Semin Neurol 2014; 34(03): 312-320
DOI: 10.1055/s-0034-1386769
Thieme Medical Publishers 333 Seventh Avenue, New York, NY 10001, USA.

The Leukodystrophies

Hannah B. Gordon
1   Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah
,
Anthea Letsou
1   Department of Human Genetics, University of Utah School of Medicine, Salt Lake City, Utah
,
Joshua L. Bonkowsky
2   Division of Pediatric Neurology, Department of Pediatrics, University of Utah School of Medicine, Salt Lake City, Utah
3   Department of Neurology, University of Utah School of Medicine, Salt Lake City, Utah
› Author Affiliations
Further Information

Address for correspondence

Josh Bonkowsky, MD, PhD
Department of Pediatrics
295 Chipeta Way, Salt Lake City
UT 84108   

Publication History

Publication Date:
05 September 2014 (online)

 

Abstract

Leukodystrophies are a group of genetically determined disorders that affect development or maintenance of central nervous system myelin. Leukodystrophies have a reported incidence of 1 in 7500 live births, but fewer than half of patients receive a specific diagnosis. In this review, the authors discuss types of leukodystrophies: their prevalence, clinical presentation, symptoms, and diagnosis, as well as current and future treatments. Diagnosis is based on a combination of history, exam, radiological, and laboratory findings, including genetic testing. Leukodystrophies can present at any age from infancy to adulthood, with variability in disease progression and clinical presentation, ranging from developmental delay to seizures to spasticity. Although there are few cures, there are significant opportunities for care and improvements in patient well-being. Their high prevalence, combined with rapid advances in imaging, genetics, and potential treatments, makes an understanding of the leukodystrophies necessary for care providers in genetics and neurology.


#

Leukodystrophies are a group of genetically determined disorders that affect development or maintenance of central nervous system (CNS) myelin.[1] [2] [3] [4] Leukodystrophies have a reported incidence of 1 in 7500 live births, but fewer than half of patients receive a specific diagnosis.[5] Diagnosis can be based on a combination of history, expected prevalence, physical and neurologic examination, radiological features, or laboratory findings, including genetic testing.[5] [6] Leukodystrophies can present at any age from infancy to adulthood, with variability in disease progression and clinical presentation ranging from developmental delay to seizures to spasticity. Although there are few cures, there are significant opportunities for care and improvements in patient well-being. Their high prevalence, combined with rapid advances in imaging, genetics, and potential treatments, makes an understanding of the leukodystrophies necessary for care providers in genetics, bone marrow transplantation, and neurology.

One confusing issue is the definition of the term “leukodystrophy.” For the purposes of this review, a leukodystrophy will refer to a presumed genetically determined abnormality in myelin development, maintenance, or turnover. In contrast, a “leukoencephalopathy” will be defined more broadly as any abnormality of the white matter, including not only leukodystrophies,[2] but also abnormalities of the white matter, such as caused by trauma, toxicity, or insult, for example, periventricular leukomalacia secondary to premature birth or white matter abnormalities caused by chemotherapy or meningitis.

Myelin Development

Myelin development begins during fetal life and continues through adulthood. It involves a complex developmental orchestration of genes, proteins, and different cell types. Production of myelin requires oligodendrocytes, the glial cells that produce myelin and the myelin sheath, and interactions with other cell types, particularly neurons. Myelin is necessary for normal action potential propagation and nervous system function, maintenance of axonal health, and nutritional support of axons.[7]

Myelin is produced by oligodendrocytes (also known as oligodendroglia). Oligodendrocytes are generated during fetal and postnatal life by oligodendrocyte precursor cells (OPCs). Oligodendrocyte precursor cells can persist into adult life,[8] and there is evidence that new oligodendrocytes formed during adult life generate new myelin.[9] Each oligodendrocyte extends many myelin sheet-forming processes that envelope axonal projections. Different processes from a single oligodendrocyte typically envelope different axons.[10] Thus, on any axon, different oligodendrocytes can be contributing to neighboring stretches of myelin. Myelin is composed of proteins (30%) and lipids (70%), and its production is highly energy-dependent. The most abundant protein components are myelin basic protein (MBP) and proteolipid protein (PLP). The lipid components of myelin are cholesterol, phospholipids, and glycolipids; glycosphingolipids, particularly galactocerebrosides, are the predominant glycolipids.[11] [12]

In addition to the oligodendrocyte-specific determinants of myelin formation, neuron-oligodendrocyte interactions are also necessary for normal myelination.[13] Oligodendrocytes must migrate to the correct locations in the brain to effect axonal ensheathment by myelin,[14] and the extent of myelination is affected by neuronal activity.[15]

During brain development, myelination proceeds in an inside–outside and caudal-to-rostral fashion, with myelination of deeper structures occurring first. Myelination begins by the fourth month of gestation. Essentially all areas of the brain are myelinated by the second year of life, although the total myelin content of the brain does not peak until the third decade of life.[16]

It is apparent, then, that a large number of insults or processes, during development or in adulthood, could affect myelin development or the maintenance of myelin, and hence lead to a leukodystrophy. The biological complexity of myelin development requires both neurons and glia, and requires their interactions. Therefore, problems with any of these three could produce a leukodystrophy. In addition, leukodystrophies can arise from defects in intrinsic/biochemical pathways that are common to all cells, but that are magnified in glia due to their high energy requirements and extensive membrane turnover. For example, mutations in the intrinsic myelin protein PLP1 lead to Pelizaeus-Merzbacher disease, and vanishing white matter disease (VWMD) is caused by mutations in the ubiquitously expressed translation initiation factor subunits EIF2B1–5 that are required in all cells for protein production.

