Skip to main content
Erschienen in: Orphanet Journal of Rare Diseases 1/2012

Open Access 01.12.2012 | Research

Screening for primary creatine deficiencies in French patients with unexplained neurological symptoms

verfasst von: David Cheillan, Marie Joncquel-Chevalier Curt, Gilbert Briand, Gajja S Salomons, Karine Mention-Mulliez, Dries Dobbelaere, Jean-Marie Cuisset, Laurence Lion-François, Vincent Des Portes, Allel Chabli, Vassili Valayannopoulos, Jean-François Benoist, Jean-Marc Pinard, Gilles Simard, Olivier Douay, Kumaran Deiva, Alexandra Afenjar, Delphine Héron, François Rivier, Brigitte Chabrol, Fabienne Prieur, François Cartault, Gaëlle Pitelet, Alice Goldenberg, Soumeya Bekri, Marion Gerard, Richard Delorme, Marc Tardieu, Nicole Porchet, Christine Vianey-Saban, Joseph Vamecq

Erschienen in: Orphanet Journal of Rare Diseases | Ausgabe 1/2012

Abstract

A population of patients with unexplained neurological symptoms from six major French university hospitals was screened over a 28-month period for primary creatine disorder (PCD). Urine guanidinoacetate (GAA) and creatine:creatinine ratios were measured in a cohort of 6,353 subjects to identify PCD patients and compile their clinical, 1H-MRS, biochemical and molecular data. Six GAMT [N-guanidinoacetatemethyltransferase (EC 2.1.1.2)] and 10 X-linked creatine transporter (SLC6A8) but no AGAT (GATM) [L-arginine/glycine amidinotransferase (EC 2.1.4.1)] deficient patients were identified in this manner. Three additional affected sibs were further identified after familial inquiry (1 brother with GAMT deficiency and 2 brothers with SLC6A8 deficiency in two different families). The prevalence of PCD in this population was 0.25% (0.09% and 0.16% for GAMT and SLC6A8 deficiencies, respectively). Seven new PCD-causing mutations were discovered (2 nonsense [c.577C > T and c.289C > T] and 1 splicing [c.391 + 15G > T] mutations for the GAMT gene and, 2 missense [c.1208C > A and c.926C > A], 1 frameshift [c.930delG] and 1 splicing [c.1393-1G > A] mutations for the SLC6A8 gene). No hot spot mutations were observed in these genes, as all the mutations were distributed throughout the entire gene sequences and were essentially patient/family specific. Approximately one fifth of the mutations of SLC6A8, but not GAMT, were attributed to neo-mutation, germinal or somatic mosaicism events. The only SLC6A8-deficient female patient in our series presented with the severe phenotype usually characterizing affected male patients, an observation in agreement with recent evidence that is in support of the fact that this X-linked disorder might be more frequent than expected in the female population with intellectual disability.
Begleitmaterial
Additional file 1: Updated view of creatine metabolism and transport. Two enzymes are involved in the biosynthesis of creatine: arginine-glycineamidinotransferase (AGAT) and guanidinoacetatemethyltransferase (GAMT). A third protein, a plasma membrane transporter (SLC6A8), supplies the cell with creatine. A deficiency of any of these three biosynthesis and transport steps (namely, AGAT deficiency, GAMT deficiency and creatine transporter deficiency) leads to a deficiency of creatine in the brain (collapse of brain content in creatine). Biosynthesis of creatine happens in organs that contain both AGAT and GAMT. However, particularly in the brain, AGAT and GAMT may be distributed in different cells, so the guanidinoacetate (GAA) formed in one cell can be further transformed into creatine in another cell. This requires, as illustrated in the figure, the creatine transporter which can supply cells not only with creatine but also with GAA. The reason for collapse in brain creatine in either AGAT or GAMT deficiency is easily understood by a severe impairment of one of the two creatine biosynthesis steps. In creatine transporter deficiency, several facts concur including : (i) heterogeneity of brain cells for their content in AGAT and GAMT, (ii) most cerebral cells contain only of one of these two creatine biosynthesis enzymes; (iii) the blood–brain barrier is weakly permeable to systemic creatine and the brain must secure most of its creatine requirements by endogenous synthesis; and (iv) the creatine transporter contributes in the brain not only to supplying the cells with creatine but also directly to creatine biosynthesis by bridging the formation of an intermediate (GAA) in one cerebral cell to its utilization (towards creatine synthesis) in another cerebral cell. Another protein, the taurine transporter has also been shown to be active on GAA but in this case it leads to its removal from brain structures and CSF into blood, a feature having GAA detoxifying properties, notably in GAMT deficiency. Also depicted in the figure is the creatine kinase (CK) system (based on phosphocreatine as a stored form of energy produced from and capable of restoring ATP) and the non-enzymatic formation from creatine and phosphocreatine of creatinine (an end product of creatine metabolism removed in body fluids). Not depicted in the figure is the transit via body fluids which may take place in intercellular transfer and removal of creatine metabolites. For additional considerations and references, the reader is kindly referred to the recent review of Olivier Braissant (Ref [14] in the text). (DOC 50 KB)
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1186/​1750-1172-7-96) contains supplementary material, which is available to authorized users.
David Cheillan, Marie Joncquel-Chevalier Curt contributed equally to this work.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

DC and MJCC contributed equally to this work and are first co-authors. Authors (DC, MJCC, GB, KMM, DD, JMC; AC, VV; JFB; JMP, GSI, OD, AA, LL, VD, MG, RD, NP, JV) working in the six major French university hospitals (including those of Lille, Lyon and Angers and the Hôpital Necker Enfants Malades, Hôpital Robert Debré and Hôpital Raymond Poincaré from the Public Assistance of Parisian Hospitals) collaborated to group and to generate clinical biological, biochemical and genetic data. The other authors were also involved in the clinical follow-up (AG, GP, BC, FR, KD, MT) or biological chemistry (SB) of some of the patients and in the gene studies (FC, FP, DH and GSA). DC, MJCC, GB, NP and JV analyzed and integrated the clinical and paraclinical data. JV coordinated the writing of the manuscript with DC, MJCC and GB. All authors read and approved the final manuscript.

Introduction

Creatine is a physiological compound that was first isolated by the French chemist Eugène Chevreul in the 1830s [1]. It is supplied to the body both via endogenous biosynthesis and the diet [24]. Its name (the Greek ‘kreas’ means meat) originates from its abundance in muscle in which its energetic role has previously been documented [58]. Creatine can be phosphorylated by creatine kinases, leading to the storage of a high-energy phosphate bond of ATP in the form of phosphocreatine [3]. Upon demand, and due to the reversible activity of creatine kinases which can transform ADP back into ATP, phosphocreatine can release the stored energy [3]. This creatine/phosphocreatine cycle represents a vital reserve of energy in tissues with high energetic needs such as muscles and the brain [3]. In recent years, the physiological role of brain creatine has been extended to include the modulatory control of neurotransmission which, in addition to guanidinoacetate (GAA) toxicity, has improved our understanding of brain function and its severe impairment in disorders affecting creatine biosynthesis and transport [912].
The first step of creatine biosynthesis occurs essentially but not exclusively in the kidney and involves the transfer of an amidino group from arginine to glycine by the enzyme L-arginine/glycine amidinotransferase (AGAT) (EC 2.1.4.1) to form GAA and L-ornithine. Subsequently, N-guanidinoacetatemethyltransferase (GAMT) (EC 2.1.1.2) catalyzes the transfer of a methyl group from S-adenosyl methionine (SAM) on GAA. This second step occurs notably in the liver and leads to the formation of creatine and S-adenosylhomocysteine (SAH) [3]. Once synthesized, creatine may be released into the bloodstream. It can be internalized by cells via a specific plasma membrane Na+/Cl- dependent creatine transporter (SLC6A8) and stored as phosphocreatine [3]. Finally, about 1.7% of the creatine/phosphocreatine pool undergoes spontaneous degradation each day forming creatinine that is excreted in urine [3]. Urinary excretion of creatinine is a function of body muscle mass and in practice is a useful marker of kidney function. Little or no creatine is taken up by the brain from the systemic bloodstream because the blood brain barrier is practically impermeable to creatine. The brain must therefore secure most of its requirements in creatine by endogenous synthesis [10, 11, 13]. The crucial role of SLC6A8, which has been reviewed recently [14], provides ground for understanding how its deficiency results in a creatine collapse in brain.
Extracerebral and intracerebral routes for creatine supplies concur and their respective contributions differ in the immature (i.e. fetal and perinatal) and mature brain. In the immature brain, the extracerebral supply is favored due to a better efficacy of SLC6A8 in the blood–brain barrier (BBB). This means that extraction of creatine from the blood is predominant over intracerebral creatine biosynthesis which is physiologically limited by low GAMT activity in the immature brain [14]. SLC6A8 deficiency affects the immature brain creatine content through impairment of BBB uptake. In the mature brain, the contribution of BBB SLC6A8 becomes physiologically lower and intracerebral biosynthesis becomes the major pathway for the supply of brain creatine [14]. SLC6A8 deficiency therefore affects the mature brain creatine content through impairment of intracerebral creatine biosynthesis as a result of reduced SLC6A8-driven transfer of GAA from brain GAA to creatine-synthesizing cells. Differences existing between the extracellular/intracellular routes for the brain creatine supply explain why creatine supplementation is more efficient in two other creatine disorders, AGAT and GAMT deficiencies, when given at a presymptomatic stage of the disease when BBB uptake of creatine is physiologically optimal [14, 15]. An illustrated account of creatine biosynthesis and transport is provided in the Additional file 1.
Primary creatine deficiency disorders (PCD) are a new class of inborn errors of metabolism to which belong AGAT (OMIM 602360) and GAMT (OMIM 601240) deficiencies, both inherited as an autosomal recessive trait, and SLC6A8 [CT1, CRTR] deficiency (OMIM 300036), an X-linked disorder [1012, 1620]. The three disorders share a relative reduction of brain creatine and phosphocreatine levels [19], and accordingly, the clinical presentation is predominated by neurological signs [1012, 1620]. However, these disorders still remain under-diagnosed whether considered together [1620] or individually (AGAT [2125], GAMT [26, 27] or CRTR [2833] deficiency).
In this paper we present the results of a large retrospective study of urinary screening for PCD on 6,353 patients in whom the etiology of the underlying neurological disease was unknown at the time of urinary analysis of creatine and its metabolites.

