Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 13 de 13
Filter
2.
J Inherit Metab Dis ; 38(6): 1075-83, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25896882

ABSTRACT

Glutamine synthetase (GS) deficiency is an ultra-rare inborn error of amino acid metabolism that has been described in only three patients so far. The disease is characterized by neonatal onset of severe encephalopathy, low levels of glutamine in blood and cerebrospinal fluid, chronic moderate hyperammonemia, and an overall poor prognosis in the absence of an effective treatment. Recently, enteral glutamine supplementation was shown to be a safe and effective therapy for this disease but there are no data available on the long-term effects of this intervention. The amino acid glutamine, severely lacking in this disorder, is central to many metabolic pathways in the human organism and is involved in the synthesis of nicotinamide adenine dinucleotide (NAD(+)) starting from tryptophan or niacin as nicotinate, but not nicotinamide. Using fibroblasts, leukocytes, and immortalized peripheral blood stem cells (PBSC) from a patient carrying a GLUL gene point mutation associated with impaired GS activity, we tested whether glutamine deficiency in this patient results in NAD(+) depletion and whether it can be rescued by supplementation with glutamine, nicotinamide or nicotinate. The present study shows that congenital GS deficiency is associated with NAD(+) depletion in fibroblasts, leukocytes and PBSC, which may contribute to the severe clinical phenotype of the disease. Furthermore, it shows that NAD(+) depletion can be rescued by nicotinamide supplementation in fibroblasts and leukocytes, which may open up potential therapeutic options for the treatment of this disorder.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Glutamate-Ammonia Ligase/deficiency , Glutamine/blood , Hyperammonemia/genetics , NAD/blood , NAD/deficiency , B-Lymphocytes/cytology , Cell Culture Techniques , Dietary Supplements , Fibroblasts/cytology , Glutamate-Ammonia Ligase/genetics , Humans , Point Mutation
3.
Nat Genet ; 44(10): 1152-5, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22922874

ABSTRACT

Inherited disorders of vitamin B12 (cobalamin) have provided important clues to how this vitamin, which is essential for hematological and neurological function, is transported and metabolized. We describe a new disease that results in failure to release vitamin B12 from lysosomes, which mimics the cblF defect caused by LMBRD1 mutations. Using microcell-mediated chromosome transfer and exome sequencing, we identified causal mutations in ABCD4, a gene that codes for an ABC transporter, which was previously thought to have peroxisomal localization and function. Our results show that ABCD4 colocalizes with the lysosomal proteins LAMP1 and LMBD1, the latter of which is deficient in the cblF defect. Furthermore, we show that mutations altering the putative ATPase domain of ABCD4 affect its function, suggesting that the ATPase activity of ABCD4 may be involved in intracellular processing of vitamin B12.


Subject(s)
ATP-Binding Cassette Transporters/genetics , Abnormalities, Multiple/genetics , Metabolism, Inborn Errors/genetics , Mutation , Vitamin B 12/metabolism , ATP-Binding Cassette Transporters/metabolism , Abnormalities, Multiple/enzymology , Case-Control Studies , Cells, Cultured , DNA Mutational Analysis , Fibroblasts/metabolism , Gene Expression , Genes, Recessive , Genetic Association Studies , Humans , Infant, Newborn , Lysosomal Membrane Proteins/metabolism , Metabolism, Inborn Errors/enzymology , Nucleocytoplasmic Transport Proteins/metabolism , Protein Structure, Tertiary , Protein Transport
4.
Orphanet J Rare Dis ; 7: 31, 2012 May 29.
Article in English | MEDLINE | ID: mdl-22642865

