El Hattab2015

You might also like

Download as pdf or txt
Download as pdf or txt
You are on page 1of 9

Molecular Genetics and Metabolism 116 (2015) 4–12

Contents lists available at ScienceDirect

Molecular Genetics and Metabolism

journal homepage: www.elsevier.com/locate/ymgme

Minireview

MELAS syndrome: Clinical manifestations, pathogenesis, and


treatment options
Ayman W. El-Hattab a, Adekunle M. Adesina b, Jeremy Jones c, Fernando Scaglia d,⁎
a
Division of Clinical Genetics and Metabolic Disorders, Department of Pediatrics, Tawam Hospital, Al-Ain, United Arab Emirates
b
Department of Pathology and Immunology, Baylor College of Medicine, Houston, TX, USA.
c
Singleton Department of Radiology, Texas Children's Hospital, Houston, TX, USA
d
Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, TX, USA

a r t i c l e i n f o a b s t r a c t

Article history: Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like episodes (MELAS) syndrome is one of the
Received 13 May 2015 most frequent maternally inherited mitochondrial disorders. MELAS syndrome is a multi-organ disease with
Received in revised form 14 June 2015 broad manifestations including stroke-like episodes, dementia, epilepsy, lactic acidemia, myopathy, recurrent
Accepted 14 June 2015
headaches, hearing impairment, diabetes, and short stature. The most common mutation associated with
Available online 15 June 2015
MELAS syndrome is the m.3243ANG mutation in the MT-TL1 gene encoding the mitochondrial tRNALeu(UUR).
Keywords:
The m.3243ANG mutation results in impaired mitochondrial translation and protein synthesis including the mi-
Mitochondrial diseases tochondrial electron transport chain complex subunits leading to impaired mitochondrial energy production. The
Encephalomyopathy inability of dysfunctional mitochondria to generate sufficient energy to meet the needs of various organs results
Lactic acidosis in the multi-organ dysfunction observed in MELAS syndrome. Energy deficiency can also stimulate mitochondrial
Nitric oxide deficiency proliferation in the smooth muscle and endothelial cells of small blood vessels leading to angiopathy and
Arginine impaired blood perfusion in the microvasculature of several organs. These events will contribute to the compli-
Citrulline cations observed in MELAS syndrome particularly the stroke-like episodes. In addition, nitric oxide deficiency
Angiopathy
occurs in MELAS syndrome and can contribute to its complications. There is no specific consensus approach for
Endothelial dysfunction
treating MELAS syndrome. Management is largely symptomatic and should involve a multidisciplinary team.
Unblinded studies showed that L-arginine therapy improves stroke-like episode symptoms and decreases the
frequency and severity of these episodes. Additionally, carnitine and coenzyme Q10 are commonly used in
MELAS syndrome without proven efficacy.
© 2015 Elsevier Inc. All rights reserved.

Contents

1. Introduction . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2. Clinical manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.1. Neurological manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 5
2.2. Muscular manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.3. Lactic acidemia . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.4. Cardiac manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.5. Gastrointestinal manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.6. Endocrine manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
2.7. Renal, pulmonary, dermatological, and hematological manifestations . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 7
3. Pathogenesis . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.1. Molecular genetic defects . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.2. Energy deficiency and angiopathy . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.3. Nitric oxide deficiency . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.4. Pathogenesis of various complications . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 8
3.5. Phenotype variability . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4. Management . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9

⁎ Corresponding author at: Department of Molecular and Human Genetics, Baylor College of Medicine, One Baylor Plaza, MS BCM225, Houston, TX 77030, USA.
E-mail address: fscaglia@bcm.edu (F. Scaglia).

http://dx.doi.org/10.1016/j.ymgme.2015.06.004
1096-7192/© 2015 Elsevier Inc. All rights reserved.
A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12 5

4.1. Evaluation of multi-organ involvement . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9


4.2. Management of complications. . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 9
4.3. Medications to avoid . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
5. Conclusions . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10
References . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . . 10

1. Introduction myopathy, recurrent headaches, hearing impairment, diabetes, and


short stature. Childhood is the typical age of onset with 65–76% of
Mitochondria are double membrane organelles found in all nucleated affected individuals presenting at or before the age of 20 years. Only
human cells and perform a variety of essential functions, including the 5–8% of individuals present before the age of 2 years and 1–6% after the
generation of most cellular energy in the form of adenosine triphosphate age of 40 years [6,9–11].
(ATP). The inner mitochondrial membrane harbors the electron trans- Individuals with MELAS syndrome frequently present with more
port chain (ETC) complexes that transfer electrons, translocate protons, than one initial clinical manifestation. Table 1 summarizes the initial
and produce ATP. Mitochondria contain extra-chromosomal DNA (mito- manifestations in affected individuals [6,9,10]. Table 2 summarizes the
chondrial DNA, mtDNA). However, only a very small proportion of mito- clinical manifestations of MELAS syndrome organized according to
chondrial proteins are encoded by that DNA; whereas the majority of their prevalence [6,9–11]. Below the manifestations of MELAS syndrome
mitochondrial proteins are encoded by nuclear DNA (nDNA). Mutations are presented according to the organ or system involved.
in mtDNA or mitochondria-related nDNA genes can result in mitochon-
drial dysfunction leading to mitochondrial diseases. Dysfunctional mito-
chondria are unable to generate sufficient ATP to meet the energy needs 2.1. Neurological manifestations
of various organs, particularly those with high energy demand, including
the nervous system, skeletal and cardiac muscles, kidneys, liver, and Stroke-like episodes are one of the cardinal features of MELAS
endocrine systems. Some patients with mitochondrial diseases display syndrome that occur in 84–99% of affected individuals [6,9,10]. These
a cluster of clinical features that fall into a discrete clinical syndrome. episodes present clinically with partially reversible aphasia, cortical
However, there is often considerable clinical variability, and many af- vision loss, motor weakness, headaches, altered mental status, and
fected individuals do not fit into one particular syndrome [1]. seizures with the eventual progressive accumulation of neurological
Mitochondrial encephalomyopathy, lactic acidosis, and stroke-like deficits. The affected areas in neuroimaging do not correspond to classic
episodes (MELAS) syndrome is one of the most frequent maternally vascular distribution (hence called “stroke-like”), are asymmetric,
inherited mitochondrial disorders which was first delineated in 1984 involve predominantly the temporal, parietal, and occipital lobes, and
[2]. The molecular basis of MELAS syndrome was initially discovered can be restricted to cortical areas or involve subcortical white matter
in 1990 when adenine to guanine transition at position 3243 of [5,11] (Fig. 1). Brain magnetic resonance (MR) angiography is usually
mtDNA (m.3243ANG) in the MT-TL1 gene encoding tRNALeu(UUR) was normal; whereas MR spectroscopy shows decreased N-acetylaspartate
found to be associated with this syndrome [3,4]. In 1992, clinical diag- signals and accumulation of lactate [11]. The high ventricular lactate
nostic criteria for MELAS syndrome were published indicating that the measured using MR spectroscopy was found to correlate with the de-
clinical diagnosis of this syndrome is based on the following three gree of the neurological impairment in individuals with MELAS syn-
invariant criteria: 1) stroke-like episodes before age 40 years, 2) enceph- drome [12].
alopathy characterized by seizures and/or dementia, and 3) mitochon- Dementia occurs in 40–90% of affected individuals [6,9,10]. Both
drial myopathy evident by lactic acidosis and/or ragged-red fibers the underlying neurological dysfunction and the accumulating cortical
(RRFs). The diagnosis is considered confirmed if there are also at least injuries due to stroke-like episodes contribute to the observed dementia
two of the following criteria: 1) normal early psychomotor develop- which affects intelligence, language, perception, attention, and memory
ment, 2) recurrent headaches, and 3) recurrent vomiting episodes [5]. function [11]. Additionally, executive function deficits have been ob-
More recently, the MELAS study group committee in Japan published served despite the relative sparing of the frontal lobe in neuroimaging
other diagnostic criteria by which the diagnosis is considered definitive suggesting an additional diffuse neurodegenerative process besides
with at least two category A criteria (headaches with vomiting, seizures, the damage caused by the stroke-like episodes [11].
hemiplegia, cortical blindness, and acute focal lesions in neuroimaging) Epilepsy is another common neurological manifestation occurring
and two category B criteria (high plasma or cerebrospinal fluid (CSF) in 71–96% of individuals with MELAS syndrome [6,9,10]. Epilepsy in
lactate, mitochondrial abnormalities in muscle biopsy, and a MELAS-
related gene mutation) [6]. The prevalence of MELAS syndrome has
been estimated to be 0.2:100,000 in Japan [6]. Other mtDNA mutations
were subsequently found to cause MELAS syndrome; however, the Table 1
m.3243ANG remained the commonest universally. The m.3243ANG, Initial manifestations of MELAS syndrome.
which was subsequently found to be associated with other phenotypes Frequency Manifestations
that collectively constitute a wide spectrum ranging from MELAS
N25% Seizure
syndrome at the severe end to asymptomatic carrier status, was found Recurrent headaches
to be relatively common with a prevalence of 16–18:100,000 in Stroke-like episode
Finland [7,8]. In this review, we summarize the clinical manifestations Cortical vision loss
of MELAS syndrome along with its pathogenic mechanisms and manage- Muscle weakness
Recurrent vomiting
ment options.
Short stature
10–24% Altered consciousness
Impaired mentation
2. Clinical manifestations Hearing impairment
Diabetes
MELAS syndrome is a multi-organ disease with broad manifesta- b10% Developmental delay
Fever
tions including stroke-like episodes, dementia, epilepsy, lactic acidemia,
6 A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12