Despite improvements in our fundamental understanding of myelin development, there remain numerous confusing aspects to the pathogenesis of many of the leukodystrophies. Why can VWMD be asymptomatic until initiated by a sudden precipitant?[17] Why can the same mutation in X-linked adrenoleukodystrophy be asymptomatic, or cause an adult-onset neuropathy, or lead to a fatal childhood cerebral inflammatory demyelination?[18] Research into these topics will lead to improved treatment for patients and significant insights into myelin biology.


#

Categories and Diseases

Significant confounders for discussing the different leukodystrophies are several. First, there is disagreement over the exact definition of a leukodystrophy.[5] [19] Second, only about half of all causes of leukodystrophies are known.[5] [20] Finally, limited understanding of the pathogenesis of the leukodystrophies makes classification difficult.

Currently, leukodystrophies appear to fall into three broad categories ([Table 1]): (1) hypomyelination, in which there is absent or diminished myelin production; (2) dysmyelination, in which there is abnormal myelin development; and (3) demyelination, in which there is loss and/or destruction of previously established myelin. Although widely accepted (or perhaps tacitly assumed) that demyelinating inflammatory diseases such as multiple sclerosis or neuromyelitis optica constitute a different diagnostic category than demyelinating leukodystrophies,[21] it will be interesting in the next decade to see if shared common elements of pathogenesis are discovered.[22] This being said, trials of immune modulatory therapy for demyelinating leukodystrophies have not shown efficacy.[23]

Table 1

Categories, disease, and gene name abbreviations of representative leukodystrophies

Category

Gene

Diagnosis

MRI Features

Hypomyelinating

Pelizaeus-Merzbacher disease

PLP1

G

Pelizaeus-Merzbacher-like disease

GJA12

G

4H syndrome

POLR3A, POLR3B

G

Cerebellar atrophy

Salla disease/sialuria

SLC17A5

Urine sialic acid

Fucosidosis

FUCA1

Urine oligosaccharides, LLE

HABC/Other

TUBB4A

G

Atrophy of basal ganglia, cerebellum

Dysmyelination/demyelination[a]

Metachromatic leukodystrophy

ARSA

Urine sulfatides, LLE

Frontal onset, symmetric

MLD-like

PSAP

Urine sulfatides, G

frontal onset, symmetric

SUMF1

Urine mucopolysaccharides

Frontal onset, symmetric

Krabbe (Globoid cell leukodystrophy)

GALC1

LLE

Optic nerve enlargement, DGM

Canavan disease

ASPA

Urine NAA, G

Macrocephaly, increased NAA on MRS

Alexander disease

GFAP

G

Frontal onset, megalencephaly, contrast enhancement, BG, T, BS

Aicardi-Goutieres syndrome

TREX1, RNASEH2A, RNASEH2B, RNASEH2C, SAMHD1, ADAR

CSF pleocytosis, CSF IFN-α, G

Calcifications, temporal cysts

Megalencephalic leukodystrophy with cysts

MLC1, HEPACAM

G

Megalencephaly, cysts

X-linked adrenoleukodystrophy

ABCD1

Plasma VLCFA

Contrast Enhancement, parieto-occipital onset

Mitochondrial diseases

various

Lactate (serum, CSF), G

BG, BS, DGM

Vanishing white matter disease (VWMD)

EIF2B, subunits 1–5

G

Stranding in white matter, white matter isointense with CSF

VWMD-like disease

LMNB1

G

Stranding in white matter, white matter isointense with CSF

Abbreviations: BG, basal ganglia involvement; BS, brainstem involvement; CSF, cerebrospinal fluid; DGM, involvement of deep gray matter; G, gene test; LLE, leukocyte lysosomal enzymes; MRS, magnetic resonance spectroscopy; T, thalamus involvement; VLCFA, serum very-long chain fatty acids.


Note. “Diagnosis” indicates the primary initial method for diagnosis; “MRI features” indicate any particularly unique and/or distinguishing features observed on MRI associated with the disease.


a Dysmyelination and demyelination disorders are included together, as many of these diseases may have both characteristics at different points in their natural history.


However, certain leukodystrophies do not fit clearly into the above categories. For example, Rett syndrome and Batten disease can have white matter changes and clinical features that would initially suggest a leukodystrophy before subsequent testing leads to their ultimate diagnosis. Or some patients have syndromic dysmorphic physical features, and are found to have chromosome microdeletions or microduplications, such as in the 18q- syndrome[24]; yet the primary CNS pathology can consist of leukodystrophy.

An alternative method of classification is based on the primary site of action of the gene product implicated in the leukodystrophy. In this regard, leukodystrophies such as metachromatic leukodystrophy or Krabbe disease can be linked to defects in the lysosome, while X-linked adrenoleukodystrophy can be linked to defects in the peroxisome.[4] A drawback to the site-of-action classification approach is the imperfect correlation between gene function and pathology of the leukodystrophy. For example, X-linked adrenoleukodystrophy is caused by mutation of the ABCD1 gene producing a peroxisomal transporter protein, yet levels and presence of very-long chain fatty acids correlate with neither the development of disease nor the timing of disease onset.


