Skip to main content
Erschienen in: Pediatric Nephrology 5/2017

Open Access 11.06.2016 | Review

Renal thrombotic microangiopathy in patients with cblC defect: review of an under-recognized entity

verfasst von: Bodo B. Beck, FrancJan van Spronsen, Arjan Diepstra, Rolf M. F. Berger, Martin Kömhoff

Erschienen in: Pediatric Nephrology | Ausgabe 5/2017

Abstract

Methylmalonic aciduria and homocystinuria, cobalamin C (cblC) type, is the most common genetic type of functional cobalamin (vitamin B12) deficiency. This metabolic disease is characterized by marked heterogeneity of neurocognitive disease (microcephaly, seizures, developmental delay, ataxia, hypotonia) and variable extracentral nervous system involvement (failure to thrive, cardiovascular, renal, ocular) manifesting predominantly early in life, sometimes during gestation. To enhance awareness and understanding of renal disease associated with cblC defect, we studied biochemical, genetic, clinical, and histopathological data from 36 patients. Consistent clinical chemistry features of renal disease were intravascular hemolysis, hematuria, and proteinuria in all patients, with nephrotic-range proteinuria observed in three. Renal function ranged from normal to renal failure, with eight patients requiring (intermittent) dialysis. Two thirds were diagnosed with atypical (diarrhea-negative) hemolytic uremic syndrome (HUS). Renal histopathology analyses of biopsy samples from 16 patients revealed glomerular lesions typical of thrombotic microangiopathy (TMA). Treatment with hydroxycobalamin improved renal function in the majority, including three in whom dialysis could be withdrawn. Neurological sequelae were observed in 44 % and cardiopulmonary involvement in 39 % of patients, with half of the latter group demonstrating pulmonary hypertension. Mortality reached 100 % in untreated patients and 79 and 56 % in those with cardiopulmonary or neurological involvement, respectively. In all patients presenting with unclear intravascular hemolysis, hematuria, and proteinuria, cblC defect should be ruled out by determination of blood/plasma homocysteine levels and/or genetic testing, irrespective of actual renal function and neurological status, to ensure timely diagnosis and treatment.
Begleitmaterial
Hinweise

Electronic supplementary material

The online version of this article (doi:10.​1007/​s00467-016-3399-0) contains supplementary material, which is available to authorized users.

Introduction

Vitamin B12, also known as cobalamin (Cbl), is a complex water-soluble organic compound that is essential to all animals and numerous microorganisms. From an evolutionary perspective, vitamin B12 is an extremely old molecule, which has even been suggested to be synthesized prebiotically [1]. Synthesis is limited to several archaea and eubacteria and requires more than 25 steps [2]. The standard Western diet contains 5–7 µg of Cbl per day, which comes exclusively from dairy products and meat. The liver stores sufficient Cbl for several years, even after intake has completely ceased. The absorption, transport, storage, and intracellular processing of Cbl is complex, as is reflected by the high number of at least 21 genes that are known to affect Cbl absorption, storage, and metabolism in humans [3]. Vitamin B12 deficiency can thus result from numerous primary (genetic) and secondary conditions.
The most common disorder of intracellular vitamin B12 metabolism is methylmalonic aciduria and homocystinuria, cblC complementation type (MMACHC; phenotype MIM# 277400), which accounts for ~80 % of all cases. MMACHC has also been designated as cblC defect, cobalamin deficiency, C type, or cobalamin C deficiency. The last two terms could be confused with mere Cbl deficiency/absence (i.e., nutritional Cbl/B12 deficiency). To avoid erroneous exclusion of MMACHC based on the observation of normal or even elevated plasma B12 levels typically seen in affected individuals, the designations MMACHC or cblC defect is used throughout this review for the autosomal recessive metabolic disorder [4]. MMACHC was first described clinically in 1969 in a child with severe neurological compromise, hyperhomocysteinemia (−uria) with low methionine concentrations, and methylmalonic acidemia/aciduria, who died at 8 weeks of age [5]. Patients with MMACHC show downstream cytoplasmic blocking of Cbl conversion, resulting in combined deficiency of two important cofactors: adenosylcobalamin (AdoCbl) and methylcobalamin (MeCbl) [6]. MeCbl is the coenzyme for methionine synthase (MTR; defective in autosomal recessive cblG complementation type) required for the conversion of homocysteine to methionine in the cytoplasm. When this reaction is impaired, deranged folate metabolism causes—via defective DNA synthesis—the megaloblastic maturation pattern observed in these patients. AdoCbl is the cofactor of the mitochondrial enzyme methylmalonyl coenzyme A (CoA) mutase (itself defective in autosomal recessive methylmalonic acidemia, cblA, or cblB complementation type caused by mutations in either the MMAA or MMAB gene), which converts L-methylmalonyl-CoA to succinyl-CoA. In the absence of AdoCbl, methylmalonic acid is generated from excess of L-methylmalonyl-CoA (Fig. 1).
This explains the biochemical hallmarks of MMACHC (Table 1): hyperhomocysteinemia/hyperhomocystinuria with low methioninemia, and methylmalonic acidemia/aciduria (which is also the biochemical phenotype seen in the rarer complementation types cblD, cblF, cblJ, and cblX variants [3]).
Table 1
Key laboratory parameters in cobalamin C (cblC) defect
Test
Normal range
Patients
Plasma tHCY (μm)
5–15
23–186
Plasma MMA (μm)
<0.27
1.6–14.5
Plasma B12 levels (pg/ml)
170–800
611–932
tHCY total homocysteine, MMA methylmalonic acid
The clinical findings are summarized in Table 2 and are described in detail in a recent review [4].
Table 2
Clinical synopsis of myriad methylmalonic aciduria and homocystinuria, cobalamin C (cblC) complementation type (MMACHC) manifestations (modified from [8], used with permission)
System
Clinical manifestations
Growth and habitus
Prenatal growth retardation
Postnatal failure to thrive
Microcephaly
Hydrops fetalis
Hydrocephalus
Marfanoid habitus
Dysmorphic facial features
Central nervous system
Developmental delay
Seizures
Ataxia
Hypotonia
Lethargy, progressive encephalopathy
Regression, dementia
Cognitive impairment ranging from executive dysfunction to severe mental retardation
Neuropsychiatric disturbances
Subdural hematoma
Demyelinating neuropathy
Leukoencephalopathy
Basal ganglia lesions (less frequent)
Eye
Maculopathy
Retinal degeneration
Optic atrophy
Nystagmus
Blood
Anemia, thrombocytopenia and/or neutropenia, megaloblastosis
Vascular
Recurrent venous thrombosis
Cor pulmonale or subclinical pulmonary thrombosis
Cerebrovascular complications, stroke
Renal
Hemolytic-uremic syndrome
Chronic thrombotic microangiopathy
Nephrotic syndrome
Renal failure
Heart
Fetal dilated cardiomyopathy
Congenital heart defects
Pulmonary arterial hypertension
Left ventricular noncompaction
The gene MMACHC (MIM# 609831) was identified in 2006 [9] and is located on chromosome 1p34 and contains only four coding exons. The complementary DNA (cDNA) has an 846-bp open reading frame encoding a protein of 282 amino acids with a predicted molecular weight of 31.7 kD. Contrary to the N-terminal domain of the protein (MMACHC), which lacks homology to any known protein, homology exists between the C-terminal domain and the bacterial protein TonB [9], a Cbl-binding protein [10].
The MMACHC protein (Uniprot KB entry Q9Y4U1) possesses decyanase, dealkylase, and flavin reductase activities. Decyanation of cyanocobalamin is essential for its subsequent conversion into the active cofactors adenosylcobalamin and methylcobalamin (Fig. 1) [11]. This explains why MMACHC patients do not respond to therapy with orally administered cyanocobalamin and should be treated with hydroxycobalamin i.m., which can be metabolized into adenosylcobalamin and methylcobalamin. MMACHC knockout mice die from pre-implantation defect [12]. During midgestation in murine development, MMACHC is expressed in many tissues, including the mesonephric mesenchyme of the kidney and the endothelium of blood vessels and heart [13, 14]. MMACHC represents the prototypic and most common disorder of intracellular Cbl metabolism; currently, 81 different mutations [Human Gene Mutation Database (HGMD) professional 2015.4), and well over 500 patients have been reported worldwide [9, 1517]. The frameshift mutation c.271dupA (p.R91KfsX14) accounts for about 40 % of disease-causing alleles in the Caucasian population [15], while c.609G>A (p.R161X) has been identified as the most frequent mutation in a study from China [16]. Early-onset disease, defined as manifestation within the first year of life, has been associated with the truncating alleles c.271dupA or c.331C>T (p.R111X) [15]. Two recent studies found incidence rates ranging from 1:100,000 births in the New York, USA, region [13] to 1: 60,000 in California, USA and up to 1:37,000 in the Hispanic population. French Canadian, Cajun, Indian, Chinese, Middle Eastern, and European populations are believed to be at an increased risk for cblC type [8].
Although MMACHC constitutes a multisystemic disease that can affect almost any organ system, including the kidney (see overview in Table 2), cblC defect is largely regarded as a disease affecting central nervous system (CNS) development and neurocognition [18]. In 1979, Baumgartner and her colleagues described the first patient with renal disease and MMACHC defect [19]. The boy from unrelated Italian parents presented in the neonatal phase with failure to thrive and anemia. At 3 months of age, he was diagnosed with elevated methylmalonic acid and homocysteine concentrations in blood and urine. Treatment with cyanocobalamin was unsuccessful. Severe metabolic acidosis and uremia were noted. He developed progressive arterial hypertension, hepatomegaly, and tachypnea. On chest X-ray, cardiomegaly and pulmonary edema were noted. Digoxin and furosemide led to some improvement, but at the age of 4 months, he had a second episode of heart failure and died a few hours later from intractable pulmonary edema. From the first report until to date, at least 36 patients with cblC defect and thrombotic microangiopathy (TMA) have been identified worldwide. The purpose of this review is to increase awareness of the renal manifestation and outcome in MMACHC.
MMACHC as a treatable form of TMA has received comparatively little attention in the field, as is reflected by its omission in several reviews on TMAs. This is very unfortunate, as the mortality rate of patients with MMACHC is high, while initial screening for elevated homocysteine plasma/blood levels is widely available, fast, and inexpensive (~US $30). Unlike in other rare kidney diseases, a causal therapeutic option, hydroxycobalamin, is readily available. Treatment with hydroxycobalamin is effective in many patients when started early, it has minimal side effects, and it is available at negligible costs (US $1/day).

