Entry - #251000 - METHYLMALONIC ACIDURIA DUE TO METHYLMALONYL-CoA MUTASE DEFICIENCY - OMIM

# 251000

METHYLMALONIC ACIDURIA DUE TO METHYLMALONYL-CoA MUTASE DEFICIENCY


Alternative titles; symbols

METHYLMALONIC ACIDEMIA DUE TO METHYLMALONYL-CoA MUTASE DEFICIENCY
MMA DUE TO MCM DEFICIENCY
METHYLMALONIC ACIDURIA, mut TYPE


Other entities represented in this entry:

METHYLMALONIC ACIDURIA, mut(0) TYPE, INCLUDED
METHYLMALONIC ACIDURIA, mut(-) TYPE, INCLUDED

Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
6p12.3 Methylmalonic aciduria, mut(0) type 251000 AR 3 MMUT 609058
Clinical Synopsis
 

INHERITANCE
- Autosomal recessive
GROWTH
Weight
- Failure to thrive
CARDIOVASCULAR
Heart
- Cardiomyopathy
ABDOMEN
Liver
- Hepatomegaly
Pancreas
- Pancreatitis
Gastrointestinal
- Recurrent episodes of vomiting
GENITOURINARY
Kidneys
- Interstitial nephritis
- Chronic renal failure
NEUROLOGIC
Central Nervous System
- Lethargy
- Hypotonia
- Developmental delay
- Coma
- Severe involvement of globus pallidus
- Delay in myelination
- Cerebellar hemorrhage (rare)
- Ischemic stroke in the basal ganglia (rare)
METABOLIC FEATURES
- Dehydration
- Neonatal or infantile metabolic ketoacidosis
HEMATOLOGY
- Leukopenia
- Thrombocytopenia
LABORATORY ABNORMALITIES
- Normal serum cobalamin
- Metabolic ketoacidosis
- Hyperammonemia
- Hyperglycinemia
- Methymalonicaciduria
- Methylmalonyl-CoA mutase deficiency
- Low plasma free and total carnitine
MISCELLANEOUS
- mut-0 denotes individuals with cultured fibroblast mutase activity that is undetectable secondary to no functional mutase
- mut- denotes individuals with structurally altered mutase with reduced affinity for adenosylcobalamin (AdoCbl)
- Incidence of 1/50,000 births
MOLECULAR BASIS
- Caused by mutation in the methylmalonyl-CoA mutase gene (MMUT, 609058.0001)

TEXT

A number sign (#) is used with this entry because methylmalonic aciduria (MMA) of the complementation group 'mut' is caused by homozygous or compound heterozygous mutation in the gene encoding methylmalonyl-CoA mutase (MUT; 609058) on chromosome 6p12.


Description

Methylmalonic aciduria is a genetically heterogeneous disorder of methylmalonate and cobalamin (cbl; vitamin B12) metabolism. Isolated methylmalonic aciduria is found in patients with mutations in the MUT gene causing partial, mut(-), or complete, mut(0), enzyme deficiency. This form is unresponsive to B12 therapy. Various forms of isolated methylmalonic aciduria also occur in a subset of patients with defects in the synthesis of the MUT coenzyme adenosylcobalamin (AdoCbl) and are classified according to complementation group: cblA (251100), caused by mutation in the MMAA gene (607481) on chromosome 4q31, and cblB (251110), caused by mutation in the MMAB gene (607568) on 12q24.

Combined methylmalonic aciduria and homocystinuria may be seen in complementation groups cblC (277400), cblD (277410), and cblF (277380).

See the comprehensive review of Ledley (1990).


Clinical Features

The clinical spectrum of methylmalonic aciduria is wide, ranging from a benign condition (Ledley et al., 1984) to fatal neonatal disease (Matsui et al., 1983).

Oberholzer et al. (1967) reported an inborn error of metabolism characterized by methylmalonic aciduria associated with developmental retardation and chronic metabolic acidosis. Treatment with cobalamin was ineffective. Barness and Morrow (1968) noted that some cases of methylmalonic aciduria responded to vitamin B12. Of those not responsive to B12, only a subset had hyperglycinemia. Morrow et al. (1969) provided enzymatic proof of 2 forms of the disease with regard to response to cobalamin treatment: methylmalonyl-CoA mutase activity was essentially absent from the liver in a vitamin B12-unresponsive case, whereas residual enzyme activity increased to normal with added coenzyme in a vitamin B12-responsive case. The latter case was interpreted as having a defect in AdoCbl synthesis.

Gravel et al. (1975) confirmed the genetic heterogeneity of mut, cblA, cblB, and cblC. In vitro complementation studies measuring C14 incorporation into propionate showed that each of the mutants failed to incorporate C14 alone, whereas heterokaryons produced by fusing members of each of the 4 mutant classes with any other class produced results comparable to controls.

Willard and Rosenberg (1977) found that the mutase enzyme in cells from some MMA patients showed decreased affinity for AdoCbl with abnormally high Km for the coenzyme. These cases were considered to represent a structurally abnormal enzyme and were characteristic of the mut(-) phenotype. By immunohistochemical analysis of the mutase enzyme, Kolhouse et al. (1981) found that cell lines from mut(-) patients had detectable crossreacting material (CRM) ranging from 20 to 100% of control, whereas cells from mut(0) patients had CRM ranging from no detectable protein to 40% of controls.

Matsui et al. (1983) collected detailed information on 45 patients with MMA: 15 with mut(0) type, 5 with mut(-), 14 with cblA, and 11 with cblB. The most common presenting symptoms at onset were lethargy, failure to thrive, recurrent vomiting, dehydration, respiratory distress, and hypotonia. Other common features included hepatomegaly, developmental delay, and coma. Mut(0) patients presented earlier in infancy than the 3 other groups. All patients had methylmalonic acidemia and normal serum cobalamin, and most had metabolic acidosis, ketonuria, hyperammonemia, and hyperglycinemia. Approximately half of all the patients had pancytopenia. Most cblA and nearly half of cblB patients showed a decrease in urine and blood concentrations of methylmalonic acid in response to vitamin B12 supplementation, whereas none of the mut(0) or mut(-) responded. Most cblA, cblB, and mut(-) patients were still living at the time of the report; most mut(0) patients died during the first few months of life.

Shevell et al. (1993) compared the clinical features in 11 mut(0) patients with those in 9 mut(-) patients. All 11 mut(0) patients had an early neonatal presentation; 6 of these patients died in infancy and 3 of 5 survivors had a poor neurologic outcome as evidenced by severe developmental delay or spastic quadriparesis with dystonia. The 2 other survivors included a 27-month-old child with mild delay in verbal and fine motor skills and an adolescent with low normal intelligence. Of the 9 mut(-) patients, 7 became symptomatic in late infancy or childhood and 2 were found on screening. No episode of metabolic decompensation had occurred in 2 of the 9, yet both were neurologically compromised, one being severely retarded and autistic and the other mildly delayed. Four mut(-) patients had had episodic acidosis and were neurologically moderately affected, while 3 had had episodic acidosis but were neurologically intact. Although a broad correlation was found between mutase class and phenotype, survival with good outcome was possible among mut(0) patients and, conversely, significant morbidity occurred among mut(-) patients. Acidosis and metabolic imbalance were not necessary preconditions for significant morbidity. van den Bergh et al. (1992) reported sudden death in a child with MMA.

