ArticlePDF Available

Cardiac Manifestations of Congenital Generalized Lipodystrophy

Authors:

Abstract and Figures

Congenital lipodystrophy is a rare genetic disorder characterized by a near-complete absence of fat cells, hypoleptinemia leading to a voracious appetite, and marked insulin resistance. This article focuses on the known cardiovascular manifestations of patients with congenital lipodystrophy, including cardiomyopathy, cardiac arrhythmias, and accelerated atherosclerosis arising from a markedly deranged metabolic milieu. Future research that targets leptin deficiency (metreleptin) and apoC3 mRNA (antisense oligonucleotide) could open a window for potential pharmacological treatment of this challenging disorder.
No caption available
… 
No caption available
… 
Content may be subject to copyright.
VOLUME 3 4, NUMBER 4, FA LL 2016 181
FEATURE ARTICLE
Cardiac Manifestations of Congenital
Generalized Lipodystrophy
Vani P. Sanon,1 Yehuda Handelsman,2 Son V. Pham,1 and Robert Chilton1
Congenital generalized lipo-
dystrophy (CGL), also called
Berardinelli-Seip congenital li-
podystrophy, is a rare genetic disorder
characterized by loss of adipose tissue
and marked insulin resistance. A hall-
mark of this disorder is a low leptin
level, leading to a voracious appetite
in aected individuals. ere appear
to be ~300–500 cases of CGL world-
wide (1). Mutations in a variety of
genes can lead to the various subtypes
of congenital lipodystrophy (1). At
the cellular level, dysfunctional adipo-
cytes are unable to store triglycerides,
leading to high chylomicron levels.
e disorder is characterized by pro-
tean clinical manifestations, including
diabetes, pancreatitis, fatty liver, and
endothelial cell dysfunction (2).
Berardinelli (3) first described
CGL in a 2.5-year-old boy in Brazil
in 1954. Subsequently, in 1959,
Seip (4) described three more cases
in Norway. CGL has an autosomal
recessive pattern of inheritance. It
has multisystemic manifestations,
including muscular hypertrophy,
hypertrophic cardiomyopathy, hep-
atomegaly, steatohepatitis, acanthosis
nigricans, hypertriglyceridemia, and
diabetes (5). Aected patients develop
a hypertrophic “lipotoxic” cardio-
myopathy, with both diastolic and
systolic dysfunction, and have a pre-
dilection for cardiac dysrhythmias
and early sudden cardiac death (6).
In this article, we summarize the
literature on this rare genetic dis-
order, focusing on the translational
biology of the disease, its cardiac
manifestations, and novel treatment
considerations.
Genetics and Translational
Biology of CGL
CGL has four subclinical phenotypes
(CGL1, -2, -3, and -4) secondary to
mutations in the acylglycerol-3-phos-
phate-O-acyltransferase (AGPAT2)
gene, the gamma-3–linked gene lo-
cated at chromosome 11q13 (BSCL2),
caveolin-1 (CAV1), and RNA poly-
merase 1 and transcript release fac-
tor (PTRF), respectively (7–12)
(Figure 1). Agarwal et al. (9) report-
ed mutations in many genes, one of
which is the AGPAT2 gene found on
chromosome 9 in humans. is gene
has been found to code the protein lo-
cated in the endoplasmic reticulum,
aecting phospholipid biosynthesis.
IN BRIEF Congenital lipodystrophy is a rare genetic disorder characterized
by a near-complete absence of fat cells, hypoleptinemia leading to a voracious
appetite, and marked insulin resistance. This article focuses on the known car-
diovascular manifestations of patients with congenital lipodystrophy, including
cardiomyopathy, cardiac arrhythmias, and accelerated atherosclerosis arising
from a markedly deranged metabolic milieu. Future research that targets
leptin deciency (metreleptin) and apoC3 mRNA (antisense oligonucleotide)
could open a window for potential pharmacological treatment of this chal-
lenging disorder.
1Division of Cardiology, University of Texas
Health Science Center at San Antonio and
Audie L. Murphy VA Hospital, San Antonio,
TX
2Metabolic Institute of America, Tarzana, CA
Corresponding author: Robert Chilton,
chiltonr@gmail.com
DOI: 10.2337/cd16-0002
©2016 by the American Di abetes A ssoci ation .
Readers may us e this article as l ong as th e work
is properly cited , the use i s educ ation al and n ot
for prot, and the work is not altered . See http://
creativecomm ons.org/lice nses/by-nc-nd/3.0
for det ails .
182 CLINICAL.DIABETESJOURNALS.ORG
FEATURE ARTICLE
Severeal other mutations associated
with CGL have been described in the
literature (1).
At the cellular level, the dys-
functional adipocytes in CGL are
unable to store triglycerides. Figure
2 illustrates dierences between an
adipocyte of an obese individual
and one of a person with CGL. It
is noteworthy that, in obesity, the
enlarged adipocyte is no longer able
to store triglycerides and, in CGL,
the abnormal adipocyte is unable to
store triglycerides. Both conditions
lead to increased circulating levels
of free fatty acids (FFAs). It is well
known from numerous studies that
high levels of FFAs increase cardio-
vascular risk (13). Cardiovascular
interest in CGL comes, in part, from
the marked increase in circulating
FFAs found in this condition.
Cardiac Manifestations of CGL
e cardiac manifestations of CGL
were described as early as 1959 (4). A
variety of cardiac issues have been de-
scribed, ranging from dilated cardio-
myopathy to hypertrophic cardiomy-
opathy and severe biventricular heart
failure. In addition, patients have
been noted to have both brady- and
tachyarrhythmias, particularly long
QT syndrome and sudden cardiac
death. Following is a summary of
the literature on CGL as it pertains
to the heart; ~200 cases of CGL (or
Berardinelli-Seip syndrome) have
been reported (14,15).
Rajab et al. (16) described the
cardiac manifestations of CGL4.
Patients with CGL4 have a wide
variety of clinical manifestations,
including cardiac dysrhythmias, joint
involvement, skeletal and smooth
muscle hypertrophy, and myopa-
thies. Individuals with CGL4 are
described to have mutations in the
PT R F - C AVIN gene, which in turn
interferes with the functioning of
CAV1 and CAV3 , manifesting as
lipodystrophy or myopathy, respec-
tively (17). At the cardiac physiology
level, these mutations adversely aect
the functioning of various ion chan-
nels in the heart, including the nodal
pacemaker channel hyperpolariza-
tion-activated cyclic nucleotide-gated
potassium channel 4 (HCN4 ), volt-
age-gated Na+ channel (Nav1.5,
SCN5A), and the L-type Ca2+ chan-
nels (C ACN A1C ), leading to various
dysrhythmias. Mutations in HCN4
can cause sinus node dysfunction/sick
sinus syndrome, whereas mutations
in SCN5A, CAV3, and CAC NA1C
are linked to sudden cardiac death,
ventricular tachycardia, and long QT
syndromes (LQT3, -8, and -9) (17)
(Figure 3).