ABSTRACT

BACKGROUND: Isolated 3-methylcrotonyl-CoA carboxylase (MCC) deficiency is an autosomal recessive disorder of leucine metabolism caused by mutations in MCCC1 or MCCC2 encoding the α and ß subunit of MCC, respectively. The phenotype is highly variable ranging from acute neonatal onset with fatal outcome to asymptomatic adults. METHODS: We report clinical, biochemical, enzymatic and mutation data of 88 MCC deficient individuals, 53 identified by newborn screening, 26 diagnosed due to clinical symptoms or positive family history and 9 mothers, identified following the positive newborn screening result of their baby. RESULTS: Fifty-seven percent of patients were asymptomatic while 43% showed clinical symptoms, many of which were probably not related to MCC deficiency but due to ascertainment bias. However, 12 patients (5 of 53 identified by newborn screening) presented with acute metabolic decompensations. We identified 15 novel MCCC1 and 16 novel MCCC2 mutant alleles. Additionally, we report expression studies on 3 MCCC1 and 8 MCCC2 mutations and show an overview of all 132 MCCC1 and MCCC2 variants known to date. CONCLUSIONS: Our data confirm that MCC deficiency, despite low penetrance, may lead to a severe clinical phenotype resembling classical organic acidurias. However, neither the genotype nor the biochemical phenotype is helpful in predicting the clinical course.


Subject(s)
Urea Cycle Disorders, Inborn/metabolism , Carbon-Carbon Ligases/deficiency , Carbon-Carbon Ligases/genetics , Carbon-Carbon Ligases/metabolism , Cell Line , Child , Child, Preschool , Female , Humans , Infant , Male , Polymerase Chain Reaction , Surveys and Questionnaires , Urea Cycle Disorders, Inborn/genetics , Urea Cycle Disorders, Inborn/pathology , Urea Cycle Disorders, Inborn/physiopathology
5.
Hum Mol Genet ; 21(6): 1410-8, 2012 Mar 15.
Article in English | MEDLINE | ID: mdl-22156578

ABSTRACT

The cblD defect of intracellular vitamin B(12) metabolism can lead to isolated methylmalonic aciduria (cblD-MMA) or homocystinuria (cblD-HC), or combined methylmalonic aciduria and homocystinuria (cblD-MMA/HC). We studied the mechanism whereby MMADHC mutations can lead to three phenotypes. The effect of various expression vectors containing MMADHC modified to contain an enhanced mitochondrial leader sequence or mutations changing possible downstream sites of reinitiation of translation or mutations introducing stop codons on rescue of adenosyl- and methylcobalamin (MeCbl) formation was studied. The constructs were transfected into cell lines derived from various cblD patient's fibroblasts. Expression of 10 mutant alleles from 15 cblD patients confirmed that the nature and location of the mutations correlate with the biochemical phenotype. In cblD-MMA/HC cells, improving mitochondrial targeting of MMADHC clearly increased the formation of adenosylcobalamin (AdoCbl) with a concomitant decrease in MeCbl formation. In cblD-MMA cells, this effect was dependent on the mutation and showed a negative correlation with endogenous MMADHC mRNA levels. These findings support the hypothesis that a single protein exists with two different functional domains that interact with either cytosolic or mitochondrial targets. Also a delicate balance exists between cytosolic MeCbl and mitochondrial AdoCbl synthesis, supporting the role of cblD protein as a branch point in intracellular cobalamin trafficking. Furthermore, our data indicate that the sequence after Met116 is sufficient for MeCbl synthesis, whereas the additional sequence between Met62 and Met116 is required for AdoCbl synthesis. Accordingly, western blot studies reveal proteins of the size expected from the stop codon position with subsequent reinitiation of translation.


Subject(s)
Amino Acid Metabolism, Inborn Errors/metabolism , Homocystinuria/metabolism , Mitochondrial Membrane Transport Proteins/genetics , Mitochondrial Membrane Transport Proteins/metabolism , Mutation/genetics , Vitamin B 12 Deficiency/metabolism , Vitamin B 12/metabolism , Amino Acid Metabolism, Inborn Errors/genetics , Amino Acid Metabolism, Inborn Errors/pathology , Blotting, Western , Cells, Cultured , Cytoplasm/metabolism , Fibroblasts/cytology , Fibroblasts/metabolism , Homocystinuria/genetics , Homocystinuria/pathology , Humans , Intracellular Signaling Peptides and Proteins , Mitochondria/metabolism , Phenotype , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Vitamin B 12 Deficiency/genetics , Vitamin B 12 Deficiency/pathology
6.
J Inherit Metab Dis ; 33(1): 17-24, 2010 Feb.
Article in English | MEDLINE | ID: mdl-20127417

