Download as pdf or txt
Download as pdf or txt
You are on page 1of 6

37

Carnitine transporter deficiency

Introduction 246 Genetics and pathogenesis 248


Clinical abnormalities 246 Treatment 250

MAJOR PHENOTYPIC EXPRESSION


Hypoketotic hypoglycemia, seizures, vomiting, lethargy progressive to coma; cardiomyopathy; chronic muscle weakness;
carnitine deficiency in plasma and muscle and increased excretion of free-carnitine in urine; and defective transport of
carnitine into cultured fibroblasts.

INTRODUCTION CLINICAL ABNORMALITIES

The inborn errors of fatty acid oxidation, including carnitine The classic, and frequently the initial, presentation of carni-
transporter deficiency [1], represent a newly recognized area tine transporter deficiency (CTD) is hypoketotic hypo-
of human disease. The rate of discovery of distinct disorders glycemia, as in most disorders of fatty acid oxidation. The
has increased rapidly since the discovery of medium-chain first patient reported [1] (Figure 37.2) presented at 3 months
acyl CoA dehydrogenase (MCAD) deficiency in 1982 (Chapter comatose, limp and unresponsive in the afternoon after a
40). Deficiency of carnitine is common in these disorders prolonged overnight fast. She was acidotic; the serum bicar-
in which fatty acyl CoA compounds accumulate which bonate was 16 mmol/L and the arterial pH 7.17. The blood
then form esters with carnitine and are preferentially excreted concentration of glucose was 0.39 mmol/L (7 mg/dL) and that
in the urine. Carnitine deficiency may also be profound in of the cerebrospinal fluid (CSF) was 0.2 mmol/L (4mg/dL).
organic acidemias such as propionic acidemia for the same Resuscitation required intubation, assisted ventilation and
reason. The transport of carnitine into fibroblasts is inhibited parenteral glucose and saline. Acute episodes of hypoketotic
by long and medium chain acylcarnitines [2], and this may be hypoglycemia are potentially fatal (Figure 37.3) [4] and may
an additional factor in carnitine deficiency in disorders of be sudden and unexpected. An infant of a vegetarian mother
fatty acid oxidation. Primary carnitine deficiency resulting died at 5 days of life [5]. Episodes usually occur before 2 years
from an abnormality in the synthesis of carnitine from protein- of age and follow fasting [2,6].
bound lysine has not yet been observed. Many of the patients Modest hepatomegaly is characteristic of this condition.
reported early as primary carnitine deficiency have turned Biopsy of the liver shows microvesicular lipid [7], a finding,
out to have MCAD deficiency. Deficiency of carnitine as a like the rest of this clinical picture, that might lead to a diag-
result of abnormality in the transporter (Figure 37.1) that nosis of Reye syndrome.
facilitates its entry into certain cells has been referred to as Clinical chemistry is also consistent with Reye syndrome,
primary carnitine deficiency [1]. The gene for the carnitine with hyperammonemia and increased levels of transaminases.
transporter SLC22A5 has been cloned, and a small number of The initial patient had an ammonia of 338
mol/L, slightly
mutations have been defined [3]. prolonged prothrombin time, an aspartate transaminase
Clinical abnormalities 247

Fatty acyl CoA


Carnitine Carnitine Acylcarnitine Acylcarnitine
CPT I
Transporter
Fatty acyl CoA Carnitine

-Oxidation

Figure 37.1 The carnitine transporter


and its role in fatty acid metabolism.