Patients and methods

We retrospectively collected the metabolic results of urinary screening for PCD (GAA and creatine:creatinine ratios) from 6,353 patients with neurological disease of unknown origin (4,426 male patients and 1,927 female patients) hospitalized in six French university hospitals (Angers, Lille, Lyon and Paris (Hôpital Necker Enfants Malades, Hôpital Robert Debré and Hôpital Raymond Poincaré)) over a period of 28 months. Data with regard to GAA and creatine levels in plasma were also collected when available. For all patients with an abnormal result, a second sample was analyzed after a 24-hour diet devoid of meat and fish according to the recommendation of Arias et al. [34]. A patient was considered “at risk of PCD” (AGAT, GAMT or SLC6A8 deficiency) when the biochemical abnormality was confirmed on this second sample. Subsequently a molecular study and/or a functional test were performed to confirm the diagnosis. For the genetic study, informed consent was obtained from each patient or from their parents if probands were under 18. For patients with a confirmed diagnosis of PCD, extensive clinical data were collected, including familial, obstetrical and personal histories, main symptoms having led to consult, the specialty of the physician who requested metabolic investigations, the patient’s age at diagnosis, clinical signs present at diagnosis and results of 1H-MRS exploration. All patients with other diagnosed conditions (in particular, urea cycle disorders and remethylation defects) were discarded from the study.

Methods for metabolite measurements

Creatine and GAA levels were measured in urine and plasma by tandem mass spectrometry (LC-MS/MS) using stable isotopes as internal standards (13C2-GAA and 2H3-creatine) according to a method described previously [35]. Urinary creatinine was measured using LC-MS/MS using a specific internal standard (2H3-creatinine), except in two university hospitals in Paris (Necker Enfants Malades and Robert Debré) where this measurement was performed using the colorimetric Jaffé method [36, 37]. The control values and age groups used for plasma and urine GAA and creatine concentrations were those published by Verhoeven et al. [38].

Measurement of protein activities

GAMT activity was determined in lymphoblasts by assaying the transfer of methyl groups from 2H3-S-adenosylmethionine to 13C2-labelled GAA through the production of 13C22H3-creatine quantitated by an isotope dilution electrospray tandem spectrometry assay [39].
Creatine transporter activity was assayed by determining the intracellular incorporation of creatine in cultured skin fibroblasts as previously described [32].

Genetic studies

Genomic DNA was isolated from white blood cells or cultured fibroblasts collected from patients after informed consent. The coding regions and adjacent intronic splice sites of the GAMT and SLC6A8 genes were analyzed by direct sequencing. The pathogenicity of previously non-described mutations was predicted using the Alamut v2.02 software (Interactive Biosoftware, Rouen France) and compared to the LOVD database [40].

Results

Results of the screening for PCD in the French patients with unexplained neurological symptoms

Among the 6,353 patients screened, 16 patients were diagnosed to have PCD: 6 with GAMT deficiency (4 male and 2 female patients) and 10 with SLC6A8 deficiency (9 male and 1 female patients). Three additional affected sibs were identified after familial inquiries (1 brother affected with GAMT deficiency and 2 brothers with SLC6A8 deficiency in two different families). Diagnosis was confirmed by genetic analysis in 17 patients. For the other 2 patients (1 with GAMT deficiency and 1 with SLC6A8 deficiency), DNA analysis was rejected by the parents and the diagnosis was confirmed by the absence of a creatine peak in brain 1H-MRS, the urinary profile and the deficiency of protein activities. No patients with AGAT deficiency were identified in our cohort. The medical investigations leading to the diagnosis of these pathologies were requested by pediatric neurologists (42%), geneticists (32%), pediatricians (10.5%), adult neurologists (10.5%) and child psychiatrists (5%). Clinical descriptions and the results of the diagnostic explorations performed for the index patients and for the affected sibs are summarized in Table 1. The global prevalence of PCD in the screened population was 0.25% (16/6,353; CI95: 0.13% - 0.38%), the prevalence of GAMT deficiency was 0.09% (6/6,353; CI95: 0.02% - 0.17%) and the prevalence of SLC6A8 deficiency was 0.16% (12/6,353; CI95: 0.06% - 0.26%). Regarding our data, the prevalence of AGAT deficiency would be lower than 1/6,353, i.e. < 0.02% since no AGAT patient was diagnosed in the present study. The prevalence of PCD was 0.29% for the male patients (4 GAMT and 9 SLC6A8 deficiencies, 13/4,426; CI95: 0.14% - 0.46%) and 0.16% for the female patients (2 GAMT and 1 SLC6A8 deficiencies, 3/1,927; CI95: 0.0% - 0.34%). For the X-linked disorder SLC6A8 deficiency, the prevalence was 0.20% for the male patients (9/4,426; CI95: 0.08% - 0.34%) and 0.05% (1/1,927; CI95: 0.0% - 0.16%) for the female patients.
Table 1
Main clinical, biological, genetic and 1 H-MRS features of the patients affected with PCD
 
GAMT (n = 7)
CRTR (n = 12)
CLINICAL DATA
  
Sex
  
 Male
5/7
11/12
 Female
2/7
1/12
Ratio (M/F)
2.5
11
Consanguinity
+++
-
Age of onset (years)
 ≤ 2 years
6/7
12/12
 > 2 years
1/7
0/12
Age at diagnosis(years)
 ≤ 2 years
1/7
0/12
 [2–5 years]
3/7
3/12
 [5–10 years]
0/7
4/12
 > 10 years
3/7
5/12
Main clinical signs
 Intellectual disability
++++
++++
 Speech delay
++++
+++
 Failure to thrive
+
+
 Hypotonia
++
+
 Myopathy
+
-
 Motor delay
++
+++
 Epilepsy
++
+
 Extrapyramidal signs
+
-
 Attention deficit
++++
+++
 Sleeping disturbances
++
+
 Agressive behaviour
+++
-
 Autistic behaviour
++
+
Signs leading to consult
 Combined motor and speech delay
+++
+++
 Epilepsy
++
+
 Intellectual disability
+
-
 Autistic behavior
-
++
 Familial exploration
+
+
METABOLIC DATA
Plasma GAA (μmol/l)
 < 15 years: [0.35-1.8] a
[14–23]
[0.8 - 2.5]
 > 15 years: [1.0-3.5] a
[20-23]
2.9
Plasma creatine (μmol/l)
 < 10 years: [17109] a
[3.3 - 10]
59; 65
 > 10 years: [6.0-50] a
[4–6]
100; 112
Urine GAA/creatinine (mmol/mol)
 < 15 years: [2220] a
[399–1319]
[30–219]
 > 15 years: [378] a
242; 558
37; 38
Urine creatine/creatinine (mmol/mol)
 < 4 years: [61208] a
11; 33
2762
412 years: [17721] a
18; 26
[1638–3015]
 > 12 years: [11244] a
[12–32]
[1181–3195]
Brain 1 H-MRS investigation
Absence of creatine peak (n = 5)
Absence of creatine peak (n = 6)
GENETIC DATA
 Non-sense mutation
2/6
0/11
 Missense mutation
1/6
2/11
 Insertion
0/6
0/11
 Deletion
0/6
6/11
 Duplication
2/6
0/11
 Splicing mutation
1/6
3/11
FUNCTIONNAL TESTS
GAMT Activity (lymphoblasts)
Deficiency (2/2)
/
Creatine transport assay (fibroblasts)
/
Deficit of transport (3/3)
This table groups data from the diagnosed PCD patients and affected siblings. Fraction numbers refer to the number of positive patients (numerator) on the total number of patients studied (denominator) for the indicated item. Frequencies of signs in patients are expressed as + and – symbols: ++++, presence of signs in all patients; -, absence of signs in all patients; +++, ++ and +, presence of the sign in more than 75%, between 25 to 75%, and less than 25% of patients, respectively. a Reference laboratory values and age ranges are those published by Verhoeven et al. [38], patient data being expressed as either range or individual values. GAMT, guanidinoacetate methyltransferase; CRTR, creatine transporter SLC6A8.

Patients with GAMT deficiency

Seven patients from 6 unrelated families were diagnosed with GAMT deficiency. The sex ratio (M/F) was 2.5. Five of the 7 patients were born of a consanguineous union. Most of the patients were of Caucasian origin. Obstetrical history was available for 5 of the 7 patients; pregnancies were normal without pre-term birth. The mean ± SD values for birth-related parameters were: gestational term, 39.1 ± 0.5 weeks; birth weight, 3192 ± 135 g; birth length, 49.4 ± 1.4 cm and head circumference, 34.0 ± 1.2 cm. The predominant clinical signs at diagnosis were intellectual disability (100%) and speech delay (100%) (for general information on speech development and delay at a young age, see [41, 42]), and were associated with epilepsy (57%), motor delay (43%), hypotonia (43%) and extrapyramidal symptoms (29%). Behavioral disturbances were noticed in all patients essentially under the form of attention deficit (100%) with aggressive behaviors (hetero- and/or auto-mutilating episodes, anger and intolerance to frustration) (86%), autistic behavior (43%) and sleeping disturbances (43%). Most of these signs were usually present before 2 years of age and signs leading to consult are detailed in Table 1.
Diagnosis of GAMT deficiency was established at a median patient age of 5 years, with age values ranging between 2 and 29 years. In urine, the GAA:creatinine ratio (U-GAA/CTN) was increased for all patients and exceeded the upper control limits by 1.8- to 7.2-fold whereas the creatine:creatinine ratio (U-CT/CTN) was normal. When assayed, plasma GAA levels were systematically increased and creatine levels were below or equal to the lowest values of control limits. Cerebral 1H-MRS was performed in 5 patients and showed that creatine was undetectable in the brain in all the cases. In 2 of the 7 patients, GAMT activity was measured in lymphoblasts and these enzyme studies validated GAMT deficiency. The GAMT gene was studied in 6 of the 7 patients. In each case, patients had presumed (parents not available for study) or confirmed (parents were heterozygous for the mutation) homozygous mutations (Table 2). Mutations were either missense (n = 1), nonsense (n = 2), frame shift (n = 2) or splicing (n = 1) mutations.
Table 2
Mutations in the GAMT gene
Mutations
Number of patients identified in this study
Reference
Exon (e)/Intron (i)
Nucleotide
Amino acid
Homozygous
Heterozygous
e2
c.289C > T
p.Q97X
1
-
This study
e2
c.299_311dup13
p.R105GfsX26
2a
-
Dhar et al., 2009 [27]
i3
c.391 + 15G > T
p.(?)
1
-
This study
e5
c.506G > A
p.C169Y
1
-
Caldeira Araujo et al., 2005 [63]
e6
c.577C > T
p.Q193X
1
-
This study
Nucleotide numbering starting at the first adenine of the translation initiation codon ATG.
a Two affected siblings.