ABSTRACT

In the cblF defect of vitamin B(12) (cobalamin) metabolism, cobalamin is trapped in lysosomes. Consequently, cobalamin coenzyme synthesis is blocked, and cofactors for methionine synthase and methylmalonyl-coenzyme A (CoA) mutase are deficient. We recently identified LMBRD1 as the causative gene located on chromosome 6q13 and showed that 18 out of 24 alleles in unrelated patients carried the deletion c.1056delG (p.L352fsX18) (Rutsch et al. (Nat Genet 41:234-239, 2009). LMBRD1 encodes the lysosomal membrane protein LMBD1, which presumably facilitates lysosomal cobalamin export. Our patient is the second child of consanguineous Turkish parents. He presented on the second day of life with cerebral seizures due to intraventricular hemorrhage. Plasma homocysteine and urinary methylmalonic acid levels were elevated, and serum cobalamin level was decreased. Synthesis of both cobalamin coenzymes was deficient in cultured skin fibroblasts. The cblF defect was confirmed by somatic complementation analysis. Sequencing of LMBRD1 revealed the novel deletion c.1405delG (p.D469fsX38) on both alleles. Real-time polymerase chain reaction (PCR) revealed reduced messenger RNA (mRNA) levels in patient fibroblasts compared with controls. Transfection of patient fibroblasts with the LMBD1 wild-type complement DNA (cDNA) rescued coenzyme synthesis and function, confirming this new deletion as an additional cause of the cblF defect. This case adds to the spectrum of clinical presentations and mutations of this rare disorder of lysosomal transport.


Subject(s)
Mutation , Nucleocytoplasmic Transport Proteins/genetics , Vitamin B 12/metabolism , 5-Methyltetrahydrofolate-Homocysteine S-Methyltransferase/metabolism , Alleles , Female , Fibroblasts/metabolism , Homocysteine/blood , Humans , Lysosomes/metabolism , Male , Methylmalonic Acid/urine , Methylmalonyl-CoA Mutase/genetics , Turkey , Vitamin B 12/blood
7.
J Biol Chem ; 284(42): 28953-7, 2009 Oct 16.
Article in English | MEDLINE | ID: mdl-19706617

ABSTRACT

3-Methylcrotonyl-CoA carboxylase (MCC) deficiency is an autosomal recessive disorder of leucine catabolism. MCC is a heteromeric mitochondrial enzyme composed of biotin-containing alpha (MCCA) and smaller beta (MCCB) subunits encoded by MCCA and MCCB, respectively. We report studies of the c.1054G-->A mutation in exon 11 of MCCB detected in the homozygous state in a patient with MCC deficiency. Sequence analysis of MCCB cDNA revealed two overlapping transcripts, one containing the normal 73 bp of exon 11 including the missense mutation c.1054G-->A (p.G352R), the other with exon 11 replaced by a 64-bp sequence from intron 10 (cryptic exon 10a) that maintains the reading frame and is flanked by acceptable splice consensus sites. In expression studies, we show that both transcripts lack detectable MCC activity. Western blot analysis showed slightly reduced levels of MCCB using the transcript containing the missense mutation, whereas no MCCB was detected with the transcript containing the cryptic exon 10a. Analysis of the region harboring the mutation revealed that the c.1054G-->A mutation is located in an exon splice enhancer sequence. Using MCCB minigene constructs to transfect MCCB-deficient fibroblasts, we demonstrate that the reduction in utilization of exon 11 associated with the c.1054G-->A mutation is due to alteration of this exon splice enhancer. Further, we show that optimization of the weak splice donor site of exon 11 corrects the splicing defect. To our knowledge, this is the first demonstration of a point mutation disrupting an exon splice enhancer that causes exon skipping along with utilization of a cryptic exon.


