Bookshelf » GeneReviews » Mitochondrial Neurogastrointestinal Encephalopathy Disease
 
gene
GeneReviews
PagonRoberta A
BirdThomas C
DolanCynthia R
SmithRichard JH
StephensKaren
University of Washington, Seattle2009
geneticspublic health

GeneTests Home Page About GeneTests Search GeneReviews on the GeneTests web site Laboratory Directory Clinic Directory Educational Materials Illustrated Glossary

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Genetics clinics are a source of information for individuals and families regarding the natural history, treatment, mode of inheritance, and genetic risks to other family members as well as information about available consumer-oriented resources. See the GeneTests Clinic Directory.

Support groups have been established for individuals and families to provide information, support, and contact with other affected individuals. The Resources section may include disease-specific and/or umbrella support organizations.

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mitochondrial Neurogastrointestinal Encephalopathy Disease
[MNGIE Syndrome, Myoneurogastrointestinal Encephalopathy Syndrome, Thymidine Phosphorylase Deficiency, Mitochondrial Neurogastrointestinal Encephalopathy Syndrome]

John M Shoffner, MD
Neurology, Biochemical Genetics, Molecular Genetics
Horizon Molecular Medicine, Georgia State University
22042005mngie
Initial Posting: April 22, 2005.

*

*

*

Summary

Disease characteristics. Mitochondrial neurogastrointestinal encephalopathy (MNGIE) disease is characterized by progressive gastrointestinal dysmotility and cachexia manifesting as early satiety, nausea, dysphagia, gastroesophageal reflux, post-prandial emesis, episodic abdominal pain and/or distention, and diarrhea; ptosis/ophthalmoplegia or ophthalmoparesis; hearing loss; and demyelinating peripheral neuropathy manifesting as paresthesias (tingling, numbness, and pain) and symmetrical and distal weakness more prominently affecting the lower extremities. The order in which manifestations appear is unpredictable. Onset is usually between the first and fifth decades; in about 60% of individuals, symptoms begin before age 20 years.

Diagnosis/testing. The clinical diagnosis of MNGIE disease is based on the presence of severe gastrointestinal dysmotility, cachexia, ptosis, external ophthalmoplegia, sensorimotor neuropathy, asymptomatic leukoencephalopathy as observed on brain MRI, and family history consistent with autosomal recessive inheritance. Direct evidence of MNGIE disease is provided by increase in plasma thymidine concentration greater than 3 µmol/L and increase in plasma deoxyuridine concentration greater than than 5 µmol/L. Thymidine phosphorylase enzyme activity in leukocytes is less than 10% of the control mean. ECGF1, the gene encoding thymidine phosphorylase, is the only gene known to be associated with MNGIE disease. Molecular genetic testing of ECGF1 sequence alterations detects approximately 100% of affected individuals and is available in clinical laboratories.

Management. Management of MNGIE disease is supportive and includes attention to swallowing difficulties and airway protection; dromperidone for nausea and vomiting; celiac plexus block with bupivicaine to reduce pain; bolus feedings, gastrostomy, and parenteral feeding for nutritional support; antibiotics for intestinal bacterial overgrowth; morphine, amitriptyline, gabapentin, and phenytoin for neuropathic symptoms; specialized schooling arrangements; and physical and occupational therapy. Attention to swallowing abnormalities and diverticulosis, respectively, may help prevent aspiration pneumonia and ruptured diverticula. Drugs that interfere with mitochondrial function should be avoided and medications that are primarily metabolized in the liver used with caution.

Genetic counseling. MNGIE disease is inherited in an autosomal recessive manner. The parents of an affected child are obligate heterozygotes and therefore carry one mutant allele; heterozygotes are asymptomatic. Unless an individual with MNGIE disease has offspring with either an affected individual or a carrier, his/her offspring will be obligate heterozygotes for a disease-causing mutation in the ECGF1 gene. Carrier detection for at-risk family members is available on a clinical basis once the ECGF1 mutations have been identified in the proband. Prenatal testing may be available through laboratories offering custom prenatal testing.

Diagnosis

Clinical Diagnosis

The diagnosis of MNGIE (Mitochondrial NeuroGastroIntestinal Encephalopathy) disease is based on the presence of the following clinical findings [Hirano et al 1994, Nishino et al 1999, Nishino et al 2000]:

  • Severe gastrointestinal (GI) dysmotility

  • Cachexia

  • Ptosis

  • External ophthalmoplegia

  • Sensorimotor neuropathy (usually mixed axonal and demyelinating)

  • Asymptomatic leukoencephalopathy manifest as diffusely abnormal brain white matter (increased Flair or T2-weighted signal) on brain MRI. Relative sparing of the corpus callosum is reported in some individuals [Vissing et al 2002, Hirano et al 2004].


    Note: In the absence of leukoencephalopathy, MNGIE disease is very unlikely.

  • Family history consistent with autosomal recessive inheritance.

Note: Although magnetic resonance spectroscopy (MRS) can show increases in lactate within the white matter, it is not a reliable diagnostic test.

Testing

Direct evidence of MNGIE disease is provided by one of the following:

  • Increase in plasma thymidine concentration greater than 3 µmol/L and increase in plasma deoxyuridine concentration greater than 5 µmol/L are sufficient to make the diagnosis of MNGIE disease [Marti et al 2004].

  • Thymidine phosphorylase enzyme (E.C.2.4.2.4) activity in leukocytes is less than 10% of the control mean [Nishino et al 1999].


    Note: Although unaffected, heterozygotes display about 30% to 35% residual thymidine phosphorylase activity.