the height of the hypoglycemic illness, there may be mis-


leading ketonuria in any disorder of fatty acid oxidation.
Quantification of the plasma concentration of 3-hydroxy-
butyric acid or of acetoacetic and 3-hydroxybutyric acids at
this time will provide definitive evidence of impaired ketoge-
nesis, but this information is not usually available to the clini-
cian. Dicarboxylic aciduria is usually notably absent [1,4].
Cardiomyopathy is the other classic way in which this dis-
order presents [2,7] and may be expected in any patient not
given the benefit of diagnosis and treatment with carnitine [4].
It was the most common presenting complaint in 15 patients
[3] and present in 100 percent of 20 reported patients. The
patient of Waber and colleagues [7] reported with progressive
cardiomyopathy and cardiac failure successfully treated with
carnitine has been shown to have the transporter defect. The
median age of onset of cardiac symptoms was 3 years [2].
Onset may be with rapidly progressive heart failure [2] or a
murmur, and cardiomegaly may be found on routine physi-
cal examination or examination at the time of hypoglycemia.
Roentgenograms and echocardiography reveal cardiac enlarge-
ment and increased thickness of the left ventricular wall.
Figure 37.2 A 17-month-old who was the initial reported patient [1] Electrocardiogram (EKG) reveals left ventricular hypertrophy.
with transporter defect. She presented at 3 months with hypoglycemic Nevertheless, the cardiomyopathy has been described as char-
coma precipitated by an intercurrent illness and prolonged fasting. acteristically dilated [8], and this has repeatedly been confirmed
This episode left her with severe brain damage reflected in her vacant by cardiac catheterization. This may be expected to be a lethal
expression. She died shortly after the picture was taken because of disease in which patients without carnitine supplementation
complications of a gastrostomy. (Illustration was kindly provided by display cardiac failure progressive to death. Death in a sibling
Dr. Charles Stanley of the Children’s Hospital of Philadelphia.) has been recorded in 8 families [2]. A 12-year-old boy who died
suddenly of cardiomyopathy following a routine surgical proce-
dure was found to have a low concentration of carnitine in
(AST) of 248 and alanine transaminase (ALT) of 149 IU/L [1]. plasma and defective transport of carnitine in fibroblasts [9].
Uric acid concentrations are also elevated at the time of the Muscle weakness or hypotonia is the third major manifes-
episode. This, along with the elevation of creatine phosphok- tation of disease. It may be present along with other features,
inase (CK) [2] should strongly suggest a disorder of fatty acid particularly those of the heart, but in two patients it was the
oxidation. only manifestation [2]. The picture may be that of a progressive
Examination of the urine may reveal no ketonuria [1,2], proximal myopathy [8]. Biopsy of muscle reveals lipid storage
and this should strongly suggest the diagnosis. However, in myopathy [1].
248 Carnitine transporter deficiency

Delay in diagnosis has been another characteristic of this


disease. In nine patients, the delay was 1–6 years after the onset
of symptoms [2], and in this time all developed cardiomyopa-
thy, and all but one had muscle weakness. In some patients
mild muscle weakness may not have been noted because of the
attention devoted to the major cardiac manifestations.