Rajab et al. (16) described 11
cases of CGL, of which 6 experi-
enced sudden cardiac death at an
early age. ey reported the case of
a 14-year-old boy with CGL who
was found to have had multiple epi-
sodes of palpitations and syncope
secondary to both ventricular and
supraventricular tachycardia, as well
as sinus bradycardia. Electrocardio-
gram (ECG) revealed a prolonged
corrected QT of ~ 450–480 ms, for
indicating long QT syndrome. Long
QT syndrome is a congenital disor-
der characterized by ECG evidence of
a prolonged QT interval, which may
lead to ventricular tachyarrhythmias,
FIGURE 1. Subtypes of congenital lipodystrophy. Adapted from Ref. 1.
FIGURE 2. An adipocyte in an obese individual is contrasted with a dysfunctional
adipocyte in congenital lipodystrophy. It is noteworthy that in obesity, the enlarged
adipocyte is no longer able to store triglycerides, and in CGL, the abnormal adipo-
cyte is unable to store triglycerides. Both conditions lead to increased circulating
FFAs. TRG, triglycerides.
VOLUME 3 4, NUMBER 4, FA LL 2016 183
s a n o n e t a l .
FEATURE ARTICLE
syncope, cardiac arrest, or sudden
cardiac death. Another patient was
noted to have syncope, with a cardiac
loop recorder revealing supraven-
tricular and ventricular tachycardia
and frequent ventricular extrasysto-
les; eventually, at the age of 13 years,
this patient sustained cardiac arrest
secondary to ventricular brillation
that was refractory to debrillation.
Another ve patients reported by
Rajab et al. sustained sudden cardiac
death in their teens. Postmortem
examinations of these children were
precluded because they lived in rural
areas of Oman.
Shastry et al. (18) described ve
patients with CGL4, associated with
mutations in the PTRF gene. Two
of these patients showed catechol-
aminergic polymorphic ventricular
tachycardia on exercise treadmill
testing, three had normal left ven-
tricular (LV) size and function on
transthoracic echocardiography, and
one had hypertension.
Rahman et al. (19) described
CGL in two Pakistani children.
From the cardiac standpoint, both
children demonstrated echocardio-
graphic evidence of LV hypertrophy,
although LV function was found to
be normal. One child had a small
patent foramen ovale, and the other
had a small secundum atrial septal
defect. Electrocardiography of the
aected children showed a right bun-
dle branch pattern.
Bjornstad et al. (6) reported
the cardiac manifestations of CGL
in seven patients. ere were four
deaths in their case series, in patients
ranging in age from 24 to 37 years,
with a mean age of death of 32
years. Biventricular hypertrophy
and dysfunction with pulmonary
hypertension was also described.
Echocardiographically, the thick-
ened myocardium showed evidence
of vacuolization or intramyocardial
channels. Moreover, on histopatho-
logical assessment of endomyocardial
biopsies, diuse interstitial deposition
of collagen was noted in addition to
marked subendocardial brosis. ere
was no disarray in terms of myocyte
arrangement; however, the myocytes
were hypertrophic, with increases in
the diameters of the nuclei and bers.
Rheuban et al. (20) described the
cardiac manifestations of total gener-
alized lipodystrophy in four patients.
These patients were between the
ages of 16 and 23 years, and echo-
cardiography revealed symmetric
hypertrophic cardiomyopathy and
preserved systolic function. Autopsy
of one patient showed hypertrophy of
the myocardial bers.
Klar et al. (21) described a case
of CGL in a 13-year-old girl who
was found on echocardiography to
have asymmetric septal hypertrophy
without evidence of outow tract
obstruction. Marked myocardial
hypertrophy in a 6-month-old child
with insulin resistance and CGL
was described by Gener et al. (22).
Viegas et al. (23) described the cardio-
vascular manifestations of a woman
with total generalized lipodystrophy
who presented with decompensated
heart failure. Transthoracic echocar-
diography revealed severe concentric
LV hypertrophy with hyperdynamic
LV function and advanced diastolic
dysfunction with a restrictive mitral
inow pattern. Cardiac MRI con-
rmed LV hypertrophy. e patient
underwent diuresis and was started
on calcium channel blockers, with
marked clinical improvement.
Cardiac Imaging in CGL
Lupsa et al. (24) described the echocar-
diographic features of 25 patients with
CGL; 18 had an increased LV mass, 4
had LV dysfunction, 1 had a patent
ductus arteriosus, 1 had concentric
remodeling, and 1 had moderate LV
dysfunction and dilation. Nelson et
al. (25) used MRI and localized pho-
ton spectroscopy to demonstrate that
patients with CGL had a myocardial
triglyceride content that was three-
fold higher than in control subjects
(0.6–0.2 vs. 0.2–0.1%, P = 0.004).
In addition, patients with CGL were
found to have increased LV mass in-
dex, LV concentricity, and LV hyper-
trophy independent of blood pressure.
Interestingly, this group described
MRI evidence of pericardial fat in
CGL patients that was in contrast to
previous autopsy studies showing an
absence of pericardial fat (26).
Cardiac Autopsy Findings
in CGL
Autopsy studies from Bjornstad’s
CGL group (6) revealed “moderate-
ly symmetrically enlarged heart with
FIGURE 3. CGL4 (PTRF mutation) has been associated with cardiac arrhyth-
mias. Rajab et al. (16) characterized patients with fatal cardiac arrhythmias and long
QT syndrome and reported patients with sudden death in the teenage years. The
treatment of these patients is best evaluated by an electrophysiologist because of the
complexity related to polymorphic ventricular tachycardia seen in CGL4 patients.
CGL2 patients also are at increased risk, and avoidance of exercise might be consid-
ered, but this remains unclear. Napolitano and Priori (33) reported on the diagnosis
and treatment of patients with catecholaminergic polymorphic ventricular tachycar-
dia, which has been seen in lipodystrophy.
184 CLINICAL.DIABETESJOURNALS.ORG
FEATURE ARTICLE
normal cusps.” e histopathological
examination showed “mild thickening
of the smaller intramural coronary ar-
teries with intimal brosis, as well as
a moderate subendocardial collagen
deposition. Larger intramural arter-
ies were normal, and the epicardial
branches revealed only sparse focal
intimal brosis. e myocytes were
slightly hypertrophic and normally
arranged. e content of perivascular
and subepicardial fat was decreased.
One case in particular was described
as having concentric hypertrophy
with the posterior wall and the inter-
ventricular septum, both measuring
up to 16 mm, whereas another case
had asymmetrical septal hypertrophy.