Methods

We reviewed clinical data from patients referred to our institutions as well as from published case reports and case series of renal disease in patients with MMACHC. The supplemental table lists biochemical data and provides clinical information of patients with MMACHC for whom individual clinical details of renal disease were available. PubMed search terms included cblC, cobalamin, MMACHC, renal, and kidney.

Results

We identified 36 patients ([14, 2138], including one unpublished patient) with established cblC defect and renal involvement. Diagnosis of MMACHC was confirmed biochemically in 34 patients and genetically in 18 (Table 3; for individual data, see Table S1). The most frequent MMACHC mutations in our cohort were c.271dupA (36 % allelic frequency), c.276G>T (17 % allelic frequency), and c.565C>A (11 % allelic frequency, Fig. 2). Most MMACHC cases were sporadic, while familial disease was noted in three sets of siblings (patient numbers 18 and, 24, 26 and 30, 27 and 28 in Table S1). Infantile onset (<12 months of age) was observed in 16 patients, and 20 patients presented with TMA beyond infancy (Table 4).
Table 3
Disease onset and diagnosis of cobalamin C (cblC) and complement data: findings in 36 patients with cblC defect and thrombotic microangiopathy
 Variables
 Findings
cblC defect
 Biochemical
94 % (n = 34/36)
 Genetic
50 % (n = 18/36)
Mutations (allelic frequency)
 c.271dupA
36 % (n = 13/36)
 c.276G>T
17 % (n = 6/36)
 c.565C>A
11 % (n = 4/36)
Complement system
Normal
87 % (n = 13/15)
 CFH-autoantibodies
6.6 % (n = 1/15)
CFH mutation
6.6 % (n = 1/15)
Table 4
Methylmalonic aciduria and homocystinuria, cobalamin C (cblC) complementation type (MMACHC): general, renal, and extrarenal characteristics
Manifestation data
Onset of disease
 Infantile onset
44 %, (n = 16/36)
 Noninfantile onset
56 %, (n = 20/36)
Characteristics of renal disease
 Proteinuria and hematuria
100 %, (n = 27/27)
  Hemolytic uremic syndrome
66 %, (n = 24/36)
 Normotensive
10 %, (n = 2/20)
 Hypertensive
90 %, (n = 18/20)
 Nephrotic syndrome
11 %, (n = 3/27)
 Chronic kidney disease (CKD) 1
11 %, (n = 4)
 CKD 2–5
89 %, (n =32)
 Dialysis
22 %, ( n = 8)
Kidney transplant
6 %, (n = 2)
Extrarenal manifestations
 Neurological impairment
44 %, (n = 16/36)
 Cardiopulmonary involvement
39 %, (n = 14/36)
 Pulmonary hypertension
17 %, (n = 6/36)
All patients were clinically diagnosed with TMA characterized by (microvascular thrombosis with) thrombocytopenia, hemolytic anemia, and red blood cell fragmentation [39]. Twenty-four of the 36 patients met (diarrhea-negative) hemolytic uremic syndrome (HUS) criteria defined by the classic triad of hemolytic anemia, uremia, and thrombopenia [40]. Renal biopsies or autopsies were performed in 16 patients and revealed TMA in all of them (in two patients who were initially classified as membranoproliferative glomerulonephritis, biopsy reanalysis was consistent with TMA [31]). In general, a uniform histomorphological pattern was noted (Fig. 3, Table 5): glomerular thrombi and swelling were described in 13 cases. Duplication of the glomerular basement membrane was noted in all but one patient and intra-arterial thrombi was seen in nine.
Table 5
Renal histology
Patient no.
1
2
9
10
16
18
20
22
24
26
27
28
25
30
31
34
 Ischemia
          