Giorgio et al. (1976) reported 2 French-Canadian brothers, aged 62 and 70 years, who had a benign form of MMA due to methylmalonyl-CoA mutase deficiency. Neither had anemia or hepatic dysfunction. Serum vitamin B12 was normal and the methylmalonic aciduria was unaffected by administration of vitamin B12 in large dosage. The brothers had presented with adult-onset diabetes mellitus. Ledley et al. (1984) reported a benign form of MMA due to deficiency of methylmalonyl-CoA mutase in 8 children identified through routine neonatal screening or screening of infants with affected sibs. Despite lack of dietary or vitamin therapy, the children had normal growth and development (age range, 18 months to 13 years) and performed as well as their unaffected sibs on psychometric tests. None responded to vitamin B12 treatment and there was no other evidence of a cofactor defect. In 2 sibs, complementation studies showed a defect in the mutase apoenzyme.

Renal insufficiency is frequently reported in mutase-deficient methylmalonic acidemia. Van Calcar et al. (1998) reported a patient with mut(-) MMA who developed chronic tubulointerstitial nephropathy during adolescence. After 24 years of age, she developed end-stage renal failure and underwent renal transplantation. Both plasma and urine methylmalonic acid levels decreased significantly with improved renal function following transplantation. Renal, metabolic, and clinical status remained improved at 3 years after the kidney transplant.

In a review of inherited metabolic disorders and stroke, Testai and Gorelick (2010) noted that patients with branched-chain organic aciduria, including isovaleric aciduria (243500), propionic aciduria (606054), and methylmalonic aciduria can rarely have strokes. Cerebellar hemorrhage has been described in all 3 disorders, and basal ganglia ischemic stroke has been described in propionic aciduria and methylmalonic aciduria. These events may occur in the absence of metabolic decompensation.

Kruszka et al. (2013) studied renal growth in isolated MMA. Fifty patients with MMA (35 mut subtype, 9 cblA subtype, and 6 cblB subtype), followed from 2004 to 2011, were classified by molecular genetics and studied using a combined cross-sectional and longitudinal design that included renal ultrasound examinations, anthropometric measurements, and metabolic phenotyping. Renal length was compared with that of healthy controls and modeled to other clinical parameters using multiple regression analyses. Comparisons with age-matched controls showed that renal length in subjects with MMA was significantly decreased (p less than 0.05). Stepwise regression modeling found that combinations of height, serum cystatin C (604312), and serum methylmalonic acid concentrations best predicted kidney size. Kruszka et al. (2013) concluded that renal length, reflective of kidney growth, significantly decreased in patients with MMA over time as compared with controls and was predictable with select clinical parameters. Cystatin C and serum methylmalonic acid concentrations were highly correlated with smaller kidneys and decreased renal function in this patient population.


Clinical Management

Kaplan et al. (2006) reported the long-term (9 years) outcome for the first patient with severe methylmalonic acidopathy transplanted in the United States and provided new biochemical data that indicated why transplanted patients remain susceptible to 'metabolic strokes.' In their 10-year-old male patient, there was clear evidence that the de novo synthesis of propionyl-CoA within the central nervous system led to brain methylmalonate accumulation that was largely unaffected by transplantation. Kaplan et al. (2006) concluded that liver transplantation is not a cure for methylmalonic acidopathy.

Niemi et al. (2015) reported the outcome of 14 MMA patients who underwent liver transplantation (6 patients) or liver-kidney transplantation (8 patients) at a mean age of 8.2 years (range 0.8 to 20.7 years). At mean follow-up of 3.25 years, survival was 100%, liver allograft survival was 93% (1 patient required retransplantation due to hepatic artery thrombosis) and renal allograft survival was 100%. Following transplantation, there were no metabolic decompensations, and neurodevelopmental abilities were maintained or improved.


Pathogenesis

Using 3D organotypic brain cell cultures derived from embryos of a brain-specific Mut -/- mouse, Remacle et al. (2018) investigated mechanisms leading to brain damage in methylmalonic aciduria. The in vitro model was challenged with the catabolic stress of temperature shift. Remacle et al. (2018) found typical metabolites for methylmalonic aciduria as well as a massive ammonia increase in the media of mutant mouse brain cultures. Investigation of pathways involved in intracerebral ammonia production revealed increased expression of glutaminase-2 (GLS2; 606365) and diminished expression of glutamate dehydrogenase-1 (GLUD1; 138130) in Mut -/- aggregates. Astrocytes showed swollen fibers and cell bodies, and oligodendrocytes showed inhibited axonal elongation and delayed myelination. Most effects were even more pronounced after 48 hours at 39 degrees C. Microglia activation and an increased apoptosis rate suggested degeneration of Mut -/- brain cells.


Other Features

Because of improvements in therapy, many patients with MMA reach childbearing age. Wasserstein et al. (1999) reported a successful pregnancy and delivery of a healthy baby to a 20-year-old woman with vitamin B12-unresponsive methylmalonic acidemia complicated by moderate renal insufficiency, chronic pancreatitis, anemia, and optic atrophy. Strict metabolic control was maintained throughout her pregnancy. The patient remained clinically asymptomatic during and after delivery, and her metabolic condition remained stable after discharge except for a slight decline in her renal function.


Diagnosis

Wilkemeyer et al. (1991) showed that the mut and cbl forms of MMA can be differentiated not only by somatic cell complementation but also by DNA-mediated gene transfer of a methylmalonyl CoA mutase cDNA clone. Transfer of the MUT clone into mut fibroblasts reconstituted holoenzyme activity, whereas the same process had no effect on cbl fibroblasts.


Cytogenetics

Abramowicz et al. (1994) studied a newborn female with a mut(0) form of MMA and complete absence of insulin-producing beta cells in otherwise normal-appearing pancreatic islets, causing insulin-dependent diabetes mellitus (IDDM; 222100). The patient died 2 weeks after birth. Serotyping of the HLA antigens, DNA typing of HLA-B and HLA class II loci, study of polymorphic DNA markers of chromosome 6, and cytogenetic analysis demonstrated paternal uniparental isodisomy, involving at least a 25-cM portion of chromosome 6 that encompasses the major histocompatibility complex. Duplication of the mutated allele on chromosome 6 inherited from the father was thought to be responsible for methylmalonic acidemia. It was also considered likely that isodisomy was etiologically related to the agenesis of beta cells, and the authors postulated the existence of a gene on chromosome 6 involved in beta-cell differentiation.


Molecular Genetics

In a patient with MMA mut(0), defined as having no residual enzyme activity, Jansen and Ledley (1990) identified compound heterozygosity for 2 mutations in the MUT gene (609058.0001 and 609058.0002).

In a patient with MMA mut(-), defined as having some residual enzyme activity, who had been reported by Ledley et al. (1990), Crane et al. (1992) identified a homozygous mutation in the MUT gene (609058.0005).

Acquaviva et al. (2001) reported a novel MUT missense mutation (609058.0010) in 5 unrelated families of French and Turkish descent from a population of 19 patients with MCM apoenzyme deficiency. All the patients exhibited a severe mut(0) methylmalonic acidemia phenotype, and 3 of them were homozygous for the mutation. The findings represented the first frequent MUT mutation reported in the Caucasian population.