Haque et al. (27) described the
autopsy findings of two patients
with familial partial lipodystrophy,
Dunnigan variety. e rst case was
of a white female who died at the age
of 66 years. e heart was enlarged
(weight 670 g vs. normal weight of
250–300 g in adult women). e
authors reported nding old myo-
cardial scars, but no new infarct was
apparent. Moderate to severe ath-
erosclerosis of the coronary arteries,
proximal aorta, and carotid arteries
was noted.
Accelerated Atherosclerosis
in CGL
Atherosclerotic and vascular disease in
patients with CGL may stem from a
number of metabolic derangements.
Marked elevations in triglyceride and
insulin levels with low HDL choles-
terol levels have been well document-
ed to be associated with increased ath-
erosclerosis, and people with CGL t
this metabolic prole (28). In addi-
tion, fatty liver, which is frequently
associated with lipodystrophy, is also
known to be associated with an in-
creased risk for cardiovascular disease
(CVD) (29).
e National Institutes of Health
(NIH) and FHA101 trials were
open-label, single-arm, U.S. Food
and Drug Administration regulatory
trials that focused on the long-term
safety and ecacy of metreleptin for
the treatment of patients with CGL
(30,31). Goldens (31) described the
burden of atherosclerotic and CVD
in this small cohort of CGL patients:
72 patients with CGL in the NIH tri-
als and 28 patients with CGL in the
FHA101 trial. In the NIH trials, two
patients (2.8%) had a medical history
of coronary artery disease (CAD),
and four patients (5.6%) had cardio-
myopathy (two with hypertrophic
cardiomyopathy, one with cardio-
myopathy, and one with decreased
ejection fraction). In the FHA101
trial, seven (28%) of 25 patients
had CVD (four with CAD, one
with aortic atherosclerosis, one with
myocardial infarction, and one with
ischemic cardiomyopathy). Needless
to say, older patients with CGL may
accrue a higher risk for atherosclerotic
cardiovascular events over time. e
FHA101 trial had a high proportion
of adults with CGL in contrast to
the NIH trials, in which more than
half of the patients were <18 years of
age, which likely explains the higher
proportion of CVD in the FHA101
cohort. However, further studies and
longer-term follow-up of individuals
with CGL are needed to better under-
FIGURE 4. The cardiac manifestations of CGL are wide-ranging. Involvement
of the cardiovascular system is very apparent, ranging from sudden death to likely
increased atherosclerotic risk.
TABLE 1. Treatment Considerations in Lipodystrophy
Diagnosis can lead to major anxiety for patients and their families.
Cosmetic surgery to address fat loss can be considered with caution.
Dietary considerations: high-carbohydrate, low-fat diet
Improves marked chylomicronemia but may increase VLDL cholesterol
Fibric acid agents and peroxisome proliferator–activated receptor
agents may be helpful but are untested
Fish oil supplementation should be considered
There are special cardiovascular concerns with CGL4 because of
arrhythmia risk:
Avoid strenguous exercise
Possible role for β-blockers
Electrophysiology consultation should be given strong consideration
Newest consideration: metreleptin
Leads to improved metabolic parameters
Glucose, triglyercides, fat ty organ disease
VOLUME 3 4, NUMBER 4, FA LL 2016 185
s a n o n e t a l .
FEATURE ARTICLE
stand these patients’ predilection for
atherosclerotic vascular disease.
Conclusion
CGL is a rare genetic disorder with
unusual cardiac manifestations
(Figure 4). Cardiac involvement
may include electrophysiological ab-
normalities resulting from long QT
syndrome and a predisposition to
tachyarrhythmias, including cate-
cholaminergic polymorphic ventric-
ulartachycardia and sudden cardiac
death. Patients with CGL may devel-
op heart failure and advanced diastolic
dysfunction from marked LV hyper-
trophy that may be either symmet-
rical or asymmetrical. Histologically,
the absence of rearrangement of
myocardial bers in CGL dierenti-
ates this condition from hypertrophic
cardiomyopathy. In addition, because
of increased circulating FFAs and low
HDL cholesterol levels, such patients
may develop accelerated atherosclero-
sis. Patients with this rare genetic dis-
order require cardiac care focused on
the diagnosis and treatment of heart
failure, appropriate screening and
management of rhythm disorders,
and aggressive treatment of dyslipid-
emia (Table 1). Since leptin deciency
is a key abnormality in lipodystrophy,
leptin replacement has been found to
ameliorate insulin resistance, hyper-
glycemia, hypertriglyceridemia, and
hepatic steatosis (32). Future direc-
tions for treatment could include
novel pharmacological therapies that
target leptin deciency (metreleptin)
and hypertriglyceridemia via inhibi-
tion of apoC3 mRNA (antisense oli-
gonucleotide) (32–34).
Duality of Interest
No potential con icts of interest relevant to
this article were reported.
References
1. Patni N, Garg A. Congen ital generalized
lipodystroph ies: new insig hts into meta-
bolic dysfunction. Nat Rev Endocr inol
2015;11:52 2–534
2. Fioren za CG, Chou SH, Mantzoros
CS. Lipodystrophy: pathophysiology and
advanc es in treatment. Nat Rev Endocrinol
2011;7:137–150
3. Berardinelli W. An undiagnosed endo-
crinometabolic syndrome: report of 2 cases.
J Clin Endocr inol Met ab 1954;14:193–204
4. Seip M. Lipodystrophy and gigantism
with associated endocrine manifestations: a
new diencephalic syndrome? Acta Paediatr
1959;48:555 574
5. Fu M, Kazlauskaite R, Baracho Mde F,
et al. Mutations in Gng3lg and AGPAT2 in
Berardinelli-Seip congenital lipodystrophy
and Bru nzell syndrome: phenoty pe variabi l-
ity suggests i mportant modier effects. J
Clin Endocr inol Met ab 2004;89:2916–2922
6. Bjornstad PG, Foerster A, Ihlen H.
Cardiac nd ings i n generali zed l ipodystro-
phy. Acta Paed iatr 1996;413(Suppl.):39– 43
7. Van Maldergem L, Magre J, Khallouf TE,
et al. Genotyp e-phenotype relationsh ips in
Berardinelli-Seip congenital lipodystrophy.