Table 2
Overall manifestations of MELAS syndrome.

Frequency Manifestations

≥90% Stroke-like episodes


Dementia
Epilepsy
Lactic acidemia
Ragged red fibers
Exercise intolerance
75–89% Hemiparesis
Cortical vision loss
Recurrent headaches
Hearing impairment
Muscle weakness
50–74% Peripheral neuropathy
Learning disability
Memory impairment
Recurrent vomiting
Short stature
25–49% Basal ganglia calcification
Myoclonus
Ataxia
Episodic altered consciousness
Gait disturbance
Depression
Anxiety
Psychotic disorders
Diabetes
b25% Optic atrophy
Pigmentary retinopathy
Progressive external ophthalmoplegia
Motor developmental delay
Cardiomyopathy
Cardiac conduction abnormalities
Nephropathy
Vitiligo

individuals with MELAS syndrome is heterogeneous. Despite the


frequent focal nature of the brain insult by the stroke-like episodes,
not only focal, but also primary generalized seizures can occur. Primary
generalized seizures in MELAS syndrome can occur with normal neuro-
imaging or abnormalities including stroke-like episodes, white matter
lesions, cortical atrophy, and corpus callosum agenesis or hypogenesis.
Seizures can occur in MELAS syndrome as a manifestation of a stroke-
like episode or independently, and may even induce a stroke-like
episode [13].
Recurrent headaches occur in 54–91% of individuals with MELAS
syndrome [6,9–11]. Migrainous headaches in the form of recurrent
attacks of severe pulsatile headaches with frequent vomiting are typical
in individuals with MELAS syndrome and can precipitate stroke-like
episodes [14]. On the other hand, these headache episodes are often
more severe during the stroke-like episodes [10].
Hearing impairment occurs in 71–77% of individuals with MELAS
syndrome [9–11]. Sensorineural hearing loss in MELAS syndrome is typ-
ically mild, insidiously progressive, and often an early clinical manifes-
tation [11].
Peripheral neuropathy is another common manifestation of MELAS
syndrome occurring in 22–77% of affected individuals. The neuropathy
in MELAS syndrome is usually a chronic and progressive, sensorimotor, Fig. 1. Neuroimaging for a 9 year old girl with MELAS syndrome who presented with headache:
A) axial CT image demonstrates basal ganglia calcification as well as diffuse decreased
and distal polyneuropathy. Nerve conduction studies typically show an attenuation in the right occipital lobe involving both gray and white matter, B) sagittal recon-
axonal or mixed axonal and demyelinating neuropathy [11,15,16]. structed image reveals vermian atrophy, C) axial FLAIR image reveals increased signal in the
Other neurological manifestations in MELAS syndrome include cortex and subcortical white matter of the right occipital lobe as well as a smaller focus in the
learning disability, memory impairment, myoclonus, ataxia, episodes right lateral temporal lobe, D) axial diffusion weighted images demonstrated restriction
in the subcortical white matter of the right occipital lobe, E) axial ADC (apparent diffusion
of altered consciousness, basal ganglia calcifications in neuroimaging,
coefficient) map confirms restricted diffusion, and F) axial post contrast T1-weighted image re-
and elevated protein in CSF analysis [9,10]. Ophthalmological complica- veals increased sulcal enhancement, which may represent hyperemia or luxury perfusion. Six
tions include optic atrophy, pigmentary retinopathy, and ophthal- months later, the same girl presented with left sided weakness: G & H) FLAIR images demon-
moplegia [9,10]. Psychiatric illnesses can occur in MELAS syndrome strate resolution of abnormal signal at the right occipital pole with evolving encephalomalacia
and include depression, bipolar disorder, anxiety, psychosis, and per- in the lateral occipital lobe. New extensive cortical/subcortical signal abnormalities are
appreciated in the right temporal lobe, bilateral frontal lobes, and bilateral parietal lobes.
sonality changes [17].
A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12 7

2.2. Muscular manifestations

Myopathy is a cardinal manifestation of MELAS syndrome. Exercise


intolerance and muscle weakness occurs in 73–100% and 42–89% of
affected individuals, respectively [6,9–11]. Motor developmental delay
was reported in 23% of affected children [18].
Histologic examination of muscle tissue from individuals with MELAS
syndrome shows scattered vacuolated muscle fibers with clear sur-
rounding rim using hematoxylin and eosin (H&E) staining. Using the
Gomori trichrome stain, the RRFs can be seen which represent mito-
chondrial proliferation below the plasma membrane of the muscular
fibers causing the contour of the muscle fiber to become irregular.
These proliferated mitochondria in RRFs also stain strongly with the suc-
cinate dehydrogenase (SDH) stain giving the appearance of ragged blue
fibers. Although RRFs are present in many other mitochondrial diseases
e.g., MERRF (myoclonic epilepsy with ragged red fibers), most of the
RRFs in MELAS stain positively with the cytochrome c oxidase (COX)
histochemical stain unlike other mitochondrial diseases where RRFs do
not react with COX. In MELAS syndrome, the COX stain of muscle tissue
can be decreased, normal, or increased which may reflect variable
m.3243ANG heteroplasmy in different muscle fibers (Fig. 2). Another
characteristic feature in MELAS syndrome is the excessive mitochondrial
proliferation observed in smooth muscle and endothelial cells in intra-
muscular blood vessels revealed with the SDH stain and called strong
SDH reactive blood vessels (SSVs). Biochemical analysis of respiratory
chain enzymes in muscle extracts usually shows multiple partial defects,
especially involving complex I and complex IV [10,11].