Subject(s)
Amino Acid Metabolism, Inborn Errors/genetics , Carbon-Carbon Ligases/deficiency , Carbon-Carbon Ligases/genetics , Alternative Splicing , Consanguinity , DNA Mutational Analysis , DNA, Complementary/metabolism , Exons , Failure to Thrive/genetics , Fibroblasts/metabolism , Humans , Models, Genetic , Mutation , Mutation, Missense , Sequence Analysis, DNA , Skin/metabolism
8.
Hum Mutat ; 30(7): 1072-81, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19370762

ABSTRACT

Methylmalonic aciduria and homocystinuria, cblC type, is a rare disorder of intracellular vitamin B(12) (cobalamin [Cbl]) metabolism caused by mutations in the MMACHC gene. MMACHC was sequenced from the gDNA of 118 cblC individuals. Eleven novel mutations were identified, as well as 23 mutations that were observed previously. Six sequence variants capture haplotype diversity in individuals across the MMACHC interval. Genotype-phenotype correlations of common mutations were apparent; individuals with c.394C>T tend to present with late-onset disease whereas patients with c.331C>T and c.271dupA tend to present in infancy. Other missense variants were also associated with late- or early-onset disease. Allelic expression analysis was carried out on human cblC fibroblasts compound heterozygous for different combinations of mutations including c.271dupA, c.331C>T, c.394C>T, and c.482G>A. The early-onset c.271dupA mutation was consistently underexpressed when compared to control alleles and the late-onset c.394C>T and c.482G>A mutations. The early-onset c.331C>T mutation was also underexpressed when compared to control alleles and the c.394C>T mutation. Levels of MMACHC mRNA transcript in cell lines homozygous for c.271dupA, c.331C>T, and c.394C>T were assessed using quantitative real-time RT-PCR. Cell lines homozygous for the late onset c.394C>T mutation had significantly higher levels of transcript when compared to cell lines homozygous for the early-onset mutations. Differential or preferential MMACHC transcript levels may provide a clue as to why individuals carrying c.394C>T generally present later in life.


Subject(s)
Alleles , Carrier Proteins/genetics , Gene Expression Regulation , Mutation , Age of Onset , Canada , DNA Mutational Analysis , Female , Genotype , Homocystinuria/genetics , Humans , Italy , Male , Oxidoreductases , Phenotype , RNA, Messenger/analysis , Vitamin B 12 Deficiency/genetics
9.
Nat Genet ; 41(2): 234-9, 2009 Feb.
Article in English | MEDLINE | ID: mdl-19136951

ABSTRACT

Vitamin B(12) (cobalamin) is essential in animals for metabolism of branched chain amino acids and odd chain fatty acids, and for remethylation of homocysteine to methionine. In the cblF inborn error of vitamin B(12) metabolism, free vitamin accumulates in lysosomes, thus hindering its conversion to cofactors. Using homozygosity mapping in 12 unrelated cblF individuals and microcell-mediated chromosome transfer, we identified a candidate gene on chromosome 6q13, LMBRD1, encoding LMBD1, a lysosomal membrane protein with homology to lipocalin membrane receptor LIMR. We identified five different frameshift mutations in LMBRD1 resulting in loss of LMBD1 function, with 18 of the 24 disease chromosomes carrying the same mutation embedded in a common 1.34-Mb haplotype. Transfection of fibroblasts of individuals with cblF with wild-type LMBD1 rescued cobalamin coenzyme synthesis and function. This work identifies LMBRD1 as the gene underlying the cblF defect of cobalamin metabolism and suggests that LMBD1 is a lysosomal membrane exporter for cobalamin.