GENETICS AND PATHOGENESIS

Transmission of the disorder is autosomal recessive. Affected


siblings of both sexes have been observed, and consanguinity
has been present in at least five families [2]. Concentrations
of carnitine in plasma were low in 11 mothers studied and 10
of 11 fathers [6]. Prevalence is unknown, but there were 10
patients in the series of 107 with disorders of fatty acid oxida-
tion in the Saudubray Paris experience [8]. Among 313 patients
with an autopsy diagnosis of sudden infant death syndrome
(SIDS), 3 were designated as transporter defects on the basis
of hepatic steatosis and very low hepatic carnitine along with
Figure 37.3 J.S., a12-year-old boy with carnitine transporter
low esterified carnitine [10].
deficiency. The disease is exquisitely responsive to carnitine. His death
The diagnosis is usually suspected initially on the basis of
at 13 years highlights the dangerous nature of the disease and the
a low concentration of free-carnitine in plasma. In the first
importance of close follow-up of carnitine status and expert
patient, the total plasma carnitine ranged from 0 to 2.2
mol/L
management.
and no free-carnitine could be detected. In 20 patients [2]
total plasma carnitine ranged from 0 to 9
mol/L, with 18
having values less than 4.2
mol/L. In controls total carnitine 1
mol/L uptake was negligible. Uptake in patients at high
was 40–60
mol/L. The acylcarnitine profile reveals a decrease concentrations, such as 10 or 20
mol/L reflect a second low-
in free and esterfied carnitines. affinity transporter [11] or passive diffusion [12]. Transport of
Concentrations of carnitine in muscle are also quite low. carnitine in control fibroblasts is sodium-dependent [1]. The
In 13 patients studied, the range for total carnitine was from uptake of carnitine by fibroblasts at 5
mol/L showed no over-
0.05 to 17 percent of the normal mean. In liver, the total was lap among patients, parents and controls with the exception of
five percent of normal. The excretion of carnitine in the urine one father. The velocity of carnitine uptake can be measured in
is inappropriately high, consistent with defective renal tubu- lymphoblasts as well as fibroblasts [13]. Patients display rates
lar reabsorption [7]. At a time when the plasma carnitine below 10 percent of control. Heterozygosity can be demon-
approximated zero, the renal excretion was 121
mol/g crea- strated in some patients by rates below 40 percent of control.
tinine (normal 17–425) [1], and following a dose of 100 mg/kg Low uptake of carnitine has also been demonstrated in cul-
of oral carnitine the plasma carnitine rose only to 21
mol/L, tured myocytes derived from patients [14]. Prenatal diagnosis
but urinary excretion increased to 2911 mol/g creatinine. has been accomplished by demonstration of defective uptake
After four months of carnitine treatment, the plasma concen- of carnitine from aminocytes of an affected fetus [15].
trations reached the low normal range, while urinary excre- In response to the administration of carnitine, levels in
tion was four to five times normal. The fractional excretion liver return to normal, while those in muscle respond poorly,
rate for free-carnitine was nearly 100 percent of the filtered indicating that the transport defect includes muscle and kid-
load. On withdrawal of treatment the fractional excretion ney, but not liver. Consistent with this, the low Km and prefer-
exceeded the filtered load. ence for L-isomer that characterize the uptake of carnitine by
The nature of the defect has been demonstrated by study fibroblasts is shared by heart and muscle [11,12,16], but not
of the uptake of carnitine in vitro by cultured fibroblasts [1,2] by liver [17].
(Figure 37.3). In control cells the uptake of 14C-labeled carni- The gene for the carnitine transporter, SLC22A5, has
tine was via a high-affinity, carrier-mediated transport process been mapped to chromosome 5q31, the locus for carnitine
with an apparent Km of 3.24 0.5 and a Vmax of 1.67 0.19 transporter deficiency in a large Japanese kindred. It codes
[1] consistent with previous reports [11]. Fibroblasts from for an organic cation transporter OCTN2 [18,19]. This is one
patients have shown little uptake of carnitine; at a concentra- of a family of organic cation (OCTN) sodium ion dependent
tion of carnitine of 5
mol/L, control uptake was 0.94 and transporters. The protein contains 557 amino acids and has
a patient uptake was 0.1 pmol/min/mg protein [1]. High- the properties of a high affinity transporter. A number of
affinity transport is best shown at lower concentrations; up to mutations have now been identified in patients with this disease
Genetics and pathogenesis 249

Table 37.1 Differential diagnosis of disorders involving carnitine

Plasma total Carnitine esterfied Urinary


␮mol/L % of total carnitine

Control 40–60 30 Normal


Carnitine transporter deficiency 5 30 Paradoxically high free
Carnitine palmitoyl transferase (CPT) I deficiency 60–100 20 Normal or high
Carnitine translocase deficiency 5–30 80–100 High ester
Carnitine palmitoyl transferase (CPT) II deficiency 10–40 40–80 Normal or high ester
Defects in -oxidation 10–30 30–60 High ester
3-Hydroxy-3-methylglutaryl CoA lyase deficiency 10–30 30–60 High ester