J Med Genet 2002;39:722–733
8. Simha V, Garg A. Phenoty pic heteroge-
neity in body fat distribution in patients
with congenital generalized lipodystrophy
caused by mutations in the AGPAT2 or
seipi n genes. J Clin Endocrinol Metab
2003;88:5 433–5437
9. Agarwal A K, Arioglu E, De Almeida S, et
al. AGPAT2 is mutated in congenital gener-
alized lipodystrophy linked to chromosome
9q34. Nat Genet 2002;31:21–23
10. Magre J, Delepine M, K hallouf E, et
al. Identication of the gene altered in
Berardinelli-Seip congenital lipodystro-
phy on chromosome 11q13. Nat Genet
20 01;28:365–370
11. Kim CA, Delepine M, Boutet E, et al.
Association of a homozygous nonsense
caveolin-1 mutation with Berardinel li-Seip
congen ital l ipodystrophy. J Clin Endocrinol
Me t ab 2 008 ;93:112 9 –1134
12. Hayashi YK. Human PTRF mutations
cause secondar y deciency of caveolins
resulting in muscular dystrophy with
generalized lipodystrophy. J Clin I nvest
2009;119:2623–2633
13. Pilz S, Marz W. Free fatty acids as a
cardiovascular risk factor. Clin Chem Lab
Med 2008;46:429– 434
14. Agarwal AK, Garg A. Genetic disorders
of adipose tissue development, different ia-
tion, and death. A nn Rev Genomics Hum
Ge net 2 0 06 ;7:175–199
15. Machado PV, Daxbacher EL, Obadia
DL, Cunha EF, Alves Mde F, Mann D.
Do you know this syndrome? Berardinelli-
Seip syndrome. An Bras Dermatol
2013;8 8:1011–1013
16. Rajab A, Straub V, McCann LJ, et al.
Fatal cardiac arrhythmia and long- QT
syndrome in a new form of congenit al gen-
eralized lipodystrophy w ith muscle rippling
(CGL4) due to PTRF-CAVIN mut ations.
PLo S G en et 2010 ;6:e100 08 74
17. Balijepal li RC, Kamp TJ. Caveolae, ion
channels and cardiac arrhyth mias. Prog
Biophys Mol Biol 20 08;98:149–160
18. Shastry S, Delgado MR, Dirik E,
Turkmen M, Agarwal AK, Garg A.
Congen ital generaliz ed lipodystrophy, type
4 (CGL4) assoc iated with myopathy due to
novel PTRF mutations. Am J Med G enet A
2010;152A :2245 –2 253
19. Rahman OU, Khawar N, Khan MA, et
al. Deletion mutation in BSCL2 gene under-
lies c ongen ital generaliz ed lipodystrophy in
a Pakistani family. Diagn Pathol 2013;8:78
20. Rheuban KS, Blizzard RM, Parker
MA, Car ter T, Wilson T, Gutgesell HP.
Hyper trophic cardiomyopathy in tota l lipo -
dystrophy. J Pediatr 1986;109:301–302
21. Klar A, Brand A, Hurv itz H, Gross-
Kieselstein E, Bransk i D. Cardiomyopathy
in lipodystrophy and the spec icity spillover
hypothesis. Isr J Med Sc i 1993;29:50–52
22. Gef fner ME , Golde DW. Selective
insulin action on skin, ovary, and heart
in insuli n-resistant state s. Diab etes Care
1988;11:500–505
23. Viegas RF, Diniz RV, Viega s TM,
Lira EB, A lmeida DR. Card iac involve-
ment in total general ized lipodystrophy
(Bera rdinelli-Seip syndrome). Arq Bras
Cardiol 2000;75:243–248
24. Lupsa BC, Sachdev V, Lungu AO,
Rosing DR, Gorden P. Cardiomyopathy in
congen ital a nd acquire d generali zed lipo-
dystrophy: a clin ical asse ssment. Medicine
(Baltimore) 2010;89:245–250
25. Nelson MD, Victor RG, Szczepaniak
EW, Simha V, Garg A, Szczepa niak LS.
Cardiac steatosis a nd left ventricular
hypertrophy in patients with generalized
lipodystrophy as deter mined by magnetic
resonance spectrosc opy and imagi ng. Am J
Ca rd iol 2013;112:1019–10 24
26. Chandalia M, Garg A, Vuitch F, Nizzi
F. Postmortem ndi ngs in c ongen ital gen-
eralized lipodystrophy. J Clin Endocrinol
Metab 1995;80:3077–3081
27. Haque WA, Vuitch F, Garg A. Post-
mortem ndings in familial partial
lipodystrophy, Dun niga n variety. Diabet
Med 2002;19:1022–1025
28. Oral EA, Chan JL. Rationale for
lepti n-replacement therapy for severe lipo-
dystrophy. Endocr Pract 2010;16:324–333
29. Rodriguez A, Mastronard i CA, Paz-
Filho GJ. New advance s in the treatment
of generalized lipodystrophy: role of
metreleptin. Ther Clin Risk Manag
2015;11:1391–140 0
30. Colman C. Myalept (metreleptin).
Presentation at a U.S. Food and Dr ug
Administration Adv isor y Committee
Meeting, 11 December 2013. Available
online from htt p://ww w.fda.gov/dow nloads /
AdvisoryComm ittees/CommitteesMeeting
Materials/Drugs/Endocrinologicand
MetabolicDr ugsAdvisoryCommittee/
UCM379648.pdf. Acc ess ed 3 May 2016
186 CLINICAL.DIABETESJOURNALS.ORG
FEATURE ARTICLE
31. Goldens J. Metreleptin clinical efcacy
and safety review. Presentation at a U.S.
Food and Drug Administration Advisory
Comm itte e Meeti ng, 11 Dece mber 2013.
Available on line from http://www.fda.gov/
downloads/AdvisoryCommittees/
CommitteesMeetingMaterials/Drugs/
EndocrinologicandMetabolicDrugs
Advi soryCom mittee/ UCM379648.pdf.
Accessed 3 May 2016
32. Oral EA, Simha V, Ruiz E, et al. Leptin-
replacement therapy for lipodystrophy. N
Engl J Med 2002;346:570–578
33. ClinicalTrials.gov. Safety, tolerabil ity,
and pharmacokinetic study of ISIS ApoC-
III Rx in hypertriglyceride mia. Available
from http://clinicaltrials.gov/ct2/show/
NCT01529424. Accessed 22 March 2016
34. Meehan CA, Coch ran E, Kassai A,
Brown RJ, Gorden P. Metrelepti n for
injection to treat the complications of leptin
deciency in patients with congenital or
acquired generalized lipodystrophy. Exper t
Rev Clin Pharmacol 2016;9:59–68
... The pathological changes characterized by LV hypertrophy, cardiac fibrosis, diastolic and systolic dysfunction, and metabolic disorders originating from obesity alone are also known as obesity cardiomyopathy [6,7]. Interestingly, lipodystrophy manifested by generalized or partial loss of adipose tissue, can also cause cardiomyopathy, regardless of whether the etiology of lipodystrophy is congenital or acquired [8][9][10]. Just like obesity cardiomyopathy, myocardial hypertrophy is the most prominent pathological change of lipodystrophy cardiomyopathy. ...