#

Clinical Presentation

There are few reliable clinical indicators of leukodystrophies: The symptoms can be nonspecific, and the age of onset can vary from prenatal to adult. After history and examination, magnetic resonance imaging (MRI) is the key diagnostic test that should prompt evaluation for a specific leukodystrophy diagnosis.

A few leukodystrophies can have classic presentations, and a specific diagnosis should be considered. In a child with acute deterioration of neurologic status, mitochondrial disease or VWMD should be considered. Infants with Pelizaeus-Merzbacher disease often present with nystagmus and/or head titubation. Macrocephaly is a common feature in Canavan disease and Alexander disease, whereas microcephaly is a feature of Aicardi-Goutieres disease. Skin color changes and/or adrenal insufficiency (e.g., Addison disease) in a school-age boy should lead to evaluation for X-linked adrenoleukodystrophy.

In neonates and infants, presenting symptoms can include encephalopathy or developmental delay. In children, teens, and adults, symptoms can be more insidious, ranging from behavioral or psychiatric changes, loss of formerly achieved milestones or deterioration in skills, vision changes, ataxia, or gait problems (often from spasticity). Although not necessarily a presenting symptom, seizures are much more common than previously realized in children with leukodystrophies, and affect up to 49% of children.[5]


#

Incidence and Prevalence

The most recent population-based estimate for leukodystrophies shows an incidence of 1 in 7,663 live births, although this was felt to be an underestimate, with a “true” incidence closer to 1:7,500.[5] A population-based determination for X-linked adrenoleukodystrophy in Norway revealed a birth prevalence of 1.6 in 100,000.[25] In Poland, the birth prevalence of metachromatic leukodystrophy has been estimated at 4.1 in 100,000.[26] To date, there have been few large studies from Africa, the Middle East, or Asia to examine relative incidences or prevalences of the different leukodystrophies, and it will be important and interesting to learn the disease distribution from these regions.

The prevalence of different leukodystrophies has also been difficult to determine; furthermore, in most studies almost half of all patients remain without a definitive diagnosis.[5] [20] In the Utah cohort, the most common diagnoses (in descending order of prevalence) were metachromatic leukodystrophy, Pelizaeus-Merzbacher disease, mitochondrial diseases, X-linked adrenoleukodystrophy, and VWMD.[5] Most studies have not compared incidences of different leukodystrophies. For many years, leukodystrophy-specific incidences were estimated based on case determinations at tertiary referral centers. Given the referral bias in case ascertainment, these estimates must be interpreted with caution. That being said, and using a restrictive definition of leukodystrophy, an incidence of 2 in 100,000 live births was determined from referral centers in Germany.[27] X-linked adrenoleukodystrophy has been estimated at 1 in 100,000 in France, 1.6 in 100,000 in Australasia, and 1 in 30,000 in Japan.[28] [29] [30]


#

Magnetic Resonance Imaging and Neuroimaging

Magnetic resonance imaging is the current gold standard for diagnosis of a leukodystrophy. Although computed tomography (CT) can indicate abnormal signal density in the white matter, MRI is necessary for the determination of signal characteristics that can provide potentially diagnostic information. A large amount of careful work has been performed on the use of MRI and features that can be used for diagnosis.[6] [31] [32] [33] Key features on MRI include the presence of contrast enhancement; the presence of cysts, calcifications, or more subtle structural abnormalities (such as periventricular garlands in Alexander disease)[34]; hypomyelination versus high T2-signal abnormality; and the relative T1-signal hyper- or hypointensity.[6] However, even though MRI algorithms are helpful for diagnosis,[6] they are not able to provide a final diagnosis for many patients because of the need for skilled radiological interpretation and experience and limitations of their sensitivity and specificity.[31]

Complications and limitations to the use of MRI include the appearance and characteristics of myelin change with normal development,[35] and this can be confounding for the interpretation of what is normal or abnormal on MRI. In particular, until the age of 2 years children are normally relatively hypomyelinated compared with adults. Further, there is a range (or bell curve) of normal development of myelin appearance on MRI, and at age 2 years some children will not be fully myelinated. Also, hypomyelination can be a result or complication of developmental delay or other systemic disorders or illnesses.[36] [37] Another complicating feature is that the T1 and T2 signal characteristics of myelin shift between birth and 1 year; in infants, myelin is bright on T1 and dark on T2. By 1 year of life, myelin is dark on T1 and bright on T2. Despite caveats, it is important to broadly recognize leukodystrophies by certain key features on MRI, and to differentiate these from potential leukodystrophy mimics ([Fig. 1]; [Table 1]).

Zoom Image
Fig. 1 Illustrative examples of magnetic resonance images of different leukodystrophies, and of leukodystrophy mimics. Images are axial slices. (A) T1 Image of a hypomyelinating leukodystrophy, TUBB4A. (B) T1 Image with contrast of a demyelinating leukodystrophy, X-linked adrenoleukodystrophy (X-ALD). (C) T2 Fluid-attenuated inversion-recovery (FLAIR) image of metachromatic leukodystrophy (MLD). (D) T2 FLAIR image of vanishing white matter disease (VWMD). (E) T2 FLAIR image of multiple sclerosis (MS). (F) T2 FLAIR image of acute disseminated encephalomyelitis (ADEM).