y
     
 Intra-arterial thrombi
y
n
n
y
n
y
y
y
y
 
y
y
   
y
 Glomerular thrombi
y
  
y
y
y
y
y
 
y
y
y
y
y
y
y
Endothelial
 Swelling
 
y
n
 
y
  
y
y
y
y
y
y
y
y
y
 Detachment
 
y
n
 
y
           
 Duplication of GBM
 
y
n
 
y
y
y
y
y
y
y
y
y
y
y
y
 Glomerular sclerosis
 
n
n
 
n
n
n
n
n
n
n
n
y
n
n
n
y yes, n no, GBM glomerular basement membrane
Complement defects were detected in two of 15 patients screened for potential atypical HUS (aHUS): patient 20 had antifactor H autoantibodies and died from pulmonary hypertension despite treatment with hydroxycobalamin. Patient 27 had a mutation in CFH, underwent plasmapheresis and renal replacement therapy (RRT), and responded well to treatment with hydroxycobalamin (see Tables 6 and S1). Of the 32 patients with reduced renal function, eight required dialysis and two received a kidney graft. In four patients, glomerular filtration rate was within normal limits. Hematuria and proteinuria were present in all tested patients (n = 27, Table 4); three of them presented with frank nephrotic syndrome [22, 30]. Arterial hypertension was reported in 18 patients. Thirty-one patients were treated with hydroxycobalamin; 17 of them improved clinically, two stabilized, and 12 died (Table 6). In 81 % of the survivors, renal function improved. In children and adolescents, glomerular filtration rate (GFR) increased from 49.5 to 89 ml/min/1.73 m2. Two patients were treated with cyanocobalamin (which is ineffective to overcome the metabolic block in MMACHC patients): one of them died, and no improvement of renal function was observed in the other. Three patients did not receive metabolic treatment and died.
Table 6
Response to metabolic and complement-targeted therapy
Response to metabolic therapy
 Clinical recovery
54 %, n = 17
 Improvement of GFR
81 %, n = 13
 Stable GFR
19 %, n = 3
 Pretreatment GFR (ml/min/1.73 m2)
49.5 n = 10
 Posttreatment GFR (ml/min/1.73 m2)
89, n = 10
Mortality
 Overall mortality
44 % (n = 16/36)
 Infantile onset
56 %, (n = 9/16)
 Noninfantile onset
35 %, (n = 7/20)
 With neurological disease
50 %, (n = 8/16)
 With cardiopulmonary disease
79 %, (n = 11/14)
 Untreated
100 %, (n = 4/4)
Complement-targeted therapy (response)
 Plasma exchange (patients 22, 24, 27, 33)
1* out of 4
 Eculizumab (patients 33, 34)
No response
GFR glomerular filtration rate
Five patients were treated with complement-targeted therapy (Table 6), of whom, one (patient 27 with a 3254T3 C mutation in factor H) responded. In the four nonresponders, no complement defect was detected. Mortality was higher in infants (57 %) compared with later-onset disease (35 %), resulting in an overall mortality rate of 44 %. Extrarenal involvement included neurological and cardiopulmonary disease in 44 and 39 % of patients, respectively (Table 4). It is noteworthy that among the 16 nonsurvivors, 11 patients had cardiopulmonary involvement. In seven patients, pulmonary arterial hypertension was diagnosed by right ventricle catheterization and/or cardiac ultrasound; it was fatal in four.

Discussion

Clinical, histopathological, and genetic characteristics of renal disease in cblC defect

The key findings in the 36 patients with MMACHC and renal disease are as follows:
(1) In general, the first manifestation of renal disease occurs early in infancy, although onset can vary widely among affected individuals (median onset is 1.5 months, range 12 days to 40 years).
(2) Overall mortality is high (44 %).
(3) Mortality is primarily the result of cardiopulmonary problems (68 %), with pulmonary hypertension identified as a specific cause in seven patients (44 %).
(4) Renal impairment (as defined by reduced glomerular filtration) is variable and ranges from insignificant to initiation of RRT, which is necessary in 22 % of patients.
(5) Consistent features of renal disease in all patients are intravascular hemolysis, hematuria, and proteinuria.
(6) Consistently, pathological changes predominantly show glomerular thrombotic microangiopathies [41], including glomerular fibrin thrombi, endothelial swelling, and duplication of the glomerular basement membrane.
(7) The majority of patients respond to therapy with hydxroycobalamin.
(8) As could be expected from its high allelic frequency, the frameshift mutation c.271dupA represented the most frequent allele in our cohort, followed by the splice-site mutation c.276G>T and the missense mutation c.565C>A. As all patients with a c.276G>T allele originated from the Dutch “Bible belt” or adjacent regions in Germany, a founder effect is highly likely here.

Pathophysiologic considerations

Given the fact that all patients demonstrated microangiopathic hemolytic anemia [elevated lactose dehydrogenase (LDH), reduced haptoglobin levels, and/or fragmentocytes), the concept of endothelial cell damage appears to be a central pathogenic factor. This notion is clearly supported by the histological evidence of endothelial damage, i.e., endothelial swelling, detachment, and duplication of the glomerular basement membrane. The fact that the latter was observed in all but one case probably reflects the chronicity of the lesions.
The etiology of endothelial damage remains largely unknown. Homocysteine, unlike methylmalonic acid, is known to damage endothelial cells [42]. Half of the patients in this review had intermediate levels of homocysteinemia (30–100 μmol/l) at presentation; the others showed severe hyperhomocysteinemia (homocysteine levels > 100 μmol/l). Interestingly, isolated (i.e. without methylmalonic acidemia) intermediate or even severe hyperhomocysteinemia—as observed in methylenetetrahydrofolate reductase (MTHFR) deficiency (OMIM 607093) or in homocystinuria due to cystathionine beta-synthase (CBS) deficiency (OMIM 236200)—have not been reported to cause any specific renal (e.g., HUS) or cardiopulmonary (e.g., pulmonary hypertension) disease apart from thromboembolism. Possibly, additional biochemical abnormalities such as methylmalonic acidemia and/or methionine deficiency—as in cblC defect (this study) or in methylenetetrahydrofolate dehydrogenase 1 (MTHFD1) deficiency (OMIM 172460, [43, 44])—are required to develop TMA. The notion that it takes two or more toxic metabolites to trouble endothelial cells is supported by the occasional occurrence of pseudo-TMA in nutritional B12 deficiency [45], which—like MMACHC and MTHFD1 deficiency—is characterized by combined hyperhomocysteinemia and methylmalonic acidemia and low methionine levels. This would also hold true for such findings in some other Cbl defects, such as cblD, cblF, cblJ, and cblX variants, which are much more rare [3].
The fact that only a subgroup (minimum 36 of >500 reported patients) of patients with MMACHC are affected by TMA requires further investigation. Apart from the reduced penetrance seen in many mendelian disorders, other factors in addition to homocysteine levels could contribute to the manifestation of TMA. This fits with the observation of discordant families in a recent study reporting asymptomatic MMACHC cases (prior to treatment with hydroxylcobalamin) [46] and one from our hospital [clinically unaffected sister of patient 29 (homozygous for c.276G>T)] with comparable homocysteine levels (Kömhoff, and Berger, unpublished data). That TMA may be underreported because its features are too subtle in the presence of a predominant neurocognitive or cardiopulmonary disease or because the patient dies before renal disease develops cannot be excluded.
Comparison of the the study reported here with cohorts of children with suspected or proven aHUS/complement defects [47, 48] allows a number of interesting considerations: median age of presentation in MMACHC with renal disease (1.5 months) was much earlier than in the French ( 1.5 years, [47]) or Dutch (80 % older than 1 year of age at presentation, [48]) aHUS cohorts. Renal impairment, however, was much more pronounced in both the Dutch and French aHUS cohorts, with 44 % [48] and 59 % [47], respectively, of patients requiring RRT at presentation, compared with 22 % in MMACHC. Early presentation with relatively mild renal impairment probably reflects that MMACHC is a severe systemic disease resulting in early referral for multiple reasons when renal disease has not yet advanced. In contrast to the comparatively low mortality of 2 % [48] and 8 % [47] in the Dutch and French aHUS cohorts, respectively, overall mortality with cblC defect is high (44 %). In 11 of the 16 nonsurvivors, a cardiopulmonary problem was noted, with pulmonary arterial hypertension diagnosed in seven. The concept of cardiorenal syndrome with pulmonary hypertension and TMA in MMACHC deficiency is supported by hemolytic anemia in an infant with cblC defect who died from cor pulmonale [49] and by increased creatinine and LDH levels in another infant who died from pulmonary hypertension and MMACHC [50]. The increased risk for pulmonary hypertension should thus prompt a thorough evaluation of MMACHC patients by a pediatric cardiologist with expertise in pulmonary hypertension.

Diagnosis

MMACHC should be considered in all children, adolescents, and young adults with unclear intravascular hemolysis, hematuria, and proteinuria, irrespective of renal and neurological impairment. Homocysteine blood concentrations > 20 μmol/l with normal renal function and >30 with renal failure require further biochemical and genetic workup [4]. It is important to realize that total Cbl levels are unrealiable indicators of a functional Cbl deficiency [51].