Champattanachai et al. (2003) reported 2 novel mutations in a Thai patient with mut(0) methylmalonic acidemia.


History

Hoffman (1991) recounted the story of Patricia Stallings who was sentenced to life in prison for the presumed murder of her infant son with ethylene glycol, an ingredient of antifreeze. While in prison, the woman gave birth to a second son, who was found to have methylmalonic acidemia. William Sly and James Shoemaker at St. Louis University performed analyses of the first son's blood and did not detect ethylene glycol; Piero Rinaldo at Yale University demonstrated the biochemical features of methylmalonic acidemia and found no evidence of ethylene glycol in the body fluids. All charges against Patricia Stallings were dropped. Shoemaker et al. (1992) determined that the gas chromatographic peak that had been identified as ethylene glycol by a clinical laboratory was actually due to propionic acid. Woolf et al. (1992) noted that the opposite situation could occur: intentional infantile ethylene glycol poisoning being misinterpreted as an inborn error of metabolism leading to recurrent infantile metabolic acidosis.


REFERENCES

  1. Abramowicz, M. J., Andrien, M., Dupont, E., Dorchy, H., Parma, J., Duprez, L., Ledley, F. D., Courtens, W., Vamos, E. Isodisomy of chromosome 6 in a newborn with methylmalonic acidemia and agenesis of pancreatic beta cells causing diabetes mellitus. J. Clin. Invest. 94: 418-421, 1994. [PubMed: 7913714, related citations] [Full Text]

  2. Acquaviva, C., Benoist, J.-F., Callebaut, I., Guffon, N., Ogier de Baulny, H., Touati, G., Aydin, A., Porquet, D., Elion, J. N219Y, a new frequent mutation among mut-0 forms of methylmalonic acidemia in Caucasian patients. Europ. J. Hum. Genet. 9: 577-582, 2001. [PubMed: 11528502, related citations] [Full Text]

  3. Barness, L. A., Morrow, G., III. Methylmalonic aciduria--a newly discovered inborn error. Ann. Intern. Med. 69: 633-635, 1968. [PubMed: 5673182, related citations] [Full Text]

  4. Champattanachai, V., Ketudat Cairns, J. R., Shotelersuk, V., Keeratichamroen, S., Sawangareetrakul, P., Srisomsap, C., Kaewpaluek, V., Svasti, J. Novel mutations in a Thai patient with methylmalonic acidemia. Molec. Genet. Metab. 79: 300-302, 2003. [PubMed: 12948746, related citations] [Full Text]

  5. Crane, A. M., Jansen, R., Andrews, E. R., Ledley, F. D. Cloning and expression of a mutant methylmalonyl coenzyme A mutase with altered cobalamin affinity that causes mut(-) methylmalonic aciduria. J. Clin. Invest. 89: 385-391, 1992. [PubMed: 1346616, related citations] [Full Text]

  6. Fowlow, S. B., Holmes, T. M., Morgan, K., Snyder, F. F. Screening for methylmalonic aciduria in Alberta: a voluntary program with particular significance for the Hutterite brethren. Am. J. Med. Genet. 22: 513-519, 1985. [PubMed: 2865895, related citations] [Full Text]

  7. Giorgio, A. J., Trowbridge, M., Boone, A. W., Patten, R. S. Methylmalonic aciduria without vitamin B12 deficiency in an adult sibship. New Eng. J. Med. 295: 310-313, 1976. [PubMed: 6909, related citations] [Full Text]

  8. Gravel, R. A., Mahoney, M. J., Ruddle, F. H., Rosenberg, L. E. Genetic complementation in heterokaryons of human fibroblasts defective in cobalamin metabolism. Proc. Nat. Acad. Sci. 72: 3181-3185, 1975. [PubMed: 1059104, related citations] [Full Text]

  9. Hoffman, M. Scientific sleuths solve a murder mystery. Science 254: 931 only, 1991. [PubMed: 1948075, related citations] [Full Text]

  10. Hsia, Y. E., Lilljeqvist, A. C., Rosenberg, L. E. Vitamin B12-dependent methylmalonic aciduria: amino acid toxicity, long chain ketonuria, and protective effect of vitamin B12. Pediatrics 46: 497-507, 1970. [PubMed: 5503685, related citations]

  11. Jansen, R., Ledley, F. D. Heterozygous mutations at the mut locus in fibroblasts with mut-0 methylmalonic acidemia identified by polymerase-chain-reaction cDNA cloning. Am. J. Hum. Genet. 47: 808-814, 1990. [PubMed: 1977311, related citations]

  12. Kaplan, P., Ficicioglu, C., Mazur, A. T., Palmieri, M. J., Berry, G. T. Liver transplantation is not curative for methylmalonic acidopathy caused by methylmalonyl-CoA mutase deficiency. Molec. Genet. Metab. 88: 322-326, 2006. [PubMed: 16750411, related citations] [Full Text]

  13. Kolhouse, J. F., Utley, C., Fenton, W. A., Rosenberg, L. E. Immunochemical studies on cultured fibroblasts from patients with inherited methylmalonic acidemia. Proc. Nat. Acad. Sci. 78: 7737-7741, 1981. [PubMed: 6121323, related citations] [Full Text]

  14. Kruszka, P. S., Manoli, I., Sloan, J. L., Kopp, J. B., Venditti, C. P. Renal growth in isolated methylmalonic acidemia. Genet. Med. 15: 990-996, 2013. [PubMed: 23639900, images, related citations] [Full Text]

  15. Ledley, F. D., Jansen, R., Nham, S.-U., Fenton, W. A., Rosenberg, L. E. Mutation eliminating mitochondrial leader sequence of methylmalonyl-CoA mutase causes mut-0 methylmalonic acidemia. Proc. Nat. Acad. Sci. 87: 3147-3150, 1990. [PubMed: 1970180, related citations] [Full Text]

  16. Ledley, F. D., Levy, H. L., Shih, V. E., Benjamin, R., Mahoney, M. J. Benign methylmalonic aciduria. New Eng. J. Med. 311: 1015-1018, 1984. [PubMed: 6148691, related citations] [Full Text]

  17. Ledley, F. D. Perspectives on methylmalonic acidemia resulting from molecular cloning of methylmalonyl CoA mutase. BioEssays 12: 335-340, 1990. [PubMed: 1975493, related citations] [Full Text]

  18. Matsui, S. M., Mahoney, M. J., Rosenberg, L. E. The natural history of the inherited methylmalonic acidemias. New Eng. J. Med. 308: 857-861, 1983. [PubMed: 6132336, related citations] [Full Text]

  19. Morrow, G., III, Barness, L. A., Cardinale, G. J., Abeles, R. H., Flaks, J. G. Congenital methylmalonic acidemia: enzymatic evidence for two forms of the disease. Proc. Nat. Acad. Sci. 63: 191-197, 1969. [PubMed: 5257962, related citations] [Full Text]

  20. Niemi, A.-K., Kim, I. K., Krueger, C. E., Cowan, T. M., Baugh, N., Farrell, R., Bonham, C. A., Concepcion, W., Esquivel, C. O., Enns, G. M. Treatment of methylmalonic acidemia by liver or combined liver-kidney transplantation. J. Pediat. 166: 1455-1461, 2015. [PubMed: 25771389, related citations] [Full Text]