For laboratories offering biochemical testing, see graphic element.

Indirect evidence of MNGIE disease is provided by evidence of mitochondrial dysfunction manifest by any of the following:

Molecular Genetic Testing

GeneReviews designates a molecular genetic test as clinically available only if the test is listed in the GeneTests Laboratory Directory by either a US CLIA-licensed laboratory or a non-US clinical laboratory. GeneTests does not verify laboratory-submitted information or warrant any aspect of a laboratory's licensure or performance. Clinicians must communicate directly with the laboratories to verify information.—ED.

Gene. ECGF1, the gene encoding thymidine phosphorylase, is the only gene known to be associated with MNGIE disease.

Molecular genetic testing: Clinical uses

Molecular genetic testing: Clinical method

  • Sequence analysis

    • Mutations are detected in genomic DNA by sequencing the ECGF1 exons and flanking regions in 100% of individuals with enzymatically proven MNGIE disease [Nishino et al 1999, Nishino et al 2000]. Affected individuals are either homozygotes or compound heterozygotes for the identified mutations.

    • Splice-site mutations are identified by sequence analysis of genomic DNA. The pathogenicity of splice-site mutations is confirmed by identification of altered splicing in reverse transcriptase (RT) PCR assays.

Table 1 summarizes molecular genetic testing for this disorder.

Table 1. Molecular Genetic Testing Used in Mitochondrial Neurogastrointestinal Encephalopathy Disease

Test MethodMutations DetectedMutation Detection RateTest Availability
Sequence analysisECGF1 missense, microdeletions, insertions, splice-site mutations100% 1 Clinical graphic element

1. Mutation detection rate refers to those individuals with enzymatically proven MNGIE disease.

Interpretation of test results. For issues to consider in interpretation of sequence analysis results, click here.

Testing Strategy for a Proband

  • Detection of elevated plasma concentrations of thymidine and deoxyuridine is sufficient to make the diagnosis of MNGIE disease.

  • Measurement of thymidine phosphorylase enzyme activity in buffy coat samples confirms the diagnosis.

  • Sequencing ECGF1, the gene encoding thymidine phoshorylase, can identify pathogenic mutations for assessment of carrier status.

Clinical Description

Natural History

Gestation and delivery are normal. Although the earliest reported age of onset is five months, onset is usually between the first and fifth decades. In about 60% of individuals, symptoms begin before age 20 years [Nishino et al 2000, Teitelbaum et al 2002]. Prior to the onset of symptoms, many individuals with MNGIE disease are healthy, but usually have a long history of subtle fatigability. The order in which manifestations appear is unpredictable; however, in one review, the first symptoms were gastrointestinal in about 67%, ptosis/ophthalmoplegia in about 21%, hearing loss in about 12%, and neuropathic pain (most commonly in the legs) in about 9% [Teitelbaum et al 2002].

Gastrointestinal dysmotility and cachexia. Progressive GI dysmotility, caused by the combined effects of neuromuscular dysfunction and autonomic dysfunction, occurs in virtually all individuals with MNGIE disease at some point during the course of the illness. Symptoms usually progress slowly over several decades and can affect any part of the GI tract. Gastric and small bowel hypomotility are the most common. Symptoms include early satiety, nausea, dysphagia, gastroesophageal reflux, post-prandial emesis, episodic abdominal pain, episodic abdominal distention, and diarrhea.

Weight loss and cachexia coincide with the onset of GI symptoms. The average weight loss is about 15 kg [Nishino et al 2000]. Affected individuals generally have a thin body habitus and reduced muscle mass. Despite severe GI dysfunction, serum concentrations of micronutrients, vitamins E and B12, and folate are typically normal.

Histopathology. Rectal biopsy can show eosinophilic cytoplasmic inclusions, representing abnormal mitochondria, in the submucosal ganglion cells [Perez-Atayde et al 1998]. Duodenal pathology can demonstrate focal muscle atrophy or absence with increased nerve numbers, serosal granulomas, and focal loss of Auerbach's plexus with fibrosis [Teitelbaum et al 2002].

Eye findings. Ptosis and ophthalmoplegia (weakness of the extraocular muscles) or ophthalmoparesis (lack of function of the extraocular muscles) are common findings. Because of the absence of symptoms like diplopia, individuals with MNGIE disease are not usually aware of the eye movement defect. Instead, the abnormalities are usually first noted by a health care provider. Retinitis pigmentosa can also be present.

Sensorimotor neuropathy. All individuals with MNGIE disease have peripheral neuropathy. The neuropathy is demyelinating in all cases and about half also have axonal neuropathy. In some, the first symptoms are paresthesias and weakness. Paresthesias occur in a stocking-glove distribution and may be described as tingling, numbness, or even pain. The weakness is usually symmetrical and distal. Proximal weakness is less common. Lower extremities are more prominently affected than upper extremities. Unilateral or bilateral foot drop, as well as clawed hands, may occur. The severity of the neuropathic symptoms often fluctuates during the early stages of the disease.

The segmental demyelination is hypothesized to be caused by the uneven distribution of mtDNA abnormalities (depletion, point mutations, deletions, duplication) along the nerve. Areas with the highest concentration of these mutations may be predisposed to demyelination.

Electrodiagnostic features can include decreased motor and sensory nerve conduction velocities, prolonged F-wave latency, and partial conduction block. Myopathic changes are common.

Histologically, demyelination and remyelination (onion bulb formation) are observed. Loss of large myelinated fibers is common.

Leukoencephalopathy. The leukoencephalopathy is usually asymptomatic. Spasticity is not present. Although mental retardation is described in some individuals, dementia can be a rare late feature of the disease.

