is a well established risk factor for IR and DM, it has been suggested that HCV infection could directly induce IR and DM by interfering with glucose metabolism. 14 In addition, DM has been observed as a side effect during or after interferon (IFN)-based HCV therapy. ...
Article
Full-text available
Background Both human immunodeficiency virus (HIV) and hepatitis C virus (HCV) infections have been reportedly associated with a higher risk of diabetes mellitus (DM) but results are conflicting. Aims To determine whether there is an association between chronic HCV and the incidence of DM, and to study the role of factors such as cirrhosis, IFN‐based HCV therapy, sustained virologic response (SVR) and chronic HBV infection among patients living with HIV (PLHIV) followed in a large French multicentre cohort in the combination antiretroviral therapy (cART) era. Methods All PLHIV followed up in the Dat’AIDS cohort were eligible. Cox models for survival analysis were used to study the time to occurrence of DM. Results Among 28 699 PLHIV, 4004 patients had chronic HCV infection. The mean duration of HCV follow‐up was 12.5 ± 8.1 years. The rate ratio of DM was 2.74 per 1000 person‐years. By multivariate analysis, increasing age, body mass index>25, AIDS status, nadir CD4 cell count ≤200/mm³, detectable HIV viral load and cirrhosis (HR 2.26 95% CI 1.14‐1.18; P < 0.0001) were predictors of DM, whereas longer cART duration was associated with a lower risk of DM. Chronic HCV and HBV infection and IFN‐based HCV therapy were not associated with DM. In a subanalysis among HCV‐infected patients, SVR was not related to DM. Conclusions Our study shows that in the HIV population, cirrhosis is associated with an increased occurrence of DM, but not chronic HCV infection or duration of HCV infection.
Article
Full-text available
Obesity and diabetes represent two increasing and invalidating public health issues that often coexist. It is acknowledged that fat mass excess predisposes to insulin resistance and type 2 diabetes mellitus (T2D), with the increasing incidence of the two diseases significantly associated. Moreover, emerging evidence suggests that obesity might also accelerate the appearance of type 1 diabetes (T1D), which is now a relatively frequent comorbidity in patients with obesity. It is a common clinical finding that not all patients with obesity will develop diabetes at the same level of adiposity, with gender, genetic, and ethnic factors playing an important role in defining the timing of diabetes appearance. The adipose tissue (AT) expandability hypothesis explains this paradigm, indicating that the individual capacity to appropriately store energy surplus in the form of fat within the AT determines and prevents the toxic deposition of lipids in other organs, such as the pancreas. Thus, we posit that when the maximal storing capacity of AT is exceeded, individuals will develop T2D. In this review, we provide an insight into mechanisms by which the AT controls pancreas lipid content and homeostasis in case of obesity to offer an adipocentric perspective of pancreatic lipotoxicity in the pathogenesis of diabetes. Moreover, we suggest that improving AT function is a valid therapeutic approach to fighting obesity-associated complications including diabetes.
Article
Advances in the development of noninvasive imaging techniques have spurred investigations into ectopic lipid deposition in the liver and muscle and its implications in the development of metabolic diseases such as type 2 diabetes. Computed tomography and ultrasound have been applied in the past, though magnetic resonance-based methods are currently considered the gold standard as they allow more accurate quantitative detection of ectopic lipid stores. This review focuses on methodological considerations of magnetic resonance-based methods to image hepatic and muscle fat fractions, and it emphasizes anatomical and morphological aspects and how these may influence data acquisition, analysis, and interpretation.
Article
In the research setting, white adipose tissue (WAT) transplantation, also known as fat transplantation, is often used to understand the physiological function of adipocytes or associated stromal vascular cells such as macrophages in the context of local and systemic metabolism. The mouse is the most common animal model used where WAT from a donor is transferred either to a subcutaneous site of the same organism or to a subcutaneous region of a recipient. Here, we describe in detail the procedure for heterologous fat transplantation, and, given the need for survival surgery, peri- and postoperative care and subsequent histological confirmation of fat grafts will also be discussed.