  21. Oberholzer, V. G., Levin, B., Burgess, E. A., Young, W. F. Methylmalonic aciduria: an inborn error of metabolism leading to chronic metabolic acidosis. Arch. Dis. Child. 42: 492-504, 1967. [PubMed: 6061291, related citations] [Full Text]

  22. Remacle, N., Forny, P., Cudre-Cung, H.-P., Gonzalez-Melo, M., do Vale-Pereira, S., Henry, H., Teav, T., Gallart-Ayala, H., Braissant, O., Baumgartner, M., Ballhausen, D. New in vitro model derived from brain-specific Mut-/- mice confirms cerebral ammonium accumulation in methylmalonic aciduria. Molec. Genet. Metab. 124: 266-277, 2018. [PubMed: 29934063, related citations] [Full Text]

  23. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E., Rosenbloom, F. M. Vitamin B12 dependent methylmalonic-aciduria: defective B12 metabolism in cultured fibroblasts. Biochem. Biophys. Res. Commun. 37: 607-614, 1969. [PubMed: 5353892, related citations] [Full Text]

  24. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E. Methylmalonic aciduria: an inborn error leading to metabolic acidosis, long-chain ketonuria and hyperglycinemia. New Eng. J. Med. 278: 1319-1322, 1968. [PubMed: 5648598, related citations] [Full Text]

  25. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E. Methylmalonic aciduria: metabolic block localization and vitamin B12 dependency. Science 162: 805-807, 1968. [PubMed: 5686220, related citations] [Full Text]

  26. Satoh, T., Narisawa, K., Igarashi, Y., Saitoh, T., Hayasaka, K., Ichinohazama, Y., Onodera, H., Tada, K., Oohara, K. Dietary therapy in two patients with vitamin B12-unresponsive methylmalonic acidemia. Europ. J. Pediat. 135: 305-312, 1981. [PubMed: 7227387, related citations] [Full Text]

  27. Shevell, M. I., Matiaszuk, N., Ledley, F. D., Rosenblatt, D. S. Varying neurological phenotypes among mut-0 and mut- patients with methylmalonyl CoA mutase deficiency. Am. J. Med. Genet. 45: 619-624, 1993. [PubMed: 7681251, related citations] [Full Text]

  28. Shoemaker, J. D., Lynch, R. E., Hoffmann, J. W., Sly, W. S. Misidentification of propionic acid as ethylene glycol in a patient with methylmalonic acidemia. J. Pediat. 120: 417-421, 1992. [PubMed: 1538288, related citations] [Full Text]

  29. Testai, F. D., Gorelick, P. B. Inherited metabolic disorders and stroke part 2: homocystinuria, organic acidurias, and urea cycle disorders. Arch. Neurol. 67: 148-153, 2010. [PubMed: 20142522, related citations] [Full Text]

  30. Van Calcar, S. C., Harding, C. O., Lyne, P., Hogan, K., Banerjee, R., Sollinger, H., Rieselbach, R. E., Wolff, J. A. Renal transplantation in a patient with methylmalonic acidaemia. J. Inherit. Metab. Dis. 21: 729-737, 1998. [PubMed: 9819702, related citations] [Full Text]

  31. van den Bergh, F. A. J. T. M., del Canho, H., Duran, M. Methylmalonic aciduria and sudden child death. J. Inherit. Metab. Dis. 15: 897-898, 1992. [PubMed: 1293386, related citations] [Full Text]

  32. Wasserstein, M. P., Gaddipati, S., Snyderman, S. E., Eddleman, K., Desnick, R. J., Sansaricq, C. Successful pregnancy in severe methylmalonic acidaemia. J. Inherit. Metab. Dis. 22: 788-794, 1999. [PubMed: 10518278, related citations] [Full Text]

  33. Wilcken, B., Kilham, H. A., Faull, K. Methylmalonic aciduria: a variant form of methylmalonyl coenzyme A apomutase deficiency. J. Pediat. 91: 428-430, 1977. [PubMed: 19569, related citations] [Full Text]

  34. Wilkemeyer, M. F., Crane, A. M., Ledley, F. D. Differential diagnosis of mut and cbl methylmalonic aciduria by DNA-mediated gene transfer in primary fibroblasts. J. Clin. Invest. 87: 915-918, 1991. [PubMed: 1671869, related citations] [Full Text]

  35. Willard, H. F., Rosenberg, L. E. Inherited deficiencies of human methylmalonyl CaA (sic) mutase activity: reduced affinity of mutant apoenzyme for adenosylcobalamin. Biochem. Biophys. Res. Commun. 78: 927-934, 1977. [PubMed: 20894, related citations] [Full Text]

  36. Woolf, A. D., Wynshaw-Boris, A., Rinaldo, P., Levy, H. L. Intentional infantile ethylene glycol poisoning presenting as an inherited metabolic disorder. J. Pediat. 120: 421-424, 1992. [PubMed: 1538289, related citations] [Full Text]


Ada Hamosh - updated : 09/15/2021
Ada Hamosh - updated : 12/07/2018
Ada Hamosh - updated : 1/9/2014
Cassandra L. Kniffin - updated : 10/11/2010
Ada Hamosh - updated : 6/28/2007
Cassandra L. Kniffin - reorganized : 12/10/2004
Cassandra L. Kniffin - updated : 12/6/2004
Ada Hamosh - updated : 10/9/2003
Michael B. Petersen - updated : 2/8/2002
Wilson H. Y. Lo - updated : 11/17/1999
Victor A. McKusick - updated : 12/2/1998
Victor A. McKusick - updated : 4/29/1998
Victor A. McKusick - updated : 9/22/1997
Creation Date:
Victor A. McKusick : 6/4/1986
carol : 03/11/2024
carol : 06/14/2022
alopez : 09/15/2021
carol : 11/20/2020
alopez : 12/07/2018
carol : 12/30/2015
alopez : 1/9/2014
wwang : 10/29/2010
ckniffin : 10/11/2010
alopez : 7/6/2007
terry : 6/28/2007
terry : 4/20/2005
terry : 4/6/2005
tkritzer : 3/16/2005
carol : 12/10/2004
carol : 12/10/2004
ckniffin : 12/6/2004
carol : 3/17/2004
alopez : 10/9/2003
terry : 10/9/2003
alopez : 2/11/2002
alopez : 2/8/2002
alopez : 2/8/2002
carol : 11/17/1999
carol : 11/17/1999
mgross : 6/18/1999
carol : 12/8/1998
terry : 12/2/1998
carol : 5/8/1998
terry : 4/29/1998
terry : 11/10/1997
mark : 9/23/1997
terry : 9/22/1997
terry : 1/23/1997
terry : 1/21/1997
mark : 6/24/1996
terry : 6/12/1996
terry : 10/27/1994
jason : 7/26/1994
warfield : 4/15/1994
mimadm : 4/8/1994
carol : 3/24/1993
carol : 2/10/1993

# 251000

METHYLMALONIC ACIDURIA DUE TO METHYLMALONYL-CoA MUTASE DEFICIENCY


Alternative titles; symbols

METHYLMALONIC ACIDEMIA DUE TO METHYLMALONYL-CoA MUTASE DEFICIENCY
MMA DUE TO MCM DEFICIENCY
METHYLMALONIC ACIDURIA, mut TYPE


Other entities represented in this entry:

METHYLMALONIC ACIDURIA, mut(0) TYPE, INCLUDED
METHYLMALONIC ACIDURIA, mut(-) TYPE, INCLUDED

ORPHA: 27, 289916, 79312;   DO: 0060740;  


Phenotype-Gene Relationships

Location Phenotype Phenotype
MIM number
Inheritance Phenotype
mapping key
Gene/Locus Gene/Locus
MIM number
6p12.3 Methylmalonic aciduria, mut(0) type 251000 Autosomal recessive 3 MMUT 609058

TEXT

A number sign (#) is used with this entry because methylmalonic aciduria (MMA) of the complementation group 'mut' is caused by homozygous or compound heterozygous mutation in the gene encoding methylmalonyl-CoA mutase (MUT; 609058) on chromosome 6p12.