#

Diagnosis

How does one diagnose a new patient with a leukodystrophy? This continues to be a vexing problem for the clinician. High costs of genetic testing, combined with the likelihood that many causes of leukodystrophy have not yet been discovered, continue to limit the ability to make diagnoses. There is no universally accepted algorithm, and no estimation of costs or sensitivity and specificity for different approaches to diagnosis.

Currently, the most sensible approach appears to combine three elements: (1) testing for treatable diseases, (2) testing based on MRI features, and (3) using relative prevalence to guide testing ([Fig. 2]). We propose an algorithm for leukodystrophy diagnosis that is rational and cost effective, with a tiered approach to testing. There is utility in making an expedient diagnosis by excluding treatable forms of leukodystrophies, curtailing other expensive and lengthy testing, and providing valuable reassurance and prognostic information to the patient and their family.

Zoom Image
Fig. 2 A proposed diagnostic algorithm for testing of leukodystrophies. CK, creatine kinase; MRI, magnetic resonance imaging; PMD, Pelizaeus-Merzbacher disease; VWMD, vanishing white matter disease.

Steady advances in next-generation sequencing technologies are making whole exome sequencing a first-tier option for diagnosis of complex genetic disorders, with reported yields of 25%.[38] Unbiased genome-wide approaches exemplified by whole exome or whole genome sequencing provide the potential for diagnosis of known diseases without step-wise ordering of multiple individual tests. Further, genome-wide sequencing can contribute to the ongoing discovery of novel disease genes.

Next-generation sequencing technologies provide the potential for unbiased diagnosis of known diseases without individual ordering of multiple individual tests, and will contribute to the discovery of novel disease genes. However, continued limitations and problems associated with this technology include (1) substantial cost—up to $15,000 to $20,000—although these numbers are rapidly decreasing; (2) potential for identifying unanticipated disease variants unrelated to the test indication; (3) potential false-negatives due to imperfect exome coverage; and (4) methodological limitations in the interpretation phase if a clear disease-associated variant is not identified. This last problem is significant due to the large number of deleterious gene variants in all humans that could plausibly be related to a phenotype (especially in the CNS), which could yield false-positive associations.

Whole-exome sequencing continues to become more accessible, and may become the method of choice for the diagnosis of leukodystrophies. A rational, cost-effective algorithm for leukodystrophy diagnosis could be developed, with a tiered approach to testing. For example, first-tier testing could include assessment for treatable leukodystrophies: leukocyte lysosomal enzyme testing for metachromatic leukodystrophy, and in males, serum very-long chain fatty acids, for adrenoleukodystrophy. The second tier could be whole-exome sequencing. However, without sufficient data on the sensitivity of whole-exome sequencing for leukodystrophies, and of the false-negative rate for missing gene deletions or duplications (such as for Pelizaeus-Merzbacher or VWMD), this approach is not yet indicated.

Newborn screening for leukodystrophies is being developed in several states.[39] [40] Screening has been piloted in New York for Krabbe disease,[41] [42] but is now also being considered in other states, including California and Minnesota. However, because of variability in disease course, variability in penetrance and disease progression, as well as limited treatment options, there continues to be debate about how widely, and for what diseases, newborn screening should be considered.[43] [44] [45]


#

Treatments

Treatment options for leukodystrophies are disappointingly sparse at the current time. Essentially, the only curative treatment option is bone marrow transplant (BMT)/hematopoietic stem cell transplantation (HSCT). Bone marrow transplant is only available for a subset of leukodystrophies, chiefly X-linked adrenoleukodystrophy, metachromatic leukodystrophy, and Krabbe disease,[46] [47] [48] and is only helpful if transplantation is performed prior to substantial disease progression. Patients who have BMT are at high risk for disease progression; first, because there is a time lag between transplant and effective “rescue,” and second, because the BMT process itself appears to accelerate disease progression in some patients. Bone marrow transplant is not always successful, and carries a substantial mortality risk, approaching 20%. Donor sources for BMT include cord blood, bone marrow, or peripheral blood stem cells.[49] [50]

Enzyme replacement therapy (ERT) is an option for some of the lysosomal disorders (Gaucher disease; Fabry disease; mucopolysaccharidosis types I, II, and VI; and Pompe disease), which can have leukodystrophy as a component.[51] Enzyme replacement therapy has shown some efficacy in animal models in other leukodystrophies.[52] Clinical trials are being pursued, but convincing effectiveness has not yet been shown.

Lorenzo's oil is a treatment that has been proposed to reduce progression of X-linked adrenoleukodystrophy in its cerebral form.[53] A 4:1 mixture of glyceryl trioleate and glyceryl trierucate, Lorenzo's oil can normalize levels of very-long chain fatty acids in plasma, although as previously noted, these do not show a correlation with disease progression, and current published data to do not demonstrate efficacy for inhibiting disease progression or altering outcomes.[54] [55] [56] In the United States, Lorenzo's oil is available only in clinical trials.