Other. Other highly variable manifestations:

  • Active hepatic cirrhosis with increased liver enzymes and macrovesicular steatosis

  • Anemia

  • Early-onset sensorineural hearing loss involving either the cochlea or eighth cranial nerve.

  • Short stature

  • Autonomic nervous system dysfunction (usually orthostatic hypotension)

  • Bladder dysfunction

  • Ventricular hypertrophy and bundle branch block in the absence of cardiac symptoms

  • Significantly increased CSF protein: typically 60 mg/dL to over 100 mg/dL (normal: 15-45 mg/dL)

  • Lactic acidemia (increased serum lactate without a change in the pH) and hyperalaninemia. Lactic acidosis (increased serum lactate associated with a decrease in blood pH) is unusual, but is more likely to occur in the presence of renal or hepatic impairment.

  • Diverticula, which may become infected (diverticulitis) or perforate, causing peritonitis, which may be fatal.

Prognosis. MNGIE disease is a progressive, degenerative disease with a poor prognosis. In the study of Nishino et al (2000), the mean age of death was 37.6 years (range 26-58 years).

Genotype-Phenotype Correlations

No relationship exists between the enzymatic activity of thymidine phosphorylase and the clinical severity of MNGIE disease.

ECGF1 mutation type does not correlate with the severity of the enzyme defect or clinical expression of the disease [Spinazzola et al 2002].

Nomenclature

MNGIE disease was first described as congenital oculo-skeletal myopathy with abnormal muscle and liver mitochondria [Okamura et al 1976]. Other acronyms for MNGIE disease include polyneuropathy, ophthalmoplegia, leukoencephalopathy, and intestinal pseudo-obstruction (POLIP) [Simon et al 1990]; oculogastrointestinal muscular dystrophy (OGIMD) [Ionasescu 1983]; and mitochondrial myopathy with sensorimotor polyneuropathy, ophthalmoplegia, and pseudo-obstruction (MEPOP) [Rowland 1992].

Prevalence

MNGIE disease is rare. The prevalence is unknown. Fewer than 70 individuals with features consistent with MNGIE disease have been reported since it was first described [Okamura et al 1976]. No ethnic predilection for MNGIE disease has been observed; it occurs in individuals of mixed European, Puerto Rican, Ashkenazi Jewish, Iranian Jewish, German-American, Asian, Spanish, and African-American heritage. Parental consanguinity is common, representing nearly half the families in some reports [Nishino et al 1999, Nishino et al 2000].

Differential Diagnosis

For current information on availability of genetic testing for disorders included in this section, see GeneTests Laboratory Directory. —ED.

MNGIE disease has been confused with anorexia nervosa and other classes of GI diseases such as intestinal pseudo-obstruction, inflammatory bowel disease, celiac disease, and irritable bowel disease. Acute abdominal pain in individuals with MNGIE disease has been misdiagnosed as superior mesenteric artery syndrome.

Because of the rapid appearance of neuropathic symptoms over several months in some individuals, chronic inflammatory demyelinating polyneuropathy (CIDP) has been misdiagnosed [Bedlack et al 2004].

Oxidative phosphorylation (OXPHOS) diseases. Because of the cumulative effects on cells of mtDNA depletion and increasing levels of mtDNA deletions and point mutations in MNGIE disease, affected individuals present with clinical and metabolic features of oxidative phosphorylation diseases, which are characterized by GI dysmotility, polyneuropathy, and leukoencephalopathy (see Mitochondrial Disorders Overview). However, when the diagnostic criteria for MNGIE disease are strictly applied, thymidine phosphorylase activity and molecular genetic testing of the ECGF1 gene are found to be normal in these other disorders [Vissing et al 2002, Hirano et al 2004].

Disorders caused by imbalance in the mitochondrial nucleotide pools or by quantitative or qualitative defects in mtDNA:

Leukodystrophy. Various leukodystrophies are distinguished from MNGIE disease by clinical features. These include metachromatic leukodystrophy, X-linked adrenoleukodystrophy, childhood ataxia with central nervous system hypomyelination/vanishing white matter disease, connexin46.6 (GJA12) mutations, PLP1-related disorders, Krabbe disease, Alexander disease, Canavan disease, congenital muscular dystrophy with merosin deficiency (see Congenital Muscular Dystrophy Overview), and Salla disease (see Free Sialic Acid Storage Defects).

Although mutations in GJB1, the gene encoding connexin 32, can be associated with transient white matter defects [Hanemann et al 2003], most individuals present with X-linked Charcot-Marie-Tooth disease (CMTX).

Management

Evaluations at Initial Diagnosis

  • EMG/NCV

  • EKG

  • Ophthalmologic evaluation

  • Audiologic evaluation

  • Developmental assessment

  • Assessment of hepatic function, renal function, plasma amino acids, and serum concentration of lactate and pyruvate

  • GI evaluation, which depends on the symptoms and may include abdominal films, abdominal CT, upper GI contrast radiography, esophagogastroduodenoscopy, sigmoidoscopy, liquid phase scintigraphy, and antroduodenal manometry. Radiologic studies may show hypoperistalsis, gastroparesis, dilated duodenum, and diverticulosis. Small bowel manometry shows reduced amplitude of contractions.

Treatment of Manifestations

Cooperation among multiple specialties including neurology, medical genetics, nutrition, gastroenterology, pain management, psychiatry, and physical/occupational therapy helps with timely detection and treatment of the various aspects of multi-organ dysfunction. Once symptoms appear, treatment is supportive.