Description

Methylmalonic aciduria is a genetically heterogeneous disorder of methylmalonate and cobalamin (cbl; vitamin B12) metabolism. Isolated methylmalonic aciduria is found in patients with mutations in the MUT gene causing partial, mut(-), or complete, mut(0), enzyme deficiency. This form is unresponsive to B12 therapy. Various forms of isolated methylmalonic aciduria also occur in a subset of patients with defects in the synthesis of the MUT coenzyme adenosylcobalamin (AdoCbl) and are classified according to complementation group: cblA (251100), caused by mutation in the MMAA gene (607481) on chromosome 4q31, and cblB (251110), caused by mutation in the MMAB gene (607568) on 12q24.

Combined methylmalonic aciduria and homocystinuria may be seen in complementation groups cblC (277400), cblD (277410), and cblF (277380).

See the comprehensive review of Ledley (1990).


Clinical Features

The clinical spectrum of methylmalonic aciduria is wide, ranging from a benign condition (Ledley et al., 1984) to fatal neonatal disease (Matsui et al., 1983).

Oberholzer et al. (1967) reported an inborn error of metabolism characterized by methylmalonic aciduria associated with developmental retardation and chronic metabolic acidosis. Treatment with cobalamin was ineffective. Barness and Morrow (1968) noted that some cases of methylmalonic aciduria responded to vitamin B12. Of those not responsive to B12, only a subset had hyperglycinemia. Morrow et al. (1969) provided enzymatic proof of 2 forms of the disease with regard to response to cobalamin treatment: methylmalonyl-CoA mutase activity was essentially absent from the liver in a vitamin B12-unresponsive case, whereas residual enzyme activity increased to normal with added coenzyme in a vitamin B12-responsive case. The latter case was interpreted as having a defect in AdoCbl synthesis.

Gravel et al. (1975) confirmed the genetic heterogeneity of mut, cblA, cblB, and cblC. In vitro complementation studies measuring C14 incorporation into propionate showed that each of the mutants failed to incorporate C14 alone, whereas heterokaryons produced by fusing members of each of the 4 mutant classes with any other class produced results comparable to controls.

Willard and Rosenberg (1977) found that the mutase enzyme in cells from some MMA patients showed decreased affinity for AdoCbl with abnormally high Km for the coenzyme. These cases were considered to represent a structurally abnormal enzyme and were characteristic of the mut(-) phenotype. By immunohistochemical analysis of the mutase enzyme, Kolhouse et al. (1981) found that cell lines from mut(-) patients had detectable crossreacting material (CRM) ranging from 20 to 100% of control, whereas cells from mut(0) patients had CRM ranging from no detectable protein to 40% of controls.

Matsui et al. (1983) collected detailed information on 45 patients with MMA: 15 with mut(0) type, 5 with mut(-), 14 with cblA, and 11 with cblB. The most common presenting symptoms at onset were lethargy, failure to thrive, recurrent vomiting, dehydration, respiratory distress, and hypotonia. Other common features included hepatomegaly, developmental delay, and coma. Mut(0) patients presented earlier in infancy than the 3 other groups. All patients had methylmalonic acidemia and normal serum cobalamin, and most had metabolic acidosis, ketonuria, hyperammonemia, and hyperglycinemia. Approximately half of all the patients had pancytopenia. Most cblA and nearly half of cblB patients showed a decrease in urine and blood concentrations of methylmalonic acid in response to vitamin B12 supplementation, whereas none of the mut(0) or mut(-) responded. Most cblA, cblB, and mut(-) patients were still living at the time of the report; most mut(0) patients died during the first few months of life.

Shevell et al. (1993) compared the clinical features in 11 mut(0) patients with those in 9 mut(-) patients. All 11 mut(0) patients had an early neonatal presentation; 6 of these patients died in infancy and 3 of 5 survivors had a poor neurologic outcome as evidenced by severe developmental delay or spastic quadriparesis with dystonia. The 2 other survivors included a 27-month-old child with mild delay in verbal and fine motor skills and an adolescent with low normal intelligence. Of the 9 mut(-) patients, 7 became symptomatic in late infancy or childhood and 2 were found on screening. No episode of metabolic decompensation had occurred in 2 of the 9, yet both were neurologically compromised, one being severely retarded and autistic and the other mildly delayed. Four mut(-) patients had had episodic acidosis and were neurologically moderately affected, while 3 had had episodic acidosis but were neurologically intact. Although a broad correlation was found between mutase class and phenotype, survival with good outcome was possible among mut(0) patients and, conversely, significant morbidity occurred among mut(-) patients. Acidosis and metabolic imbalance were not necessary preconditions for significant morbidity. van den Bergh et al. (1992) reported sudden death in a child with MMA.

Giorgio et al. (1976) reported 2 French-Canadian brothers, aged 62 and 70 years, who had a benign form of MMA due to methylmalonyl-CoA mutase deficiency. Neither had anemia or hepatic dysfunction. Serum vitamin B12 was normal and the methylmalonic aciduria was unaffected by administration of vitamin B12 in large dosage. The brothers had presented with adult-onset diabetes mellitus. Ledley et al. (1984) reported a benign form of MMA due to deficiency of methylmalonyl-CoA mutase in 8 children identified through routine neonatal screening or screening of infants with affected sibs. Despite lack of dietary or vitamin therapy, the children had normal growth and development (age range, 18 months to 13 years) and performed as well as their unaffected sibs on psychometric tests. None responded to vitamin B12 treatment and there was no other evidence of a cofactor defect. In 2 sibs, complementation studies showed a defect in the mutase apoenzyme.

Renal insufficiency is frequently reported in mutase-deficient methylmalonic acidemia. Van Calcar et al. (1998) reported a patient with mut(-) MMA who developed chronic tubulointerstitial nephropathy during adolescence. After 24 years of age, she developed end-stage renal failure and underwent renal transplantation. Both plasma and urine methylmalonic acid levels decreased significantly with improved renal function following transplantation. Renal, metabolic, and clinical status remained improved at 3 years after the kidney transplant.