#

Care for Patients

Although we have seen that treatment options for leukodystrophies are limited, there are tremendous opportunities for improving the care for patients.[57] This patient-centric approach should lead to discussions between the clinician, patient, and family about what care and treatment is most important and most helpful. As has been demonstrated for other currently incurable genetic conditions (e.g., cystic fibrosis), the strategies of routine symptomatic care can have a profound impact on both the quality and the duration of a patient's life.[58] Nationally and internationally, there are no standardized guidelines for care or treatment of leukodystrophy patients, which results in wide variability in care and costs: > sevenfold difference in costs across children's hospitals in the United States.[59]

Complications of disease, even if the disease itself is not progressive, can lead to progressive disability requiring assistance for mobility and activities of daily living, and even surgery.[60] [61] Leukodystrophy patients can have significant health care requirements and costs, driven largely by in-patient admissions.[5] [62] A relatively small subset of leukodystrophy patients (15%) have much greater health care problems, suggesting that potential interventions for these patients could have a proportionately greater impact. As expected, patients who undergo BMT have much higher costs. However, even taking into account BMT, patients with infections and patients needing mechanical ventilation have higher costs and health care needs.[62]

Building from this analysis of health care utilization, a recent study showed that infection rates in leukodystrophy patients correlate with potentially modifiable risk factors[63]: for example, failure to vaccinate against seasonal flu significantly increases the risk for hospitalization with influenza, and urinary tract infections are associated with the presence of indwelling urinary catheters. Although these are common sense issues, they also outline a path for potential clinical care guidelines that could be implemented at this time to reduce hospitalizations and improve care.


#

Future Directions

The development of new treatments is and should be a primary goal. In this regard, studies in model organisms are leading to a better understanding of the molecular underpinnings of the leukodystrophies, which will provide foundations for new therapeutic options. The best examples include X-linked adrenoleukodystrophy and Alexander disease. Adrenoleukodystrophy has been studied in mouse knockout models[64] [65] [66] [67] although failing to recapitulate CNS degeneration, mouse knockout models have proven useful for the testing of anti-inflammatory therapies.[67] Interestingly, a powerful genetic model of adrenoleukodystrophy is the Drosophila bubblegum mutant, which manifests two key hallmarks of the human disease: age-dependent CNS degeneration and very-long chain fatty acid accumulation.[68] For Alexander disease, both Drosophila and mice harboring the mutated isoform of human GFAP (glial fibrillary acidic protein) as a transgene exhibit Rosenthal fiber formation and neurodegeneration, which has been shown to originate from glia.[69] [70] Additional models are likely to emerge in the near future. It is also important to note that although animal models recapitulate key diagnostic aspects of diseases, they also have the potential to mimic the incomplete penetrance and variable expressivities that characterize human conditions. Thus, animal models will provide a platform to explore the environmental and genetic interactions that make each case presentation so unique.

From the perspective of existing therapies, two significant hurdles to their implementation are the absence of efficient delivery to the CNS, an organ system largely protected by the blood–brain barrier, and the lack of robust methodologies to deliver gene replacement in affected oligodendrocytes or neurons. In addition, significant pathology may happen early in development, in the first year of life, or even before birth. There is significant interest in using stem cells or modified induced pluripotent stem cells (iPSCs) for the leukodystrophies.[71] Although their use has great potential, their practical use still does not seem imminent. Gene therapy is also under active investigation, but with only a few examples and considerable technical challenges, it remains experimental.[72] [73] A limitation of some of the gene therapy approaches is that they use BMT to provide the source of the enzyme, which thus introduces all of the potential morbidities of BMT. Novel drug discovery, or repurposing of known drugs, is another promising avenue of current therapies for several leukodystrophies.[56] [67] [74]

A corollary to the need for improved treatments is the need for faster, definitive diagnosis. Whole exome or whole genome sequencing appears to be on the threshold of becoming the diagnostic method of choice, as it can avoid the “diagnostic odyssey” faced by many patients.[75] [76] Sequencing is as cost effective as a brain MRI, and with improved data analysis will become as rapid. However, there are unanswered questions about the disconnect between the underlying gene or biochemical defect, as in X-linked adrenoleukodystrophy or Krabbe disease, and actually developing the disease.[39] [77] Further, unresolved ethical questions surround the implementation of whole genome sequencing as a screening or general diagnostic tool.[78] [79]


#

Conclusions

The field of knowledge encompassing leukodystrophies is rapidly expanding. Within this decade new methods of diagnosis, such as next-generation sequencing, as well as new therapies will revolutionize patient care. Though exciting, we can still improve our care and improve the lives of patients today by increased attention to preventable and modifiable features of the leukodystrophy diseases.


#
#

Acknowledgments

This work was supported by NIH grant DP2 MH10008, PCORI grant PPRN-1306–04787, and a Vanishing White Matter Foundation grant to JLB; NIH grant R01 NS065474 to AL; a training fellowship from the Howard Hughes Medical Institute and the Little Red Riding Hood Research Foundation grant to HBG.