Article
Full-text available
Aim: Adipose tissue (AT) dysfunction that occurs in both obesity and lipodystrophy is associated with the development of cardiomyopathy. However, it is unclear how dysfunctional AT induces cardiomyopathy due to limited animal models available. We have identified vacuolar H⁺-ATPase subunit Vod1, encoded by Atp6v0d1, as a master regulator of adipogenesis, and adipose-specific deletion of Atp6v0d1 (Atp6v0d1AKO) in mice caused generalized lipodystrophy and spontaneous cardiomyopathy. Using this unique animal model, we explore the mechanism(s) underlying lipodystrophy-related cardiomyopathy. Methods and Results: Atp6v0d1AKO mice developed cardiac hypertrophy at 12 weeks, and progressed to heart failure at 28 weeks. The Atp6v0d1AKO mouse hearts exhibited excessive lipid accumulation and altered lipid and glucose metabolism, which are typical for obesity- and diabetes-related cardiomyopathy. The Atp6v0d1AKO mice developed cardiac insulin resistance evidenced by decreased IRS-1/2 expression in hearts. Meanwhile, the expression of forkhead box O1 (FoxO1), a transcription factor which plays critical roles in regulating cardiac lipid and glucose metabolism, was increased. RNA-seq data and molecular biological assays demonstrated reduced expression of myocardin, a transcription coactivator, in Atp6v0d1AKO mouse hearts. RNA interference (RNAi), luciferase reporter and ChIP-qPCR assays revealed the critical role of myocardin in regulating IRS-1 transcription through the CArG-like element in IRS-1 promoter. Reducing IRS-1 expression with RNAi increased FoxO1 expression, while increasing IRS-1 expression reversed myocardin downregulation-induced FoxO1 upregulation in cardiomyocytes. In vivo, restoring myocardin expression specifically in Atp6v0d1AKO cardiomyocytes increased IRS-1, but decreased FoxO1 expression. As a result, the abnormal expressions of metabolic genes in Atp6v0d1AKO hearts were reversed, and cardiac dysfunctions were ameliorated. Myocardin expression was also reduced in high fat diet-induced diabetic cardiomyopathy and palmitic acid-treated cardiomyocytes. Moreover, increasing systemic insulin resistance with rosiglitazone restored cardiac myocardin expression and improved cardiac functions in Atp6v0d1AKO mice. Conclusion: Atp6v0d1AKO mice are a novel animal model for studying lipodystrophy- or metabolic dysfunction-related cardiomyopathy. Moreover, myocardin serves as a key regulator of cardiac insulin sensitivity and metabolic homeostasis, highlighting myocardin as a potential therapeutic target for treating lipodystrophy- and diabetes-related cardiomyopathy.
... The most extreme phenotype of CGL is severe insulin resistance with the loss of nearly all the body fat at birth and early development of metabolic complications in childhood (1,2). Another four distinct genetic subtypes of CGL have been reported to date, which are associated with mutations of AGPAT2, BSCL2, CAV1, and PTRF, respectively (3). Besides the common clinical manifestations, patients with type 1 CGL might present with acromegaloid features with an enlarged mandible, hands, and feet, and bone cysts as a late complication. ...
Article
Full-text available
We herein first report the use of conventional echocardiography combined with two-dimensional speckle-tracking to diagnose and monitor the changing process of cardiac involvement in an infant with congenital lipodystrophy. An 8-month-old girl was admitted to our hospital after first presenting at the age of 3 months with abnormal facial features that had been noticed within 4 weeks of birth. Echocardiography performed at the age of 3 months showed only slightly accelerated blood flow in the right ventricular outflow tract. At the age of 5 months, echocardiography showed myocardial hypertrophy; this finding combined with the physical characteristics and other examination results led to the consideration of congenital lipodystrophy. Genetic testing at the age of 9 months confirmed type 2 congenital lipodystrophy caused by BSCL2 gene mutation, and dietary modification was initiated. Conventional echocardiography performed at the ages of 5, 8, and 14 months showed no significant changes and a normal ejection fraction. However, two-dimensional speckle-tracking performed between the ages of 5 and 8 months showed cardiac systolic abnormalities that tended to improve after treatment. This case highlights the value of echocardiography in detecting structural and early functional cardiac changes in infants with congenital lipodystrophy.
... Interventions with oils have been developed primarily in murine models, although there are clinical trials on diabetic and healthy adults (Table 1). Patients with metabolic dysregulation such as DMT2 have common complications such as cardiomyopathy, a typical outcome in certain lipodystrophic patients [36]. A study analyzed the effect of Krill oil (an oil rich in n-3 polyunsaturated acid (PUFA) of marine origin) in the prevention of cardiomyopathy in diabetic mice [37]. ...
Article
Full-text available
Lipodystrophy syndromes are rare diseases that could be of genetic or acquired origin. The main complication of lipodystrophy is the dysfunction of adipose tissue, which leads to an ectopic accumulation of triglycerides in tissues such as the liver, pancreas and skeletal muscle. This abnormal fat distribution is associated with hypertriglyceridemia, insulin resistance, liver steatosis, cardiomyopathies and chronic inflammation. Although the origin of acquired lipodystrophies remains unclear, patients show alterations in genes related to genetic lipodystrophy, suggesting that this disease could be improved or aggravated by orchestrating gene activity, for example by diet. Nowadays, the main reason for adipose tissue dysfunction is an imbalance in metabolism, caused in other pathologies associated with adipose tissue dysfunction by high-fat diets. However, not all dietary fats have the same health implications. Therefore, this article aims to summarize the main genes involved in the pathophysiology of lipodystrophy, identify connections between them and provide a systematic review of studies published between January 2017 and January 2022 of the dietary fats that can modulate the development of lipodystrophy through transcriptional regulation or the regulation of protein expression in adipocytes.
... Other presenting features include hepatomegaly, umbilical protuberance, prominent musculature, voracious appetite, and acromegaloid features (17). Hypertrophic cardiomyopathy has been reported in CGL as a substantial cause of morbidity and mortality (18). Bone abnormalities have also been described, including focal osteolysis, cyst formation, and absence of marrow fat (19). ...
Article
Lipodystrophy constitutes a spectrum of diseases characterized by a generalized or partial absence of adipose tissue. Underscoring the role of healthy fat in maintenance of metabolic homeostasis, fat deficiency in lipodystrophy typically leads to profound metabolic disturbances including insulin resistance, hypertriglyceridemia, and ectopic fat accumulation. While rare, recent genetic studies indicate that lipodystrophy is more prevalent than has been previously thought, suggesting significant underdiagnosis in clinical practice. In this article, we provide an overview of the etiology and management of generalized and partial lipodystrophy disorders. We bring together the latest scientific evidence and clinical guidelines and expose key gaps in knowledge. Through improved recognition of the lipodystrophy disorders, patients (and their affected family members) can be appropriately screened for cardiometabolic, non-cardiometabolic, and syndromic abnormalities and undergo treatment with targeted interventions. Notably, insights gained through the study of this rare and extreme phenotype can inform our knowledge of more common disorders of adipose tissue overload including generalized obesity.