Management of GI dysfunction can include:

  • Early attention to swallowing difficulties and airway protection, especially in the most severely affected individuals

  • Trial of dromperidone for nausea and vomiting

  • Celiac plexus block with bupivicaine. This has been successful in reducing pain by interrupting visceral afferent pain sensation and increasing GI motility by inhibiting sympathetic efferent activity to the upper abdominal viscera and much of the small bowel [Teitelbaum et al 2002].

  • Nutritional support including, when necessary, bolus feedings, gastrostomy tube placement, and total parenteral nutrition

  • Antibiotic therapy for intestinal bacterial overgrowth, a complication of dysmotility

  • Complex medication regimens that include morphine, amitriptyline, gabapentin, and phenytoin for relief of neuropathic symptoms, which are difficult to treat

  • Specialized schooling arrangements, typically necessary for children and young adults

  • Physical therapy and occupational therapy to help preserve mobility. Activity as tolerated should be encouraged.

Prevention of Primary Manifestations

  • No specific therapy for MNGIE disease is currently available.

  • Supplements like coenzyme Q10, vitamin K, vitamin C, riboflavin, niacin, and other compounds have no proven efficacy and do not change the natural history of the disease [Bresolin et al 1990; Matthews et al 1993; Shoffner, personal observation].

  • Although plasma concentration of thymidine can be reduced by hemodialysis, the plasma concentration becomes elevated again in about three hours [Spinazzola et al 2002].

Prevention of Secondary Complications

  • Establishing the correct diagnosis of MNGIE disease may help avoid unnecessary exploratory abdominal surgeries, risks associated with anesthesia, and inappropriate therapies.

  • The approximately 20% of individuals with MNGIE disease who have hepatopathy may be at increased risk for worsening hepatic dysfunction caused by medications metabolized by the liver and as a result of total parenteral nutrition. Therefore, medications that are primarily metabolized in the liver should be used with caution.

  • Attention to swallowing abnormalities associated with oropharyngeal muscle dysfunction may help decrease the risk for aspiration pneumonia.

  • Early attention to diverticulosis can help prevent complications such as ruptured diverticula and fatal peritonitis.

Agents/Circumstances to Avoid

  • Drugs that interfere with mitochondrial function such as valproate, phenytoin, chloramphenicol, tetracycline, and certain anti-psychotic medications

Therapies Under Investigation

Possible future treatments include reducing plasma thymidine concentration by reducing renal reabsorption of thymidine (i.e.,blocking the Na+/thymidine transporter). Normalization of intracellular thymidine concentrations could reduce the rate of the mtDNA damage, which progressively increases in an individual over time.

Search ClinicalTrials.gov for access to information on clinical studies for a wide range of diseases and conditions.

Genetic Counseling

Genetic counseling is the process of providing individuals and families with information on the nature, inheritance, and implications of genetic disorders to help them make informed medical and personal decisions. The following section deals with genetic risk assessment and the use of family history and genetic testing to clarify genetic status for family members. This section is not meant to address all personal, cultural, or ethical issues that individuals may face or to substitute for consultation with a genetics professional. To find a genetics or prenatal diagnosis clinic, see the GeneTests Clinic Directory.

Mode of Inheritance

MNGIE disease is inherited in an autosomal recessive manner.

Risk to Family Members

Parents of a proband

  • The parents of an affected child are obligate heterozygotes and therefore carry one mutant allele.

  • Heterozygotes (carriers) are asymptomatic.

Sibs of a proband

  • At conception, the sibs of an affected individual have a 25% chance of being affected, a 50% chance of being asymptomatic carriers, and a 25% chance of being unaffected and not carriers.

  • Once an at-risk sib is known to be unaffected, the risk of his/her being a carrier is 2/3.

  • Heterozygotes (carriers) are asymptomatic.

Offspring of a proband. Unless an individual with MNGIE disease has offspring with either an affected individual or a carrier, his/her offspring will be obligate heterozygotes (carriers) for a disease-causing mutation in the ECGF1 gene.

Other family members of a proband. Sibs of the proband's parents are at 50% risk of being carriers.

Carrier Detection

Carrier detection for at-risk family members is available on a clinical basis once the ECGF1 mutations have been identified in the proband.

Related Genetic Counseling Issues

Family planning. The optimal time for determination of genetic risk and clarification of carrier status is before pregnancy.

DNA banking. DNA banking is the storage of DNA (typically extracted from white blood cells) for possible future use. Because it is likely that testing methodology and our understanding of genes, mutations, and diseases will improve in the future, consideration should be given to banking DNA of affected individuals. DNA banking is particularly relevant in situations in which the sensitivity of currently available testing is less than 100%. See DNA Banking for a list of laboratories offering this service.

Prenatal Testing

No laboratories offering molecular genetic testing for prenatal diagnosis of MNGIE disease are listed in the GeneTests Laboratory Directory. However, prenatal testing may be available for families in which the disease-causing mutations have been identified in an affected family member in a research or clinical laboratory. For laboratories offering custom prenatal testing, see graphic element.

Molecular Genetics

Information in the Molecular Genetics tables is current as of initial posting or most recent update. —ED.

Table A. Molecular Genetics of Mitochondrial Neurogastrointestinal Encephalopathy Disease

Gene SymbolChromosomal LocusProtein Name
ECGF122q13.3-qterThymidine phosphorylase

Data are compiled from the following standard references: Gene symbol from HUGO; chromosomal locus, locus name, critical region, complementation group from OMIM; protein name from Swiss-Prot.