  • References

  • 1 Bielschowsky M, Henneberg R. Ueber familiaere diffuse Sklerose (Leukodystrophia cerebri progressiva hereditaria). J Psychol Neurol 1928; 36: 131-181
  • 2 Kaye EM. Update on genetic disorders affecting white matter. Pediatr Neurol 2001; 24 (1) 11-24
  • 3 Maria BL, Deidrick KM, Moser H, Naidu S. Leukodystrophies: pathogenesis, diagnosis, strategies, therapies, and future research directions. J Child Neurol 2003; 18 (9) 578-590
  • 4 Kohlschütter A, Bley A, Brockmann K , et al. Leukodystrophies and other genetic metabolic leukoencephalopathies in children and adults. Brain Dev 2010; 32 (2) 82-89
  • 5 Bonkowsky JL, Nelson C, Kingston JL, Filloux FM, Mundorff MB, Srivastava R. The burden of inherited leukodystrophies in children. Neurology 2010; 75 (8) 718-725
  • 6 Schiffmann R, van der Knaap MS. Invited article: an MRI-based approach to the diagnosis of white matter disorders. Neurology 2009; 72 (8) 750-759
  • 7 Nave KA. Myelination and support of axonal integrity by glia. Nature 2010; 468 (7321) 244-252
  • 8 Savas JN, Toyama BH, Xu T, Yates III JR, Hetzer MW. Extremely long-lived nuclear pore proteins in the rat brain. Science 2012; 335 (6071) 942
  • 9 Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 2010; 68 (4) 668-681
  • 10 Baumann N, Pham-Dinh D. Biology of oligodendrocyte and myelin in the mammalian central nervous system. Physiol Rev 2001; 81 (2) 871-927
  • 11 Jahn O, Tenzer S, Werner HB. Myelin proteomics: molecular anatomy of an insulating sheath. Mol Neurobiol 2009; 40 (1) 55-72
  • 12 Aggarwal S, Yurlova L, Simons M. Central nervous system myelin: structure, synthesis and assembly. Trends Cell Biol 2011; 21 (10) 585-593
  • 13 White R, Krämer-Albers EM. Axon-glia interaction and membrane traffic in myelin formation. Front Cell Neurosci 2014; 7: 284
  • 14 de Castro F, Bribián A, Ortega MC. Regulation of oligodendrocyte precursor migration during development, in adulthood and in pathology. Cell Mol Life Sci 2013; 70 (22) 4355-4368
  • 15 Wake H, Lee PR, Fields RD. Control of local protein synthesis and initial events in myelination by action potentials. Science 2011; 333 (6049) 1647-1651
  • 16 Bartzokis G, Beckson M, Lu PH, Nuechterlein KH, Edwards N, Mintz J. Age-related changes in frontal and temporal lobe volumes in men: a magnetic resonance imaging study. Arch Gen Psychiatry 2001; 58 (5) 461-465
  • 17 van der Knaap MS, Pronk JC, Scheper GC. Vanishing white matter disease. Lancet Neurol 2006; 5 (5) 413-423
  • 18 Berger J, Forss-Petter S, Eichler FS. Pathophysiology of X-linked adrenoleukodystrophy. Biochimie 2014; 98: 135-142
  • 19 Vanderver A, Hussey H, Schmidt JL, Pastor W, Hoffman HJ. Relative incidence of inherited white matter disorders in childhood to acquired pediatric demyelinating disorders. Semin Pediatr Neurol 2012; 19 (4) 219-223
  • 20 van der Knaap MS, Breiter SN, Naidu S, Hart AA, Valk J. Defining and categorizing leukoencephalopathies of unknown origin: MR imaging approach. Radiology 1999; 213 (1) 121-133
  • 21 Pfeifenbring S, von Baumgarten L, Schüller U , et al. Biopsy findings of symptomatic cerebral X-linked adrenoleukodystrophy and histological differentiation from multiple sclerosis. Neuropathol Appl Neurobiol 2014; 40 (5) 658-661
  • 22 Baronica KB, Mlinac K, Ozretić D, Vladić A, Bognar SK. Arylsulfatase a gene polymorphisms in relapsing remitting multiple sclerosis: genotype-phenotype correlation and estimation of disease progression. Coll Antropol 2011; 35 (Suppl. 01) 11-16
  • 23 Eichler F, Van Haren K. Immune response in leukodystrophies. Pediatr Neurol 2007; 37 (4) 235-244
  • 24 Feenstra I, Vissers LE, Orsel M , et al. Genotype-phenotype mapping of chromosome 18q deletions by high-resolution array CGH: an update of the phenotypic map. Am J Med Genet A 2007; 143A (16) 1858-1867
  • 25 Horn MA, Retterstøl L, Abdelnoor M, Skjeldal OH, Tallaksen CM. Adrenoleukodystrophy in Norway: high rate of de novo mutations and age-dependent penetrance. Pediatr Neurol 2013; 48 (3) 212-219
  • 26 Ługowska A, Ponińska J, Krajewski P, Broda G, Płoski R. Population carrier rates of pathogenic ARSA gene mutations: is metachromatic leukodystrophy underdiagnosed?. PLoS ONE 2011; 6 (6) e20218
  • 27 Heim P, Claussen M, Hoffmann B , et al. Leukodystrophy incidence in Germany. Am J Med Genet 1997; 71 (4) 475-478
  • 28 Sereni C, Paturneau-Jouas M, Aubourg P, Baumann N, Feingold J. Adrenoleukodystrophy in France: an epidemiological study. Neuroepidemiology 1993; 12 (4) 229-233