... While metabolic and hepatic effects of lipodystrophy are well studied, there is a paucity of data related to its CV effects. Based on limited data, lipodystrophy patients often have hypertrophic cardiomyopathy, CHD, diabetes, and hypertension (91, 281,366,380,510). The frequent cardiomyopathies contribute to sudden cardiac arrest and a shortened lifespan as well. ...
Chapter
Cardiovascular disease (CVD) continues to be the leading cause of death in adults, highlighting the need to develop novel strategies to mitigate cardiovascular risk. The advancing obesity epidemic is now threatening the gains in CVD risk reduction brought about by contemporary pharmaceutical and surgical interventions. There are sex differences in the development and outcomes of CVD; premenopausal women have significantly lower CVD risk than men of the same age, but women lose this advantage as they transition to menopause, an observation suggesting potential role of sex hormones in determining CVD risk. Clear differences in obesity and regional fat distribution among men and women also exist. While men have relatively high fat in the abdominal area, women tend to distribute a larger proportion of their fat in the lower body. Considering that regional body fat distribution is an important CVD risk factor, differences in how men and women store their body fat may partly contribute to sex-based alterations in CVD risk as well. This article presents findings related to the role of obesity and sex hormones in determining CVD risk. Evidence for the role of sex hormones in determining body composition in men and women is also presented. Lastly, the clinical potential for using sex hormones to alter body composition and reduce CVD risk is outlined. © 2022 American Physiological Society. Compr Physiol 12:1-45, 2022.
Article
Full-text available
Introduction Lipodystrophy syndromes are rare diseases that can present with a broad range of symptoms. Delays in diagnosis are common, which in turn, may predispose to the development of severe metabolic complications and end-organ damage. Many patients with lipodystrophy syndromes are only diagnosed after significant metabolic abnormalities arise. Prompt action by clinical teams may improve disease outcomes in lipodystrophy syndromes. The aim of the Rapid Action Plan is to serve as a set of recommendations from experts that can support clinicians with limited experience in lipodystrophy syndromes. Methods The Rapid Action Plan was developed using insights gathered through a series of advisory meetings with clinical experts in lipodystrophy syndromes. A skeleton template was used to facilitate interviews. A consensus document was developed, reviewed, and approved by all experts. Results Lipodystrophy is a clinical diagnosis. The Rapid Action Plan discusses tools that can help diagnose lipodystrophy syndromes. The roles of clinical and family history, physical exam, patient and family member photos, routine blood tests, leptin levels, skinfold measurements, imaging studies, and genetic testing are explored. Additional topics such as communicating the diagnosis to the patients/families and patient referrals are covered. A set of recommendations regarding screening and monitoring for metabolic diseases and end-organ abnormalities is presented. Finally, the treatment of lipodystrophy syndromes is reviewed. Discussion The Rapid Action Plan may assist clinical teams with the prompt diagnosis and holistic work-up and management of patients with lipodystrophy syndromes, which may improve outcomes for patients with this rare disease.
Article
Aim: To describe the Turkish generalized lipodystrophy (GL) cohort with the frequency of each complication and the death rate during the period of the follow-up. Methods: This study reports on 72 patients with GL (47 families) registered at different centres in Turkey that cover all regions of the country. The mean ± SD follow-up was 86 ± 78 months. Results: The Kaplan-Meier estimate of the median time to diagnosis of diabetes and/or prediabetes was 16 years. Hyperglycaemia was not controlled in 37 of 45 patients (82.2%) with diabetes. Hypertriglyceridaemia developed in 65 patients (90.3%). The Kaplan-Meier estimate of the median time to diagnosis of hypertriglyceridaemia was 14 years. Hypertriglyceridaemia was severe (≥ 500 mg/dl) in 38 patients (52.8%). Seven (9.7%) patients suffered from pancreatitis. The Kaplan-Meier estimate of the median time to diagnosis of hepatic steatosis was 15 years. Liver disease progressed to cirrhosis in nine patients (12.5%). Liver disease was more severe in congenital lipodystrophy type 2 (CGL2). Proteinuric chronic kidney disease (CKD) developed in 32 patients (44.4%) and cardiac disease in 23 patients (31.9%). Kaplan-Meier estimates of the median time to diagnosis of CKD and cardiac disease were 25 and 45 years, respectively. Females appeared to have a more severe metabolic disease, with an earlier onset of metabolic abnormalities. Ten patients died during the follow-up period. Causes of death were end-stage renal disease, sepsis (because of recurrent intestinal perforations, coronavirus disease, diabetic foot infection and following coronary artery bypass graft surgery), myocardial infarction, heart failure because of dilated cardiomyopathy, stroke, liver complications and angiosarcoma. Conclusions: Standard treatment approaches have only a limited impact and do not prevent the development of severe metabolic abnormalities and early onset of organ complications in GL.
Article
Purpose of review: Total ceramide levels in cardiac tissue relate to cardiac dysfunction in animal models. However, emerging evidence suggests that the fatty acyl chain length of ceramides also impacts their relationship to cardiac function. This review explores evidence regarding the relationship between ceramides and left ventricular dysfunction and heart failure. It further explores possible mechanisms underlying these relationships. Recent findings: In large, community-based cohorts, a higher ratio of specific plasma ceramides, C16 : 0/C24 : 0, related to worse left ventricular dysfunction. Increased left ventricular mass correlated with plasma C16 : 0/C24 : 0, but this relationship became nonsignificant after adjustment for multiple comparisons. Decreased left atrial function and increased left atrial size also related to C16 : 0/C24 : 0. Furthermore, increased incident heart failure, overall cardiovascular disease (CVD) mortality and all-cause mortality were associated with higher C16 : 0/C24 : 0 (or lower C24 : 0/C16 : 0). Finally, a number of possible biological mechanisms are outlined supporting the link between C16 : 0/C24 : 0 ceramides, ceramide signalling and CVD. Summary: High cardiac levels of total ceramides are noted in heart failure. In the plasma, C16 : 0/C24 : 0 ceramides may be a valuable biomarker of preclinical left ventricular dysfunction, remodelling, heart failure and mortality. Continued exploration of the mechanisms underlying these profound relationships may help develop specific lipid modulators to combat cardiac dysfunction and heart failure.