Therapy

The general principles of metabolic therapy have recently been described in detail [4]. Whenever MMACHC is suspected, we consider it mandatory to promptly initiate treatment with parenteral administration of hydroxycobalamin without waiting for confirmation by genetic testing. A practical guideline is given in Table 3. It should be kept in mind, however, that some patients are difficult to treat, even with intense therapy [52]. In the event of a patient with TMA and pulmonary hypertension in whom MMACHC and a complement defect coincide (Table S1, patient 20), we strongly advocate the use of a C5 inhibitor [53] instead of plasma exchange to inhibit complement activation, as the latter carries an extremely high risk for transfusion-related acute lung injury (TRALI) [54]. When general anesthesia is required, it should be taken into account that nitrous oxide is potentially toxic to patients with MMACHC disease and needs to be avoided, because it depletes the body stores of B12 and inhibits methionine synthase activity [55, 56] Table 7.
Table 7
Metabolic therapy in patients with cobalamin C (cblC) defect (modified from [4], used with permission)
Medication
Recommended dose
Frequency
Efficacy
Target biochemical parameter
Hydroxycobalamin: i.m., i.v. or intranasally [20]
0.3 mg/kg/day
Start once daily
Established
Reducing serum MMA and tHCY levels
Normalizing plasma MET and hematological parameters
Betaine oral
250 mg/kg/day
tid
Established
Reducing HCY, increasing MET
Folinic acid oral
5–15 mg/day
tid
Theoretical
Reducing HCY, increasing MET
Levocarnitine oral
50–100 mg/kg/day
tid
Theoretical
Unnecessary in case of normal MET
HCY homocysteine, MET methionine, MMA methylmalonic acid, tid three times a day
This retrospective analysis has significant limitations mainly resulting from (a) the long period (>35 years) over which patients were reported and during which significant advances have been made in the field of diagnostics and therapeutics; and (b) the heterogeneity of centers using different diagnostic and therapeutic approaches. We think, however, that the consistent features of MMACHC compensate for the heterogeneity and thereby allow us to draw the following valid conclusions:
Renal TMA due to CblC defect is a devastating disease with high mortality in which mild renal impairment may distract the nephrologist from detecting severe cardiopulmonary involvement. However, when recognized and treated at an early stage, complete clinical recovery is possible. Diagnosis is simple, and therapy is cheap and successful in the majority of patients. We therefore strongly recommend [57] sceening for MMACHC in all patients with intravascular hemolysis in combination with hematuria and proteinuria.

Compliance with ethical standards

Conflict of interest

The authors declare no conflict interest.
Open Access This article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made.

Unsere Produktempfehlungen

e.Med Interdisziplinär

Kombi-Abonnement

Für Ihren Erfolg in Klinik und Praxis - Die beste Hilfe in Ihrem Arbeitsalltag

Mit e.Med Interdisziplinär erhalten Sie Zugang zu allen CME-Fortbildungen und Fachzeitschriften auf SpringerMedizin.de.

e.Med Pädiatrie

Kombi-Abonnement

Mit e.Med Pädiatrie erhalten Sie Zugang zu CME-Fortbildungen des Fachgebietes Pädiatrie, den Premium-Inhalten der pädiatrischen Fachzeitschriften, inklusive einer gedruckten Pädiatrie-Zeitschrift Ihrer Wahl.

e.Med Urologie

Kombi-Abonnement

Mit e.Med Urologie erhalten Sie Zugang zu den urologischen CME-Fortbildungen und Premium-Inhalten der urologischen Fachzeitschriften.