[2,20–30]. Most individual families have had unique muta- palmitoyl-transferase I (Chapter 39) also develop hypoketotic
tions. There have been a few instances of the same mutation hypoglycemia without dicarboxylic aciduria [36]. Comparisons
in unrelated patients [22,24,25,28]. A few stop codons and of alterations of plasma carnitine in various disorders is
frame shifts have been defined [2,20]. A lack of correlation shown in Table 37.1. Low free and total carnitine in plasma
between genotype and phenotype has been discussed [2,28]. along with urinary free carnitine that is paradoxically main-
However, decisions as to severity of phenotype often rest on tained is suggestive of a transporter defect.
whether or not hypoglycemia once occurred early in life, as in The response to fasting in a patient with defective carnitine
the case of one of two sibs with the R399Q missense mutation. transporter showed hypoketosis throughout and hypoglycemia
The episode followed gastroenteritis at 2 years of age [2]. by 12 hours (Figure 37.4) [1]. The fast was stopped when the
Because of her diagnosis an older sib who had proximal limb plasma glucose reached 2.8
mol/L (51 mg/dL) at which time
girdle weakness and mild developmental delay at 4 years-of- the patient remained asymptomatic. Levels of free-fatty acids
age was tested and found to have the mutation. Differences of in plasma rose sharply to 2.22
mol/L, but the level of
this nature appear to reflect the chance occurrence of an illness 3-hydroxybutyrate remained flat at 0.27
mol/L. Blood con-
that led to fasting. centrations of ammonia rose. Treatment with carnitine cor-
Among ethnic differences an 11 bp deletion was found in rected this patient’s impaired hepatic oxidation of fatty acids;
unrelated patients from Switzerland and neighboring north- and she was able to fast for 24 hours without hypoglycemia.
ern Italy [2], and R169W was found in two unrelated families Levels of 3-hydroxybutyrate rose to 2 mmol/L, higher than
in Italy [28]. In Japan, where the disease appears relatively the free-fatty acids (1.25 mmol/L).
frequent, most families have had a few mutations [31]. In a Diet may contribute to the pathogenesis of symptoms in
survey of 973 unrelated Japanese [31], 14 were found to have this disease. A 12-year old who died suddenly following sur-
low levels of carnitine, and of these, six had mutations in the gery [9] had been exposed to an essentially vegetarian diet
gene for OCTN2: W132X, S467C, W283C and M179L. These for some time. The 3-month-old initial patient [1] had been
data gave a carrier frequency of one percent in Japan. Echo- changed from a cow’s milk protein containing formulation to
cardiographic study indicated asymptomatic cardiac hyper- a soy protein preparation that contained no carnitine, four
trophy in these heterozygotes. weeks prior to the episode of hypoketotic hypoglycemia.
An animal model of the carnitine transporter defect, the The pathogenesis of symptoms of hypoketotic hypo-
juvenile visceral steatosis (jvs) mouse [32], has autosomal glycemia reflects the role of fat in energy metabolism. Hypo-
recessive fatty infiltration of the liver, hypoglycemia and glycemia after short periods of fasting usually represent
hyperammonemia two weeks after birth, and very low levels disorders of carbohydrate metabolism. The oxidation of fatty
of carnitine in blood and muscle, along with defective renal acids is not a major source of energy until relatively late in
reabsorption of free carnitine. The hyperammonemia results fasting. It usually takes 15 to 24 hours of fasting to induce
from decreased expression of genes for enzymes of the urea hypoglycemia in a patient with a disorder of fatty acid oxida-
cycle; low levels of mRNA are associated with low levels of all tion. An individual who never fasted beyond 12 hours would
of the hepatic enzymes of the urea cycle [33]. Treatment with usually be protected against this manifestation.
carnitine corrects the abnormal expression and urea cycle The metabolism of fat begins with lipolysis; those patients
enzyme activity [34]. The jvs gene has been mapped to with defective fatty acid oxidation have high ratios of free
mouse chromosome 11, which is syntenic with the SLC22A5 fatty acids to 3-hydroxybutyrate in blood after fasting. Once
locus on human chromosome 5 [35]. transported into cells carnitine is esterified with acyl CoA esters
Analysis of the organic acids of the urine of these patients including those of fatty acids resulting from lipolysis. The
is usually normal. The absence of dicarboxylic aciduria, espe- esterifications are catalyzed by carnitine acyl transferases such
cially at times of acute illness and hypoglycemia, contrasts as carnitine palmitoyl transferase (CPT) I. Carnitine translo-
sharply with findings in patients with defects in -oxidation case then catalyzes the transfer of the fatty acylcarnitines across
such as MCAD deficiency. Patients with deficiency of carnitine the membrane into the mitochondrion, where hydrolysis to
250 Carnitine transporter deficiency

80 120 160 4.0 NH3


(A) (B)

60 90 120 3.0

Plasma BOB, FFA (mmol/L)


Plasma carnitine (
mol/L)

Blood ammonia (
mol/L)

Plasma glucose (mg/dL)

Glu
FFA Carn
40 60 80 2.0
NH3

Glu
20 30 40 1.0

FFA

BOB
Carn BOB
0 0 0 0
0 6 12 18 24 0 6 12 18 24

Duration of fast (hours)

Figure 37.4 The response to fasting in a patient with the carnitine transporter defect. (A) In the control state hypoglycemia(glu) was
prominent at 12 hours, and there was no evident ketogenesis (3-hydroxybutyrate [BOB]) despite elevation of free-fatty acids (FFA). (B) Following
treatment with carnitine, fasting for 24 hours was without hypoglycemia, and ketogenesis was evident in the rising BOB. (Reprinted with
permission from the New England Journal of Medicine [1].)