Patients with SLC6A8 deficiency

Twelve patients from 10 unrelated families were identified with SLC6A8 deficiency. The sex ratio (M/F) was 11. When available, the notion of consanguinity was absent. Obstetrical history was available for one patient with a gestational term at 39 weeks; birth weight, 3080 g; length, 48 cm, head circumference, 32 cm. At diagnosis, intellectual disability (100%) associated with language (75%) or motor (83%) delays were reported in most of the patients. Behavioral abnormalities were observed in 83% (10/12) patients and evoked those seen in GAMT deficiency patients, notably attention deficit (83%, 10/12 patients) with hyperactivity, autistic behavior (17%) and sleeping disturbances (8%). No aggressive behavior was observed. Epilepsy was reported in 17% of the patients. The main signs leading parents to consult (see Table 1 for an overview) were combined motor and speech delays; autistic behavior was the second cause of consultation. Familial inquiries led to the diagnosis of SLC6A8 deficiency in 2 of the 12 patients having an affected sibling. Anamnesis indicated that the initial signs could be present before the age of 2 years with in particular global psychomotor retardation and to a lesser extent epilepsy and autistic behavior. The median age for diagnosis was 9.5 years with age values ranging between 2.5 and 28 years.
The U-CT/CTN ratio was increased in all male patients, and exceeded the upper control limit by 2.3- to 13.6-fold. For the affected girl, the U-CT/CTN ratio was only 2.3-fold greater than the upper control limit. In plasma, creatine levels were normal or moderately increased (up to 1.3-fold greater than the upper control limit value). Plasma GAA levels were normal (or slightly affected), as were urinary U-GAA/CTN ratios. Cerebral 1H-MRS was performed in 50% (6/12) of the patients and consistently confirmed creatine deficiency in the brain in all examined patients (undetectable creatine peak). In 3 of the 12 (25%) patients, a functional study of SCL6A8 activity was carried out in cultured skin fibroblasts, and evidenced a deficiency of the creatine transporter function.
The SCL6A8 gene was studied in 92% (11/12) of the patients (Table 3). A hemizygous mutation was identified for all the affected male patients. For the female patient, a pathogenic mutation was identified at a heterozygous state and no X-inactivation biases could be identified in leucocytes. Mutations were either missense (n = 2), frame shift (n = 6) or splicing (n = 3) mutations. The underlying mutations were also detected in the mothers of the patients, except for the heterozygous female patient (c.942_944delCTT in exon 6) and a hemizygous boy (c.1208C > A in exon 8). In the latter two patients, SLC6A8 deficiencies were concluded to result from neo-mutation, germinal or somatic mosaicism.
Table 3
Mutations in the SLC6A8 gene
Mutations
Number of patients identified in this study
Reference
Exon (e)/Intron (i)
Nucleotide
Amino acid
Male
Female
Hemizigous
Heterozygous
e2
c.321_323delCTT
p.F107del
2 a
-
Degrauw et al., 2002[64]
i4
c.778-2A > G
p.(?)
1
-
Betsalel et al., 2011[61]
e6
c.926C > A
p.A309E
1
-
This study
e6
c.930delG
p.T311PfsX85
1
-
This study
e6
c.942_944delCTT
p.F314del
-
1
Fons et al., 2009[65]
e6
c.1006_1008delAAC
p.N336del
1
-
Clark et al., 2006[56]
e8
c.1208C > A
p.A403D
1
-
This study
i9
c.1393-1G > A
p.(?)
2 a
-
This study
e11
c.1519_1543del
p.I507LfsX5
1
-
Betsalel et al., 2011[61]
Nucleotide numbering starting at the first adenine of the translation initiation codon ATG.
a Two affected brothers.

Discussion

In the present study we have identified 16 new patients diagnosed with a PCD over a period of 28 months, as well as three affected siblings. Intellectual disability and attention deficit were, along with motor and speech delays, the main symptoms reported by the medical staff involved in the diagnosis of these disorders. Nevertheless, it is surprising that intellectual disability, which was a sign observed by the medical examiners in all patients with PCD, was not the main cause leading the parents to consult. This might reflect the difficulty of identifying intellectual disability in the very young child or in turn to of associating intellectual disability with PCD. These disorders are known to progress via irreversible lesions of the brain, so therapeutic measures are needed to be taken early in the life of the patients. For GAMT patients, supplementation with creatine together with a GAA lowering strategy (ornithine supplementation with or without arginine restriction) may represent an efficient treatment strategy [4346] and may provide some medical neurological benefit for GAMT patients. In these conditions, patients may recover a normal brain creatine peak as detected by 1H-MRS as well as normal urine GAA levels. They also improve clinically with better language and social development and apparently some regression of the epilepsy [47]. By contrast, in patients with SLC6A8 deficiency, supplementations with creatine and its precursors arginine or glycine, though improving muscular signs, were found to be without substantial benefit on cognitive and psychiatric signs, and failed to modify the creatine signal in 1H-MRS imaging of the brain [48], suggesting the inefficacy of these supplementations in improving brain as stated by other studies [4, 19, 30, 4952]. Though there is hope to treat efficiently this group of patients through the discovery of creatine pro-drugs capable of entering the brain and cells via a by-pass of the SCL6A8 creatine transporter, some clinical studies also question the inefficacy of the therapeutic measures mentioned above and, on the contrary, show beneficial effects with creatine and/or arginine supplementations in patients with SLC6A8 deficiency. In a heterozygous female patient with intractable epilepsy, treatment with creatine combined with arginine and glycine completely resolved seizures [53]. Creatine supplementation was also described to improve the neurological, language and behavioral status and was associated with a rise in the brain creatine peak as demonstrated by MRS in a child with SLC6A8 deficiency [54]. In a recent study, creatine deficient patients were also shown to be improved by a L-arginine-based therapy which positively impacted daily living skills, lowered the frequency of epileptic episodes and induced a mild increase in brain creatine and phosphocreatine MRS signals although normal cerebral levels of these metabolites were not recovered [55].
The calculated prevalence of PCD in our cohort was 0.25%. This result was not expected because most studies report a higher prevalence of PCD, notably SLC6A8 deficiency, which is generally estimated between 1% and 3% of the population affected with intellectual disability [5659]. However, this prevalence has been considered to be an overestimation of the real prevalence of PCD. Because PCDs are monogenic disorders, their prevalence was proposed to be no different from that of other nonsyndromic diseases such ARX (Aristaless-Related homeobox gene located on X chromosome) and therefore closer to 0.1% - 0.3% [60]. Thus, for the first time, our screening study provides strong practice-based evidence confirming this estimated prevalence for PCD. The girl diagnosed with SLC6A8 deficiency presented a severe phenotype similar to affected male patients, an observation that is not surprising in view of recent work [34, 61, 62] also describing this X-linked disorder in the female population with intellectual disability. In this respect, screening for SLC6A8 deficiency in female patients should be included in the diagnostic workup since this disorder still remains under-diagnosed. However, it should be noted that if the urinary U-CT/CTN ratio is not increased the diagnosis cannot be ruled out because it has been shown that the majority of female patients with a heterozygous mutation in SLC6A8 have a normal ratio, although the average ratio of this group is increased [62]. It might be recommended to perform brain MRS and/or SLC6A8 gene studies in female patients with suspected creatine transporter deficiency.
Five mutations were identified for GAMT gene in 6 unrelated families. These included 3 new mutations, c.289C > T, c.391 + 15G > T and c.577C > T, and two previously described mutations, c.299_311dup13 [27] and c.506G > A [63] (Table 2). The new mutation c391 + 15G > T was considered pathogenic by the creation of an alternative splicing donor site in intron 3 (Alamut software), by the segregation of this mutation in his parents and because it was linked with decreased GAMT activity in cultured fibroblasts.
The SLC6A8 gene was analyzed in 11 patients from 9 unrelated families. Nine mutations were identified throughout the gene, including 4 new mutations (2 missense (c.1208C > A and c.926C > A), 1 frameshift (c.930delG) and 1 splicing (c.1393-1G > A) mutation), and 5 previously described mutations (c.321_323delCTT [64], c.942_944delCTT [65], c.1006_1008delAAC [56], c.778-2A > G and c.1519_1543del [61]) (Table 3). In accordance with the results of Clark et al. [56], we found a fairly high proportion of frameshift and splicing mutations in our patients, and approximately one fifth of the mutations of SLC6A8, but not GAMT, were attributed to neo-mutation, germinal or somatic mosaicism events.
Interestingly, DNA studies performed in our patients clearly showed that mutations were essentially patient/family specific and highlighted the absence of hot spot mutations in these genes. Finally, as a mutation was identified in each patient for whom DNA was analyzed, the study of the target gene appears to be a key diagnostic step for PCD in suspect patients, keeping in mind that secondary creatine disorders such as deficiencies of ornithine delta-aminotransferase (EC 2.6.1.13) [66, 67], urea [6871] and homocysteine/methionine [72, 73] cycles, and succinate semialdehyde dehydrogenase (SSADH) (EC 1.2.1.24) [74, 75] may also be alternative causes of abnormal laboratory values of creatine metabolism markers.

Conclusion

Screening for PCD in the population with neurological disorders of unknown etiology has shown that these patients express important clinical signs of PCD which mainly include intellectual disability, speech delay, epilepsy, attention deficit and autistic behavior.
In this respect, some improvement might be made by conducting an information campaign among general and specialized educational staff to contribute to better recognizing intellectual disability and hence earlier referral of the child for professional medical evaluation. Improved anamnesis in regard to the familial history of possible patients might be also helpful. As suggested in the present study and highlighted in other work, both female and male patients can be concerned by severe PCD. However, and importantly, our study has shown that the prevalence of PCD remains low in the patients with unexplained neurological symptoms. Finally, the mutational spectrum of PCD has been extended by the new mutations detected during the present screening effort.
When PCD was suspected informed consent was obtained from patients and/or their parents for the DNA analysis of GAMT or SLC6A8 genes.

Acknowledgments

We would like to thank the patients for participating in this study. This work was supported by grants from the French Ministère de la Santé (PHRC 2003R/1903) and FMO (Fédération des Maladies Orphelines).
Open Access This article is published under license to BioMed Central Ltd. This is an Open Access article is distributed under the terms of the Creative Commons Attribution License ( https://​creativecommons.​org/​licenses/​by/​2.​0 ), which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properly cited.

Competing interests

The authors declare that they have no competing interests.