2.3. Lactic acidemia

Lactic acidemia is a cardinal sign that is present in 94% of affected


individuals. CSF lactate is also elevated in the majority of individuals
with MELAS syndrome [9,10]. Lactic acidemia is not specific for MELAS
syndrome as it can occur in other mitochondrial diseases, metabolic
diseases, and systemic illness. On the other hand, lactate level can be
normal in a minority of individuals with MELAS syndrome [9,10].

2.4. Cardiac manifestations

Cardiomyopathy occurs in 18–30% of individuals with MELAS


syndrome [6,9,10]. Both dilated and hypertrophic cardiomyopathies
have been observed in MELAS syndrome, however, the more typical is
a non-obstructive concentric hypertrophy [11]. Cardiac conduction
abnormalities including Wolff–Parkinson–White syndrome has been
reported in 13–27% of individuals with MELAS syndrome [9,10,18,19].

2.5. Gastrointestinal manifestations

Gastrointestinal complications are common in individuals with Fig. 2. Muscle histopathologic changes in individuals with MELAS syndrome: A–C
MELAS syndrome and occur in 64–77% of affected individuals [9–11]. show ragged red fiber with modified Gomori trichrome, NADH tetrazolium reductase,
Recurrent or cyclic vomiting is the commonest observed gastrointesti- and cytochrome c oxidase histochemistry, respectively. The arrow identifies the ragged
red fiber. The images are at 400× magnification each.
nal complaint in MELAS syndrome. Diarrhea, constipation, gastric
dysmotility, intestinal pseudo-obstruction, and recurrent pancreatitis
have also been reported in MELAS syndrome [20]. Failure to thrive has has occasionally been found in individuals with MELAS syndrome [22].
been reported in 28% of children with MELAS syndrome [6]. Hypothyroidism, hypogonadotropic hypogonadism, and hypoparathy-
roidism have also been reported in individuals with MELAS syndrome
2.6. Endocrine manifestations [23–25].

Diabetes occurs in 21–33% of individuals with MELAS syndrome [6,


11]. Diabetes in MELAS syndrome manifests at the average age of 2.7. Renal, pulmonary, dermatological, and hematological manifestations
38 years and can be type 1 or type 2 in nature. Individuals with type 2
diabetes can initially be treated by diet or sulfonylurea. Significant Renal manifestations of MELAS syndrome include Fanconi proximal
insulinopenia can develop and affected individuals may require insulin tubulopathy, proteinuria, and focal segmental glomerulosclerosis [26].
treatment [21]. Individuals with MELAS syndrome are typically shorter Pulmonary hypertension has been rarely reported in individuals with
than their unaffected family members. Short stature has been reported MELAS syndrome [27]. Dermatological complaints, including vitiligo
in 33–82% of affected individuals [6,9–11]. Growth hormone deficiency and diffuse erythema with reticular pigmentation, are infrequent
8 A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12

manifestations in MELAS syndrome [28,29]. Chronic anemia has contribution in impaired NO production. Low plasma citrulline may
been reported in individuals with MELAS syndrome [30]. result from decreased citrulline synthesis in the mitochondria of
enterocytes due to mitochondrial dysfunction [37,43]. Most of the
3. Pathogenesis citrulline flux is directed toward arginine synthesis; therefore, lower
citrulline availability can result in decreased de novo arginine synthesis
The pathogenesis of MELAS syndrome is not fully understood. The and lower intracellular arginine availability [37–39]. Decreased NO
observed phenotype of MELAS syndrome can be explained by several production can also result from impaired NOS activity due to reactive
interacting mechanisms including impaired mitochondrial energy pro- oxygen species (ROS) overproduction (oxidative stress) resulting from
duction, microvasculature angiopathy, and nitric oxide (NO) deficiency the ETC impairment [44,45]. Oxidative stress due to mitochondrial
(Fig. 3). dysfunction may also result in increased asymmetric dimethylarginine
(ADMA), which is an endogenous inhibitor of NOS [37,39,46]. In addi-
3.1. Molecular genetic defects tion to impaired NO production, postproduction NO sequestration can
contribute to the NO deficiency in MELAS syndrome. Mitochondrial
The m.3243ANG mutation in the MT-TL1 gene encoding tRNALeu(UUR) proliferation in endothelial cells in MELAS syndrome can be associated
is found in 80% of individuals with MELAS syndrome. Additional muta- with increased COX activity, which can react with and thus sequester
tions (e.g., m.3271TNC and m.3252ANG) in the MT-TL1 gene can also NO [11,43]. In addition, oxidative stress can result in decreased NO
cause MELAS syndrome. Rarely, mutations in other mitochondrial availability by shunting NO into reactive nitrogen species (RNS) forma-
genes have been reported to cause MELAS syndrome including MT-TL2 tion [47].
encoding tRNALeu(CUN), MT-TK encoding tRNALys, MT-TH encoding
tRNAHis, MT-TQ encoding tRNAGln, MT-TF encoding tRNAPhe, MT-TV 3.4. Pathogenesis of various complications
encoding tRNAVal, MT-ND1, MT-ND4, MT-ND5, and MT-ND6 encoding
subunits of complex I, MT-CO2 and MT-CO3 encoding subunits of complex During early stages of stroke-like episodes in MELAS syndrome,
IV, and MT-CYB encoding a subunit of complex III [10,31]. In addition, SPECT (single photon emission computed tomography) scanning stud-
mutations in the nuclear gene POLG encoding the mitochondrial DNA ies have demonstrated hypoperfusion in the affected regions, indicating
polymerase gamma have been associated with a MELAS-like phenotype that these episodes are due to ischemic insults [48]. It is believed that
[32]. these ischemic insults result from impaired perfusion in cerebral micro-
vasculature due to the angiopathy and NO deficiency that occur in
3.2. Energy deficiency and angiopathy MELAS syndrome [11,37–39,43,48].
The inability of dysfunctional mitochondria to generate sufficient
The m.3243ANG mutation results in impaired mitochondrial transla- ATP to meet the energy needs of muscle tissue can explain the observed
tion that leads to decreased mitochondrial protein synthesis affecting myopathy in MELAS syndrome. NO deficiency may also play a signifi-
the ETC complex subunits. Decreased synthesis of ETC complexes cant role in the myopathic manifestations of this syndrome. Normally,
results in impaired mitochondrial energy production [33,34]. The inabil- endothelial cells release basal and stimulated NO. During physical activ-
ity of dysfunctional mitochondria to generate sufficient ATP to meet the ity increased muscular blood flow stimulates endothelial NO production
energy needs of various organs results in the multi-organ dysfunction that contributes significantly to exercise-induced hyperemia in mus-
observed in MELAS syndrome (Fig. 3). cular tissue [40]. Decreased NO availability can potentially lead to
Energy deficiency can also stimulate mitochondrial proliferation. impaired muscle exercise-induced hyperemia and thus contribute to
Angiopathy due to mitochondrial proliferation in smooth muscle and the exercise intolerance reported in individuals with MELAS syndrome.
endothelial cells of small blood vessels occurs in MELAS syndrome and In addition, NO deficiency may result in decreased basal muscular
leads to impaired blood perfusion in microvasculature contributing perfusion, leading to limited availability of nutrients such as amino
significantly to the complications observed in MELAS syndrome partic- acids, and thus decreased muscle protein synthesis that may contribute
ularly stroke-like episodes [11,35,36] (Fig. 3). to the myopathy and muscle wasting observed in MELAS syndrome
[39].
3.3. Nitric oxide deficiency Lactic acidemia in MELAS syndrome results from the inability of
dysfunctional mitochondria to adequately oxidize glucose, leading to
In addition to energy depletion there has been growing evidence the accumulation of pyruvate and shunting of pyruvate to lactate [11].
that NO deficiency occurs in MELAS syndrome and can contribute signif- Moreover, hypoperfusion may result in lactic acidosis due to decreased
icantly to its complications [37–39]. NO is formed from arginine via the oxygen delivery to peripheral tissues and a shift to anaerobic glycolysis.
enzyme nitric oxide synthase (NOS), which catalyzes the conversion of NO deficiency in MELAS syndrome can result in decreased blood perfu-
arginine to citrulline. Citrulline can be converted to arginine via argin- sion and therefore may aggravate lactic acidosis [39].
inosuccinate synthase and argininosuccinate lyase. Therefore, both cit- Diabetes develops in MELAS syndrome due to multiple defects in
rulline and arginine are considered as NO donors. Citrulline is a non- insulin and glucose metabolism including insulin deficiency, increased
essential amino acid for which the main source is the de novo synthesis gluconeogenesis, and insulin resistance [49]. In pancreatic β-cells, an
in small intestine enterocytes through a number of mitochondrial en- ATP sensitive potassium channel is required for insulin release. De-
zymes [40]. creased ATP synthesis as a result of mitochondrial dysfunction can
NO produced by vascular endothelium plays a major role in vascular result in impaired insulin secretion and insulinopenia [11,21]. Two fac-
smooth muscle relaxation that is needed to maintain the patency of tors may contribute to increasing gluconeogenesis in MELAS syndrome:
small blood vessels [41,42]. Therefore, NO deficiency in MELAS syn- insulin resistance, which results in reduced insulin inhibition of hepatic
drome can result in impaired blood perfusion in the microvasculature gluconeogenesis, and lactic acidemia, which can fuel gluconeogenesis in
of different organs that can contribute to the pathogenesis of several the liver [49]. The etiology of insulin resistance in MELAS syndrome is
complications [38,39]. NO deficiency in MELAS syndrome is believed likely to be multifactorial. First, decreased glucose oxidative capacity
to be multifactorial in origin (Fig. 3). Mitochondrial proliferation in due to the mitochondrial dysfunction can result in impaired insulin
vascular endothelial cells can result in impaired normal endothelial responsiveness. Second, through enhancing endothelial NOS (eNOS)
function (endothelial dysfunction), and impaired endothelial NO syn- activity, insulin can induce vasodilation that results in an adequate
thesis can reflect one aspect of endothelial dysfunction. Decreased avail- delivery of glucose and insulin to muscle tissue [50]. Therefore, NO
ability of NO precursors, arginine and citrulline, may have a major deficiency in MELAS syndrome may lead to impaired insulin-mediated
A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12 9