Table B. OMIM Entries for Mitochondrial Neurogastrointestinal Encephalopathy Disease

131222 ENDOTHELIAL CELL GROWTH FACTOR, PLATELET-DERIVED; ECGF1
603041 MITOCHONDRIAL NEUROGASTROINTESTINAL ENCEPHALOPATHY SYNDROME; MNGIE

Table C. Genomic Databases for Mitochondrial Neurogastrointestinal Encephalopathy Disease

Gene SymbolEntrez GeneHGMD
ECGF11890 (MIM No. 131222)ECGF1

For a description of the genomic databases listed, click here.

Molecular Genetic Pathogenesis

MNGIE disease results from the mutagenic effect of thymidine phosphorylase deficiency on mitochondrial DNA (mtDNA). Thymidine phosphorylase deficiency results from mutations in the nuclear gene ECGF1. The pathologic consequences of thymidine phosphorylase deficiency are thought to be the accumulation of qualitative mtDNA defects (deletions and duplications) and quantitative mtDNA defects (depletion) in various tissues over time. Nuclear DNA damage does not appear to be a factor in the pathogenesis of MNGIE disease.

During mtDNA synthesis, polymerase gamma is unable to distinguish between dTTP and dUTP. Normally, incorporation of thymidine over uracil into replicating mtDNA is accomplished by maintaining a high dTTP/dUTP ratio (>105) in the mitochondria [Goulian et al 1980, Bestwick et al 1982]. However, in MNGIE disease, imbalances in these mitochondrial deoxynucleoside 5'-triphosphate (dNTP) pools caused by increases in deoxythymidine and deoxyuridine result in increased uracil incorporation into the mtDNA, producing mtDNA instability [Nishigaki et al 2003]. This preferential damage to mtDNA over time appears to be caused by several factors:

  • The mitochondrial dNTP pool is sequestered within the mitochondria.

  • mtDNA is more dependent on thymidine salvage than nuclear DNA, which depends primarily on de novo thymidine synthesis.

  • mtDNA has a limited capability to repair damage as compared to nuclear DNA.

Since mtDNA continues to replicate throughout an individual's life, various tissues throughout the body develop abnormalities over time as a result of progressive oxidative phosphorylation (OXPHOS) impairment. Accumulation of mtDNA mutations can be observed in fibroblasts of individuals with MNGIE disease as well in HeLa cells cultured in the presence of increased thymidine [Nishigaki et al 2003, Song et al 2003]. mtDNA depletion and mtDNA deletions are present in most individuals with MNGIE disease, but not all [Hirano et al 1994, Debouverie et al 1997, Hamano et al 1997].

Thymidine-deficient mice (TP -/-) appear normal and do not show features of MNGIE disease [Haraguchi et al 2002]. Since mice can use uridine phosphorylase to clear thymidine, deficiency in both thymidine phosphorylase and uridine phosphorylase are required to affect nucleoside metabolism. Mice that are double mutants for these two enzymes produce increased T2-weighted signal on MRI in the white matter. Muscle is normal and no mtDNA mutations are observed.

Normal allelic variants: The gene contains ten exons spanning more than 4.3 kb [Hagiwara et al 1991].

Table 2. Normal Allelic Variants Identified in MNGIE Disease

ExonMutationCodonAmino Acid ChangeReference 1
Exon 7G3336ACodon 277Synonymous
Exon 8T3570CCodon 322Synonymous Hagiwara et al 1991
Exon 8T3576CCodon 324SynonymousNishino et al 1999
Exon 8A3673GCodon 357Thr→AlaNishino et al 1999
Exon 9T3992ACodon 428SynonymousKocaefe et al 2003
Exon 10C4191TCodon 467Synonymous
Exon 10C4202TCodon 471Ser→LeuHagiwara et al 1991
Exon 10C4222TCodon 478Synonymous

1. Polymorphisms in exons are from the Single Nucleotide Polymorphism database, NCBI, geneID: 1890 except those designated with references.

Pathologic allelic variants: The nucleotide positions listed in the genomic DNA are according to Hagiwara et al (1991). No large deletions involving this gene have been described.

Table 3. Missense and Nonsense Mutations Identified in MNGIE Disease

ExonMutationCodonAmino Acid ChangeReference
Exon 2A432CCodon 43Lys→ThrHirano et al 2004
Exon 2G436ACodon 44Arg→GlnGamez et al 2002
Exon 2C466GCodon 54Ile→MetKocaefe et al 2003
Exon 3G721CCodon 87Glu→AspLabauge et al 2002
Exon 4G1419ACodon 145Gly→ArgNishino et al 1999
Exon 4G1443ACodon 153Gly→SerNishino et al 1999
Exon 4A1453GCodon 156Asp→GlyHirano et al 2004
Exon 4T1464CCodon 160Ser→ProNishino et al 2000
Exon 5T2306CCodon 177Leu→ProHirano et al 2004
Exon 5T2294GCodon 173Met→ArgNishino et al 2000
Exon 6A2744GCodon 222Lys→ArgNishino et al 1999
Exon 6A2839CCodon 254Thr→ProHirano et al 2004
Exon 7A3371CCodon 289Glu→AlaNishino et al 1999
Exon 7G3370ACodon 289Glu→LysNishino et al 2000
Exon 8T3542CCodon 313Leu→ProHirano et al 2004
Exon 8T3716CCodon 371Leu→ProKocaefe et al 2003
Exon 9G3990ACodon 428Gly→SerHirano et al 2004
Exon 10G4183ACodon 465Ala→ThrKocaefe et al 2003
Exon 10G4101ACodon 437Trp→TermWeiss et al 2004