Authors’ contributions

DC and MJCC contributed equally to this work and are first co-authors. Authors (DC, MJCC, GB, KMM, DD, JMC; AC, VV; JFB; JMP, GSI, OD, AA, LL, VD, MG, RD, NP, JV) working in the six major French university hospitals (including those of Lille, Lyon and Angers and the Hôpital Necker Enfants Malades, Hôpital Robert Debré and Hôpital Raymond Poincaré from the Public Assistance of Parisian Hospitals) collaborated to group and to generate clinical biological, biochemical and genetic data. The other authors were also involved in the clinical follow-up (AG, GP, BC, FR, KD, MT) or biological chemistry (SB) of some of the patients and in the gene studies (FC, FP, DH and GSA). DC, MJCC, GB, NP and JV analyzed and integrated the clinical and paraclinical data. JV coordinated the writing of the manuscript with DC, MJCC and GB. All authors read and approved the final manuscript.
Anhänge

Electronic supplementary material

Additional file 1: Updated view of creatine metabolism and transport. Two enzymes are involved in the biosynthesis of creatine: arginine-glycineamidinotransferase (AGAT) and guanidinoacetatemethyltransferase (GAMT). A third protein, a plasma membrane transporter (SLC6A8), supplies the cell with creatine. A deficiency of any of these three biosynthesis and transport steps (namely, AGAT deficiency, GAMT deficiency and creatine transporter deficiency) leads to a deficiency of creatine in the brain (collapse of brain content in creatine). Biosynthesis of creatine happens in organs that contain both AGAT and GAMT. However, particularly in the brain, AGAT and GAMT may be distributed in different cells, so the guanidinoacetate (GAA) formed in one cell can be further transformed into creatine in another cell. This requires, as illustrated in the figure, the creatine transporter which can supply cells not only with creatine but also with GAA. The reason for collapse in brain creatine in either AGAT or GAMT deficiency is easily understood by a severe impairment of one of the two creatine biosynthesis steps. In creatine transporter deficiency, several facts concur including : (i) heterogeneity of brain cells for their content in AGAT and GAMT, (ii) most cerebral cells contain only of one of these two creatine biosynthesis enzymes; (iii) the blood–brain barrier is weakly permeable to systemic creatine and the brain must secure most of its creatine requirements by endogenous synthesis; and (iv) the creatine transporter contributes in the brain not only to supplying the cells with creatine but also directly to creatine biosynthesis by bridging the formation of an intermediate (GAA) in one cerebral cell to its utilization (towards creatine synthesis) in another cerebral cell. Another protein, the taurine transporter has also been shown to be active on GAA but in this case it leads to its removal from brain structures and CSF into blood, a feature having GAA detoxifying properties, notably in GAMT deficiency. Also depicted in the figure is the creatine kinase (CK) system (based on phosphocreatine as a stored form of energy produced from and capable of restoring ATP) and the non-enzymatic formation from creatine and phosphocreatine of creatinine (an end product of creatine metabolism removed in body fluids). Not depicted in the figure is the transit via body fluids which may take place in intercellular transfer and removal of creatine metabolites. For additional considerations and references, the reader is kindly referred to the recent review of Olivier Braissant (Ref [14] in the text). (DOC 50 KB)
Literatur
1.
Zurück zum Zitat Chevreul E: Sur la composition chimique du bouillon de viandes (On the chemical composition of meatbroth). J Pharm Sci Access. 1835, 21: 231-242. Chevreul E: Sur la composition chimique du bouillon de viandes (On the chemical composition of meatbroth). J Pharm Sci Access. 1835, 21: 231-242.
2.
Zurück zum Zitat Balsom P, Söderland K, Ekbolm B: Creatine in humans with special reference to creatine supplementation. Sports Med. 1994, 18 (4): 268-280. 10.2165/00007256-199418040-00005.CrossRefPubMed Balsom P, Söderland K, Ekbolm B: Creatine in humans with special reference to creatine supplementation. Sports Med. 1994, 18 (4): 268-280. 10.2165/00007256-199418040-00005.CrossRefPubMed
3.
Zurück zum Zitat Wyss M, Kaddurah-Daouk R: Creatine and creatinine metabolism. Physiol Rev. 2000, 80 (3): 1107-1213.PubMed Wyss M, Kaddurah-Daouk R: Creatine and creatinine metabolism. Physiol Rev. 2000, 80 (3): 1107-1213.PubMed
4.
Zurück zum Zitat Wyss M, Schulze A: Health implications of creatine: can oral creatine supplementation protect against neurological and atherosclerotic disease?. Neuroscience. 2002, 112 (2): 243-260. 10.1016/S0306-4522(02)00088-X.CrossRefPubMed Wyss M, Schulze A: Health implications of creatine: can oral creatine supplementation protect against neurological and atherosclerotic disease?. Neuroscience. 2002, 112 (2): 243-260. 10.1016/S0306-4522(02)00088-X.CrossRefPubMed
5.
Zurück zum Zitat Lundsgaard E: Untersuchungen über Muskel kontraktion en ohne Milchsäure bildung (Muscle contraction without formation of lactic acid). Biochem Z. 1930, 217: 162-177. Lundsgaard E: Untersuchungen über Muskel kontraktion en ohne Milchsäure bildung (Muscle contraction without formation of lactic acid). Biochem Z. 1930, 217: 162-177.
6.
Zurück zum Zitat Lundsgaard E: Weitere Untersuchungen fiber Muskel kontraktion en ohne Milchsäure bildung. (Further investigation on muscle contraction without lactate formation). Biochem Z. 1930, 227: 51-83. Lundsgaard E: Weitere Untersuchungen fiber Muskel kontraktion en ohne Milchsäure bildung. (Further investigation on muscle contraction without lactate formation). Biochem Z. 1930, 227: 51-83.
7.
Zurück zum Zitat Lohman K: Über die enzymatische aufspaltung der kreatin phosphorsaure; zugleich ein beitrag zum chemismus der muskel kontraktion. (On the enzymic cleavage of creatinephosphate; also a contribution to the chemistry of the muscle contraction). Biochem Z. 1934, 271: 264-277. Lohman K: Über die enzymatische aufspaltung der kreatin phosphorsaure; zugleich ein beitrag zum chemismus der muskel kontraktion. (On the enzymic cleavage of creatinephosphate; also a contribution to the chemistry of the muscle contraction). Biochem Z. 1934, 271: 264-277.
8.
Zurück zum Zitat Engelhardt WA, Lyubimova MN: Myosin and adenosine triphosphatase. Nature. 1939, 144: 668-669.CrossRef Engelhardt WA, Lyubimova MN: Myosin and adenosine triphosphatase. Nature. 1939, 144: 668-669.CrossRef
9.
Zurück zum Zitat Andres RH, Ducray AD, Schlattner U, Wallimannb T, Widmer HF: Functions and effects of creatine in the central nervous system. Brain Res Bull. 2008, 76 (4): 329-343. 10.1016/j.brainresbull.2008.02.035.CrossRefPubMed Andres RH, Ducray AD, Schlattner U, Wallimannb T, Widmer HF: Functions and effects of creatine in the central nervous system. Brain Res Bull. 2008, 76 (4): 329-343. 10.1016/j.brainresbull.2008.02.035.CrossRefPubMed
10.
Zurück zum Zitat Braissant O, Henry H: AGAT, GAMT and SLC6A8 distribution in the central nervous system, in relation to creatine deficiency syndromes: A review. J Inherit Metab Dis. 2008, 31 (2): 230-239. 10.1007/s10545-008-0826-9.CrossRefPubMed Braissant O, Henry H: AGAT, GAMT and SLC6A8 distribution in the central nervous system, in relation to creatine deficiency syndromes: A review. J Inherit Metab Dis. 2008, 31 (2): 230-239. 10.1007/s10545-008-0826-9.CrossRefPubMed
11.
Zurück zum Zitat Braissant O, Béard E, Torrent C, Henry H: Dissociation of AGAT, GAMT and SLC6A8 in CNS: relevance to creatine deficiency syndromes. Neurobiol Dis. 2010, 37 (2): 423-433. 10.1016/j.nbd.2009.10.022.CrossRefPubMed Braissant O, Béard E, Torrent C, Henry H: Dissociation of AGAT, GAMT and SLC6A8 in CNS: relevance to creatine deficiency syndromes. Neurobiol Dis. 2010, 37 (2): 423-433. 10.1016/j.nbd.2009.10.022.CrossRefPubMed
12.
Zurück zum Zitat Béard E, Braissant O: Synthesis and transport of creatine in the CNS: importance for cerebral functions. J Neurochem. 2010, 115 (2): 297-313. 10.1111/j.1471-4159.2010.06935.x.CrossRefPubMed Béard E, Braissant O: Synthesis and transport of creatine in the CNS: importance for cerebral functions. J Neurochem. 2010, 115 (2): 297-313. 10.1111/j.1471-4159.2010.06935.x.CrossRefPubMed
13.