Subject(s)
Hyperhomocysteinemia/complications , Membrane Transport Proteins/deficiency , Methylmalonic Acid/metabolism , Proteins/genetics , Transcobalamins/genetics , Vitamin B 12 Deficiency/genetics , Vitamin B 12/metabolism , Child , Chromosome Deletion , Chromosome Mapping , Chromosomes, Human, Pair 6 , Female , HeLa Cells , Humans , Hyperhomocysteinemia/genetics , Lysosomal Membrane Proteins/metabolism , Male , Membrane Transport Proteins/genetics , Membrane Transport Proteins/metabolism , Methylmalonic Acid/urine , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/metabolism , Nucleocytoplasmic Transport Proteins/physiology , Polymorphism, Genetic , Proteins/isolation & purification , Proteins/metabolism , Tissue Distribution , Transcobalamins/isolation & purification , Transcobalamins/metabolism , Vitamin B 12 Deficiency/etiology , Vitamin B 12 Deficiency/metabolism
10.
N Engl J Med ; 358(14): 1454-64, 2008 Apr 03.
Article in English | MEDLINE | ID: mdl-18385497

ABSTRACT

BACKGROUND: Vitamin B12 (cobalamin) is an essential cofactor in several metabolic pathways. Intracellular conversion of cobalamin to its two coenzymes, adenosylcobalamin in mitochondria and methylcobalamin in the cytoplasm, is necessary for the homeostasis of methylmalonic acid and homocysteine. Nine defects of intracellular cobalamin metabolism have been defined by means of somatic complementation analysis. One of these defects, the cblD defect, can cause isolated methylmalonic aciduria, isolated homocystinuria, or both. Affected persons present with multisystem clinical abnormalities, including developmental, hematologic, neurologic, and metabolic findings. The gene responsible for the cblD defect has not been identified. METHODS: We studied seven patients with the cblD defect, and skin fibroblasts from each were investigated in cell culture. Microcell-mediated chromosome transfer and refined genetic mapping were used to localize the responsible gene. This gene was transfected into cblD fibroblasts to test for the rescue of adenosylcobalamin and methylcobalamin synthesis. RESULTS: The cblD gene was localized to human chromosome 2q23.2, and a candidate gene, designated MMADHC (methylmalonic aciduria, cblD type, and homocystinuria), was identified in this region. Transfection of wild-type MMADHC rescued the cellular phenotype, and the functional importance of mutant alleles was shown by means of transfection with mutant constructs. The predicted MMADHC protein has sequence homology with a bacterial ATP-binding cassette transporter and contains a putative cobalamin binding motif and a putative mitochondrial targeting sequence. CONCLUSIONS: Mutations in a gene we designated MMADHC are responsible for the cblD defect in vitamin B12 metabolism. Various mutations are associated with each of the three biochemical phenotypes of the disorder.


Subject(s)
Membrane Transport Proteins/genetics , Metabolism, Inborn Errors/genetics , Methylmalonic Acid/urine , Mitochondrial Proteins/genetics , Vitamin B 12/metabolism , Adolescent , Amino Acid Sequence , Child , Chromosomes, Human, Pair 2 , DNA Mutational Analysis , Female , Homocystinuria/genetics , Humans , Infant , Infant, Newborn , Intracellular Signaling Peptides and Proteins , Male , Mitochondrial Membrane Transport Proteins , Molecular Sequence Data , Sequence Analysis, Protein
11.
Biologicals ; 35(1): 35-42, 2007 Mar.
Article in English | MEDLINE | ID: mdl-16581263

ABSTRACT

Patients with immunodeficiencies or some types of autoimmune diseases rely on a safe therapy with intravenous immunoglobulins (IVIGs) manufactured from human plasma, the only available source for this therapeutic. Since plasma is predisposed to contamination by a variety of blood-borne pathogens, ascertaining and ensuring the pathogen safety of plasma-derived therapeutics is a priority among manufacturers. State-of-the-art manufacturing processes provide a high safety standard by incorporating virus elimination procedures into the manufacturing process. Based on their mechanism these procedures are grouped into three classes: partitioning, inactivation, and virusfiltration.