In a review of inherited metabolic disorders and stroke, Testai and Gorelick (2010) noted that patients with branched-chain organic aciduria, including isovaleric aciduria (243500), propionic aciduria (606054), and methylmalonic aciduria can rarely have strokes. Cerebellar hemorrhage has been described in all 3 disorders, and basal ganglia ischemic stroke has been described in propionic aciduria and methylmalonic aciduria. These events may occur in the absence of metabolic decompensation.

Kruszka et al. (2013) studied renal growth in isolated MMA. Fifty patients with MMA (35 mut subtype, 9 cblA subtype, and 6 cblB subtype), followed from 2004 to 2011, were classified by molecular genetics and studied using a combined cross-sectional and longitudinal design that included renal ultrasound examinations, anthropometric measurements, and metabolic phenotyping. Renal length was compared with that of healthy controls and modeled to other clinical parameters using multiple regression analyses. Comparisons with age-matched controls showed that renal length in subjects with MMA was significantly decreased (p less than 0.05). Stepwise regression modeling found that combinations of height, serum cystatin C (604312), and serum methylmalonic acid concentrations best predicted kidney size. Kruszka et al. (2013) concluded that renal length, reflective of kidney growth, significantly decreased in patients with MMA over time as compared with controls and was predictable with select clinical parameters. Cystatin C and serum methylmalonic acid concentrations were highly correlated with smaller kidneys and decreased renal function in this patient population.


Clinical Management

Kaplan et al. (2006) reported the long-term (9 years) outcome for the first patient with severe methylmalonic acidopathy transplanted in the United States and provided new biochemical data that indicated why transplanted patients remain susceptible to 'metabolic strokes.' In their 10-year-old male patient, there was clear evidence that the de novo synthesis of propionyl-CoA within the central nervous system led to brain methylmalonate accumulation that was largely unaffected by transplantation. Kaplan et al. (2006) concluded that liver transplantation is not a cure for methylmalonic acidopathy.

Niemi et al. (2015) reported the outcome of 14 MMA patients who underwent liver transplantation (6 patients) or liver-kidney transplantation (8 patients) at a mean age of 8.2 years (range 0.8 to 20.7 years). At mean follow-up of 3.25 years, survival was 100%, liver allograft survival was 93% (1 patient required retransplantation due to hepatic artery thrombosis) and renal allograft survival was 100%. Following transplantation, there were no metabolic decompensations, and neurodevelopmental abilities were maintained or improved.


Pathogenesis

Using 3D organotypic brain cell cultures derived from embryos of a brain-specific Mut -/- mouse, Remacle et al. (2018) investigated mechanisms leading to brain damage in methylmalonic aciduria. The in vitro model was challenged with the catabolic stress of temperature shift. Remacle et al. (2018) found typical metabolites for methylmalonic aciduria as well as a massive ammonia increase in the media of mutant mouse brain cultures. Investigation of pathways involved in intracerebral ammonia production revealed increased expression of glutaminase-2 (GLS2; 606365) and diminished expression of glutamate dehydrogenase-1 (GLUD1; 138130) in Mut -/- aggregates. Astrocytes showed swollen fibers and cell bodies, and oligodendrocytes showed inhibited axonal elongation and delayed myelination. Most effects were even more pronounced after 48 hours at 39 degrees C. Microglia activation and an increased apoptosis rate suggested degeneration of Mut -/- brain cells.


Other Features

Because of improvements in therapy, many patients with MMA reach childbearing age. Wasserstein et al. (1999) reported a successful pregnancy and delivery of a healthy baby to a 20-year-old woman with vitamin B12-unresponsive methylmalonic acidemia complicated by moderate renal insufficiency, chronic pancreatitis, anemia, and optic atrophy. Strict metabolic control was maintained throughout her pregnancy. The patient remained clinically asymptomatic during and after delivery, and her metabolic condition remained stable after discharge except for a slight decline in her renal function.


Diagnosis

Wilkemeyer et al. (1991) showed that the mut and cbl forms of MMA can be differentiated not only by somatic cell complementation but also by DNA-mediated gene transfer of a methylmalonyl CoA mutase cDNA clone. Transfer of the MUT clone into mut fibroblasts reconstituted holoenzyme activity, whereas the same process had no effect on cbl fibroblasts.


Cytogenetics

Abramowicz et al. (1994) studied a newborn female with a mut(0) form of MMA and complete absence of insulin-producing beta cells in otherwise normal-appearing pancreatic islets, causing insulin-dependent diabetes mellitus (IDDM; 222100). The patient died 2 weeks after birth. Serotyping of the HLA antigens, DNA typing of HLA-B and HLA class II loci, study of polymorphic DNA markers of chromosome 6, and cytogenetic analysis demonstrated paternal uniparental isodisomy, involving at least a 25-cM portion of chromosome 6 that encompasses the major histocompatibility complex. Duplication of the mutated allele on chromosome 6 inherited from the father was thought to be responsible for methylmalonic acidemia. It was also considered likely that isodisomy was etiologically related to the agenesis of beta cells, and the authors postulated the existence of a gene on chromosome 6 involved in beta-cell differentiation.


Molecular Genetics

In a patient with MMA mut(0), defined as having no residual enzyme activity, Jansen and Ledley (1990) identified compound heterozygosity for 2 mutations in the MUT gene (609058.0001 and 609058.0002).

In a patient with MMA mut(-), defined as having some residual enzyme activity, who had been reported by Ledley et al. (1990), Crane et al. (1992) identified a homozygous mutation in the MUT gene (609058.0005).

Acquaviva et al. (2001) reported a novel MUT missense mutation (609058.0010) in 5 unrelated families of French and Turkish descent from a population of 19 patients with MCM apoenzyme deficiency. All the patients exhibited a severe mut(0) methylmalonic acidemia phenotype, and 3 of them were homozygous for the mutation. The findings represented the first frequent MUT mutation reported in the Caucasian population.

Champattanachai et al. (2003) reported 2 novel mutations in a Thai patient with mut(0) methylmalonic acidemia.


History

Hoffman (1991) recounted the story of Patricia Stallings who was sentenced to life in prison for the presumed murder of her infant son with ethylene glycol, an ingredient of antifreeze. While in prison, the woman gave birth to a second son, who was found to have methylmalonic acidemia. William Sly and James Shoemaker at St. Louis University performed analyses of the first son's blood and did not detect ethylene glycol; Piero Rinaldo at Yale University demonstrated the biochemical features of methylmalonic acidemia and found no evidence of ethylene glycol in the body fluids. All charges against Patricia Stallings were dropped. Shoemaker et al. (1992) determined that the gas chromatographic peak that had been identified as ethylene glycol by a clinical laboratory was actually due to propionic acid. Woolf et al. (1992) noted that the opposite situation could occur: intentional infantile ethylene glycol poisoning being misinterpreted as an inborn error of metabolism leading to recurrent infantile metabolic acidosis.