Zurück zum Zitat Braissant O, Henry H, Loup M, Eilers B, Bachmann C: Endogenous synthesis and transport of creatine in the rat brain: an in situ hybridization study. Mol Brain Res. 2001, 86 (1–2): 193-201.CrossRefPubMed Braissant O, Henry H, Loup M, Eilers B, Bachmann C: Endogenous synthesis and transport of creatine in the rat brain: an in situ hybridization study. Mol Brain Res. 2001, 86 (1–2): 193-201.CrossRefPubMed
14.
Zurück zum Zitat Braissant O: Creatine and guanidinoacetate transport at blood–brain and blood-cerebrospinal fluid barriers. J Inherit Metab Dis. 2012, 35 (4): 655-664. 10.1007/s10545-011-9433-2.CrossRefPubMed Braissant O: Creatine and guanidinoacetate transport at blood–brain and blood-cerebrospinal fluid barriers. J Inherit Metab Dis. 2012, 35 (4): 655-664. 10.1007/s10545-011-9433-2.CrossRefPubMed
15.
Zurück zum Zitat Schulze A, Battini R: Pre-symptomatic treatment of creatine biosynthesis defects. Subcell Biochem. 2007, 46: 167-181. 10.1007/978-1-4020-6486-9_9.CrossRefPubMed Schulze A, Battini R: Pre-symptomatic treatment of creatine biosynthesis defects. Subcell Biochem. 2007, 46: 167-181. 10.1007/978-1-4020-6486-9_9.CrossRefPubMed
16.
Zurück zum Zitat Nasrallah F, Feki M, Kaabachi N: Creatine and creatine deficiency syndromes: biochemical and clinical aspects. Pediatr Neurol. 2010, 42 (3): 163-171. 10.1016/j.pediatrneurol.2009.07.015.CrossRefPubMed Nasrallah F, Feki M, Kaabachi N: Creatine and creatine deficiency syndromes: biochemical and clinical aspects. Pediatr Neurol. 2010, 42 (3): 163-171. 10.1016/j.pediatrneurol.2009.07.015.CrossRefPubMed
17.
18.
Zurück zum Zitat Stockler S, Schutz PW, Salomons GS: Cerebral creatine deficiency syndromes: clinical aspects, treatment and pathophysiology. Subcell Biochem. 2007, 46: 149-166. 10.1007/978-1-4020-6486-9_8.CrossRefPubMed Stockler S, Schutz PW, Salomons GS: Cerebral creatine deficiency syndromes: clinical aspects, treatment and pathophysiology. Subcell Biochem. 2007, 46: 149-166. 10.1007/978-1-4020-6486-9_8.CrossRefPubMed
19.
Zurück zum Zitat Stromberger C, Bodamer OA, Stöckler-Ipsiroglu S: Clinical characteristics and diagnostic clues in inborn errors of creatine metabolism. J Inherit Metab Dis. 2003, 26 (2–3): 299-308.CrossRefPubMed Stromberger C, Bodamer OA, Stöckler-Ipsiroglu S: Clinical characteristics and diagnostic clues in inborn errors of creatine metabolism. J Inherit Metab Dis. 2003, 26 (2–3): 299-308.CrossRefPubMed
20.
Zurück zum Zitat Sykut-Cegielska J, Gradowska W, Mercimek-Mahmutoglu S, Stöckler-Ipsiroglu S: Biochemical and clinical characteristics of creatine deficiency syndromes. Acta Biochim Pol. 2004, 51 (4): 875-882.PubMed Sykut-Cegielska J, Gradowska W, Mercimek-Mahmutoglu S, Stöckler-Ipsiroglu S: Biochemical and clinical characteristics of creatine deficiency syndromes. Acta Biochim Pol. 2004, 51 (4): 875-882.PubMed
21.
Zurück zum Zitat Battini R, Alessandrì MG, Leuzzi V, Moro F, Tosetti M, Bianchi MC, Cioni G: Arginine: glycine amidinotransferase (AGAT) deficiency in a newborn: early treatment can prevent phenotypic expression of the disease. J Pediatr. 2006, 148 (6): 828-830. 10.1016/j.jpeds.2006.01.043.CrossRefPubMed Battini R, Alessandrì MG, Leuzzi V, Moro F, Tosetti M, Bianchi MC, Cioni G: Arginine: glycine amidinotransferase (AGAT) deficiency in a newborn: early treatment can prevent phenotypic expression of the disease. J Pediatr. 2006, 148 (6): 828-830. 10.1016/j.jpeds.2006.01.043.CrossRefPubMed
22.
Zurück zum Zitat Carducci C, Birarelli M, Leuzzi V, Carducci C, Battini R, Cioni G, Antonozzi I: Guanidinoacetate and creatine plus creatinine assessment in physiologic fluids: an effective diagnostic tool for the biochemical diagnosis of arginine:glycine amidinotransferase and guanidinoacetate methyltransferase deficiencies. Clin Chem. 2002, 48 (10): 1772-1778.PubMed Carducci C, Birarelli M, Leuzzi V, Carducci C, Battini R, Cioni G, Antonozzi I: Guanidinoacetate and creatine plus creatinine assessment in physiologic fluids: an effective diagnostic tool for the biochemical diagnosis of arginine:glycine amidinotransferase and guanidinoacetate methyltransferase deficiencies. Clin Chem. 2002, 48 (10): 1772-1778.PubMed
23.
Zurück zum Zitat Item CB, Stöckler-Ipsiroglu S, Stromberger C, Mühl A, Alessandrì MG, Bianchi MC, Tosetti M, Fornai F, Cioni G: Arginine:glycine amidinotransferase deficiency: the third inborn error of creatine metabolism in humans. Am J Hum Genet. 2001, 69 (5): 1127-1133. 10.1086/323765.PubMedCentralCrossRefPubMed Item CB, Stöckler-Ipsiroglu S, Stromberger C, Mühl A, Alessandrì MG, Bianchi MC, Tosetti M, Fornai F, Cioni G: Arginine:glycine amidinotransferase deficiency: the third inborn error of creatine metabolism in humans. Am J Hum Genet. 2001, 69 (5): 1127-1133. 10.1086/323765.PubMedCentralCrossRefPubMed
24.
Zurück zum Zitat Mercimek-Mahmutoglu S, Stöckler-Ipsiroglu S: Creatine deficiency syndromes. GeneReviews. Edited by: Pagon RA, Bird TC, Dolan CR, Stephens K. 2009, Seattle (WA): University of Washington, Seattle, 1993–2010 Mercimek-Mahmutoglu S, Stöckler-Ipsiroglu S: Creatine deficiency syndromes. GeneReviews. Edited by: Pagon RA, Bird TC, Dolan CR, Stephens K. 2009, Seattle (WA): University of Washington, Seattle, 1993–2010
25.
Zurück zum Zitat Sun Q, O’Brien WE: Diagnosis of creatine metabolism disorders by determining creatine and guanidinoacetate in plasma and urine. Methods Mol Biol. 2010, 603: 175-185. 10.1007/978-1-60761-459-3_17.CrossRefPubMed Sun Q, O’Brien WE: Diagnosis of creatine metabolism disorders by determining creatine and guanidinoacetate in plasma and urine. Methods Mol Biol. 2010, 603: 175-185. 10.1007/978-1-60761-459-3_17.CrossRefPubMed
26.
Zurück zum Zitat Bodamer OA, Bloesch SM, Gregg AR, Stockler-Ipsiroglu S, O’Brien WE: Analysis of guanidinoacetate and creatine by isotope dilution electrospray tandem mass spectrometry. Clin Chim Acta. 2001, 308 (1–2): 173-178.CrossRefPubMed Bodamer OA, Bloesch SM, Gregg AR, Stockler-Ipsiroglu S, O’Brien WE: Analysis of guanidinoacetate and creatine by isotope dilution electrospray tandem mass spectrometry. Clin Chim Acta. 2001, 308 (1–2): 173-178.CrossRefPubMed
27.
Zurück zum Zitat Dhar SU, Scaglia F, Li FY, Smith L, Barshop BA, Eng CM, Haas RH, Hunter JV, Lotze T, Maranda B, Willis M, Abdenur JE, Chen E, O'Brien W, Wong LJ: Expanded clinical and molecular spectrum of guanidinoacetatemethyltransferase (GAMT) deficiency. Mol Genet Metab. 2009, 96 (1): 38-43. 10.1016/j.ymgme.2008.10.008.CrossRefPubMed Dhar SU, Scaglia F, Li FY, Smith L, Barshop BA, Eng CM, Haas RH, Hunter JV, Lotze T, Maranda B, Willis M, Abdenur JE, Chen E, O'Brien W, Wong LJ: Expanded clinical and molecular spectrum of guanidinoacetatemethyltransferase (GAMT) deficiency. Mol Genet Metab. 2009, 96 (1): 38-43. 10.1016/j.ymgme.2008.10.008.CrossRefPubMed
28.
Zurück zum Zitat Alcaide P, Merinero B, Ruiz-Sala P, Richard E, Navarrete R, Arias A, Ribes A, Artuch R, Campistol J, Ugarte M, Rodríguez-Pombo P: Defining the pathogenicity of creatine deficiency syndrome. Hum Mutat. 2011, 32 (3): 282-291. 10.1002/humu.21421.CrossRefPubMed Alcaide P, Merinero B, Ruiz-Sala P, Richard E, Navarrete R, Arias A, Ribes A, Artuch R, Campistol J, Ugarte M, Rodríguez-Pombo P: Defining the pathogenicity of creatine deficiency syndrome. Hum Mutat. 2011, 32 (3): 282-291. 10.1002/humu.21421.CrossRefPubMed
29.
Zurück zum Zitat Almeida LS, Verhoeven NM, Roos B, Valongo C, Cardoso ML, Vilarinho L, Salomons GS, Jakobs C: Creatine and guanidinoacetate: diagnostic markers for inborn errors in creatine biosynthesis and transport. Mol Genet Metab. 2004, 82 (3): 214-219. 10.1016/j.ymgme.2004.05.001.CrossRefPubMed Almeida LS, Verhoeven NM, Roos B, Valongo C, Cardoso ML, Vilarinho L, Salomons GS, Jakobs C: Creatine and guanidinoacetate: diagnostic markers for inborn errors in creatine biosynthesis and transport. Mol Genet Metab. 2004, 82 (3): 214-219. 10.1016/j.ymgme.2004.05.001.CrossRefPubMed
30.
Zurück zum Zitat Bizzi A, Bugiani M, Salomons GS, Hunneman DH, Moroni I, Estienne M, Danesi U, Jakobs C, Uziel G: X-linked creatine deficiency syndrome: a novel mutation in creatine transporter gene SLC6A8. Ann Neurol. 2002, 52 (2): 227-231. 10.1002/ana.10246.CrossRefPubMed Bizzi A, Bugiani M, Salomons GS, Hunneman DH, Moroni I, Estienne M, Danesi U, Jakobs C, Uziel G: X-linked creatine deficiency syndrome: a novel mutation in creatine transporter gene SLC6A8. Ann Neurol. 2002, 52 (2): 227-231. 10.1002/ana.10246.CrossRefPubMed
31.