Fig. 3. Pathogenesis of MELAS syndrome (NO: nitric oxide, NOS: nitric oxide synthase, ADMA: asymmetric dimethylarginine, RNS: reactive nitrogen species).

vasodilation, resulting in impaired insulin delivery and contributing to accounting for the clinical diversity seen in individuals harboring this
insulin resistance [49]. Finally, increased ROS production due to mito- mutation [1]. Interestingly, it has been demonstrated that the muscle
chondrial dysfunction can play a role in impaired insulin responsiveness m.3243ANG heteroplasmy correlates with maximum oxygen uptake
and the development of insulin resistance [51]. and workload, resting plasma lactate, and muscle morphology abnor-
Renal Fanconi syndrome is believed to result from impaired activity malities in individuals with MELAS syndrome indicating that the
of ATP-dependent sodium–potassium pumps that are needed in the threshold of muscle mutation load at which oxidative impairment
tubular reabsorption process. Growth failure may reflect the effects of occurs is about 50% [55].
a chronic systemic state of energy deficiency [11].
4. Management
3.5. Phenotype variability
There is no specific consensus approach for treating individuals with
The m.3243ANG mutation is associated with variable phenotypes MELAS syndrome. Management is largely symptomatic and should
that collectively constitute a wide spectrum, ranging from MELAS involve a multidisciplinary team that may include a neurologist, cardio-
syndrome at the severe end in only ~ 10% of individuals with the logist, endocrinologist, audiologist, ophthalmologist, physical and occu-
m.3243ANG mutation to asymptomatic carrier status in another ~10% pational therapists, psychologist, and social worker.
of m.3243ANG mutation carriers. Between these two extremes inter-
mediate phenotypes exist including single organ involvement (e.g., 4.1. Evaluation of multi-organ involvement
cardiomyopathy or DM) and multi-organ involvement with various
combinations of symptoms (e.g., myopathy, diabetes, and deafness) Upon diagnosis, a comprehensive evaluation for the multi-organ
that occasionally constitute distinctive syndromes including mater- involvement is needed. Affected individuals should also be followed at
nally inherited deafness and diabetes (MIDD) and progressive exter- regular intervals to monitor progression and screen for potential compli-
nal ophthalmoplegia (PEO) [11,52]. Other mitochondrial syndromes cations. Performing the following evaluations has been recommended at
including Leigh syndrome and MERRF have been associated with the time of diagnosis and as needed during regular follow up visits: compre-
m.3243ANG mutation as well [53,54]. hensive neurological examination with cognitive assessments, brain
The extreme variability in phenotypes associated with the MRI, audiologic and ophthalmologic examinations, growth assessment,
m.3243ANG mutation is a common observation in many mtDNA- echocardiogram and electrocardiogram, screening for hypothyroidism,
related mitochondrial diseases. Similarly to the majority of mtDNA mu- and screening for diabetes by fasting blood glucose and glucose tolerance
tations, the m.3243ANG is a heteroplasmic mutation, i.e., present in test [10].
some copies of mtDNA, and therefore the cells harbor a mixture of
mutant and normal mtDNA. During cell division, mutant mtDNAs are 4.2. Management of complications
distributed randomly among daughter cells. Therefore, the percentage
of mutant mtDNAs differs in different tissues and organs within the Sensorineural hearing loss has been successfully treated with
same individual. These tissues and organs have different thresholds in cochlear implants [56]. Seizures respond to traditional anticonvulsant
heteroplasmy percentage before clinical phenotypes manifest thus therapy and standard analgesics can be used for migraine headaches.
10 A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12