See Also:

Fowlow et al. (1985); Hsia et al. (1970); Rosenberg et al. (1969); Rosenberg et al. (1968); Rosenberg et al. (1968); Satoh et al. (1981); Wilcken et al. (1977)

REFERENCES

  1. Abramowicz, M. J., Andrien, M., Dupont, E., Dorchy, H., Parma, J., Duprez, L., Ledley, F. D., Courtens, W., Vamos, E. Isodisomy of chromosome 6 in a newborn with methylmalonic acidemia and agenesis of pancreatic beta cells causing diabetes mellitus. J. Clin. Invest. 94: 418-421, 1994. [PubMed: 7913714] [Full Text: https://doi.org/10.1172/JCI117339]

  2. Acquaviva, C., Benoist, J.-F., Callebaut, I., Guffon, N., Ogier de Baulny, H., Touati, G., Aydin, A., Porquet, D., Elion, J. N219Y, a new frequent mutation among mut-0 forms of methylmalonic acidemia in Caucasian patients. Europ. J. Hum. Genet. 9: 577-582, 2001. [PubMed: 11528502] [Full Text: https://doi.org/10.1038/sj.ejhg.5200675]

  3. Barness, L. A., Morrow, G., III. Methylmalonic aciduria--a newly discovered inborn error. Ann. Intern. Med. 69: 633-635, 1968. [PubMed: 5673182] [Full Text: https://doi.org/10.7326/0003-4819-69-3-633_2]

  4. Champattanachai, V., Ketudat Cairns, J. R., Shotelersuk, V., Keeratichamroen, S., Sawangareetrakul, P., Srisomsap, C., Kaewpaluek, V., Svasti, J. Novel mutations in a Thai patient with methylmalonic acidemia. Molec. Genet. Metab. 79: 300-302, 2003. [PubMed: 12948746] [Full Text: https://doi.org/10.1016/s1096-7192(03)00106-9]

  5. Crane, A. M., Jansen, R., Andrews, E. R., Ledley, F. D. Cloning and expression of a mutant methylmalonyl coenzyme A mutase with altered cobalamin affinity that causes mut(-) methylmalonic aciduria. J. Clin. Invest. 89: 385-391, 1992. [PubMed: 1346616] [Full Text: https://doi.org/10.1172/JCI115597]

  6. Fowlow, S. B., Holmes, T. M., Morgan, K., Snyder, F. F. Screening for methylmalonic aciduria in Alberta: a voluntary program with particular significance for the Hutterite brethren. Am. J. Med. Genet. 22: 513-519, 1985. [PubMed: 2865895] [Full Text: https://doi.org/10.1002/ajmg.1320220309]

  7. Giorgio, A. J., Trowbridge, M., Boone, A. W., Patten, R. S. Methylmalonic aciduria without vitamin B12 deficiency in an adult sibship. New Eng. J. Med. 295: 310-313, 1976. [PubMed: 6909] [Full Text: https://doi.org/10.1056/NEJM197608052950604]

  8. Gravel, R. A., Mahoney, M. J., Ruddle, F. H., Rosenberg, L. E. Genetic complementation in heterokaryons of human fibroblasts defective in cobalamin metabolism. Proc. Nat. Acad. Sci. 72: 3181-3185, 1975. [PubMed: 1059104] [Full Text: https://doi.org/10.1073/pnas.72.8.3181]

  9. Hoffman, M. Scientific sleuths solve a murder mystery. Science 254: 931 only, 1991. [PubMed: 1948075] [Full Text: https://doi.org/10.1126/science.1948075]

  10. Hsia, Y. E., Lilljeqvist, A. C., Rosenberg, L. E. Vitamin B12-dependent methylmalonic aciduria: amino acid toxicity, long chain ketonuria, and protective effect of vitamin B12. Pediatrics 46: 497-507, 1970. [PubMed: 5503685]

  11. Jansen, R., Ledley, F. D. Heterozygous mutations at the mut locus in fibroblasts with mut-0 methylmalonic acidemia identified by polymerase-chain-reaction cDNA cloning. Am. J. Hum. Genet. 47: 808-814, 1990. [PubMed: 1977311]

  12. Kaplan, P., Ficicioglu, C., Mazur, A. T., Palmieri, M. J., Berry, G. T. Liver transplantation is not curative for methylmalonic acidopathy caused by methylmalonyl-CoA mutase deficiency. Molec. Genet. Metab. 88: 322-326, 2006. [PubMed: 16750411] [Full Text: https://doi.org/10.1016/j.ymgme.2006.04.003]

  13. Kolhouse, J. F., Utley, C., Fenton, W. A., Rosenberg, L. E. Immunochemical studies on cultured fibroblasts from patients with inherited methylmalonic acidemia. Proc. Nat. Acad. Sci. 78: 7737-7741, 1981. [PubMed: 6121323] [Full Text: https://doi.org/10.1073/pnas.78.12.7737]

  14. Kruszka, P. S., Manoli, I., Sloan, J. L., Kopp, J. B., Venditti, C. P. Renal growth in isolated methylmalonic acidemia. Genet. Med. 15: 990-996, 2013. [PubMed: 23639900] [Full Text: https://doi.org/10.1038/gim.2013.42]

  15. Ledley, F. D., Jansen, R., Nham, S.-U., Fenton, W. A., Rosenberg, L. E. Mutation eliminating mitochondrial leader sequence of methylmalonyl-CoA mutase causes mut-0 methylmalonic acidemia. Proc. Nat. Acad. Sci. 87: 3147-3150, 1990. [PubMed: 1970180] [Full Text: https://doi.org/10.1073/pnas.87.8.3147]

  16. Ledley, F. D., Levy, H. L., Shih, V. E., Benjamin, R., Mahoney, M. J. Benign methylmalonic aciduria. New Eng. J. Med. 311: 1015-1018, 1984. [PubMed: 6148691] [Full Text: https://doi.org/10.1056/NEJM198410183111604]

  17. Ledley, F. D. Perspectives on methylmalonic acidemia resulting from molecular cloning of methylmalonyl CoA mutase. BioEssays 12: 335-340, 1990. [PubMed: 1975493] [Full Text: https://doi.org/10.1002/bies.950120706]

  18. Matsui, S. M., Mahoney, M. J., Rosenberg, L. E. The natural history of the inherited methylmalonic acidemias. New Eng. J. Med. 308: 857-861, 1983. [PubMed: 6132336] [Full Text: https://doi.org/10.1056/NEJM198304143081501]

  19. Morrow, G., III, Barness, L. A., Cardinale, G. J., Abeles, R. H., Flaks, J. G. Congenital methylmalonic acidemia: enzymatic evidence for two forms of the disease. Proc. Nat. Acad. Sci. 63: 191-197, 1969. [PubMed: 5257962] [Full Text: https://doi.org/10.1073/pnas.63.1.191]

  20. Niemi, A.-K., Kim, I. K., Krueger, C. E., Cowan, T. M., Baugh, N., Farrell, R., Bonham, C. A., Concepcion, W., Esquivel, C. O., Enns, G. M. Treatment of methylmalonic acidemia by liver or combined liver-kidney transplantation. J. Pediat. 166: 1455-1461, 2015. [PubMed: 25771389] [Full Text: https://doi.org/10.1016/j.jpeds.2015.01.051]

  21. Oberholzer, V. G., Levin, B., Burgess, E. A., Young, W. F. Methylmalonic aciduria: an inborn error of metabolism leading to chronic metabolic acidosis. Arch. Dis. Child. 42: 492-504, 1967. [PubMed: 6061291] [Full Text: https://doi.org/10.1136/adc.42.225.492]