Zurück zum Zitat Mercimek-Mahmutoglu S, Muehl A, Salomons GS, Neophytou B, Moeslinger D, Struys E, Bodamer OA, Jakobs C, Stockler-Ipsiroglu S: Screening for X-linked creatine transporter (SLC6A8) deficiency via simultaneous determination of urinary creatine to creatinine ratio by tandem mass-spectrometry. Mol Genet Metab. 2009, 96 (4): 273-275. 10.1016/j.ymgme.2008.12.020.CrossRefPubMed Mercimek-Mahmutoglu S, Muehl A, Salomons GS, Neophytou B, Moeslinger D, Struys E, Bodamer OA, Jakobs C, Stockler-Ipsiroglu S: Screening for X-linked creatine transporter (SLC6A8) deficiency via simultaneous determination of urinary creatine to creatinine ratio by tandem mass-spectrometry. Mol Genet Metab. 2009, 96 (4): 273-275. 10.1016/j.ymgme.2008.12.020.CrossRefPubMed
32.
Zurück zum Zitat Salomons GS, van Dooren SJ, Verhoeven NM, Cecil KM, Ball WS, Degrauw TJ, Jakobs C: X-linked creatine-transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome. Am J Hum Genet. 2001, 68 (6): 1497-1500. 10.1086/320595.PubMedCentralCrossRefPubMed Salomons GS, van Dooren SJ, Verhoeven NM, Cecil KM, Ball WS, Degrauw TJ, Jakobs C: X-linked creatine-transporter gene (SLC6A8) defect: a new creatine-deficiency syndrome. Am J Hum Genet. 2001, 68 (6): 1497-1500. 10.1086/320595.PubMedCentralCrossRefPubMed
33.
Zurück zum Zitat Sempere A, Fons C, Arias A, Rodríguez-Pombo P, Colomer R, Merinero B, Alcaide P, Capdevila A, Ribes A, Artuch R, Campistol J: Creatine transporter deficiency in two adult patients with static encephalopathy. J Inherit Metab Dis. 2009, Short Report #, 158 10.1007/s10545-009-1083-2. Sempere A, Fons C, Arias A, Rodríguez-Pombo P, Colomer R, Merinero B, Alcaide P, Capdevila A, Ribes A, Artuch R, Campistol J: Creatine transporter deficiency in two adult patients with static encephalopathy. J Inherit Metab Dis. 2009, Short Report #, 158 10.1007/s10545-009-1083-2.
34.
Zurück zum Zitat Arias A, Corbella M, Fons C, Sempere A, García-Villoria J, Ormazabal A, Poo P, Pineda M, Vilaseca MA, Campistol J, Briones P, Pàmpols T, Salomons GS, Ribes A, Artuch R: Creatine transporter deficiency: prevalence among patients with mental retardation and pitfalls in metabolite screening. Clin Biochem. 2007, 40 (16–17): 1328-1331.CrossRefPubMed Arias A, Corbella M, Fons C, Sempere A, García-Villoria J, Ormazabal A, Poo P, Pineda M, Vilaseca MA, Campistol J, Briones P, Pàmpols T, Salomons GS, Ribes A, Artuch R: Creatine transporter deficiency: prevalence among patients with mental retardation and pitfalls in metabolite screening. Clin Biochem. 2007, 40 (16–17): 1328-1331.CrossRefPubMed
35.
Zurück zum Zitat Cognat S, Cheillan D, Piraud M, Roos B, Jakobs C, Vianey-Saban C: Determination of guanidinoacetate and creatine in urine and plasma by liquid chromatography-tandem mass spectrometry. Clin Chem. 2004, 50 (8): 1459-1461. 10.1373/clinchem.2004.034538.CrossRefPubMed Cognat S, Cheillan D, Piraud M, Roos B, Jakobs C, Vianey-Saban C: Determination of guanidinoacetate and creatine in urine and plasma by liquid chromatography-tandem mass spectrometry. Clin Chem. 2004, 50 (8): 1459-1461. 10.1373/clinchem.2004.034538.CrossRefPubMed
36.
Zurück zum Zitat Jaffe M: Uber den niederschlag, welchen pikrinsaure in normalen ham erzeugt und ubereineneue reaction des kreatinins. Z Physiol Chem. 1886, 10: 391-400. Jaffe M: Uber den niederschlag, welchen pikrinsaure in normalen ham erzeugt und ubereineneue reaction des kreatinins. Z Physiol Chem. 1886, 10: 391-400.
37.
Zurück zum Zitat Tsikas D, Wolf A, Mitschke A, Gutzki FM, Will W, Bader M: GC-MS determination of creatinine in human biological fluids as pentafluorobenzyl derivative in clinical studies and biomonitoring: Inter-laboratory comparison in urine with Jaffé, HPLC and enzymatic assays. J Chromatogr B Analyt Technol Biomed Life Sci. 2010, 878 (27): 2582-2592. 10.1016/j.jchromb.2010.04.025.CrossRefPubMed Tsikas D, Wolf A, Mitschke A, Gutzki FM, Will W, Bader M: GC-MS determination of creatinine in human biological fluids as pentafluorobenzyl derivative in clinical studies and biomonitoring: Inter-laboratory comparison in urine with Jaffé, HPLC and enzymatic assays. J Chromatogr B Analyt Technol Biomed Life Sci. 2010, 878 (27): 2582-2592. 10.1016/j.jchromb.2010.04.025.CrossRefPubMed
38.
Zurück zum Zitat Verhoeven NM, Salomons GS, Jakobs C: Laboratory diagnosis of defects of creatine biosynthesis and transport. Clin Chim Acta. 2005, 361 (1–2): 1-9.CrossRefPubMed Verhoeven NM, Salomons GS, Jakobs C: Laboratory diagnosis of defects of creatine biosynthesis and transport. Clin Chim Acta. 2005, 361 (1–2): 1-9.CrossRefPubMed
39.
Zurück zum Zitat Verhoeven NM, Roos B, Struys EA, Salomons GS, Van Der Knaap MS, Jakobs C: Enzyme assay for diagnosis of guanidinoacetatemethyltransferase deficiency. Clin Chem. 2004, 50 (2): 441-443. 10.1373/clinchem.2003.022764.CrossRefPubMed Verhoeven NM, Roos B, Struys EA, Salomons GS, Van Der Knaap MS, Jakobs C: Enzyme assay for diagnosis of guanidinoacetatemethyltransferase deficiency. Clin Chem. 2004, 50 (2): 441-443. 10.1373/clinchem.2003.022764.CrossRefPubMed
40.
Zurück zum Zitat Fokkema IF, Taschner PE, Schaafsma GC, Celli J, Laros JF, den Dunnen JT: LOVD v.2.0: the next generation in gene variant databases. Hum Mutat. 2011, 32 (5): 557-563. 10.1002/humu.21438.CrossRefPubMed Fokkema IF, Taschner PE, Schaafsma GC, Celli J, Laros JF, den Dunnen JT: LOVD v.2.0: the next generation in gene variant databases. Hum Mutat. 2011, 32 (5): 557-563. 10.1002/humu.21438.CrossRefPubMed
41.
Zurück zum Zitat Lung AK, Kao CP: Evaluation and management of the child with speech delay. Am Fam Physician. 1999, 59 (11): 3121-3128. Lung AK, Kao CP: Evaluation and management of the child with speech delay. Am Fam Physician. 1999, 59 (11): 3121-3128.
42.
Zurück zum Zitat Wankoff LS: Warning signs in the development of speech, language, and communication: when to refer to a speech-language pathologist. J Child Adolesc Psychiatr Nurs. 2011, 24 (3): 175-184. 10.1111/j.1744-6171.2011.00292.x.CrossRefPubMed Wankoff LS: Warning signs in the development of speech, language, and communication: when to refer to a speech-language pathologist. J Child Adolesc Psychiatr Nurs. 2011, 24 (3): 175-184. 10.1111/j.1744-6171.2011.00292.x.CrossRefPubMed
43.
Zurück zum Zitat Bianchi MC, Tosetti M, Battini R, Leuzzi V, Alessandri MG, Carducci C, Antonozzi I, Cioni G: Treatment monitoring of brain creatine deficiency syndromes: a 1H- and 31P-MR spectroscopy study. Am J Neuroradiol. 2007, 28 (3): 548-554.PubMed Bianchi MC, Tosetti M, Battini R, Leuzzi V, Alessandri MG, Carducci C, Antonozzi I, Cioni G: Treatment monitoring of brain creatine deficiency syndromes: a 1H- and 31P-MR spectroscopy study. Am J Neuroradiol. 2007, 28 (3): 548-554.PubMed
44.
Zurück zum Zitat Ensenauer R, Thiel T, Schwab KO, Tacke U, Stöckler S: Guanidinoacetate methyltransferase deficiency: differences of creatine uptake in human brain and muscle. Mol Genet Metab. 2004, 82 (3): 208-213. 10.1016/j.ymgme.2004.04.005.CrossRefPubMed Ensenauer R, Thiel T, Schwab KO, Tacke U, Stöckler S: Guanidinoacetate methyltransferase deficiency: differences of creatine uptake in human brain and muscle. Mol Genet Metab. 2004, 82 (3): 208-213. 10.1016/j.ymgme.2004.04.005.CrossRefPubMed
45.
Zurück zum Zitat Gordon N: Guanidinoacetate methyltransferase deficiency (GAMT). Brain Dev. 2010, 32 (2): 79-81. 10.1016/j.braindev.2009.01.008.CrossRefPubMed Gordon N: Guanidinoacetate methyltransferase deficiency (GAMT). Brain Dev. 2010, 32 (2): 79-81. 10.1016/j.braindev.2009.01.008.CrossRefPubMed
46.
Zurück zum Zitat Schulze A, Hoffmann GF, Bachert P, Kirsch S, Salomons GS, Verhoeven NM, Mayatepek E: Presymptomatic treatment of neonatal guanidinoacetate methyltransferase deficiency. Neurology. 2006, 67 (4): 719-721. 10.1212/01.wnl.0000230152.25203.01.CrossRefPubMed Schulze A, Hoffmann GF, Bachert P, Kirsch S, Salomons GS, Verhoeven NM, Mayatepek E: Presymptomatic treatment of neonatal guanidinoacetate methyltransferase deficiency. Neurology. 2006, 67 (4): 719-721. 10.1212/01.wnl.0000230152.25203.01.CrossRefPubMed
47.
Zurück zum Zitat Blanc E: Approche thérapeutique d'une maladie génétique rare le déficit de synthèse en créatine par déficit en guanidinoacétate méthyltransferase. Thèse d’Exercice de Pharmacie (Cheillan D, Thesis Director). 2009, Université Claude Bernard (Lyon, France), http://www.sudoc.fr/145878910, 82 pages Blanc E: Approche thérapeutique d'une maladie génétique rare le déficit de synthèse en créatine par déficit en guanidinoacétate méthyltransferase. Thèse d’Exercice de Pharmacie (Cheillan D, Thesis Director). 2009, Université Claude Bernard (Lyon, France), http://​www.​sudoc.​fr/​145878910, 82 pages