Cardiac manifestations can also benefit from standard pharmacologic Additionally, a randomized, double-blind, placebo-controlled study
therapy. Diabetes can be managed by dietary modifications with or showed that a combination therapy including creatine monohydrate,
without oral hypoglycemic agents. However, insulin therapy is often CoQ10, and lipoic acid resulted in improved muscle strength and lower
required [10]. Nutrition support is needed for children failing to thrive plasma lactate in individuals with MELAS syndrome and other mito-
and rehabilitation with physical and occupational therapy is needed chondrial cytopathies [65].
after stroke-like episodes. Psychiatric evaluation and treatment are CoQ10 does not cross the blood–brain barrier; therefore, it may have
needed for individuals with psychiatric manifestations. limited effect on the central nervous system. Idebenone is a CoQ10
Regular exercise can improve exercise capacity in individuals with analog that can cross the blood–brain barrier and has been shown to
MELAS syndrome and other mitochondrial myopathies. Endurance improve neurological complications in some case reports [66,67].
training can induce mitochondrial biogenesis whereas resistance train- L-Carnitine is required for long-chain fatty acid transportation to
ing can induce transferring normal mitochondrial templates from satel- the mitochondrial matrix where it undergoes β-oxidation. Secondary
lite cells to mature muscle that may lower the mutation heteroplasmy carnitine deficiency can be rarely observed in MELAS syndrome [58,
[57]. 68]. Carnitine supplementation can potentially enhance β-oxidation
Several supplementations, including antioxidants and cofactors, and replenish the intracellular pools of coenzyme A [58]. The carnitine
are being used in MELAS syndrome based on limited clinical trials can be given at doses of 3 g daily in three divided doses for adults and
[58]. Unblinded studies have showed that L-arginine therapy can 100 mg/kg/day for children in three divided doses [58].
be beneficial in stroke-like episode treatment and prevention. Intrave-
nous L-arginine infusion during the acute phase has been shown to 4.3. Medications to avoid
improve stroke-like episode symptoms whereas oral L-arginine supple-
mentation during the interictal phase has been shown to decrease the Valproic acid should be avoided in the treatment of seizure because
frequency and severity of stroke-like episodes [48,59]. The suggested of its deleterious effects on mitochondrial function. Clinically, valproic
intravenous L-arginine dose is 0.5 g/kg for children or 10 g/m2 body acid may result in worsening or triggering seizures in individuals
surface area for adults with a similar daily dose to be given orally in with MELAS syndrome [69,70]. Other antiepileptic drugs which may af-
three divided doses during the interictal phase [58]. The therapeutic fect the mitochondrial metabolism, include phenobarbital, carbamaze-
effect of arginine in stroke-like episodes in MELAS syndrome is pro- pine, phenytoin, oxcarbazepine, ethosuximide, zonisamide, topiramate,
posed to be due to increase NO availability leading to improve intrace- gabapentin and vigabatrin [71]. Metformin should be avoided in individ-
rebral vasodilation and blood flow. This potential mechanism has been uals with MELAS syndrome because of its propensity to cause lactic
supported by the demonstration that arginine supplementation to sub- acidosis [11]. Dichloroacetate, which reduces lactate by activating the
jects with MELAS syndrome results in increased NO production and im- pyruvate dehydrogenase enzyme, should be avoided in MELAS syn-
proved flow-mediated dilation (FMD), which is considered a measure of drome. A study evaluating the effect of dichloroacetate in individuals
NO synthesized by endothelial cells in response to re-perfusion [37,60]. with MELAS syndrome was terminated because of onset or worsening
Although the clinical effects of citrulline administration in MELAS syn- of peripheral neuropathy indicating that dichloroacetate can be associ-
drome have not been studied, a stable isotope study demonstrated ated with peripheral nerve toxicity [72]. Individuals with MELAS syn-
that citrulline supplementation to subjects with MELAS syndrome re- drome should also avoid agents with potential mitochondrial toxicity
sulted in increased NO production. Interestingly, citrulline supplemen- including aminoglycosides, linezolid, and alcohol [10]. Cigarette smoke
tation was found to induce a greater increase in the NO synthesis rate contains hundreds of compounds many of which can accumulate in mi-
than that associated with arginine supplementation, indicating that cit- tochondria and disturb the function of the ETC including phenolic com-
rulline is a more effective NO precursor than arginine. Therefore, citrul- pounds, aldehydes, heavy metals, carbon monoxide, nicotine, and aromatic
line may have a better therapeutic effect than arginine [37]. compounds [73]. Therefore, smoking can aggravate mitochondrial dysfunc-
Additionally, increasing NO availability with arginine or citrulline sup- tion in individuals with mitochondrial diseases.
plementation will potentially improve perfusion in all microvasculature
compartments. Therefore, the effect of arginine and citrulline 5. Conclusions
supplementation may not be limited to improving stroke-like episodes,
but may also lead to improvements in other clinical features of MELAS MELAS syndrome, which is a frequent maternally inherited mito-
syndrome, including muscle weakness, exercise intolerance, and lactic chondrial disorder, is a multi-organ disease with broad manifestations
acidosis. Interestingly, arginine and citrulline supplementation has including stroke-like episodes, dementia, epilepsy, lactic acidemia, and
been reported to result in a reduction in plasma alanine and lactate myopathy. The m.3243ANG mutation in the MT-TL1 gene occurs in
concentrations, suggesting that such supplementation may improve 80% of individuals with MELAS syndrome. Several mechanisms can
lactic acidemia in MELAS syndrome by increasing NO production and interact to result in the multi-organ phenotype of MELAS syndrome
improving perfusion and oxygen delivery [37,61]. Additional clinical including impaired mitochondrial energy production, microvasculature
studies assessing the clinical effects of citrulline and arginine supple- angiopathy, and NO deficiency. Management of MELAS syndrome is
mentations on different manifestations of MELAS syndrome are needed largely symptomatic and should involve a multidisciplinary team. Sev-
to determine their potential therapeutic utility in this syndrome. eral supplementations, including antioxidants and cofactors, are being
Coenzyme Q10 (CoQ10) facilitates electron transfer from complexes used in MELAS syndrome based on limited clinical trials. Valproic acid,
I and II to complex III of ETC and stabilizes the ETC complexes by provid- metformin, and dichloroacetate should be avoided in individuals with
ing protective antioxidant effects. Some studies showed beneficial MELAS syndrome.
effects on muscle weakness, fatigability, and lactate level for CoQ10 in
individuals with MELAS syndrome [62,63]. The recommended doses
References
are 5–10 mg/kg/day for children and 200–400 mg/day for adults [58].
Creatine, which is mainly stored in muscle, heart, and brain, is [1] A.W. El-Hattab, F. Scaglia, Mitochondrial disorders, in: B. Lee, F. Scaglia (Eds.), Inborn
Errors of Metabolism: From Neonatal Screening to Metabolic Pathways, Oxford
metabolized to phosphocreatine which is an essential phosphate
University Press, New York, NY, USA 2015, pp. 180–202.
donor for ATP regeneration in muscle and brain. Creatine monohydrate [2] S.G. Pavlakis, P.C. Phillips, S. DiMauro, D.C. De Vivo, L.P. Rowland, Mitochondrial
supplementation was shown to increase the strength of high-intensity myopathy, encephalopathy, lactic acidosis, and strokelike episodes: a distinctive
anaerobic and aerobic activities in individuals with MELAS syndrome clinical syndrome, Ann. Neurol. 16 (1984) 481–488.
[3] Y. Goto, I. Nonaka, S. Horai, A mutation in the tRNA(Leu)(UUR) gene associated with
and other mitochondrial cytopathies [64]. The recommended doses the MELAS subgroup of mitochondrial encephalomyopathies, Nature 348 (1990)
are 100 mg/kg/day for children and 2–5 g/day for adults [58]. 651–653.
A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12 11