Article
White adipocytes play a key role in maintaining whole body energy homeostasis by forming white adipose tissue (WAT). The impairment of WAT formation or WAT dysfunction is clearly associated with severe metabolic disorders. Mature adipocytes are derived from differentiated preadipocytes and are pivotal in energy storage and metabolism. Nuclear factor erythroid 2-related factor 2 (Nrf2) is a member of a family of CNC-bZIP proteins which exert their transcriptional control on genes harboring antioxidant response elements (ARE) in partnership with small musculoaponeurotic fibrosarcoma proteins. The activation of Nrf2-ARE coordinated by specific repressor Kelch-like ECH-associated protein 1 (Keap1) regulates networks of genes controlling diverse homeostatic processes involving adaptive antioxidant response and detoxification among many other adaptive responses. Interestingly, accumulating evidence indicates that Nrf2 may act as a transcription factor in regulating the formation and function of adipose tissues, including adipogenesis, lipid metabolism and insulin sensitivity. In this mini-review, an overview on the distinct roles of Nrf2 in adipocytes is provided. While highlighting the regulatory role of Nrf2 in adipogenesis, recent key findings on Nrf2 in insulin signal transduction and energy metabolism of adipocytes are also summarized.
Article
Full-text available
Laminopathies, due to mutations in LMNA, encoding A type-lamins, can lead to premature ageing and/or lipodystrophic syndromes, showing that these diseases could have close physiopathological relationships. We show here that lipodystrophy and extreme insulin resistance can also reveal the adult progeria Werner syndrome linked to mutations in WRN, encoding a RecQ DNA helicase. We analysed the clinical and biological features of two women, aged 32 and 36, referred for partial lipodystrophic syndrome which led to the molecular diagnosis of Werner syndrome. Cultured skin fibroblasts from one patient were studied. Two normal-weighted women presented with a partial lipodystrophic syndrome with hypertriglyceridemia and liver steatosis. One of them had also diabetes. Both patients showed a peculiar, striking lipodystrophic phenotype with subcutaneous lipoatrophy of the four limbs contrasting with truncal and abdominal fat accumulation. Their oral glucose tolerance tests showed extremely high levels of insulinemia, revealing major insulin resistance. Low serum levels of sex-hormone binding globulin and adiponectin suggested a post-receptor insulin signalling defect. Other clinical features included bilateral cataracts, greying hair and distal skin atrophy. We observed biallelic WRN null mutations in both women (p.Q748X homozygous, and compound heterozygous p.Q1257X/p.M1329fs). Their fertility was decreased, with preserved menstrual cycles and normal follicle-stimulating hormone levels ruling out premature ovarian failure. However undetectable anti-mullerian hormone and inhibin B indicated diminished follicular ovarian reserve. Insulin-resistance linked ovarian hyperandrogenism could also contribute to decreased fertility, and the two patients became pregnant after initiation of insulin-sensitizers (metformin). Both pregnancies were complicated by severe cervical incompetence, leading to the preterm birth of a healthy newborn in one case, but to a second trimester-abortion in the other. WRN-mutated fibroblasts showed oxidative stress, increased lamin B1 expression, nuclear dysmorphies and premature senescence. We show here for the first time that partial lipodystrophy with severe insulin resistance can reveal WRN-linked premature aging syndrome. Increased expression of lamin B1 with altered lamina architecture observed in WRN-mutated fibroblasts could contribute to premature cellular senescence. Primary alterations in DNA replication and/or repair should be considered as possible causes of lipodystrophic syndromes.
Article
Full-text available
DNA polymerase δ, whose catalytic subunit is encoded by POLD1, is responsible for lagging-strand DNA synthesis during DNA replication. It carries out this synthesis with high fidelity owing to its intrinsic 3'- to 5'-exonuclease activity, which confers proofreading ability. Missense mutations affecting the exonuclease domain of POLD1 have recently been shown to predispose to colorectal and endometrial cancers. Here we report a recurring heterozygous single-codon deletion in POLD1 affecting the polymerase active site that abolishes DNA polymerase activity but only mildly impairs 3'- to 5'-exonuclease activity. This mutation causes a distinct multisystem disorder that includes subcutaneous lipodystrophy, deafness, mandibular hypoplasia and hypogonadism in males. This discovery suggests that perturbing the function of the ubiquitously expressed POLD1 polymerase has unexpectedly tissue-specific effects in humans and argues for an important role for POLD1 function in adipose tissue homeostasis.