Anhänge

Electronic supplementary material

Below is the link to the electronic supplementary material.
Literatur
1.
Zurück zum Zitat Eschenmoser A (2011) Etiology of potentially primordial biomolecular structures: from vitamin B12 to the nucleic acids and an inquiry into the chemistry of life’s origin: a retrospective. Angew Chem Int Ed Engl 50:12412–12472CrossRefPubMed Eschenmoser A (2011) Etiology of potentially primordial biomolecular structures: from vitamin B12 to the nucleic acids and an inquiry into the chemistry of life’s origin: a retrospective. Angew Chem Int Ed Engl 50:12412–12472CrossRefPubMed
2.
Zurück zum Zitat Roth JR, Lawrence JG, Bobik TA (1996) Cobalamin (coenzyme B12): synthesis and biological significance. Annu Rev Microbiol 50:137–181CrossRefPubMed Roth JR, Lawrence JG, Bobik TA (1996) Cobalamin (coenzyme B12): synthesis and biological significance. Annu Rev Microbiol 50:137–181CrossRefPubMed
3.
Zurück zum Zitat Froese DS, Gravel RA (2010) Genetic disorders of vitamin B(1)(2) metabolism: eight complementation groups—eight genes. Expert Rev Mol Med 12:e37CrossRefPubMedPubMedCentral Froese DS, Gravel RA (2010) Genetic disorders of vitamin B(1)(2) metabolism: eight complementation groups—eight genes. Expert Rev Mol Med 12:e37CrossRefPubMedPubMedCentral
4.
Zurück zum Zitat Carrillo-Carrasco N, Chandler RJ, Venditti CP (2012) Combined methylmalonic acidemia and homocystinuria, cblC type I. Clinical presentations, diagnosis and management. J Inherit Metab Dis 35:91–102CrossRefPubMed Carrillo-Carrasco N, Chandler RJ, Venditti CP (2012) Combined methylmalonic acidemia and homocystinuria, cblC type I. Clinical presentations, diagnosis and management. J Inherit Metab Dis 35:91–102CrossRefPubMed
5.
Zurück zum Zitat Mudd SH, Levy HL, Abeles RH, Jennedy JP Jr (1969) A derangement in B 12 metabolism leading to homocystinemia, cystathioninemia and methylmalonic aciduria. Biochem Biophys Res Commun 35:121–126CrossRefPubMed Mudd SH, Levy HL, Abeles RH, Jennedy JP Jr (1969) A derangement in B 12 metabolism leading to homocystinemia, cystathioninemia and methylmalonic aciduria. Biochem Biophys Res Commun 35:121–126CrossRefPubMed
6.
Zurück zum Zitat Gravel RA, Mahoney MJ, Ruddle FH, Rosenberg LE (1975) Genetic complementation in heterokaryons of human fibroblasts defective in cobalamin metabolism. Proc Natl Acad Sci U S A 72:3181–3185CrossRefPubMedPubMedCentral Gravel RA, Mahoney MJ, Ruddle FH, Rosenberg LE (1975) Genetic complementation in heterokaryons of human fibroblasts defective in cobalamin metabolism. Proc Natl Acad Sci U S A 72:3181–3185CrossRefPubMedPubMedCentral
7.
Zurück zum Zitat Harding CO, Pillers DA, Steiner RD, Bottiglieri T, Rosenblatt DS, Debley J, Michael Gibson K (2003) Potential for misdiagnosis due to lack of metabolic derangement in combined methylmalonic aciduria/hyperhomocysteinemia (cblC) in the neonate. J Perinatol 23:384–386CrossRefPubMed Harding CO, Pillers DA, Steiner RD, Bottiglieri T, Rosenblatt DS, Debley J, Michael Gibson K (2003) Potential for misdiagnosis due to lack of metabolic derangement in combined methylmalonic aciduria/hyperhomocysteinemia (cblC) in the neonate. J Perinatol 23:384–386CrossRefPubMed
8.
Zurück zum Zitat Carrillo-Carrasco N, Venditti CP (2012) Combined methylmalonic acidemia and homocystinuria, cblC type. II. Complications, pathophysiology, and outcomes. J Inherit Metab Dis 35:103–114CrossRefPubMed Carrillo-Carrasco N, Venditti CP (2012) Combined methylmalonic acidemia and homocystinuria, cblC type. II. Complications, pathophysiology, and outcomes. J Inherit Metab Dis 35:103–114CrossRefPubMed
9.
Zurück zum Zitat Lerner-Ellis JP, Tirone JC, Pawelek PD, Doré C, Atkinson JL, Watkins D, Morel CF, Fujiwara TM, Moras E, Hosack AR, Dunbar GV, Antonicka H, Forgetta V, Dobson CM, Leclerc D, Gravel RA, Shoubridge EA, Coulton JW, Lepage P, Rommens JM, Morgan K, Rosenblatt DS (2006) Identification of the gene responsible for methylmalonic aciduria and homocystinuria, cblC type. Nat Genet 38:93–100CrossRefPubMed Lerner-Ellis JP, Tirone JC, Pawelek PD, Doré C, Atkinson JL, Watkins D, Morel CF, Fujiwara TM, Moras E, Hosack AR, Dunbar GV, Antonicka H, Forgetta V, Dobson CM, Leclerc D, Gravel RA, Shoubridge EA, Coulton JW, Lepage P, Rommens JM, Morgan K, Rosenblatt DS (2006) Identification of the gene responsible for methylmalonic aciduria and homocystinuria, cblC type. Nat Genet 38:93–100CrossRefPubMed
10.
Zurück zum Zitat Banerjee R (2006) B12 trafficking in mammals: a for coenzyme escort service. ACS Chem Biol 1:149–159CrossRefPubMed Banerjee R (2006) B12 trafficking in mammals: a for coenzyme escort service. ACS Chem Biol 1:149–159CrossRefPubMed
11.
12.
Zurück zum Zitat Moreno-Garcia MA, Pupavac M, Rosenblatt DS, Tremblay ML, Jerome-Majewska LA (2014) The Mmachc gene is required for pre-implantation embryogenesis in the mouse. Mol Genet Metab 112:198–204CrossRefPubMed Moreno-Garcia MA, Pupavac M, Rosenblatt DS, Tremblay ML, Jerome-Majewska LA (2014) The Mmachc gene is required for pre-implantation embryogenesis in the mouse. Mol Genet Metab 112:198–204CrossRefPubMed
13.
Zurück zum Zitat Weisfeld-Adams JD, Morrissey MA, Kirmse BM, Salveson BR, Wasserstein MP, McGuire PJ, Sunny S, Cohen-Pfeffer JL, Yu C, Caggana M, Diaz GA (2010) Newborn screening and early biochemical follow-up in combined methylmalonic aciduria and homocystinuria, cblC type, and utility of methionine as a secondary screening analyte. Mol Genet Metab 99:116–123CrossRefPubMed Weisfeld-Adams JD, Morrissey MA, Kirmse BM, Salveson BR, Wasserstein MP, McGuire PJ, Sunny S, Cohen-Pfeffer JL, Yu C, Caggana M, Diaz GA (2010) Newborn screening and early biochemical follow-up in combined methylmalonic aciduria and homocystinuria, cblC type, and utility of methionine as a secondary screening analyte. Mol Genet Metab 99:116–123CrossRefPubMed
14.
Zurück zum Zitat Sonsino E, Ogier H, Mercier JC, Devictor D, Ferkdadji L, Bocquet L (1985) Congenital anomaly of the metabolism of vitamin B 12. Histopathological study. Arch Anat Cytol Pathol 33:98–99PubMed Sonsino E, Ogier H, Mercier JC, Devictor D, Ferkdadji L, Bocquet L (1985) Congenital anomaly of the metabolism of vitamin B 12. Histopathological study. Arch Anat Cytol Pathol 33:98–99PubMed
15.
Zurück zum Zitat Lerner-Ellis JP, Anastasio N, Liu J, Coelho D, Suormala T, Stucki M, Loewy AD, Gurd S, Grundberg E, Morel CF, Watkins D, Baumgartner MR, Pastinen T, Rosenblatt DS, Fowler B (2009) Spectrum of mutations in MMACHC, allelic expression, and evidence for genotype–phenotype correlations. Hum Mutat 30:1072–1081CrossRefPubMed Lerner-Ellis JP, Anastasio N, Liu J, Coelho D, Suormala T, Stucki M, Loewy AD, Gurd S, Grundberg E, Morel CF, Watkins D, Baumgartner MR, Pastinen T, Rosenblatt DS, Fowler B (2009) Spectrum of mutations in MMACHC, allelic expression, and evidence for genotype–phenotype correlations. Hum Mutat 30:1072–1081CrossRefPubMed
16.
Zurück zum Zitat Liu MY, Yang YL, Chang YC, Chiang SH, Lin SP, Han LS, Qi Y, Hsiao KJ, Liu TT (2010) Mutation spectrum of MMACHC in Chinese patients with combined methylmalonic aciduria and homocystinuria. J Hum Genet 55:621–626CrossRefPubMed Liu MY, Yang YL, Chang YC, Chiang SH, Lin SP, Han LS, Qi Y, Hsiao KJ, Liu TT (2010) Mutation spectrum of MMACHC in Chinese patients with combined methylmalonic aciduria and homocystinuria. J Hum Genet 55:621–626CrossRefPubMed
17.
Zurück zum Zitat Watkins D, Rosenblatt DS (2011) Inborn errors of cobalamin absorption and metabolism. Am J Med Genet C: Semin Med Genet 157C:33–44CrossRef Watkins D, Rosenblatt DS (2011) Inborn errors of cobalamin absorption and metabolism. Am J Med Genet C: Semin Med Genet 157C:33–44CrossRef
18.
Zurück zum Zitat Fischer S, Huemer M, Baumgartner M, Deodato F, Ballhausen D, Boneh A, Burlina AB, Cerone R, Garcia P, Gökçay G, Grünewald S, Häberle J, Jaeken J, Ketteridge D, Lindner M, Mandel H, Martinelli D, Martins EG, Schwab KO, Gruenert SC, Schwahn BC, Sztriha L, Tomaske M, Trefz F, Vilarinho L, Rosenblatt DS, Fowler B, Dionisi-Vici C (2014) Clinical presentation and outcome in a series of 88 patients with the cblC defect. J Inherit Metab Dis 37:831–840CrossRefPubMed Fischer S, Huemer M, Baumgartner M, Deodato F, Ballhausen D, Boneh A, Burlina AB, Cerone R, Garcia P, Gökçay G, Grünewald S, Häberle J, Jaeken J, Ketteridge D, Lindner M, Mandel H, Martinelli D, Martins EG, Schwab KO, Gruenert SC, Schwahn BC, Sztriha L, Tomaske M, Trefz F, Vilarinho L, Rosenblatt DS, Fowler B, Dionisi-Vici C (2014) Clinical presentation and outcome in a series of 88 patients with the cblC defect. J Inherit Metab Dis 37:831–840CrossRefPubMed
19.
Zurück zum Zitat Baumgartner ER, Wick H, Maurer R, Egli N, Steinmann B (1979) Congenital defect in intracellular cobalamin metabolism resulting in homocysteinuria and methylmalonic aciduria. I. Case report and histopathology. Helv Paediatr Acta 34:465–482PubMed Baumgartner ER, Wick H, Maurer R, Egli N, Steinmann B (1979) Congenital defect in intracellular cobalamin metabolism resulting in homocysteinuria and methylmalonic aciduria. I. Case report and histopathology. Helv Paediatr Acta 34:465–482PubMed