[4] Y. Kobayashi, M.Y. Momoi, K. Tominaga, T. Momoi, K. Nihei, M. Yanagisawa, Y. [31] L.J. Wong, Pathogenic mitochondrial DNA mutations in protein-coding genes,
Kagawa, S. Ohta, A point mutation in the mitochondrial tRNA(Leu)(UUR) gene in Muscle Nerve 36 (2007) 279–293.
MELAS (mitochondrial myopathy, encephalopathy, lactic acidosis and stroke-like [32] A. Cheldi, D. Ronchi, A. Bordoni, B. Bordo, S. Lanfranconi, M.G. Bellotti, S. Corti, V.
episodes), Biochem. Biophys. Res. Commun. 173 (1990) 816–822. Lucchini, M. Sciacco, M. Moggio, P. Baron, G.P. Comi, A. Colombo, A. Bersano,
[5] M. Hirano, E. Ricci, M.R. Koenigsberger, R. Defendini, S.G. Pavlakis, D.C. DeVivo, S. Lombardia GENS collaborators, POLG1 mutations and stroke like episodes: a
DiMauro, L.P. Rowland, MELAS: an original case and clinical criteria for diagnosis, distinct clinical entity rather than an atypical MELAS syndrome, BMC Neurol.
Neuromuscul. Disord. 2 (1992) 125–135. 13 (2013) 8.
[6] S. Yatsuga, N. Povalko, J. Nishioka, K. Katayama, N. Kakimoto, T. Matsuishi, T. [33] M.P. King, Y. Koga, M. Davidson, E.A. Schon, Defects in mitochondrial protein synthe-
Kakuma, Y. Koga, Taro Matsuoka for MELAS Study Group in Japan, MELAS: a nation- sis and respiratory chain activity segregate with the tRNA(Leu(UUR)) mutation
wide prospective cohort study of 96 patients in Japan, Biochim. Biophys. Acta 1820 associated with mitochondrial myopathy, encephalopathy, lactic acidosis, and
(2012) 619–624. strokelike episodes, Mol. Cell. Biol. 12 (1992) 480–490.
[7] K. Majamaa, J.S. Moilanen, S. Uimonen, A.M. Remes, P.I. Salmela, M. Kärppä, K.A. [34] A. Chomyn, J.A. Enriquez, V. Micol, P. Fernandez-Silva, G. Attardi, The mitochondrial
Majamaa-Voltti, H. Rusanen, M. Sorri, K.J. Peuhkurinen, I.E. Hassinen, Epidemiology myopathy, encephalopathy, lactic acidosis, and stroke-like episode syndrome-
of A3243G, the mutation for mitochondrial encephalomyopathy, lactic acidosis, and associated human mitochondrial tRNALeu(UUR) mutation causes aminoacylation
strokelike episodes: prevalence of the mutation in an adult population, Am. J. Hum. deficiency and concomitant reduced association of mRNA with ribosomes, J. Biol.
Genet. 63 (1998) 447–454. Chem. 275 (2000) 19198–19209.
[8] J. Uusimaa, J.S. Moilanen, L. Vainionpää, P. Tapanainen, P. Lindholm, M. Nuutinen, T. [35] H. Hasegawa, T. Matsuoka, Y. Goto, I. Nonaka, Strongly succinate dehydrogenase-
Löppönen, E. Mäki-Torkko, H. Rantala, K. Majamaa, Prevalence, segregation, and reactive blood vessels in muscles from patients with mitochondrial myopathy, en-
phenotype of the mitochondrial DNA 3243ANG mutation in children, Ann. Neurol. cephalopathy, lactic acidosis, and stroke-like episodes, Ann. Neurol. 29 (1991)
62 (2007) 278–287. 601–605.
[9] M. Hirano, S.G. Pavlakis, Mitochondrial myopathy, encephalopathy, lactic acidosis, [36] Y. Goto, S. Horai, T. Matsuoka, Y. Koga, K. Nihei, M. Kobayashi, I. Nonaka, Mitochon-
and strokelike episodes (MELAS): current concepts, J. Child Neurol. 9 (1994) 4–13. drial myopathy, encephalopathy, lactic acidosis, and stroke-like episodes (MELAS):
[10] S. DiMauro, M. Hirano, MELAS, in: R.A. Pagon, M.P. Adam, H.H. Ardinger, et al., (Eds.), a correlative study of the clinical features and mitochondrial DNA mutation, Neurol-
GeneReviews®, University of Washington, Seattle, 2013 [Internet]. ogy 42 (1992) 545–550.
[11] D.M. Sproule, P. Kaufmann, Mitochondrial encephalopathy, lactic acidosis, and [37] A.W. El-Hattab, J.W. Hsu, L.T. Emrick, L.J. Wong, W.J. Craigen, F. Jahoor, F. Scaglia,
strokelike episodes: basic concepts, clinical phenotype, and therapeutic manage- Restoration of impaired nitric oxide production in MELAS syndrome with citrulline
ment of MELAS syndrome, Ann. N. Y. Acad. Sci. 1142 (2008) 133–158. and arginine supplementation, Mol. Genet. Metab. 105 (2012) 607–614.
[12] P. Kaufmann, D.C. Shungu, M.C. Sano, S. Jhung, K. Engelstad, E. Mitsis, X. Mao, S. [38] A.W. El-Hattab, L.T. Emrick, W.J. Craigen, F. Scaglia, Citrulline and arginine utility in
Shanske, M. Hirano, S. DiMauro, D.C. De Vivo, Cerebral lactic acidosis correlates treating nitric oxide deficiency in mitochondrial disorders, Mol. Genet. Metab. 107
with neurological impairment in MELAS, Neurology 62 (2004) 1297–1302. (2012) 247–252.
[13] J. Finsterer, S. Zarrouk-Mahjoub, Focal and generalized seizures may occur in mito- [39] A.W. El-Hattab, L.T. Emrick, S. Chanprasert, W.J. Craigen, F. Scaglia, Mitochondria:
chondrial encephalomyopathy, lactic acidosis, and strokelike episodes (MELAS) role of citrulline and arginine supplementation in MELAS syndrome, Int. J. Biochem.
patients, J. Child. Neurol. (2015) (in press). Cell Biol. 48 (2014) 85–91.
[14] K. Ohno, E. Isotani, K. Hirakawa, MELAS presenting as migraine complicated by [40] G. Wu, S.M. Morris, Arginine metabolism: nitric oxide and beyond, Biochem. J. 336
stroke: case report, Neuroradiology 39 (1997) 781–784. (1998) 1–17.
[15] M. Kärppä, P. Syrjälä, U. Tolonen, K. Majamaa, Peripheral neuropathy in pa- [41] D.J. Green, A. Maiorana, G. O'Driscoll, R. Taylor, Effect of exercise training on
tients with the 3243ANG mutation in mitochondrial DNA, J. Neurol. 250 (2003) endothelium-derived nitric oxide function in humans, J. Physiol. 561 (2004) 1–25.
216–221. [42] N. Toda, T. Okamura, The pharmacology of nitric oxide in the peripheral nervous
[16] P. Kaufmann, J.M. Pascual, Y. Anziska, C.L. Gooch, K. Engelstad, S. Jhung, S. DiMauro, system of blood vessels, Pharmacol. Rev. 55 (2003) 271–324.
D.C. De Vivo, Nerve conduction abnormalities in patients with MELAS and the [43] A. Naini, P. Kaufmann, S. Shanske, K. Engelstad, D.C. De Vivo, E.A. Schon,
A3243G mutation, Arch. Neurol. 63 (2006) 746–748. Hypocitrullinemia in patients with MELAS: an insight into the “MELAS paradox”, J.
[17] R.E. Anglin, S.L. Garside, M.A. Tarnopolsky, M.F. Mazurek, P.I. Rosebush, The psychi- Neurol. Sci. 229–230 (2005) 187–193.
atric manifestations of mitochondrial disorders: a case and review of the literature, J. [44] C.H. Tengan, B.H. Kiyomoto, R.O. Godinho, J. Gamba, A.C. Neves, B. Schmidt, A.S.