Table 4. Mutations Affecting Exon Splicing in MNGIE Disease

IntronMutationSplice SiteReference
Intron 1G294CAcceptor siteSzigeti et al 2004
Intron 2G675CAcceptor siteHirano et al 2004
Intron 4T1504CDonor siteNishino et al 1999
Intron 7G3532AAcceptor siteKocaefe et al 2003
Intron 8G3867AAcceptor siteNishino et al 2000
Intron 8G3867CAcceptor siteNishino et al 1999
Intron 8T3765ADonor siteKocaefe et al 2003
Exon 9T4007ADonor siteKocaefe et al 2003
Intron 9G4090AAcceptor siteNishino et al 1999

Table 5. Small Exon Insertions in MNGIE Disease

ExonMutationReference
Exon 93919T insertionHirano et al 2004
Exon 104009G insertionWeiss et al 2004
Exon 104196C insertionNishino et al 1999

Table 6. Small Deletions in MNGIE Disease

Exon/IntronNucleotide Positions of MutationDeletionReference
Exon 2355-3662-base pair deletion (CT)Nishino et al 2000
Exon 627991-base pair deletion (C)Labauge et al 2002
Intron 73527-35304-base pair deletion (CCGC)Nishino et al 1999
Exon 93895-39006-base pair deletion (CGCTGG)Nishino et al 1999

Normal gene product: Thymidine phosphorylase is a homodimer that catalyzes the conversion of thymidine to thymine and 2-deoxy-D-ribose 1-phosphate [Brown & Bicknell 1998]. The forward reaction (thymidine to thymine) is important to nucleoside catabolism. Although the reverse reaction is possible (thymidine to thymidine triphosphate), only the forward reaction appears important physiologically. Thymidine phosphorylase is expressed in the GI system, brain, peripheral nerves, autonomic nerves, spleen, bladder, and lungs and is not expressed in muscle, kidney, gallbladder, aorta, or fat [Yoshimura et al 1990].

Thymidine phosphorylase was originally mistakenly identified as a "growth factor" abundant in platelets; therefore, it was named "platelet-derived endothelial cell growth factor " (PD-ECGF or ECGF1). The misconception that thymidine phosphorylase (TP) is a growth factor is based on [3H]-labeled thymidine incorporation assays [Miyazono et al 1987]. Purified "ECGF" was added to cell culture medium 18 hours prior to addition of [3H]-thymidine, which was rapidly incorporated by cultured endothelial cells. This result was misinterpreted as stimulation of mitosis. In reality, the addition of TP degraded thymidine in the culture medium, and subsequently the thymidine-starved endothelial cells rapidly incorporated the [3H]-thymidine. TP may be angiogenic indirectly because ribose liberated from the degradation of thymidine may stimulate cell division and migration [Brown & Bicknell 1998]. In addition to its function in angiogenesis, it also limits glial cell proliferation.

Abnormal gene product: See Molecular Genetic Pathogenesis.

Resources

GeneReviews provides information about selected national organizations and resources for the benefit of the reader. GeneReviews is not responsible for information provided by other organizations. Information that appears in the Resources section of a GeneReview is current as of initial posting or most recent update of the GeneReview. Search GeneTests for this disorder and select graphic element for the most up-to-date Resources information.—ED.

United Mitochondrial Disease Foundation
8085 Saltsburg Road, Suite 201
Pittsburg, PA 15239
Phone: 412-793-8077
Fax: 412-793-6477
Email: info@umdf.org
www.umdf.org

Muscular Dystrophy Association (MDA)
3300 East Sunrise Drive
Tucson, AZ 85718-3208
Phone: 800-572-1717; 520-529-2000
Fax: 520-529-5300
Email: mda@mdausa.org
www.mdausa.org

Muscular Dystrophy Campaign
7-11 Prescott Place
London SW4 6BS, United Kingdom
Phone: (+44) 0 020 7720 8055
Fax: (+44) 0 020 7498 0670
Email: info@muscular-dystrophy.org
www.muscular-dystrophy.org

References

Medical Genetic Searches: A specialized PubMed search designed for clinicians that is located on the PubMed Clinical Queries page. graphic element