20.
Zurück zum Zitat Slot WB, Merkus FW, Van Deventer SJ, Tytgat GN (1997) Normalization of plasma vitamin B12 concentration by intranasal hydroxocobalamin in vitamin B12-deficient patients. Gastroenterology 113:430–433CrossRefPubMed Slot WB, Merkus FW, Van Deventer SJ, Tytgat GN (1997) Normalization of plasma vitamin B12 concentration by intranasal hydroxocobalamin in vitamin B12-deficient patients. Gastroenterology 113:430–433CrossRefPubMed
21.
Zurück zum Zitat Li QL, Song WQ, Peng XX, Liu XR, He LJ, Fu LB (2015) Clinical characteristics of hemolytic uremic syndrome secondary to cobalamin C disorder in Chinese children. World J Pediatr 11:276–280CrossRefPubMed Li QL, Song WQ, Peng XX, Liu XR, He LJ, Fu LB (2015) Clinical characteristics of hemolytic uremic syndrome secondary to cobalamin C disorder in Chinese children. World J Pediatr 11:276–280CrossRefPubMed
22.
Zurück zum Zitat Koenig JC, Rutsch F, Bockmeyer C, Baumgartner M, Beck BB, Kranz B, Konrad M (2015) Nephrotic syndrome and thrombotic microangiopathy caused by cobalamin C deficiency. Pediatr Nephrol 30:1203–1206CrossRefPubMed Koenig JC, Rutsch F, Bockmeyer C, Baumgartner M, Beck BB, Kranz B, Konrad M (2015) Nephrotic syndrome and thrombotic microangiopathy caused by cobalamin C deficiency. Pediatr Nephrol 30:1203–1206CrossRefPubMed
23.
Zurück zum Zitat Carmel R, Bedros AA, Mace JW, Goodman SI (1980) Congenital methylmalonic aciduria—homocystinuria with megaloblastic anemia: observations on response to hydroxocobalamin and on the effect of homocysteine and methionine on the deoxyuridine suppression test. Blood 55:570–579PubMed Carmel R, Bedros AA, Mace JW, Goodman SI (1980) Congenital methylmalonic aciduria—homocystinuria with megaloblastic anemia: observations on response to hydroxocobalamin and on the effect of homocysteine and methionine on the deoxyuridine suppression test. Blood 55:570–579PubMed
24.
Zurück zum Zitat Jiménez Varo I, Bueno Delgado M, Dios Fuentes E, Delgado Pecellin C, González Meneses A, Soto Moreno A, Venegas Moreno E (2015) Combined methylmalonic acidemia and homocystinuria; a case report. Nutr Hosp 31:1885–1888PubMed Jiménez Varo I, Bueno Delgado M, Dios Fuentes E, Delgado Pecellin C, González Meneses A, Soto Moreno A, Venegas Moreno E (2015) Combined methylmalonic acidemia and homocystinuria; a case report. Nutr Hosp 31:1885–1888PubMed
25.
Zurück zum Zitat Cornec-Le Gall E, Delmas Y, De Parscau L, Doucet L, Ogier H, Benoist JF, Fremeaux-Bacchi V, Le Meur Y (2014) Adult-onset eculizumab-resistant hemolytic uremic syndrome associated with cobalamin C deficiency. Am J Kidney Dis 63:119–123CrossRefPubMed Cornec-Le Gall E, Delmas Y, De Parscau L, Doucet L, Ogier H, Benoist JF, Fremeaux-Bacchi V, Le Meur Y (2014) Adult-onset eculizumab-resistant hemolytic uremic syndrome associated with cobalamin C deficiency. Am J Kidney Dis 63:119–123CrossRefPubMed
26.
Zurück zum Zitat Kömhoff M, Roofthooft MT, Westra D, Teertstra TK, Losito A, van de Kar NC, Berger RM (2013) Combined pulmonary hypertension and renal thrombotic microangiopathy in cobalamin C deficiency. Pediatrics 132:e540–e544CrossRefPubMed Kömhoff M, Roofthooft MT, Westra D, Teertstra TK, Losito A, van de Kar NC, Berger RM (2013) Combined pulmonary hypertension and renal thrombotic microangiopathy in cobalamin C deficiency. Pediatrics 132:e540–e544CrossRefPubMed
27.
Zurück zum Zitat Menni F, Testa S, Guez S, Chiarelli G, Alberti L, Esposito S (2012) Neonatal atypical hemolytic uremic syndrome due to methylmalonic aciduria and homocystinuria. Pediatr Nephrol 27:1401–1405CrossRefPubMed Menni F, Testa S, Guez S, Chiarelli G, Alberti L, Esposito S (2012) Neonatal atypical hemolytic uremic syndrome due to methylmalonic aciduria and homocystinuria. Pediatr Nephrol 27:1401–1405CrossRefPubMed
28.
Zurück zum Zitat Bouts AH, Roofthooft MT, Salomons GS, Davin JC (2010) CD46-associated atypical hemolytic uremic syndrome with uncommon course caused by cblC deficiency. Pediatr Nephrol 25:2547–2548CrossRefPubMedPubMedCentral Bouts AH, Roofthooft MT, Salomons GS, Davin JC (2010) CD46-associated atypical hemolytic uremic syndrome with uncommon course caused by cblC deficiency. Pediatr Nephrol 25:2547–2548CrossRefPubMedPubMedCentral
29.
Zurück zum Zitat Sharma AP, Greenberg CR, Prasad AN, Prasad C (2007) Hemolytic uremic syndrome (HUS) secondary to cobalamin C (cblC) disorder. Pediatr Nephrol 22:2097–2103CrossRefPubMed Sharma AP, Greenberg CR, Prasad AN, Prasad C (2007) Hemolytic uremic syndrome (HUS) secondary to cobalamin C (cblC) disorder. Pediatr Nephrol 22:2097–2103CrossRefPubMed
30.
Zurück zum Zitat Guigonis V, Frémeaux-Bacchi V, Giraudier S, Favier R, Borderie D, Massy Z, Mougenot B, Rosenblatt DS, Deschênes G (2005) Late-onset thrombocytic microangiopathy caused by cblC disease: association with a factor H mutation. Am J Kidney Dis 45:588–595CrossRefPubMed Guigonis V, Frémeaux-Bacchi V, Giraudier S, Favier R, Borderie D, Massy Z, Mougenot B, Rosenblatt DS, Deschênes G (2005) Late-onset thrombocytic microangiopathy caused by cblC disease: association with a factor H mutation. Am J Kidney Dis 45:588–595CrossRefPubMed
31.
Zurück zum Zitat Van Hove JL, Van Damme-Lombaerts R, Grünewald S, Peters H, Van Damme B, Fryns JP, Arnout J, Wevers R, Baumgartner ER, Fowler B (2002) Cobalamin disorder Cbl-C presenting with late-onset thrombotic microangiopathy. Am J Med Genet 111:195–201CrossRefPubMed Van Hove JL, Van Damme-Lombaerts R, Grünewald S, Peters H, Van Damme B, Fryns JP, Arnout J, Wevers R, Baumgartner ER, Fowler B (2002) Cobalamin disorder Cbl-C presenting with late-onset thrombotic microangiopathy. Am J Med Genet 111:195–201CrossRefPubMed
32.
Zurück zum Zitat Kind T, Levy J, Lee M, Kaicker S, Nicholson JF, Kane SA (2002) Cobalamin C disease presenting as hemolytic-uremic syndrome in the neonatal period. J Pediatr Hematol Oncol 24:327–329CrossRefPubMed Kind T, Levy J, Lee M, Kaicker S, Nicholson JF, Kane SA (2002) Cobalamin C disease presenting as hemolytic-uremic syndrome in the neonatal period. J Pediatr Hematol Oncol 24:327–329CrossRefPubMed
33.
Zurück zum Zitat Chenel C, Wood C, Gourrier E, Zittoun J, Casadevall I, Ogier H (1993) Neonatal hemolytic-uremic syndrome, methylmalonic aciduria and homocystinuria caused by intracellular vitamin B 12 deficiency. Value of etiological diagnosis. Arch Fr Pediatr 50:749–754PubMed Chenel C, Wood C, Gourrier E, Zittoun J, Casadevall I, Ogier H (1993) Neonatal hemolytic-uremic syndrome, methylmalonic aciduria and homocystinuria caused by intracellular vitamin B 12 deficiency. Value of etiological diagnosis. Arch Fr Pediatr 50:749–754PubMed
34.
Zurück zum Zitat Geraghty MT, Perlman EJ, Martin LS, Hayflick SJ, Casella JF, Rosenblatt DS, Valle D (1992) Cobalamin C defect associated with hemolytic-uremic syndrome. J Pediatr 120:934–937CrossRefPubMed Geraghty MT, Perlman EJ, Martin LS, Hayflick SJ, Casella JF, Rosenblatt DS, Valle D (1992) Cobalamin C defect associated with hemolytic-uremic syndrome. J Pediatr 120:934–937CrossRefPubMed
35.
Zurück zum Zitat Huemer M, Scholl-Bürgi S, Hadaya K, Kern I, Beer R, Seppi K, Fowler B, Baumgartner MR, Karall D (2014) Three new cases of late-onset cblC defect and review of the literature illustrating when to consider inborn errors of metabolism beyond infancy. Orphanet J Rare Dis 9:161CrossRefPubMedPubMedCentral Huemer M, Scholl-Bürgi S, Hadaya K, Kern I, Beer R, Seppi K, Fowler B, Baumgartner MR, Karall D (2014) Three new cases of late-onset cblC defect and review of the literature illustrating when to consider inborn errors of metabolism beyond infancy. Orphanet J Rare Dis 9:161CrossRefPubMedPubMedCentral
36.
Zurück zum Zitat Richard E, Jorge-Finnigan A, Garcia-Villoria J, Merinero B, Desviat LR, Gort L, Briones P, Leal F, Pérez-Cerdá C, Ribes A, Ugarte M, Pérez B, MMACHC Working Group (2009) Genetic and cellular studies of oxidative stress in methylmalonic aciduria (MMA) cobalamin deficiency type C (cblC) with homocystinuria (MMACHC). Hum Mutat 30:1558–1566CrossRefPubMed Richard E, Jorge-Finnigan A, Garcia-Villoria J, Merinero B, Desviat LR, Gort L, Briones P, Leal F, Pérez-Cerdá C, Ribes A, Ugarte M, Pérez B, MMACHC Working Group (2009) Genetic and cellular studies of oxidative stress in methylmalonic aciduria (MMA) cobalamin deficiency type C (cblC) with homocystinuria (MMACHC). Hum Mutat 30:1558–1566CrossRefPubMed
37.
Zurück zum Zitat Thauvin-Robinet C, Roze E, Couvreur G, Horellou MH, Sedel F, Grabli D, Bruneteau G, Tonneti C, Masurel-Paulet A, Perennou D, Moreau T, Giroud M, de Baulny HO, Giraudier S, Faivre L (2008) The adolescent and adult form of cobalamin C disease: clinical and molecular spectrum. J Neurol Neurosurg Psychiatry 79:725–728CrossRefPubMed Thauvin-Robinet C, Roze E, Couvreur G, Horellou MH, Sedel F, Grabli D, Bruneteau G, Tonneti C, Masurel-Paulet A, Perennou D, Moreau T, Giroud M, de Baulny HO, Giraudier S, Faivre L (2008) The adolescent and adult form of cobalamin C disease: clinical and molecular spectrum. J Neurol Neurosurg Psychiatry 79:725–728CrossRefPubMed