Article
Background Glucommander™ (GM), an electronic glycemic management system, was implemented across a multi-hospital health system as the standard of care for glycemic control. GM provides insulin dosing recommendations based on patient-specific blood glucose (BG) trends after providers select either a custom dose or weight-based multiplier as the initial dosing strategy for the first 24 hours. This study evaluated the impact of initial subcutaneous (SC) GM insulin dosing strategies on glycemic management. Methods Non-intensive care unit patients treated with SC GM using either initial custom (based on provider discretion) or weight-based multiplier settings (0.3, 0.5, or 0.7 units/kg/day) were evaluated in this retrospective chart review. The primary endpoint was time to target BG range defined as time to first two consecutive in range point of care BG. Secondary endpoints included percentage of BG values in target range, percentage of orders following institutional recommendations, length of stay (LOS), average BG, and incidence of hypoglycemia and hyperglycemia. Results A review of 348 patients showed time to target BG was not significantly different between custom and multiplier groups (55 vs 64 hours, P = .07). Target BG was achieved in less than half of patients in both groups (47% vs 44%, respectively). There were no differences in hospital LOS, proportion of BG in target range, rates of hypo/hyperglycemia, and average BG. Conclusions Custom initial SC GM insulin dosing settings showed a nonsignificant decrease in time to target BG range compared to pre-defined multiplier settings. Future studies evaluating the impact of compliance with institutional recommendations on BG control are warranted.
Article
Résumé Les lipodystrophies congénitales généralisées, aussi connues sous le nom de lipodystrophie congénitale de Berardinelli-Seip (BSCL), représentent le phénotype le plus extrême des lipodystrophies. Elles se caractérisent par la perte quasi-totale du tissu adipeux et une prédisposition à développer des complications métaboliques, telles qu’un diabète, une hypertriglycéridémie et une hépatopathie stéatosique dysmétabolique. Quatre sous-types de BSCL sont distingués en fonction du gène en cause : AGPAT2 codant l’enzyme 1-acylglycérol-3-phosphate O-acyltransférase 2 et BSCL2 codant la Seipine, et plus rarement, CAV1 et PTRF codant des protéines associées aux cavéoles, respectivement la cavéoline-1, et la polymérase 1 et facteur de libération de la transcription appelée aussi Cavin-1. L’étude de la physiopathologie de la BSCL, par des travaux cliniques et par la génération de modèles animaux et cellulaires originaux a permis de comprendre comment l’absence de ces protéines entraîne des dysfonctions adipocytaires primaires et sévères. Ces études soulignent l’importance cruciale du tissu adipeux dans l’homéostasie glucido-lipidique, tant par son rôle de stockage des molécules énergétiques que par son action endocrine. Dans cette revue, nous nous concentrerons sur les caractéristiques cliniques, les mécanismes moléculaires, et le traitement des lipodystrophies congénitales. La BSCL est un phénotype extrême qui donne l’opportunité unique d’étudier les conséquences physiopathologiques de la vie sans tissu adipeux.
Article
Full-text available
The lipodystrophies represent a class of diseases characterized by leptin deficiency. Leptin deficiency is associated with a severe form of the metabolic syndrome characterized by dyslipidemia, insulin resistance, diabetes, and ovarian dysfunction. Metreleptin is the pharmaceutical derived product that has been approved by the Food and Drug Administration (FDA) to treat the severe metabolic abnormalities of the generalized forms of lipodystrophy. Herein we describe the properties of metreleptin, its use in patients, which includes the administration of the drug and how it may be acquired by medical professionals as well as its safety, tolerability, and properties. Finally, we speculate on future uses and development of metreleptin.
Article
Full-text available
Recombinant methionyl human leptin or metreleptin is a synthetic leptin analog that has been trialed in patients with leptin-deficient conditions, such as leptin deficiency due to mutations in the leptin gene, hypothalamic amenorrhea, and lipodystrophy syndromes. These syndromes are characterized by partial or complete absence of adipose tissue and hormones derived from adipose tissue, most importantly leptin. Patients deficient in leptin exhibit a number of severe metabolic abnormalities such as hyperglycemia, hypertriglyceridemia, and hepatic steatosis, which can progress to diabetes mellitus, acute pancreatitis, and hepatic cir-rhosis, respectively. For the management of these abnormalities, multiple therapies are usually required, and advanced stages may be progressively difficult to treat. Following many successful trials, the US Food and Drug Administration approved metreleptin for the treatment of non-HIV-related forms of generalized lipodystrophy. Leptin replacement therapy with metreleptin has, in many cases, reversed these metabolic complications, with improvements in glucose-insulin-lipid homeostasis, and regression of fatty liver disease. Besides being effective, a daily subcutaneous administration of metreleptin is generally safe, but the causal association between metreleptin and immune complications (such as lymphoma) is still unclear. Moreover, further investigation is needed to elucidate mechanisms by which metreleptin leads to the development of anti-leptin antibodies. Herein, we review clinical aspects of generalized lipodystrophy and the pharmacological profile of metreleptin. Further, we examine studies that assessed the safety and efficacy of metreleptin, and outline some clinical perspectives on the drug.
Article
Full-text available
Berardinelli-Seip syndrome is a rare autosomal recessive disease characterized by inadequate metabolism and inefficient storing of lipids in fat cells, generating accumulation of fat in organs such as the liver, spleen, pancreas, heart, arterial endothelium and skin. Classically, patients manifest generalized lipoatrophy at birth or until 2 years of age, and in adolescence usually develop marked insulin resistance with rapid progression to diabetes and dyslipidemia. We report the case of a 17-year-old Berardinelli-Seip syndrome patient with eruptive xanthoma associated with severe hypertriglyceridemia. It is worth noting Eruptive xanthoma as a dermatological manifestation that is not generally highlighted in the reports of cases of this genetic metabolic disorder.
Article
Full-text available
Generalized lipodystrophy is a rare disorder characterized by marked loss of adipose tissue with reduced triglyceride storage capacity, leading to a severe form of metabolic syndrome including hypertriglyceridemia, insulin resistance, type 2 diabetes mellitus, and hepatic steatosis. Recent echocardiographic studies suggest that concentric left ventricular (LV) hypertrophy is another characteristic feature of this syndrome, but the mechanism remains unknown. It has recently been hypothesized that the LV hypertrophy could be an extreme clinical example of "lipotoxic cardiomyopathy": excessive myocyte accumulation of triglyceride leading to adverse hypertrophic signaling. To test this hypothesis, the first cardiac magnetic resonance study of patients with generalized lipodystrophy was performed, using magnetic resonance imaging and localized proton spectroscopy to detect excessive triglyceride content in the hypertrophied myocytes. Six patients with generalized lipodystrophy and 6 healthy controls matched for age, gender, and body mass index were studied. As hypothesized, myocardial triglyceride content was threefold higher in patients than controls: 0.6 ± 0.2% versus 0.2 ± 0.1% (p = 0.004). The presence of pericardial fat was also found, representing a previously undescribed adipose depot in generalized lipodystrophy. Patients with generalized lipodystrophy, compared with controls, also had a striking degree of concentric LV hypertrophy, independent of blood pressure: LV mass index 101.0 ± 18.3 versus 69.0 ± 17.7 g/m(2), respectively (p = 0.02), and LV concentricity 1.3 ± 0.3 versus 0.99 ± 0.1 g/ml, respectively (p = 0.04). In conclusion, these findings advance the lipotoxicity hypothesis as a putative underlying mechanism for the dramatic concentric LV hypertrophy found in generalized lipodystrophy.