Clin. Psychiatry 73 (2012) 506–512. Oliveira, A.A. Gabbai, The role of nitric oxide in muscle fibers with oxidative phos-
[18] D.M. Sproule, P. Kaufmann, K. Engelstad, T.J. Starc, A.J. Hordof, D.C. De Vivo, Wolff– phorylation defects, Biochem. Biophys. Res. Commun. 359 (2007) 771–777.
Parkinson–White syndrome in patients with MELAS, Arch. Neurol. 64 (2007) [45] V. Desquiret-Dumas, N. Gueguen, M. Barth, A. Chevrollier, S. Hancock, D.C. Wallace,
1625–1627. P. Amati-Bonneau, D. Henrion, D. Bonneau, P. Reynier, V. Procaccio, Metabolically in-
[19] Y. Okajima, Y. Tanabe, M. Takayanagi, H. Aotsuka, A follow up study of myocardial duced heteroplasmy shifting and L-arginine treatment reduce the energetic defect
involvement in patients with mitochondrial encephalomyopathy, lactic acidosis, in a neuronal-like model of MELAS, Biochim. Biophys. Acta 1822 (2012) 1019–1029.
and stroke-like episodes (MELAS), Heart 80 (1998) 292–295. [46] T. Teerlink, Z. Luo, F. Palm, C.S. Wilcox, Cellular ADMA: regulation and action,
[20] A. Fujii, M. Yoneda, M. Ohtani, H. Nakagawa, T. Kumano, K. Hayashi, A. Muramatsu, Pharmacol. Res. 60 (2009) 448–460.
S. Takabatake, T. Ibi, K. Sahashi, T. Azuma, M. Kuriyama, Gastric dysmotility associated [47] G. Vattemi, Y. Mechref, M. Marini, P. Tonin, P. Minuz, L. Grigoli, V. Guglielmi, I.
with accumulation of mitochondrial A3243G mutation in the stomach, Intern. Med. Klouckova, C. Chiamulera, A. Meneguzzi, M. Di Chio, V. Tedesco, L. Lovato, M.
43 (2004) 1126–1130. Degan, G. Arcaro, A. Lechi, M.V. Novotny, G. Tomelleri, Increased protein nitration
[21] J.A. Maassen, L.M. 'T Hart, E. Van Essen, R.J. Heine, G. Nijpels, R.S. Jahangir Tafrechi, in mitochondrial diseases: evidence for vessel wall involvement, Mol. Cell. Proteo-
A.K. Raap, G.M. Janssen, H.H. Lemkes, Mitochondrial diabetes: molecular mecha- mics 10 (2011) (M110.002964).
nisms and clinical presentation, Diabetes 53 (2004) S103–S109. [48] Y. Koga, Y. Akita, J. Nishioka, S. Yatsuga, N. Povalko, Y. Tanabe, S. Fujimoto, T.
[22] T. Yorifuji, M. Kawai, T. Momoi, H. Sasaki, K. Furusho, J. Muroi, K. Shimizu, Y. Matsuishi, L-Arginine improves the symptoms of strokelike episodes in MELAS,
Takahashi, M. Matsumura, M. Nambu, T. Okuno, Nephropathy and growth hormone Neurology 64 (2005) 710–712.
deficiency in a patient with mitochondrial tRNA(Leu(UUR)) mutation, J. Med. [49] A.W. El-Hattab, L.T. Emrick, J.W. Hsu, S. Chanprasert, F. Jahoor, F. Scaglia, W.J.
Genet. 33 (1996) 621–622. Craigen, Glucose metabolism derangements in adults with the MELAS m.3243ANG
[23] P. Balestri, S. Grosso, Endocrine disorders in two sisters affected by MELAS mutation, Mitochondrion 18 (2014) 63–69.
syndrome, J. Child Neurol. 15 (2000) 755–758. [50] A.M. Jonk, A.J. Houben, R.T. de Jongh, E.H. Serné, N.C. Schaper, C.D. Stehouwer CD,
[24] H. Topaloğlu, V. Seyrantepe, N. Kandemir, Z. Akçören, M. Ozgüç, mtDNA nt3243 mu- Microvascular dysfunction in obesity: a potential mechanism in the pathogenesis
tation, external ophthalmoplegia, and hypogonadism in an adolescent girl, Pediatr. of obesity-associated insulin resistance and hypertension, Physiology (Bethesda)
Neurol. 18 (1998) 429–431. 22 (2007) 252–260.
[25] K. Tanaka, Y. Takada, T. Matsunaka, S. Yuyama, S. Fujino, M. Maguchi, S. Yamashita, I. [51] J. Szendroedi, E. Phielix, M. Roden, The role of mitochondria in insulin resistance and
Yuba, Diabetes mellitus, deafness, muscle weakness and hypocalcemia in a patient type 2 diabetes mellitus, Nat. Rev. Endocrinol. 8 (2011) 92–103.
with an A3243G mutation of the mitochondrial DNA, Intern. Med. 39 (2000) 249–252. [52] V. Nesbitt, R.D. Pitceathly, D.M. Turnbull, R.W. Taylor, M.G. Sweeney, E.E.
[26] O. Hotta, C.N. Inoue, S. Miyabayashi, T. Furuta, A. Takeuchi, Y. Taguma, Clinical and Mudanohwo, S. Rahman, M.G. Hanna, R. McFarland, The UK MRC Mitochondrial
pathologic features of focal segmental glomerulosclerosis with mitochondrial Disease Patient Cohort Study: clinical phenotypes associated with the m.3243ANG
tRNALeu(UUR) gene mutation, Kidney Int. 59 (2001) 1236–1243. mutation—implications for diagnosis and management, J. Neurol. Neurosurg.
[27] D.M. Sproule, J. Dyme, J. Coku, D. de Vinck, E. Rosenzweig, W.K. Chung, D.C. De Vivo, Psychiatry 84 (2013) 936–938.
Pulmonary artery hypertension in a child with MELAS due to a point mutation of the [53] Y. Koga, Y. Akita, N. Takane, Y. Sato, H. Kato, Heterogeneous presentation in A3243G
mitochondrial tRNA((Leu)) gene (m.3243ANG), J. Inherit. Metab. Dis. 64 (2008) mutation in the mitochondrial tRNA(Leu(UUR)) gene, Arch. Dis. Child. 82 (2000)
1625–1627. 407–411.
[28] S.L. Karvonen, K.M. Haapasaari, M. Kallioinen, A. Oikarinen, I.E. Hassinen, K. [54] G.M. Fabrizi, E. Cardaioli, G.S. Grieco, T. Cavallaro, A. Malandrini, L. Manneschi, M.T.
Majamaa, Increased prevalence of vitiligo, but no evidence of premature ageing, Dotti, A. Federico, G. Guazzi, The A to G transition at nt 3243 of the mitochondrial
in the skin of patients with bp 3243 mutation in mitochondrial DNA in the mito- tRNALeu(UUR) may cause an MERRF syndrome, J. Neurol. Neurosurg. Psychiatry
chondrial encephalomyopathy, lactic acidosis and stroke-like episodes syndrome 61 (1996) 47–51.
(MELAS), Br. J. Dermatol. 140 (1999) 634–639. [55] T.D. Jeppesen, M. Schwartz, A.L. Frederiksen, F. Wibrand, D.B. Olsen, J. Vissing,
[29] Y. Kubota, T. Ishii, H. Sugihara, Y. Goto, M. Mizoguchi, Skin manifestations of a Muscle phenotype and mutation load in 51 persons with the 3243ANG mitochon-
patient with mitochondrial encephalomyopathy with lactic acidosis and strokelike drial DNA mutation, Arch. Neurol. 63 (2006) 1701–1706.
episodes (MELAS syndrome), J. Am. Acad. Dermatol. 41 (1999) 469–473. [56] M. Scarpelli, F. Zappini, M. Filosto, A. Russignan, P. Tonin, G. Tomelleri, Mitochondrial
[30] J. Finsterer, Chronic anemia as a manifestation of MELAS syndrome, Rev. Investig. sensorineural hearing loss: a retrospective study and a description of cochlear
Clin. 63 (2011) 100–103. implantation in a MELAS patient, Genet. Res. Int. 2012 (2012) 287432.
12 A.W. El-Hattab et al. / Molecular Genetics and Metabolism 116 (2015) 4–12