Literature Cited

Bedlack RS, Vu T, Hammans S, Sparr SA, Myers B, Morgenlander J, Hirano M. MNGIE neuropathy: five cases mimicking chronic inflammatory demyelinating polyneuropathy. Muscle Nerve. 2004; 29: 3648. [PubMed]
Bestwick RK, Moffett GL, Mathews CK. Selective expansion of mitochondrial nucleoside triphosphate pools in antimetabolite-treated HeLa cells. J Biol Chem. 1982; 257: 93004. [PubMed]
Bresolin N, Doriguzzi C, Ponzetto C, Angelini C, Moroni I, Castelli E, Cossutta E, Binda A, Gallanti A, Gabellini S. Ubidecarenone in the treatment of mitochondrial myopathies: a multi-center double-blind trial. J Neurol Sci. 1990; 100: 708. [PubMed]
Brown NS, Bicknell R. Thymidine phosphorylase, 2-deoxy-D-ribose and angiogenesis. Biochem J 334 (Pt. 1998; 1): 18. [PubMed]
Debouverie M, Wagner M, Ducrocq X, Grignon Y, Mousson B, Weber M. [MNGIE syndrome in 2 siblings] Rev Neurol (Paris). 1997; 153: 54753. [PubMed]
Fairbanks LD, Marinaki AM, Carrey EA, Hammans SR, Duley JA. Deoxyuridine accumulation in urine in thymidine phosphorylase deficiency (MNGIE). J Inherit Metab Dis. 2002; 25: 6034. [PubMed]
Gamez J, Ferreiro C, Accarino ML, Guarner L, Tadesse S, Marti RA, Andreu AL, Raguer N, Cervera C, Hirano M. Phenotypic variability in a Spanish family with MNGIE. Neurology. 2002; 59: 4557. [PubMed]
Goulian M, Bleile B, Tseng BY. The effect of methotrexate on levels of dUTP in animal cells. J Biol Chem. 1980; 255: 106307. [PubMed]
Hagiwara K, Stenman G, Honda H, Sahlin P, Andersson A, Miyazono K, Heldin CH, Ishikawa F, Takaku F. Organization and chromosomal localization of the human platelet-derived endothelial cell growth factor gene. Mol Cell Biol. 1991; 11: 212532. [PubMed]
Hamano H, Ohta T, Takekawa Y, Kouda K, Shinohara Y. [Mitochondrial neurogastrointestinal encephalomyopathy presenting with protein-losing gastroenteropathy and serum copper deficiency: a case report] Rinsho Shinkeigaku. 1997; 37: 91722. [PubMed]
Hanemann CO, Bergmann C, Senderek J, Zerres K, Sperfeld AD. Transient, recurrent, white matter lesions in X-linked Charcot-Marie-Tooth disease with novel connexin 32 mutation. Arch Neurol. 2003; 60: 6059. [PubMed]
Haraguchi M, Tsujimoto H, Fukushima M, Higuchi I, Kuribayashi H, Utsumi H, Nakayama A, Hashizume Y, Hirato J, Yoshida H, Hara H, Hamano S, Kawaguchi H, Furukawa T, Miyazono K, Ishikawa F, Toyoshima H, Kaname T, Komatsu M, Chen ZS, Gotanda T, Tachiwada T, Sumizawa T, Miyadera K, Osame M, Yoshida H, Noda T, Yamada Y, Akiyama S. Targeted deletion of both thymidine phosphorylase and uridine phosphorylase and consequent disorders in mice. Mol Cell Biol. 2002; 22: 521221. [PubMed]
Hirano M, Nishigaki Y, Marti R. Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE): a disease of two genomes. Neurologist. 2004; 10: 817. [PubMed]
Hirano M, Silvestri G, Blake DM, Lombes A, Minetti C, Bonilla E, Hays AP, Lovelace RE, Butler I, Bertorini TE. Mitochondrial neurogastrointestinal encephalomyopathy (MNGIE): clinical, biochemical, and genetic features of an autosomal recessive mitochondrial disorder. Neurology. 1994; 44: 7217. [PubMed]
Holt IJ, Harding AE, Morgan-Hughes JA. Deletions of muscle mitochondrial DNA in patients with mitochondrial myopathies. Nature. 1988; 331: 7179. [PubMed]
Kaukonen J, Juselius JK, Tiranti V, Kyttala A, Zeviani M, Comi GP, Keranen S, Peltonen L, Suomalainen A. Role of adenine nucleotide translocator 1 in mtDNA maintenance. Science. 2000; 289: 7825. [PubMed]
Kocaefe YC, Erdem S, Ozguc M, Tan E. Four novel thymidine phosphorylase gene mutations in mitochondrial neurogastrointestinal encephalomyopathy syndrome (MNGIE) patients. Eur J Hum Genet. 2003; 11: 1024. [PubMed]
Labauge P, Durant R, Castelnovo G, Dubois A. MNGIE: diarrhea and leukoencephalopathy. Neurology. 2002; 58: 1862. [PubMed]
Mandel H, Szargel R, Labay V, Elpeleg O, Saada A, Shalata A, Anbinder Y, Berkowitz D, Hartman C, Barak M, Eriksson S, Cohen N. The deoxyguanosine kinase gene is mutated in individuals with depleted hepatocerebral mitochondrial DNA. Nat Genet. 2001; 29: 33741. [PubMed]
Marti R, Nishigaki Y, Hirano M. Elevated plasma deoxyuridine in patients with thymidine phosphorylase deficiency. Biochem Biophys Res Commun. 2003; 303: 148. [PubMed]
Marti R, Nishigaki Y, Vila MR, Hirano M. Alteration of nucleotide metabolism: a new mechanism for mitochondrial disorders. Clin Chem Lab Med. 2003; 41: 84551. [PubMed]
Marti R, Spinazzola A, Tadesse S, Nishino I, Nishigaki Y, Hirano M. Definitive diagnosis of mitochondrial neurogastrointestinal encephalomyopathy by biochemical assays. Clin Chem. 2004; 50: 1204. [PubMed]
Matthews PM, Ford B, Dandurand RJ, Eidelman DH, O'Connor D, Sherwin A, Karpati G, Andermann F, Arnold DL. Coenzyme Q10 with multiple vitamins is generally ineffective in treatment of mitochondrial disease. Neurology. 1993; 43: 88490. [PubMed]