fatty acyl CoA and free or recycled carnitine is catalyzed function may be unaffected until the intracellular muscle
by CPT II. Fatty acyl CoA compounds then undergo - concentration of carnitine falls below 30–50
mol/L or two to
oxidation in which there is successive shortening by two carbon four percent of normal. Biopsied muscle revealed a decrease
atoms releasing acetyl CoA. In muscle, this is largely oxidized of stored lipid with treatment, but not a disappearance [1].
via the citric acid cycle, while in liver ketogenesis proceeds via
the successive action of 3-hydroxymethylglutaryl (HMG) CoA References
synthase and lyase-yielding acetoacetate, which is converted
to 3-hydroxybutyrate. 1 Treem WR, Stanley CA, Finegold DN, et al. Primary carnitine deficiency due
to a failure of carnitine transport in kidney muscle and fibroblasts. N Engl
J Med 1988;319:1331.
2 Stanley CA, DeLeeuw S, Coates PM, et al. Chronic cardiomyopathy and
TREATMENT weakness of acute coma in children with a defect in carnitine uptake. Ann
Neurol 1991;30:709.
Treatment of this disease with carnitine has been highly 3 Wang Y, Horman SH, Ye J, et al. Phenotype and genotype variation in
successful [2,7]. Levels of free-carnitine in plasma and liver primary carnitine deficiency. Genet Med 2001;3:387.
are readily restored, preventing further attacks of hypoketotic 4 Brivet M, Boutron A, Slama A, et al. Defects in activation and transport of
hypoglycemia. Developmental delay or seizures induced by fatty acids. J Inherit Metab Dis 1999;22:428.
hypoglycemic attacks prior to treatment persist, but are not 5 Rinaldo P, Stanley CA, Hsu B, et al. Sudden neonatal death in carnitine
progressive. transporter deficiency. J Pediatr 1997;131:304.
Cardiomyopathy and failure respond dramatically to treat- 6 Tein I, De Vivo DC, Bierman F, et al. Impaired skin fibroblast carnitine uptake
in primary systemic carnitine deficiency manifested by childhood carnitine-
ment [6,7]. Heart size is reduced to normal within months.
responsive cardiomyopathy. Pediatr Res 1990;28:217.
Doses have ranged from 50–120 mg/kg p.o. Skeletal muscle 7 Waber LJ, Valle D, Neill C, et al. Carnitine deficiency presenting as familial
weakness improved, although mild proximal muscle weakness cardiomyopathy: a treatable defect in carnitine transport. J Pediatr
has occasionally persisted. However, muscle concentrations 1982;101:700.
of carnitine were documented to increase only slightly to 8 Saudubray JM, Martin D, De Lonlay P, et al. Recognition and management
22–80 mmol/g; control levels are 2500 to 3500 mmol/g. These of fatty acid oxidation defects: A series of 107 patients. Arch Neurol
observations suggested that muscle oxidation of fat and muscle 2004;61:570.
References 251