48.
Zurück zum Zitat Valayannopoulos V, Boddaert N, Chabli A, Barbier V, Desguerre I, Philippe A, Afenjar A, Mazzuca M, Cheillan D, Munnich A, de Keyzer Y, Jakobs C, Salomons GS, de Lonlay P: Treatment by oral creatine, L-arginine and L-glycine in six severely affected patients with creatine transporter defect. J Inherit Metab Dis. 2012, 35 (1): 151-157. 10.1007/s10545-011-9358-9.CrossRefPubMed Valayannopoulos V, Boddaert N, Chabli A, Barbier V, Desguerre I, Philippe A, Afenjar A, Mazzuca M, Cheillan D, Munnich A, de Keyzer Y, Jakobs C, Salomons GS, de Lonlay P: Treatment by oral creatine, L-arginine and L-glycine in six severely affected patients with creatine transporter defect. J Inherit Metab Dis. 2012, 35 (1): 151-157. 10.1007/s10545-011-9358-9.CrossRefPubMed
49.
Zurück zum Zitat Braissant O, Henry H, Béard E, Uldry J: Creatine deficiency syndromes and the importance of creatine synthesis in the brain. Amino Acids. 2011, 40 (5): 1315-1324. 10.1007/s00726-011-0852-z.CrossRefPubMed Braissant O, Henry H, Béard E, Uldry J: Creatine deficiency syndromes and the importance of creatine synthesis in the brain. Amino Acids. 2011, 40 (5): 1315-1324. 10.1007/s00726-011-0852-z.CrossRefPubMed
50.
Zurück zum Zitat Evangeliou A, Vasilaki K, Karagianni P, Nikolaidis N: Clinical applications of creatine supplementation on paediatrics. Curr Pharm Biotechnol. 2009, 10 (7): 683-690. 10.2174/138920109789542075.CrossRefPubMed Evangeliou A, Vasilaki K, Karagianni P, Nikolaidis N: Clinical applications of creatine supplementation on paediatrics. Curr Pharm Biotechnol. 2009, 10 (7): 683-690. 10.2174/138920109789542075.CrossRefPubMed
51.
Zurück zum Zitat Fons C, Sempere A, Arias A, López-Sala A, Póo P, Pineda M, Mas A, Vilaseca MA, Salomons GS, Ribes A, Artuch R, Campistol J: Arginine supplementation in four patients with X-linked creatine transporter defect. J Inherit Metab Dis. 2008, 31 (6): 724-728. 10.1007/s10545-008-0902-1.CrossRefPubMed Fons C, Sempere A, Arias A, López-Sala A, Póo P, Pineda M, Mas A, Vilaseca MA, Salomons GS, Ribes A, Artuch R, Campistol J: Arginine supplementation in four patients with X-linked creatine transporter defect. J Inherit Metab Dis. 2008, 31 (6): 724-728. 10.1007/s10545-008-0902-1.CrossRefPubMed
52.
Zurück zum Zitat Salomons GS, van Dooren SJ, Verhoeven NM, Marsden D, Schwartz C, Cecil KM, De Grauw TJ, Jakobs C: X-linked creatine transporter defect: an overview. J Inherit Metab Dis. 2003, 26 (2–3): 309-318.CrossRefPubMed Salomons GS, van Dooren SJ, Verhoeven NM, Marsden D, Schwartz C, Cecil KM, De Grauw TJ, Jakobs C: X-linked creatine transporter defect: an overview. J Inherit Metab Dis. 2003, 26 (2–3): 309-318.CrossRefPubMed
53.
Zurück zum Zitat Mercimek-Mahmutoglu S, Connolly MB, Poskitt KJ, Horvath GA, Lowry N, Salomons GS, Casey B, Sinclair G, Davis C, Jakobs C, Stockler-Ipsiroglu S: Treatment of intractable epilepsy in a female with SLC6A8 deficiency. Mol Genet Metab. 2010, 101 (4): 409-412. 10.1016/j.ymgme.2010.08.016.CrossRefPubMed Mercimek-Mahmutoglu S, Connolly MB, Poskitt KJ, Horvath GA, Lowry N, Salomons GS, Casey B, Sinclair G, Davis C, Jakobs C, Stockler-Ipsiroglu S: Treatment of intractable epilepsy in a female with SLC6A8 deficiency. Mol Genet Metab. 2010, 101 (4): 409-412. 10.1016/j.ymgme.2010.08.016.CrossRefPubMed
54.
Zurück zum Zitat Chilosi A, Leuzzi V, Battini R, Tosetti M, Ferretti G, Comparini A, Casarano M, Moretti E, Alessandri MG, Bianchi MC, Cioni G: Treatment with L-arginine improves neuropsychological disorders in a child with creatine transporter defect. Neurocase. 2008, 14 (2): 151-161. 10.1080/13554790802060821.CrossRefPubMed Chilosi A, Leuzzi V, Battini R, Tosetti M, Ferretti G, Comparini A, Casarano M, Moretti E, Alessandri MG, Bianchi MC, Cioni G: Treatment with L-arginine improves neuropsychological disorders in a child with creatine transporter defect. Neurocase. 2008, 14 (2): 151-161. 10.1080/13554790802060821.CrossRefPubMed
55.
Zurück zum Zitat Chilosi A, Casarano M, Comparini A, Battaglia FM, Mancardi MM, Schiaffino C, Tosetti M, Leuzzi V, Battini R, Cioni G: Neuropsychological profile and clinical effects of arginine treatment in children with creatine transport deficiency. Orphanet J Rare Dis. 2012, 7 (1): 43-10.1186/1750-1172-7-43.PubMedCentralCrossRefPubMed Chilosi A, Casarano M, Comparini A, Battaglia FM, Mancardi MM, Schiaffino C, Tosetti M, Leuzzi V, Battini R, Cioni G: Neuropsychological profile and clinical effects of arginine treatment in children with creatine transport deficiency. Orphanet J Rare Dis. 2012, 7 (1): 43-10.1186/1750-1172-7-43.PubMedCentralCrossRefPubMed
56.
Zurück zum Zitat Clark AJ, Rosenberg EH, Almeida LS, Wood TC, Jakobs C, Stevenson RE, Schwartz CE, Salomons GS: X-linked CT (SLC6A8) mutations in about 1% of males with mental retardation of unknown etiology. Hum Genet. 2006, 119 (6): 604-610. 10.1007/s00439-006-0162-9.CrossRefPubMed Clark AJ, Rosenberg EH, Almeida LS, Wood TC, Jakobs C, Stevenson RE, Schwartz CE, Salomons GS: X-linked CT (SLC6A8) mutations in about 1% of males with mental retardation of unknown etiology. Hum Genet. 2006, 119 (6): 604-610. 10.1007/s00439-006-0162-9.CrossRefPubMed
57.
Zurück zum Zitat Lion-François L, Cheillan D, Pitelet G, Acquaviva-Bourdain C, Bussy G, Cotton F, Guibaud L, Gérard D, Rivier C, Vianey-Saban C, Jakobs C, Salomons GS, des Portes V: High frequency of creatine deficiency syndromes in patients with unexplained mental retardation. Neurology. 2006, 67 (9): 1713-1714. 10.1212/01.wnl.0000239153.39710.81.CrossRefPubMed Lion-François L, Cheillan D, Pitelet G, Acquaviva-Bourdain C, Bussy G, Cotton F, Guibaud L, Gérard D, Rivier C, Vianey-Saban C, Jakobs C, Salomons GS, des Portes V: High frequency of creatine deficiency syndromes in patients with unexplained mental retardation. Neurology. 2006, 67 (9): 1713-1714. 10.1212/01.wnl.0000239153.39710.81.CrossRefPubMed
58.
Zurück zum Zitat Newmeyer A, Cecil KM, Schapiro M, Clark JF, Degrauw TJ: Incidence of brain CT deficiency in males with developmental delay referred for brain magnetic resonance imaging. J Dev Behav Pediatr. 2005, 26 (4): 276-282. 10.1097/00004703-200508000-00003.CrossRefPubMed Newmeyer A, Cecil KM, Schapiro M, Clark JF, Degrauw TJ: Incidence of brain CT deficiency in males with developmental delay referred for brain magnetic resonance imaging. J Dev Behav Pediatr. 2005, 26 (4): 276-282. 10.1097/00004703-200508000-00003.CrossRefPubMed
59.
Zurück zum Zitat Rosenberg EH, Almeida LS, Kleefstra T, De Grauw RS, Yntema HG, Bahi N, Moraine C, Ropers HH, Fryns JP, De Grauw TJ, Jakobs C, Salomons GS: High prevalence of SLC6A8 deficiency in X-linked mental retardation. Am J Hum Genet. 2004, 75 (1): 97-105. 10.1086/422102.PubMedCentralCrossRefPubMed Rosenberg EH, Almeida LS, Kleefstra T, De Grauw RS, Yntema HG, Bahi N, Moraine C, Ropers HH, Fryns JP, De Grauw TJ, Jakobs C, Salomons GS: High prevalence of SLC6A8 deficiency in X-linked mental retardation. Am J Hum Genet. 2004, 75 (1): 97-105. 10.1086/422102.PubMedCentralCrossRefPubMed
60.
Zurück zum Zitat Mandel JL: Comparative frequency of fragile-X (FMR1) and creatine transporter (SLC6A8) mutations in X-linked mental retardation. Am J Hum Genet. 2004, 75 (4): 730-731. 10.1086/424821.PubMedCentralCrossRefPubMed Mandel JL: Comparative frequency of fragile-X (FMR1) and creatine transporter (SLC6A8) mutations in X-linked mental retardation. Am J Hum Genet. 2004, 75 (4): 730-731. 10.1086/424821.PubMedCentralCrossRefPubMed
61.
Zurück zum Zitat Betsalel OT, Rosenberg EH, Almeida LS, Kleefstra T, Schwartz CE, Valayannopoulos V, Abdul-Rahman O, Poplawski N, Vilarinho L, Wolf P, den Dunnen JT, Jakobs C, Salomons GS: Characterization of novel SLC6A8 variants with the use of splice-site analysis tools and implementation of a newly developed LOVD database. Eur J Hum Genet. 2011, 19 (1): 56-63. 10.1038/ejhg.2010.134.PubMedCentralCrossRefPubMed Betsalel OT, Rosenberg EH, Almeida LS, Kleefstra T, Schwartz CE, Valayannopoulos V, Abdul-Rahman O, Poplawski N, Vilarinho L, Wolf P, den Dunnen JT, Jakobs C, Salomons GS: Characterization of novel SLC6A8 variants with the use of splice-site analysis tools and implementation of a newly developed LOVD database. Eur J Hum Genet. 2011, 19 (1): 56-63. 10.1038/ejhg.2010.134.PubMedCentralCrossRefPubMed
62.