  • 29 Kirk EP, Fletcher JM, Sharp P, Carey B, Poulos A. X-linked adrenoleukodystrophy: the Australasian experience. Am J Med Genet 1998; 76 (5) 420-423
  • 30 Takemoto Y, Suzuki Y, Tamakoshi A , et al. Epidemiology of X-linked adrenoleukodystrophy in Japan. J Hum Genet 2002; 47 (11) 590-593
  • 31 Steenweg ME, Vanderver A, Blaser S , et al. Magnetic resonance imaging pattern recognition in hypomyelinating disorders. Brain 2010; 133 (10) 2971-2982
  • 32 La Piana R, Tonduti D, Gordish Dressman H , et al. Brain magnetic resonance imaging (MRI) pattern recognition in Pol III-related leukodystrophies. J Child Neurol 2014; 29 (2) 214-220
  • 33 Yang E, Prabhu SP. Imaging manifestations of the leukodystrophies, inherited disorders of white matter. Radiol Clin North Am 2014; 52 (2) 279-319
  • 34 van der Knaap MS, Ramesh V, Schiffmann R , et al. Alexander disease: ventricular garlands and abnormalities of the medulla and spinal cord. Neurology 2006; 66 (4) 494-498
  • 35 Welker KM, Patton A. Assessment of normal myelination with magnetic resonance imaging. Semin Neurol 2012; 32 (1) 15-28
  • 36 van der Knaap MS, Valk J, Bakker CJ , et al. Myelination as an expression of the functional maturity of the brain. Dev Med Child Neurol 1991; 33 (10) 849-857
  • 37 Pujol J, López-Sala A, Sebastián-Gallés N , et al. Delayed myelination in children with developmental delay detected by volumetric MRI. Neuroimage 2004; 22 (2) 897-903
  • 38 Yang Y, Muzny DM, Reid JG , et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013; 369 (16) 1502-1511
  • 39 Raymond GV, Jones RO, Moser AB. Newborn screening for adrenoleukodystrophy: implications for therapy. Mol Diagn Ther 2007; 11 (6) 381-384
  • 40 Orsini JJ, Martin MM, Showers AL , et al. Lysosomal storage disorder 4+1 multiplex assay for newborn screening using tandem mass spectrometry: application to a small-scale population study for five lysosomal storage disorders. Clin Chim Acta 2012; 413 (15-16) 1270-1273
  • 41 Duffner PK, Caggana M, Orsini JJ , et al. Newborn screening for Krabbe disease: the New York State model. Pediatr Neurol 2009; 40 (4) 245-252 , discussion 253–255
  • 42 Orsini JJ, Morrissey MA, Slavin LN , et al. Implementation of newborn screening for Krabbe disease: population study and cutoff determination. Clin Biochem 2009; 42 (9) 877-884
  • 43 Waggoner DJ, Tan CA. Expanding newborn screening for lysosomal disorders: opportunities and challenges. Dev Disabil Res Rev 2011; 17 (1) 9-14
  • 44 Lantos JD. Dangerous and expensive screening and treatment for rare childhood diseases: the case of Krabbe disease. Dev Disabil Res Rev 2011; 17 (1) 15-18
  • 45 Nakamura K, Hattori K, Endo F. Newborn screening for lysosomal storage disorders. Am J Med Genet C Semin Med Genet 2011; 157C (1) 63-71
  • 46 Boelens JJ. Trends in haematopoietic cell transplantation for inborn errors of metabolism. J Inherit Metab Dis 2006; 29 (2-3) 413-420
  • 47 Sevin C, Aubourg P, Cartier N. Enzyme, cell and gene-based therapies for metachromatic leukodystrophy. J Inherit Metab Dis 2007; 30 (2) 175-183
  • 48 Orchard PJ, Tolar J. Transplant outcomes in leukodystrophies. Semin Hematol 2010; 47 (1) 70-78
  • 49 Escolar ML, Poe MD, Provenzale JM , et al. Transplantation of umbilical-cord blood in babies with infantile Krabbe's disease. N Engl J Med 2005; 352 (20) 2069-2081
  • 50 Martin PL, Carter SL, Kernan NA , et al. Results of the cord blood transplantation study (COBLT): outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with lysosomal and peroxisomal storage diseases. Biol Blood Marrow Transplant 2006; 12 (2) 184-194
  • 51 Valayannopoulos V. Enzyme replacement therapy and substrate reduction therapy in lysosomal storage disorders with neurological expression. Handb Clin Neurol 2013; 113: 1851-1857
  • 52 Stroobants S, Gerlach D, Matthes F , et al. Intracerebroventricular enzyme infusion corrects central nervous system pathology and dysfunction in a mouse model of metachromatic leukodystrophy. Hum Mol Genet 2011; 20 (14) 2760-2769
  • 53 Moser HW, Raymond GV, Lu SE , et al. Follow-up of 89 asymptomatic patients with adrenoleukodystrophy treated with Lorenzo's oil. Arch Neurol 2005; 62 (7) 1073-1080
  • 54 Aubourg P, Adamsbaum C, Lavallard-Rousseau MC , et al. A two-year trial of oleic and erucic acids (“Lorenzo's oil”) as treatment for adrenomyeloneuropathy. N Engl J Med 1993; 329 (11) 745-752
  • 55 Moser HW, Moser AB, Hollandsworth K, Brereton NH, Raymond GV. “Lorenzo's oil” therapy for X-linked adrenoleukodystrophy: rationale and current assessment of efficacy. J Mol Neurosci 2007; 33 (1) 105-113