Subject(s)
Autoimmune Diseases/therapy , Immunoglobulins, Intravenous/isolation & purification , Immunoglobulins, Intravenous/standards , Viruses/isolation & purification , Drug Contamination , Hot Temperature , Humans , Plasma/chemistry , Prion Diseases/prevention & control , Prions/isolation & purification , Severe Combined Immunodeficiency/therapy , Viral Load , Virus Inactivation
12.
Antimicrob Agents Chemother ; 50(4): 1183-94, 2006 Apr.
Article in English | MEDLINE | ID: mdl-16569828

ABSTRACT

Carbon catabolite protein A (CcpA) is known to function as a major regulator of gene expression in different gram-positive organisms. Deletion of the ccpA homologue (saCOL1786) in Staphylococcus aureus was found to affect growth, glucose metabolization, and transcription of selected virulence determinants. In liquid culture, deletion of CcpA decreased the growth rate and yield; however, the effect was only transient during the exponential-growth phase as long as glucose was present in the medium. Depletion of glucose and production of lactate was delayed, while the level of excretion of acetate was less affected and was even higher in the mutant culture. On solid medium, in contrast, growth of the DeltaccpA mutant resulted in smaller colonies containing a lower number of CFU per colony. Deletion of CcpA had an effect on the expression of important virulence factors of S. aureus by down-regulating RNAIII, the effector molecule of the agr locus, and altering the transcription patterns of hla, encoding alpha-hemolysin, and spa, encoding protein A. CcpA inactivation markedly reduced the oxacillin resistance levels in the highly methicillin-resistant S. aureus strain COLn and the teicoplanin resistance level in a glycopeptide-intermediate-resistant S. aureus strain. The presence of CcpA in the capsular polysaccharide serotype 5 (CP5)-producing strain Newman abolished capsule formation and decreased cap operon transcription in the presence of glucose. The staphylococcal CcpA thus not only is involved in the regulation of carbon metabolism but seems to function as a modulator of virulence gene expression as well.


Subject(s)
Bacterial Proteins/physiology , DNA-Binding Proteins/physiology , Repressor Proteins/physiology , Staphylococcus aureus/drug effects , Staphylococcus aureus/pathogenicity , Virulence Factors/biosynthesis , Bacterial Capsules/biosynthesis , Carbohydrate Metabolism , Drug Resistance, Bacterial , Glucose/pharmacology , Hydrogen-Ion Concentration , RNA, Antisense/genetics , RNA, Bacterial/genetics , Response Elements/physiology , Staphylococcus aureus/growth & development , Virulence
13.
Clin Rev Allergy Immunol ; 29(3): 333-44, 2005 Dec.
Article in English | MEDLINE | ID: mdl-16391410

ABSTRACT

Patients with immunodeficiencies or some types of autoimmune diseases are dependent on safe therapy with intravenous immunoglobulins. State-of-the-art manufacturing processes provide a high safety standard by incorporating virus elimination procedures into the manufacturing process. Based on their mechanism, these procedures are grouped into three classes: partitioning, inactivation, and removal based on size. Because of current socioeconomic and ecological changes, emerging pathogens continue to be expected. Such pathogens may spread very quickly because of increased intercontinental traffic. Severe acute respiratory syndrome-coronavirus and the West Nile virus are recent examples. Currently, it is not possible to predict the impact such a pathogen will have on blood safety because the capacity for a globally coordinated reaction to such a threat is also evolving. The worst-case scenario would be the emergence of a transmissible, small, nonenveloped virus in the blood donor population. Examples of small nonenveloped viruses, which change host and tissue tropism, are discussed, with focus on parvoviridae. Although today's immunoglobulins are safer than ever, in preparation for future challenges it is a high priority for the plasma industry to proactively investigate such viruses on a molecular and cellular level to identify their vulnerabilities.


Subject(s)
Consumer Product Safety/standards , Drug Contamination/prevention & control , Immunoglobulins, Intravenous , Virus Diseases/prevention & control , Virus Inactivation , Animals , Disease Transmission, Infectious , Humans , Virus Diseases/transmission
SELECTION OF CITATIONS
SEARCH DETAIL
...