38.
Zurück zum Zitat Brunelli SM, Meyers KE, Guttenberg M, Kaplan P, Kaplan BS (2002) Cobalamin C deficiency complicated by an atypical glomerulopathy. Pediatr Nephrol 17:800–803CrossRefPubMed Brunelli SM, Meyers KE, Guttenberg M, Kaplan P, Kaplan BS (2002) Cobalamin C deficiency complicated by an atypical glomerulopathy. Pediatr Nephrol 17:800–803CrossRefPubMed
39.
Zurück zum Zitat Benz K, Amann K (2010) Thrombotic microangiopathy: new insights. Curr Opin Nephrol Hypertens 19:242–247CrossRefPubMed Benz K, Amann K (2010) Thrombotic microangiopathy: new insights. Curr Opin Nephrol Hypertens 19:242–247CrossRefPubMed
40.
Zurück zum Zitat Gasser C, Gautier E, Steck A, Siebenmann RE, Oechslin R (1955) Hemolytic-uremic syndrome: bilateral necrosis of the renal cortex in acute acquired hemolytic anemia. Schweiz Med Wochenschr 85:905–909PubMed Gasser C, Gautier E, Steck A, Siebenmann RE, Oechslin R (1955) Hemolytic-uremic syndrome: bilateral necrosis of the renal cortex in acute acquired hemolytic anemia. Schweiz Med Wochenschr 85:905–909PubMed
41.
Zurück zum Zitat Renaud C, Niaudet P, Gagnadoux MF, Broyer M, Habib R (1995) Haemolytic uraemic syndrome: prognostic factors in children over 3 years of age. Pediatr Nephrol 9:24–29CrossRefPubMed Renaud C, Niaudet P, Gagnadoux MF, Broyer M, Habib R (1995) Haemolytic uraemic syndrome: prognostic factors in children over 3 years of age. Pediatr Nephrol 9:24–29CrossRefPubMed
43.
Zurück zum Zitat Keller MD, Ganesh J, Heltzer M, Paessler M, Bergqvist AG, Baluarte HJ, Watkins D, Rosenblatt DS, Orange JS (2013) Severe combined immunodeficiency resulting from mutations in MTHFD1. Pediatrics 131:e629–e634CrossRefPubMed Keller MD, Ganesh J, Heltzer M, Paessler M, Bergqvist AG, Baluarte HJ, Watkins D, Rosenblatt DS, Orange JS (2013) Severe combined immunodeficiency resulting from mutations in MTHFD1. Pediatrics 131:e629–e634CrossRefPubMed
44.
Zurück zum Zitat Watkins D, Schwartzentruber JA, Ganesh J, Orange JS, Kaplan BS, Nunez LD, Majewski J, Rosenblatt DS (2011) Novel inborn error of folate metabolism: identification by exome capture and sequencing of mutations in the MTHFD1 gene in a single proband. J Med Genet 48:590–592CrossRefPubMed Watkins D, Schwartzentruber JA, Ganesh J, Orange JS, Kaplan BS, Nunez LD, Majewski J, Rosenblatt DS (2011) Novel inborn error of folate metabolism: identification by exome capture and sequencing of mutations in the MTHFD1 gene in a single proband. J Med Genet 48:590–592CrossRefPubMed
45.
Zurück zum Zitat Andrès E, Affenberger S, Zimmer J, Vinzio S, Grosu D, Pistol G, Maloisel F, Weitten T, Kaltenbach G, Blicklé JF (2006) Current hematological findings in cobalamin deficiency. A study of 201 consecutive patients with documented cobalamin deficiency. Clin Lab Haematol 28:50–56CrossRefPubMed Andrès E, Affenberger S, Zimmer J, Vinzio S, Grosu D, Pistol G, Maloisel F, Weitten T, Kaltenbach G, Blicklé JF (2006) Current hematological findings in cobalamin deficiency. A study of 201 consecutive patients with documented cobalamin deficiency. Clin Lab Haematol 28:50–56CrossRefPubMed
46.
Zurück zum Zitat Han L, Wu S, Ye J, Qiu W, Zhang H, Gao X, Wang Y, Gong Z, Jin J, Gu X (2015) Biochemical, molecular and outcome analysis of eight chinese asymptomatic individuals with methyl malonic acidemia detected through newborn screening. Am J Med Genet A 167A:2300–2305CrossRefPubMed Han L, Wu S, Ye J, Qiu W, Zhang H, Gao X, Wang Y, Gong Z, Jin J, Gu X (2015) Biochemical, molecular and outcome analysis of eight chinese asymptomatic individuals with methyl malonic acidemia detected through newborn screening. Am J Med Genet A 167A:2300–2305CrossRefPubMed
47.
Zurück zum Zitat Fremeaux-Bacchi V, Fakhouri F, Garnier A, Bienaimé F, Dragon-Durey MA, Ngo S, Moulin B, Servais A, Provot F, Rostaing L, Burtey S, Niaudet P, Deschênes G, Lebranchu Y, Zuber J, Loirat C (2013) Genetics and outcome of atypical hemolytic uremic syndrome: a nationwide French series comparing children and adults. Clin J Am Soc Nephrol 8:554–562CrossRefPubMedPubMedCentral Fremeaux-Bacchi V, Fakhouri F, Garnier A, Bienaimé F, Dragon-Durey MA, Ngo S, Moulin B, Servais A, Provot F, Rostaing L, Burtey S, Niaudet P, Deschênes G, Lebranchu Y, Zuber J, Loirat C (2013) Genetics and outcome of atypical hemolytic uremic syndrome: a nationwide French series comparing children and adults. Clin J Am Soc Nephrol 8:554–562CrossRefPubMedPubMedCentral
48.
Zurück zum Zitat Geerdink LM, Westra D, van Wijk JA, Dorresteijn EM, Lilien MR, Davin JC, Kömhoff M, Van Hoeck K, van der Vlugt A, van den Heuvel LP, van de Kar NC (2012) Atypical hemolytic uremic syndrome in children: complement mutations and clinical characteristics. Pediatr Nephrol 27:1283–1291CrossRefPubMedPubMedCentral Geerdink LM, Westra D, van Wijk JA, Dorresteijn EM, Lilien MR, Davin JC, Kömhoff M, Van Hoeck K, van der Vlugt A, van den Heuvel LP, van de Kar NC (2012) Atypical hemolytic uremic syndrome in children: complement mutations and clinical characteristics. Pediatr Nephrol 27:1283–1291CrossRefPubMedPubMedCentral
49.
Zurück zum Zitat Brandstetter Y, Weinhouse E, Splaingard ML, Tang TT (1990) Cor pulmonale as a complication of methylmalonic acidemia and homocystinuria (Cbl-C type). Am J Med Genet 36:167–171CrossRefPubMed Brandstetter Y, Weinhouse E, Splaingard ML, Tang TT (1990) Cor pulmonale as a complication of methylmalonic acidemia and homocystinuria (Cbl-C type). Am J Med Genet 36:167–171CrossRefPubMed
50.
Zurück zum Zitat Iodice FG, Di Chiara L, Boenzi S, Aiello C, Monti L, Cogo P, Dionisi-Vici C (2013) Cobalamin C defect presenting with isolated pulmonary hypertension. Pediatrics 132:e248–e251CrossRefPubMed Iodice FG, Di Chiara L, Boenzi S, Aiello C, Monti L, Cogo P, Dionisi-Vici C (2013) Cobalamin C defect presenting with isolated pulmonary hypertension. Pediatrics 132:e248–e251CrossRefPubMed
51.
Zurück zum Zitat Solomon LR (2007) Disorders of cobalamin (vitamin B12) metabolism: emerging concepts in pathophysiology, diagnosis and treatment. Blood Rev 21:113–130CrossRefPubMed Solomon LR (2007) Disorders of cobalamin (vitamin B12) metabolism: emerging concepts in pathophysiology, diagnosis and treatment. Blood Rev 21:113–130CrossRefPubMed
52.
Zurück zum Zitat Carrillo-Carrasco N, Sloan J, Valle D, Hamosh A, Venditti CP (2009) Hydroxocobalamin dose escalation improves metabolic control in cblC. J Inherit Metab Dis 32:728–731CrossRefPubMedPubMedCentral Carrillo-Carrasco N, Sloan J, Valle D, Hamosh A, Venditti CP (2009) Hydroxocobalamin dose escalation improves metabolic control in cblC. J Inherit Metab Dis 32:728–731CrossRefPubMedPubMedCentral
53.
Zurück zum Zitat Zuber J, Fakhouri F, Roumenina LT, Loirat C, Frémeaux-Bacchi V, French Study Group for aHUS/C3G (2012) Use of eculizumab for atypical haemolytic uraemic syndrome and C3 glomerulopathies. Nat Rev Nephrol 8:643–657CrossRefPubMed Zuber J, Fakhouri F, Roumenina LT, Loirat C, Frémeaux-Bacchi V, French Study Group for aHUS/C3G (2012) Use of eculizumab for atypical haemolytic uraemic syndrome and C3 glomerulopathies. Nat Rev Nephrol 8:643–657CrossRefPubMed
54.
Zurück zum Zitat MacLennan S, Barbara JA (2006) Risks and side effects of therapy with plasma and plasma fractions. Best Pract Res Clin Haematol 19:169–189CrossRefPubMed MacLennan S, Barbara JA (2006) Risks and side effects of therapy with plasma and plasma fractions. Best Pract Res Clin Haematol 19:169–189CrossRefPubMed
55.
Zurück zum Zitat Abels J, Kroes AC, Ermens AA, van Kapel J, Schoester M, Spijkers LJ, Lindemans J (1990) Anti-leukemic potential of methyl-cobalamin inactivation by nitrous oxide. Am J Hematol 34:128–131CrossRefPubMed Abels J, Kroes AC, Ermens AA, van Kapel J, Schoester M, Spijkers LJ, Lindemans J (1990) Anti-leukemic potential of methyl-cobalamin inactivation by nitrous oxide. Am J Hematol 34:128–131CrossRefPubMed
56.
Zurück zum Zitat Drummond JT, Matthews RG (1994) Nitrous oxide degradation by cobalamin-dependent methionine synthase: characterization of the reactants and products in the inactivation reaction. Biochemistry 33:3732–3741CrossRefPubMed Drummond JT, Matthews RG (1994) Nitrous oxide degradation by cobalamin-dependent methionine synthase: characterization of the reactants and products in the inactivation reaction. Biochemistry 33:3732–3741CrossRefPubMed
57.
Zurück zum Zitat Komhoff M, Roofthooft MT, Spronsen F (2014) Syndromes of thrombotic microangiopathy. N Engl J Med 371:1846–1847CrossRefPubMed Komhoff M, Roofthooft MT, Spronsen F (2014) Syndromes of thrombotic microangiopathy. N Engl J Med 371:1846–1847CrossRefPubMed
Metadaten
Titel
Renal thrombotic microangiopathy in patients with cblC defect: review of an under-recognized entity
verfasst von
Bodo B. Beck
FrancJan van Spronsen
Arjan Diepstra
Rolf M. F. Berger
Martin Kömhoff
Publikationsdatum
11.06.2016
Verlag
Springer Berlin Heidelberg
Erschienen in
Pediatric Nephrology / Ausgabe 5/2017
Print ISSN: 0931-041X
Elektronische ISSN: 1432-198X
DOI
https://doi.org/10.1007/s00467-016-3399-0