[57] T. Taivassalo, R.G. Haller, Implications of exercise training in mtDNA defects—use it [66] A. Napolitano, S. Salvetti, M. Vista, V. Lombardi, G. Siciliano, C. Giraldi, Long-term
or lose it? Biochim. Biophys. Acta 1659 (2004) 221–231. treatment with idebenone and riboflavin in a patient with MELAS, Neurol. Sci. 21
[58] F. Scaglia, J.L. Northrop, The mitochondrial myopathy encephalopathy, lactic acidosis (2000) S981–S982.
with stroke-like episodes (MELAS) syndrome: a review of treatment options, CNS [67] A. Lekoubou, A.E. Kouamé-Assouan, T.H. Cho, J. Luauté, N. Nighoghossian, L. Derex, Effect
Drugs 20 (2006) 443–464. of long-term oral treatment with L-arginine and idebenone on the prevention of stroke-
[59] Y. Koga, Y. Akita, J. Nishioka, S. Yatsuga, N. Povalko, K. Katayama, T. Matsuishi, like episodes in an adult MELAS patient, Rev. Neurol. (Paris) 167 (2011) 852–855.
MELAS and L-arginine therapy, Mitochondrion 2007 (7) (2007) 133–139. [68] C.C. Hsu, Y.H. Chuang, J.L. Tsai, H.J. Jong, Y.Y. Shen, H.L. Huang, H.L. Chen, H.C. Lee,
[60] Y. Koga, Y. Akita, N. Junko, S. Yatsuga, N. Povalko, R. Fukiyama, M. Ishii, T. Matsuishi, C.Y. Pang, Y.H. Wei, et al., CPEO and carnitine deficiency overlapping in MELAS
Endothelial dysfunction in MELAS improved by L-arginine supplementation, Neurol- syndrome, Acta Neurol. Scand. 92 (1995) 252–255.
ogy 66 (2006) 1766–1769. [69] C.W. Lam, C.H. Lau, J.C. Williams, Y.W. Chan, L.J. Wong, Mitochondrial myopathy,
[61] A.W. El-Hattab, L.T. Emrick, K.C. Williamson, W.J. Craigen, F. Scaglia, The effect of encephalopathy, lactic acidosis and stroke-like episodes (MELAS) triggered by
citrulline and arginine supplementation on lactic acidemia in MELAS syndrome, valproate therapy, Eur. J. Pediatr. 156 (1997) 562–564.
Meta Gene 1 (2013) 8–14. [70] C.M. Lin, P. Thajeb, Valproic acid aggravates epilepsy due to MELAS in a patient with
[62] R.S. Chen, C.C. Huang, N.S. Chu, Coenzyme Q10 treatment in mitochondrial an A3243G mutation of mitochondrial DNA, Metab. Brain Dis. 22 (2007) 105–109.
encephalomyopathies. Short-term double-blind, crossover study, Eur. Neurol. 37 [71] J. Finsterer, S. Zarrouk Mahjoub, Mitochondrial toxicity of antiepileptic drugs and
(1997) 212–218. their tolerability in mitochondrial disorders, Expert Opin. Drug Metab. Toxicol. 8
[63] E.I. Glover, J. Martin, A. Maher, R.E. Thornhill, G.R. Moran, M.A. Tarnopolsky, A (2012) 71–79.
randomized trial of coenzyme Q10 in mitochondrial disorders, Muscle Nerve 42 [72] P. Kaufmann, K. Engelstad, Y. Wei, S. Jhung, M.C. Sano, D.C. Shungu, W.S. Millar, X.
(2010) 739–748. Hong, C.L. Gooch, X. Mao, J.M. Pascual, M. Hirano, P.W. Stacpoole, S. DiMauro, D.C.
[64] M.A. Tarnopolsky, B.D. Roy, J.R. MacDonald, A randomized, controlled trial of crea- De Vivo, Dichloroacetate causes toxic neuropathy in MELAS: a randomized, con-
tine monohydrate in patients with mitochondrial cytopathies, Muscle Nerve 20 trolled clinical trial, Neurology 66 (2006) 324–330.
(1997) 1502–1509. [73] M. van der Toorn, D.J. Slebos, H.G. de Bruin, H.G. Leuvenink, S.J. Bakker, R.O. Gans,
[65] M.C. Rodriguez, J.R. MacDonald, D.J. Mahoney, G. Parise, M.F. Beal, M.A. Tarnopolsky, G.H. Koëter, A.J. van Oosterhout, H.F. Kauffman, Cigarette smoke-induced blockade
Beneficial effects of creatine, CoQ10, and lipoic acid in mitochondrial disorders, of the mitochondrial respiratory chain switches lung epithelial cell apoptosis into
Muscle Nerve 35 (2007) 235–242. necrosis, Am. J. Physiol. Lung Cell. Mol. Physiol. 292 (2007) L1211–L1218.

You might also like