  22. Remacle, N., Forny, P., Cudre-Cung, H.-P., Gonzalez-Melo, M., do Vale-Pereira, S., Henry, H., Teav, T., Gallart-Ayala, H., Braissant, O., Baumgartner, M., Ballhausen, D. New in vitro model derived from brain-specific Mut-/- mice confirms cerebral ammonium accumulation in methylmalonic aciduria. Molec. Genet. Metab. 124: 266-277, 2018. [PubMed: 29934063] [Full Text: https://doi.org/10.1016/j.ymgme.2018.06.008]

  23. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E., Rosenbloom, F. M. Vitamin B12 dependent methylmalonic-aciduria: defective B12 metabolism in cultured fibroblasts. Biochem. Biophys. Res. Commun. 37: 607-614, 1969. [PubMed: 5353892] [Full Text: https://doi.org/10.1016/0006-291x(69)90853-5]

  24. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E. Methylmalonic aciduria: an inborn error leading to metabolic acidosis, long-chain ketonuria and hyperglycinemia. New Eng. J. Med. 278: 1319-1322, 1968. [PubMed: 5648598] [Full Text: https://doi.org/10.1056/NEJM196806132782404]

  25. Rosenberg, L. E., Lilljeqvist, A. C., Hsia, Y. E. Methylmalonic aciduria: metabolic block localization and vitamin B12 dependency. Science 162: 805-807, 1968. [PubMed: 5686220] [Full Text: https://doi.org/10.1126/science.162.3855.805]

  26. Satoh, T., Narisawa, K., Igarashi, Y., Saitoh, T., Hayasaka, K., Ichinohazama, Y., Onodera, H., Tada, K., Oohara, K. Dietary therapy in two patients with vitamin B12-unresponsive methylmalonic acidemia. Europ. J. Pediat. 135: 305-312, 1981. [PubMed: 7227387] [Full Text: https://doi.org/10.1007/BF00442109]

  27. Shevell, M. I., Matiaszuk, N., Ledley, F. D., Rosenblatt, D. S. Varying neurological phenotypes among mut-0 and mut- patients with methylmalonyl CoA mutase deficiency. Am. J. Med. Genet. 45: 619-624, 1993. [PubMed: 7681251] [Full Text: https://doi.org/10.1002/ajmg.1320450521]

  28. Shoemaker, J. D., Lynch, R. E., Hoffmann, J. W., Sly, W. S. Misidentification of propionic acid as ethylene glycol in a patient with methylmalonic acidemia. J. Pediat. 120: 417-421, 1992. [PubMed: 1538288] [Full Text: https://doi.org/10.1016/s0022-3476(05)80909-6]

  29. Testai, F. D., Gorelick, P. B. Inherited metabolic disorders and stroke part 2: homocystinuria, organic acidurias, and urea cycle disorders. Arch. Neurol. 67: 148-153, 2010. [PubMed: 20142522] [Full Text: https://doi.org/10.1001/archneurol.2009.333]

  30. Van Calcar, S. C., Harding, C. O., Lyne, P., Hogan, K., Banerjee, R., Sollinger, H., Rieselbach, R. E., Wolff, J. A. Renal transplantation in a patient with methylmalonic acidaemia. J. Inherit. Metab. Dis. 21: 729-737, 1998. [PubMed: 9819702] [Full Text: https://doi.org/10.1023/a:1005493015489]

  31. van den Bergh, F. A. J. T. M., del Canho, H., Duran, M. Methylmalonic aciduria and sudden child death. J. Inherit. Metab. Dis. 15: 897-898, 1992. [PubMed: 1293386] [Full Text: https://doi.org/10.1007/BF01800229]

  32. Wasserstein, M. P., Gaddipati, S., Snyderman, S. E., Eddleman, K., Desnick, R. J., Sansaricq, C. Successful pregnancy in severe methylmalonic acidaemia. J. Inherit. Metab. Dis. 22: 788-794, 1999. [PubMed: 10518278] [Full Text: https://doi.org/10.1023/a:1005597722237]

  33. Wilcken, B., Kilham, H. A., Faull, K. Methylmalonic aciduria: a variant form of methylmalonyl coenzyme A apomutase deficiency. J. Pediat. 91: 428-430, 1977. [PubMed: 19569] [Full Text: https://doi.org/10.1016/s0022-3476(77)81313-9]

  34. Wilkemeyer, M. F., Crane, A. M., Ledley, F. D. Differential diagnosis of mut and cbl methylmalonic aciduria by DNA-mediated gene transfer in primary fibroblasts. J. Clin. Invest. 87: 915-918, 1991. [PubMed: 1671869] [Full Text: https://doi.org/10.1172/JCI115098]

  35. Willard, H. F., Rosenberg, L. E. Inherited deficiencies of human methylmalonyl CaA (sic) mutase activity: reduced affinity of mutant apoenzyme for adenosylcobalamin. Biochem. Biophys. Res. Commun. 78: 927-934, 1977. [PubMed: 20894] [Full Text: https://doi.org/10.1016/0006-291x(77)90511-3]

  36. Woolf, A. D., Wynshaw-Boris, A., Rinaldo, P., Levy, H. L. Intentional infantile ethylene glycol poisoning presenting as an inherited metabolic disorder. J. Pediat. 120: 421-424, 1992. [PubMed: 1538289] [Full Text: https://doi.org/10.1016/s0022-3476(05)80910-2]


Contributors:
Ada Hamosh - updated : 09/15/2021
Ada Hamosh - updated : 12/07/2018
Ada Hamosh - updated : 1/9/2014
Cassandra L. Kniffin - updated : 10/11/2010
Ada Hamosh - updated : 6/28/2007
Cassandra L. Kniffin - reorganized : 12/10/2004
Cassandra L. Kniffin - updated : 12/6/2004
Ada Hamosh - updated : 10/9/2003
Michael B. Petersen - updated : 2/8/2002
Wilson H. Y. Lo - updated : 11/17/1999
Victor A. McKusick - updated : 12/2/1998
Victor A. McKusick - updated : 4/29/1998
Victor A. McKusick - updated : 9/22/1997

Creation Date:
Victor A. McKusick : 6/4/1986

Edit History:
carol : 03/11/2024
carol : 06/14/2022
alopez : 09/15/2021
carol : 11/20/2020
alopez : 12/07/2018
carol : 12/30/2015
alopez : 1/9/2014
wwang : 10/29/2010
ckniffin : 10/11/2010
alopez : 7/6/2007
terry : 6/28/2007
terry : 4/20/2005
terry : 4/6/2005
tkritzer : 3/16/2005
carol : 12/10/2004
carol : 12/10/2004
ckniffin : 12/6/2004
carol : 3/17/2004
alopez : 10/9/2003
terry : 10/9/2003
alopez : 2/11/2002
alopez : 2/8/2002
alopez : 2/8/2002
carol : 11/17/1999
carol : 11/17/1999
mgross : 6/18/1999
carol : 12/8/1998
terry : 12/2/1998
carol : 5/8/1998
terry : 4/29/1998
terry : 11/10/1997
mark : 9/23/1997
terry : 9/22/1997
terry : 1/23/1997
terry : 1/21/1997
mark : 6/24/1996
terry : 6/12/1996
terry : 10/27/1994
jason : 7/26/1994
warfield : 4/15/1994
mimadm : 4/8/1994
carol : 3/24/1993
carol : 2/10/1993