Weitere Artikel der Ausgabe 5/2017

Pediatric Nephrology 5/2017 Zur Ausgabe

Kinder mit anhaltender Sinusitis profitieren häufig von Antibiotika

30.04.2024 Rhinitis und Sinusitis Nachrichten

Persistieren Sinusitisbeschwerden bei Kindern länger als zehn Tage, ist eine Antibiotikatherapie häufig gut wirksam: Ein Therapieversagen ist damit zu über 40% seltener zu beobachten als unter Placebo.

Neuer Typ-1-Diabetes bei Kindern am Wochenende eher übersehen

23.04.2024 Typ-1-Diabetes Nachrichten

Wenn Kinder an Werktagen zum Arzt gehen, werden neu auftretender Typ-1-Diabetes und diabetische Ketoazidosen häufiger erkannt als bei Arztbesuchen an Wochenenden oder Feiertagen.

Neue Studienergebnisse zur Myopiekontrolle mit Atropin

22.04.2024 Fehlsichtigkeit Nachrichten

Augentropfen mit niedrig dosiertem Atropin können helfen, das Fortschreiten einer Kurzsichtigkeit bei Kindern zumindest zu verlangsamen, wie die Ergebnisse einer aktuellen Studie mit verschiedenen Dosierungen zeigen.

Spinale Muskelatrophie: Neugeborenen-Screening lohnt sich

18.04.2024 Spinale Muskelatrophien Nachrichten

Seit 2021 ist die Untersuchung auf spinale Muskelatrophie Teil des Neugeborenen-Screenings in Deutschland. Eine Studie liefert weitere Evidenz für den Nutzen der Maßnahme.

Update Pädiatrie

Bestellen Sie unseren Fach-Newsletter und bleiben Sie gut informiert.