Zurück zum Zitat van de Kamp JM, Mancini GM, Pouwels PJ, Betsalel OT, van Dooren SJ, de Koning I, Steenweg ME, Jakobs C, van der Knaap MS, Salomons GS: Clinical features and X-inactivation in females heterozygous for creatine transporter defect. Clin Genet. 2011, 79 (3): 264-272. 10.1111/j.1399-0004.2010.01460.x.CrossRefPubMed van de Kamp JM, Mancini GM, Pouwels PJ, Betsalel OT, van Dooren SJ, de Koning I, Steenweg ME, Jakobs C, van der Knaap MS, Salomons GS: Clinical features and X-inactivation in females heterozygous for creatine transporter defect. Clin Genet. 2011, 79 (3): 264-272. 10.1111/j.1399-0004.2010.01460.x.CrossRefPubMed
63.
Zurück zum Zitat Caldeira Araújo H, Smit W, Verhoeven NM, Salomons GS, Silva S, Vasconcelos R, Tomás H, Tavares De Almeida I, Jakobs C, Duran M: Guanidinoacetate methyltransferase deficiency identified in adults and a child with mental retardation. Am J Med Genet A. 2005, 133A (2): 122-127. 10.1002/ajmg.a.30226.CrossRefPubMed Caldeira Araújo H, Smit W, Verhoeven NM, Salomons GS, Silva S, Vasconcelos R, Tomás H, Tavares De Almeida I, Jakobs C, Duran M: Guanidinoacetate methyltransferase deficiency identified in adults and a child with mental retardation. Am J Med Genet A. 2005, 133A (2): 122-127. 10.1002/ajmg.a.30226.CrossRefPubMed
64.
Zurück zum Zitat De Grauw TJ, Salomons GS, Cecil KM, Chuck G, Newmeyer A, Schapiro MB, Jakobs C: Congenital creatine transporter deficiency. Neuropediatrics. 2002, 33 (5): 232-238. 10.1055/s-2002-36743.CrossRef De Grauw TJ, Salomons GS, Cecil KM, Chuck G, Newmeyer A, Schapiro MB, Jakobs C: Congenital creatine transporter deficiency. Neuropediatrics. 2002, 33 (5): 232-238. 10.1055/s-2002-36743.CrossRef
65.
Zurück zum Zitat Fons C, Sempere A, Sanmartí FX, Arias A, Póo P, Pineda M, Ribes A, Merinero B, Vilaseca MA, Salomons GS, Artuch R, Campistol J: Epilepsy spectrum in cerebral creatine transporter deficiency. Epilepsia. 2009, 50 (9): 2168-2170. 10.1111/j.1528-1167.2009.02142.x.CrossRefPubMed Fons C, Sempere A, Sanmartí FX, Arias A, Póo P, Pineda M, Ribes A, Merinero B, Vilaseca MA, Salomons GS, Artuch R, Campistol J: Epilepsy spectrum in cerebral creatine transporter deficiency. Epilepsia. 2009, 50 (9): 2168-2170. 10.1111/j.1528-1167.2009.02142.x.CrossRefPubMed
66.
Zurück zum Zitat Sipilä I: Inhibition of arginine-glycine amidinotransferase by ornithine. A possible mechanism for the muscular and chorioretinal atrophies in gyrate atrophy of the choroid and retina with hyperornithinemia. Biochim Biophys Acta. 1980, 613 (1): 79-84. 10.1016/0005-2744(80)90194-1.CrossRefPubMed Sipilä I: Inhibition of arginine-glycine amidinotransferase by ornithine. A possible mechanism for the muscular and chorioretinal atrophies in gyrate atrophy of the choroid and retina with hyperornithinemia. Biochim Biophys Acta. 1980, 613 (1): 79-84. 10.1016/0005-2744(80)90194-1.CrossRefPubMed
67.
Zurück zum Zitat Valayannopoulos V, Boddaert N, Mention K, Touati G, Barbier V, Chabli A, Sedel F, Kaplan J, Dufier JL, Seidenwurm D, Rabier D, Saudubray JM, de Lonlay P: Secondary creatine deficiency in ornithine delta-aminotransferase deficiency. Mol Genet Metab. 2009, 97 (2): 109-113. 10.1016/j.ymgme.2008.12.010.CrossRefPubMed Valayannopoulos V, Boddaert N, Mention K, Touati G, Barbier V, Chabli A, Sedel F, Kaplan J, Dufier JL, Seidenwurm D, Rabier D, Saudubray JM, de Lonlay P: Secondary creatine deficiency in ornithine delta-aminotransferase deficiency. Mol Genet Metab. 2009, 97 (2): 109-113. 10.1016/j.ymgme.2008.12.010.CrossRefPubMed
68.
Zurück zum Zitat Arias A, Garcia-Villoria J, Ribes A: Guanidinoacetate and creatine/creatinine levels in controls and patients with urea cycle defects. Mol Genet Metab. 2004, 82 (3): 220-223. 10.1016/j.ymgme.2004.04.009.CrossRefPubMed Arias A, Garcia-Villoria J, Ribes A: Guanidinoacetate and creatine/creatinine levels in controls and patients with urea cycle defects. Mol Genet Metab. 2004, 82 (3): 220-223. 10.1016/j.ymgme.2004.04.009.CrossRefPubMed
69.
Zurück zum Zitat Braissant O: Ammonia toxicity to the brain: effects on creatine metabolism and transport and protective roles of creatine. Mol Genet Metab. 2010, 100 (Suppl 1): S53-S58.CrossRefPubMed Braissant O: Ammonia toxicity to the brain: effects on creatine metabolism and transport and protective roles of creatine. Mol Genet Metab. 2010, 100 (Suppl 1): S53-S58.CrossRefPubMed
70.
Zurück zum Zitat Brosnan JT, Brosnan ME: Creatine metabolism and the urea cycle. Mol Genet Metab. 2010, 100 (Suppl 1): S49-S52.CrossRefPubMed Brosnan JT, Brosnan ME: Creatine metabolism and the urea cycle. Mol Genet Metab. 2010, 100 (Suppl 1): S49-S52.CrossRefPubMed
71.
Zurück zum Zitat Roze E, Azuar C, Menuel C, Haberle J, Guillevin R: Usefulness of magnetic resonance spectroscopy in urea cycle disorders. Pediatr Neurol. 2007, 37 (3): 222-225. 10.1016/j.pediatrneurol.2007.05.003.CrossRefPubMed Roze E, Azuar C, Menuel C, Haberle J, Guillevin R: Usefulness of magnetic resonance spectroscopy in urea cycle disorders. Pediatr Neurol. 2007, 37 (3): 222-225. 10.1016/j.pediatrneurol.2007.05.003.CrossRefPubMed
72.
Zurück zum Zitat Baric I, Fumic K, Glenn B, Cuk M, Schulze A, Finkelstein JD, James SJ, Mejaski-Bosnjak V, Pazanin L, Pogribny IP, Rados M, Sarnavka V, Scukanec-Spoljar M, Allen RH, Stabler S, Uzelac L, Vugrek O, Wagner C, Zeisel S, Mudd SH: S-adenosylhomocysteine hydrolase deficiency in a human: a genetic disorder of methionine metabolism. Proc Natl Acad Sci USA. 2004, 101 (12): 4234-4239. 10.1073/pnas.0400658101.PubMedCentralCrossRefPubMed Baric I, Fumic K, Glenn B, Cuk M, Schulze A, Finkelstein JD, James SJ, Mejaski-Bosnjak V, Pazanin L, Pogribny IP, Rados M, Sarnavka V, Scukanec-Spoljar M, Allen RH, Stabler S, Uzelac L, Vugrek O, Wagner C, Zeisel S, Mudd SH: S-adenosylhomocysteine hydrolase deficiency in a human: a genetic disorder of methionine metabolism. Proc Natl Acad Sci USA. 2004, 101 (12): 4234-4239. 10.1073/pnas.0400658101.PubMedCentralCrossRefPubMed
73.
Zurück zum Zitat Bodamer OA, Sahoo T, Beaudet AL, O'Brien WE, Bottiglieri T, Stockler-Ipsiroglu S, Wagner C, Scaglia F: Creatine metabolism in combined methylmalonicaciduria and homocystinuria. Ann Neurol. 2005, 57 (4): 557-560. 10.1002/ana.20419.CrossRefPubMed Bodamer OA, Sahoo T, Beaudet AL, O'Brien WE, Bottiglieri T, Stockler-Ipsiroglu S, Wagner C, Scaglia F: Creatine metabolism in combined methylmalonicaciduria and homocystinuria. Ann Neurol. 2005, 57 (4): 557-560. 10.1002/ana.20419.CrossRefPubMed
74.
Zurück zum Zitat Jansen EE, Verhoeven NM, Jakobs C, Schulze A, Senephansiri H, Gupta M, Snead OC, Gibson KM: Increased guanidino species in murine and human succinate semialdehyde dehydrogenase (SSADH) deficiency. Biochim Biophys Acta. 2006, 1762 (4): 494-498. 10.1016/j.bbadis.2006.01.006.CrossRefPubMed Jansen EE, Verhoeven NM, Jakobs C, Schulze A, Senephansiri H, Gupta M, Snead OC, Gibson KM: Increased guanidino species in murine and human succinate semialdehyde dehydrogenase (SSADH) deficiency. Biochim Biophys Acta. 2006, 1762 (4): 494-498. 10.1016/j.bbadis.2006.01.006.CrossRefPubMed
75.
Zurück zum Zitat Vamecq J, Joncquel-Chevalier Curt M, Mention-Mulliez K, Dobbelaere D, Briand G: Rise in brain GABA to further stress the metabolic link between valproate and creatine. Mol Genet Metab. 2011, 102 (2): 232-234. 10.1016/j.ymgme.2010.10.006.CrossRefPubMed Vamecq J, Joncquel-Chevalier Curt M, Mention-Mulliez K, Dobbelaere D, Briand G: Rise in brain GABA to further stress the metabolic link between valproate and creatine. Mol Genet Metab. 2011, 102 (2): 232-234. 10.1016/j.ymgme.2010.10.006.CrossRefPubMed
Metadaten
Titel
Screening for primary creatine deficiencies in French patients with unexplained neurological symptoms
verfasst von
David Cheillan
Marie Joncquel-Chevalier Curt
Gilbert Briand
Gajja S Salomons
Karine Mention-Mulliez
Dries Dobbelaere
Jean-Marie Cuisset
Laurence Lion-François
Vincent Des Portes
Allel Chabli
Vassili Valayannopoulos
Jean-François Benoist
Jean-Marc Pinard
Gilles Simard
Olivier Douay
Kumaran Deiva
Alexandra Afenjar
Delphine Héron
François Rivier
Brigitte Chabrol
Fabienne Prieur
François Cartault
Gaëlle Pitelet
Alice Goldenberg
Soumeya Bekri
Marion Gerard
Richard Delorme
Marc Tardieu
Nicole Porchet
Christine Vianey-Saban
Joseph Vamecq
Publikationsdatum
01.12.2012
Verlag
BioMed Central
Erschienen in
Orphanet Journal of Rare Diseases / Ausgabe 1/2012
Elektronische ISSN: 1750-1172
DOI
https://doi.org/10.1186/1750-1172-7-96

Weitere Artikel der Ausgabe 1/2012

Orphanet Journal of Rare Diseases 1/2012 Zur Ausgabe