9 Pollitt RJ, Olpin SE, Bonham JR, et al. Late-presenting carnitine transport mutation and a recurrent Arg282ter mutation associated with an
defect. Enzyme Protein 1993;47:175. unconventional splicing abnormality. Biochem Biophys Res Commun
10 Boles RG, Buck EA, Blitzer MG, et al. Retrospective biochemical screening 1999;161:484.
of fatty acid oxidation disorders in postmortem livers of 418 cases of 25 Vaz FM, Scholte HR, Ruiter J, et al. Identification of two novel mutations
sudden death in the first year of life. J Pediat 1998;132:924. in OCTN2 of three patients with systemic carnitine deficiency. Hum Genet
11 Rebouche CJ, Engel AG. Carnitine transport in cultured muscle cells and 1999;105:157.
skin fibroblasts from patients with primary systemic carnitine deficiency. 26 Mayatepek E, Nezu J, Tamai I, et al. Two novel missense mutations of
In Vitro 1982;18:495. the OCTN2 gene (W282R and V446F) in a patient with primary systemic
12 Vary TC, Nealy JR. Characterization of carnitine transport in isolated carnitine deficiency. Hum Mutat 2000;15:118.
perfused adult rate hearts. Am J Physiol 1982;242:H585. 27 Wang Y, Kelly MA, Cowan TM, Longo N. A missense mutation in the OCTN2
13 Tein I, Xie ZQ. The human plasmalemmal carnitine transporter defect gene associated with residual carnitine transport activity. Hum Mutat
is expressed in cultured lymphoblasts: A new non-invasive method for 2000;15:238.
diagnosis. Clin Chim Acta 1996;252:201. 28 Wang Y, Taroni F, Garavaglia B, Longo N. Functional analysis of
14 Pons R, Carrozzo R, Tein I, et al. Deficient muscle carnitine transport in mutations in the OCTN2 transporter causing primary carnitine
primary carnitine deficiency. Pediatr Res 1997;42:583. deficiency: Lack of genotype-phenotype correlation. Hum Mutat
15 Christodoulou J, Teo SH, Hammond J, et al. First prenatal diagnosis of the 2000;16:401.
carnitine transporter defect. Am J Med Genet 1996;66:21. 29 Cederbaum S, Dipple K, Vilain E, et al. Clinical follow-up and molecular
16 Rebouche CJ. Carnitine movement across muscle cell membranes: Studies etiology of the original case of carnitine transporter deficiency. J Inherit
in isolated rat muscle. Biochim Biophys Acta 1977;471:145. Metab Dis 2000;23:(suppl 1) 119.
17 Christiansen RZ, Bremer J. Active transport of butyrobetaine and carnitine 30 Christensen E, Holm J, Hansen SH, et al. Sudden infant death following
into isolated liver cells. Biochim Biophys Acta 1976;448:562. pivampicillin treatment in a patient with carnitine transporter deficiency.
18 Wu X, Prasad PD, Leibach FH, Ganapathy V. cDNA sequence transport J Inherit Metab Dis 2000;23:(suppl 1) 117.
function and genomic organization of human OCTN2 a new member of 31 Koizumi A, Nozaki J, Ohura T, et al. Genetic epidemiology of the carnitine
the organic cation transporter family. Biochem Biophys Res Commun transporter OCTN2 gene in a Japanese population and phenotypic
1998;246:589. characterization in Japanese pedigrees with primary systemic carnitine
19 Tamai I, Ohashi R, Nezu J, et al. Molecular and functional identification of deficiency. Hum Mol Genet 1999;8:2247.
sodium ion-dependent high affinity human carnitine transporter OCTN2. 32 Horiuchi M, Hayakawa J, Yamaguchi S, Saheki T. Possible primary defect
J Biol Chem 1998;273:20378. of juvenile visceral steatosis (jvs) mouse with systemic carnitine deficiency.
20 Lamhonwah AH, Tein I. Carnitine uptake defect: Frameshift mutations in the (Abstr) First IUBMB Conference Biochemistry of Diseases 2-a-05-p8;
human plasmalemmal carnitine transporter gene Biochem Biophys Res 1992;Nagoya Congress Center.
Commun 1998;252:396. 33 Tomomura M, Yasushi I, Horiuchi M, et al. Abnormal expression of urea
21 Nezu J, Tamai I, Oku A, et al. Primary systemic carnitine deficiency is cycle enzyme genes in juvenile visceral steatosis (jvs) mice. Biochim
caused by mutations in a gene encoding sodium ion-dependent carnitine Biophys Acta 1992;1138:167.
transporter. Nat Genet 1999;21:91. 34 Horiuchi M, Kobayashi K, Tomomura M, et al. Carnitine administration to
22 Wang Y, Ye J, Ganapathy V, Longo N. Mutations in the organic juvenile visceral steatosis mice corrects the suppressed expression of urea
cation/carnitine transporter OCTN2 in primary carnitine deficiency. Proc cycle enzyme by normalizing their transcription. J Biol Chem
Natl Acad Sci USA 1999;96:2356. 1992;267:5032.
23 Tang NL, Ganapathy V, Wu X, et al. Mutations of OCTN2 an organic 35 Nikaido H, Horiuchi M, Hashimoto N, et al. Mapping of jvs (juvenile visceral
cation/carnitine transporter lead to deficient cellular carnitine uptake in steatosis) gene which causes systemic carnitine deficiency in mice on
primary carnitine deficiency. Hum Mol Genet 1999;8:655. chromosome 11. Mamm Genome 1995;6:369.
24 Burwinkel B, Kreuder J, Schweitzer S, et al. Carnitine transporter OCTN2 36 Bonnefont JP, Haas R, Wolff J, et al. Deficiency of carnitine
mutations in systemic primary carnitine deficiency: A novel Arg169Gln palmitoyltransferase I. J Child Neurol 1989;4:197.

You might also like