Article
Full-text available
Congenital generalized lipodystrophy (CGL) also known as Berardinelli-Seip Congenital Lipodystrophy (BSCL) is a genetically heterogeneous disorder characterized by loss of adipose tissues, Acanthosis nigricans, diabetes mellitus, muscular hypertrophy, hepatomegaly and hypertriglyceridemia. There are four subclinical phenotypes of CGL (CGL1-4) and mutations in four genes AGPAT2, BSCL2, CAV1 and PTRF have been assigned to each type. The study included clinical and molecular investigations of CGL disease in a consanguineous Pakistani family. For mutation screening all the coding exons including splice junctions of AGPAT2, BSCL2, CAV1 and PTRF genes were PCR amplified and sequenced directly using an automated DNA sequencer ABI3730. Sequence analysis revealed a single base pair deletion mutation (c.636delC; p.Tyr213ThrfsX20) in exon 5 of BSCL2 gene causing a frame shift and premature termination codon. Mutation identified here in BSCL2 gene causing congenital generalized lipodystrophy is the first report in Pakistani population. The patients exhibited characteristic features of generalized lipodystrophy, Acanthosis nigricans, diabetes mellitus and hypertrophic cardiomyopathy. Virtual Slides The virtual slide(s) for this article can be found here: http://www.diagnosticpathology.diagnomx.eu/vs/1913913076864247.
Article
Congenital generalized lipodystrophy (CGL) is a heterogeneous autosomal recessive disorder characterized by a near complete lack of adipose tissue from birth and, later in life, the development of metabolic complications, such as diabetes mellitus, hypertriglyceridaemia and hepatic steatosis. Four distinct subtypes of CGL exist: type 1 is associated with AGPAT2 mutations; type 2 is associated with BSCL2 mutations; type 3 is associated with CAV1 mutations; and type 4 is associated with PTRF mutations. The products of these genes have crucial roles in phospholipid and triglyceride synthesis, as well as in the formation of lipid droplets and caveolae within adipocytes. The predominant cause of metabolic complications in CGL is excess triglyceride accumulation in the liver and skeletal muscle owing to the inability to store triglycerides in adipose tissue. Profound hypoleptinaemia further exacerbates metabolic derangements by inducing a voracious appetite. Patients require psychological support, a low-fat diet, increased physical activity and cosmetic surgery. Aside from conventional therapy for hyperlipidaemia and diabetes mellitus, metreleptin replacement therapy can dramatically improve metabolic complications in patients with CGL. In this Review, we discuss the molecular genetic basis of CGL, the pathogenesis of the disease's metabolic complications and therapeutic options for patients with CGL.
Article
Lipodystrophy is a medical condition characterized by complete or partial loss of adipose tissue. Not infrequently, lipodystrophy occurs in combination with pathological accumulation of adipose tissue at distinct anatomical sites. Patients with lipodystrophy exhibit numerous metabolic complications, which indicate the importance of adipose tissue as an active endocrine organ. Not only the total amount but also the appropriate distribution of adipose tissue depots contribute to the metabolic state. Genetic and molecular research has improved our understanding of the mechanisms underlying lipodystrophy. Circulating levels of hormones secreted by the adipose tissue, such as leptin and adiponectin, are greatly reduced in distinct subpopulations of patients with lipodystrophy. This finding rationalizes the use of these adipokines or of agents that increase their circulating levels, such as peroxisome proliferator-activated receptor γ (PPARγ) agonists, for therapeutic purposes. Other novel therapeutic approaches, including the use of growth hormone and growth-hormone-releasing factors, are also being studied as potential additions to the therapeutic armamentarium. New insights gained from research and clinical trials could potentially revolutionize the management of this difficult-to-treat condition.
Article
Congenital generalized lipodystrophy (CGL) is a rare autosomal recessive disorder characterized by near total absence of body fat since birth with predisposition to insulin resistance, diabetes, hypertriglyceridemia, and hepatic steatosis. Three CGL loci, AGPAT2, BSCL2, and CAV1, have been identified previously. Recently, mutations in polymerase I and transcript release factor (PTRF) were reported in five Japanese patients presenting with myopathy and CGL (CGL4). We report novel PTRF mutations and detailed phenotypes of two male and three female patients with CGL4 belonging to two pedigrees of Mexican origin (CGL7100 and CGL178) and one pedigree of Turkish origin (CGL180). All patients had near total loss of body fat and congenital myopathy manifesting as weakness, percussion-induced muscle mounding, and high serum creatine kinase levels. Four of them had hypertriglyceridemia. Three of them had atlantoaxial instability. Two patients belonging to CGL178 pedigree required surgery for pyloric stenosis in the first month of life. None of them had prolonged QT interval on electrocardiography but both siblings belonging to CGL7100 had exercise-induced ventricular arrhythmias. Three of them had mild acanthosis nigricans but had normal glucose tolerance. Two of them had hepatic steatosis. All patients had novel null mutations in PTRF gene. In conclusion, mutations in PTRF result in a novel phenotype that includes generalized lipodystrophy with mild metabolic derangements, myopathy, cardiac arrhythmias, atlantoaxial instability, and pyloric stenosis. It is unclear how mutations in PTRF, which plays an essential role in formation of caveolae, affect a wide variety of tissues resulting in a variable phenotype.
Article
Lipodystrophy is a rare disorder characterized by loss of adipose tissue and low leptin levels. This condition is characterized by severe dyslipidemia, insulin resistance, diabetes mellitus, and steatohepatitis. Another phenotypic feature that occurs with considerable frequency in generalized lipodystrophy is cardiomyopathy. We report here the cardiac findings in a cohort of patients with generalized congenital and acquired lipodystrophy, and present a literature review of the cardiac findings in patients with generalized lipodystrophy. We studied 44 patients with generalized congenital and acquired lipodystrophy, most of them enrolled in a clinical trial of leptin therapy. Patients underwent electrocardiograms and transthoracic echocardiograms to evaluate their cardiac status. We followed these patients for an extended time period, some of them up to 8 years. Evaluation of our cohort of patients with generalized lipodystrophy shows that cardiomyopathy is a frequent finding in this population. Most of our patients had hypertrophic cardiomyopathy, and only a small number had features of dilated cardiomyopathy. Hypertrophic cardiomyopathy was more frequent in patients with seipin mutation, a finding consistent with the literature. The underlying mechanism for cardiomyopathy in lipodystrophy is not clear. Extreme insulin resistance and the possibility of a "lipotoxic cardiomyopathy" should be entertained as possible explanations.