Article
Leptin is important in regulating energy homeostasis. Severe lipodystrophy is associated with leptin deficiency and insulin resistance, hypertriglyceridemia, and hepatic steatosis. Leptin deficiency is also associated with abnormalities of the pituitary hormones in rodent models and patients with congenital absence of leptin. We inquired whether similar abnormalities are seen in patients with lipodystrophy and whether replacement of leptin will make an impact on the regulation of pituitary hormones. Seven female patients (aged 15-42 yr, all diabetic) with lipodystrophy and serum leptin levels less than 4 mg/liter were treated with recombinant methionyl-human leptin (recombinant leptin) in physiological doses in an open-labeled study. The following parameters were evaluated before and at 4 months of leptin treatment: menstrual history, pelvic ultrasonogram, LHRH, TRH, and CRH tests. While on recombinant leptin, mean serum leptin concentration increased from 1.3 +/- 0.3 mg/liter to 11.1 +/- 2.5 mg/liter. Only one of five patients who had intact reproductive systems was cycling normally before leptin therapy, and all five had normal menses by the fourth month of leptin therapy. Serum E2 concentrations increased (110 +/- 44 pmol/liter vs. 546 +/- 247 pmol/liter, P = 0.002), serum T concentrations decreased (3.5 +/- 3.0 nmol/liter vs. 1.3 +/- 0.7 nmol/liter, P = 0.055), and the attenuated LH response to LHRH was corrected with therapy. Serum T(3) and free T(4) were in the normal range before leptin therapy and did not change. However, serum TSH concentrations fell from 2.2 +/- 1.1 microU/ml to 1.2 +/- 0.7 microU/ml (P < 0.001). The percent increase in TSH following TRH administration was similar before (560%) and at 4 months (580%) of leptin therapy. The mean nonstimulated ACTH and cortisol concentrations were, respectively, 6.0 +/- 3.4 pmol/liter and 680 +/- 280 nmol/liter before leptin and did not change after 4 months of therapy (4.2 +/- 1.2 pmol/liter, P = 0.11 and 453 +/- 142 nmol/liter, P = 0.13, respectively). The ACTH and cortisol responses to CRH stimulation were normal both before and after therapy. Leptin replacement improved menstrual abnormalities and low E2 levels and corrected the attenuated LH response to LHRH in a group of young women with lipodystrophy and leptin deficiency. These results add to the growing body of evidence that metabolic signals such as leptin play a role in neuroendocrine regulation.
Article
DNA polymerase δ, whose catalytic subunit is encoded by POLD1, is responsible for lagging-strand DNA synthesis during DNA replication. It carries out this synthesis with high fidelity owing to its intrinsic 3'- to 5'-exonuclease activity, which confers proofreading ability. Missense mutations affecting the exonuclease domain of POLD1 have recently been shown to predispose to colorectal and endometrial cancers. Here we report a recurring heterozygous single-codon deletion in POLD1 affecting the polymerase active site that abolishes DNA polymerase activity but only mildly impairs 3'- to 5'-exonuclease activity. This mutation causes a distinct multisystem disorder that includes subcutaneous lipodystrophy, deafness, mandibular hypoplasia and hypogonadism in males. This discovery suggests that perturbing the function of the ubiquitously expressed POLD1 polymerase has unexpectedly tissue-specific effects in humans and argues for an important role for POLD1 function in adipose tissue homeostasis.
Article
Short stature, hyperextensibility of joints and/or inguinal hernia, ocular depression, Rieger anomaly, and teething delay (SHORT) syndrome is a developmental disorder with an unknown genetic cause and hallmarks that include insulin resistance and lack of subcutaneous fat. We ascertained two unrelated individuals with SHORT syndrome, hypothesized that the observed phenotype was most likely due to de novo mutations in the same gene, and performed whole-exome sequencing in the two probands and their unaffected parents. We then confirmed our initial observations in four other subjects with SHORT syndrome from three families, as well as 14 unrelated subjects presenting with syndromic insulin resistance and/or generalized lipoatrophy associated with dysmorphic features and growth retardation. Overall, we identified in nine affected individuals from eight families de novo or inherited PIK3R1 mutations, including a mutational hotspot (c.1945C>T [p.Arg649Trp]) present in four families. PIK3R1 encodes the p85α, p55α, and p50α regulatory subunits of class IA phosphatidylinositol 3 kinases (PI3Ks), which are known to play a key role in insulin signaling. Functional data from fibroblasts derived from individuals with PIK3R1 mutations showed severe insulin resistance for both proximal and distal PI3K-dependent signaling. Our findings extend the genetic causes of severe insulin-resistance syndromes and provide important information with respect to the function of PIK3R1 in normal development and its role in human diseases, including growth delay, Rieger anomaly and other ocular affections, insulin resistance, diabetes, paucity of fat, and ovarian cysts.