Miyazono K, Okabe T, Urabe A, Takaku F, Heldin CH. Purification and properties of an endothelial cell growth factor from human platelets. J Biol Chem. 1987; 262: 4098103. [PubMed]
Nishigaki Y, Marti R, Copeland WC, Hirano M. Site-specific somatic mitochondrial DNA point mutations in patients with thymidine phosphorylase deficiency. J Clin Invest. 2003; 111: 191321. [PubMed]
Nishino I, Spinazzola A, Hirano M. Thymidine phosphorylase gene mutations in MNGIE, a human mitochondrial disorder. Science. 1999; 283: 68992. [PubMed]
Nishino I, Spinazzola A, Papadimitriou A, Hammans S, Steiner I, Hahn CD, Connolly AM, Verloes A, Guimaraes J, Maillard I, Hamano H, Donati MA, Semrad CE, Russell JA, Andreu AL, Hadjigeorgiou GM, Vu TH, Tadesse S, Nygaard TG, Nonaka I, Hirano I, Bonilla E, Rowland LP, DiMauro S, Hirano M. Mitochondrial neurogastrointestinal encephalomyopathy: an autosomal recessive disorder due to thymidine phosphorylase mutations. Ann Neurol. 2000; 47: 792800. [PubMed]
Okamura K, Santa T, Nagae K, Omae T. Congenital oculoskeletal myopathy with abnormal muscle and liver mitochondria. J Neurol Sci. 1976; 27: 7991. [PubMed]
Perez-Atayde AR, Fox V, Teitelbaum JE, Anthony DA, Fadic R, Kalsner L, Rivkin M, Johns DR, Cox GF. Mitochondrial neurogastrointestinal encephalomyopathy: diagnosis by rectal biopsy. Am J Surg Pathol. 1998; 22: 11417. [PubMed]
Rosenberg MJ, Agarwala R, Bouffard G, Davis J, Fiermonte G, Hilliard MS, Koch T, Kalikin LM, Makalowska I, Morton DH, Petty EM, Weber JL, Palmieri F, Kelley RI, Schaffer AA, Biesecker LG. Mutant deoxynucleotide carrier is associated with congenital microcephaly. Nat Genet. 2002; 32: 1759. [PubMed]
Progressive External Ophthalmoplegia and Ocular Myopathies. In: Vinkens PJ, Bruyn GW, Klawans HL (eds) Handbook of Clinical Neurology. Elsevier, Amsterdam, pp 287-329. 1992
Saada A, Shaag A, Mandel H, Nevo Y, Eriksson S, Elpeleg O. Mutant mitochondrial thymidine kinase in mitochondrial DNA depletion myopathy. Nat Genet. 2001; 29: 3424. [PubMed]
Shoffner JM. Oxidative Phosphorylation Disease. In: Scriver, Beaudet, Valle, Sly, Childs, Kinzler, Vogelstein (eds) The Metabolic and Molecular Bases of Inherited Diseases. Vol II. The McGraw-Hill Companies, New York, pp 2367-2423. 2001 [PubMed]
Song S, Wheeler LJ, Mathews CK. Deoxyribonucleotide pool imbalance stimulates deletions in HeLa cell mitochondrial DNA. J Biol Chem. 2003; 278: 438936. [PubMed]
Spelbrink JN, Li FY, Tiranti V, Nikali K, Yuan QP, Tariq M, Wanrooij S, Garrido N, Comi G, Morandi L, Santoro L, Toscano A, Fabrizi GM, Somer H, Croxen R, Beeson D, Poulton J, Suomalainen A, Jacobs HT, Zeviani M, Larsson C. Human mitochondrial DNA deletions associated with mutations in the gene encoding Twinkle, a phage T7 gene 4-like protein localized in mitochondria. Nat Genet. 2001; 28: 22331. [PubMed]
Spinazzola A, Marti R, Nishino I, Andreu AL, Naini A, Tadesse S, Pela I, Zammarchi E, Donati MA, Oliver JA, Hirano M. Altered thymidine metabolism due to defects of thymidine phosphorylase. J Biol Chem. 2002; 277: 412833. [PubMed]
Szigeti K, Wong LJ, Perng CL, Saifi GM, Eldin K, Adesina AM, Cass DL, Hirano M, Lupski JR, Scaglia F. MNGIE with lack of skeletal muscle involvement and a novel TP splice site mutation. J Med Genet. 2004; 41: 1259. [PubMed]
Teitelbaum JE, Berde CB, Nurko S, Buonomo C, Perez-Atayde AR, Fox VL. Diagnosis and management of MNGIE syndrome in children: case report and review of the literature. J Pediatr Gastroenterol Nutr. 2002; 35: 37783. [PubMed]
Van Goethem G, Dermaut B, Lofgren A, Martin JJ, Van Broeckhoven C. Mutation of POLG is associated with progressive external ophthalmoplegia characterized by mtDNA deletions. Nat Genet. 2001; 28: 2112. [PubMed]
Vissing J, Ravn K, Danielsen ER, Duno M, Wibrand F, Wevers RA, Schwartz M. Multiple mtDNA deletions with features of MNGIE. Neurology. 2002; 59: 9269. [PubMed]
Weiss B, Ben-Zeev B, Hirano M, Brand N, Bujanover Y, Yaruslavski A, Sack J, Anikster Y. Cachexia as the only presenting symptom of mitochondrial neurogastrointestinal encephalomyopathy (MNGIE) without gastrointestinal involvement. J Pediatr Gastroenterol Nutr. 2004; 39: S420.
Yoshimura A, Kuwazuru Y, Furukawa T, Yoshida H, Yamada K, Akiyama S. Purification and tissue distribution of human thymidine phosphorylase; high expression in lymphocytes, reticulocytes and tumors. Biochim Biophys Acta. 1990; 1034: 10713. [PubMed]

Published Statements and Policies Regarding Genetic Testing

No specific guidelines regarding genetic testing for this disorder have been developed.

Chapter Note

Revision History

  • 22 April 2005 (me) Review posted to live Web site

  • 16 September 2004 (jms) Original submission

Next
GeneReviews2009
(navigation arrows) Go to previous chapter Go to next chapter Go to top of this page Go to bottom of this page Go to Table of Contents