  • 56 Berger J, Pujol A, Aubourg P, Forss-Petter S. Current and future pharmacological treatment strategies in X-linked adrenoleukodystrophy. Brain Pathol 2010; 20 (4) 845-856
  • 57 Van Haren K, Bonkowsky JL, Bernard G , et al , on behalf of the GLIA Consortium. Consensus statement on preventive and symptomatic care of leukodystrophy patients. Mol Genet Metab 2014; ; In press
  • 58 Cohen-Cymberknoh M, Shoseyov D, Kerem E. Managing cystic fibrosis: strategies that increase life expectancy and improve quality of life. Am J Respir Crit Care Med 2011; 183 (11) 1463-1471
  • 59 Brimley CJ, Lopez J, van Haren K , et al. National variation in costs and mortality for leukodystrophy patients in US children's hospitals. Pediatr Neurol 2013; 49 (3) 156-162 , e1
  • 60 Barrell C. Juvenile metachromatic leukodystrophy: understanding the disease and implications for nursing care. J Pediatr Oncol Nurs 2007; 24 (2) 64-69
  • 61 Holmes L, Cornes MJ, Foldi B, Miller F, Dabney K. Clinical epidemiologic characterization of orthopaedic and neurological manifestations in children with leukodystrophies. J Pediatr Orthop 2011; 31 (5) 587-593
  • 62 Nelson C, Mundorff MB, Korgenski EK, Brimley CJ, Srivastava R, Bonkowsky JL. Determinants of health care use in a population-based leukodystrophy cohort. J Pediatr 2013; 162 (3) 624-628 , e1
  • 63 Anderson HM, Wilkes J, Korgenski EK , et al. Preventable infections in children with leukodystrophy. Ann Clin Transl Neurol 2014; 1 (5) 370-374
  • 64 Lu JF, Lawler AM, Watkins PA , et al. A mouse model for X-linked adrenoleukodystrophy. Proc Natl Acad Sci U S A 1997; 94 (17) 9366-9371
  • 65 Kobayashi T, Shinnoh N, Kondo A, Yamada T. Adrenoleukodystrophy protein-deficient mice represent abnormality of very long chain fatty acid metabolism. Biochem Biophys Res Commun 1997; 232 (3) 631-636
  • 66 Forss-Petter S, Werner H, Berger J , et al. Targeted inactivation of the X-linked adrenoleukodystrophy gene in mice. J Neurosci Res 1997; 50 (5) 829-843
  • 67 Morató L, Galino J, Ruiz M , et al. Pioglitazone halts axonal degeneration in a mouse model of X-linked adrenoleukodystrophy. Brain 2013; 136 (Pt 8) 2432-2443
  • 68 Min KT, Benzer S. Preventing neurodegeneration in the Drosophila mutant bubblegum. Science 1999; 284 (5422) 1985-1988
  • 69 Hagemann TL, Connor JX, Messing A. Alexander disease-associated glial fibrillary acidic protein mutations in mice induce Rosenthal fiber formation and a white matter stress response. J Neurosci 2006; 26 (43) 11162-11173
  • 70 Wang L, Colodner KJ, Feany MB. Protein misfolding and oxidative stress promote glial-mediated neurodegeneration in an Alexander disease model. J Neurosci 2011; 31 (8) 2868-2877
  • 71 Koç ON, Day J, Nieder M, Gerson SL, Lazarus HM, Krivit W. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH). Bone Marrow Transplant 2002; 30 (4) 215-222
  • 72 Biffi A, Montini E, Lorioli L , et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 2013; 341 (6148) 1233158
  • 73 Cartier N, Hacein-Bey-Abina S, Bartholomae CC , et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009; 326 (5954) 818-823
  • 74 Saher G, Rudolphi F, Corthals K , et al. Therapy of Pelizaeus-Merzbacher disease in mice by feeding a cholesterol-enriched diet. Nat Med 2012; 18 (7) 1130-1135
  • 75 Németh AH, Kwasniewska AC, Lise S , et al; UK Ataxia Consortium. Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain 2013; 136 (Pt 10) 3106-3118
  • 76 Purnell SM, Bleyl SB, Bonkowsky JL. Clinical exome sequencing identifies a novel TUBB4A mutation in a child with static hypomyelinating leukodystrophy. Pediatr Neurol 2014; 50 (6) 608-611
  • 77 Jalal K, Carter R, Yan L, Barczykowski A, Duffner PK. Does galactocerebrosidase activity predict Krabbe phenotype?. Pediatr Neurol 2012; 47 (5) 324-329
  • 78 May T, Zusevics KL, Strong KA. On the ethics of clinical whole genome sequencing of children. Pediatrics 2013; 132 (2) 207-209
  • 79 Knoppers BM, Sénécal K, Borry P, Avard D. Whole-genome sequencing in newborn screening programs. Sci Transl Med 2014; 6 (229) cm2

Address for correspondence

Josh Bonkowsky, MD, PhD
Department of Pediatrics
295 Chipeta Way, Salt Lake City
UT 84108   

  • References

  • 1 Bielschowsky M, Henneberg R. Ueber familiaere diffuse Sklerose (Leukodystrophia cerebri progressiva hereditaria). J Psychol Neurol 1928; 36: 131-181
  • 2 Kaye EM. Update on genetic disorders affecting white matter. Pediatr Neurol 2001; 24 (1) 11-24
  • 3 Maria BL, Deidrick KM, Moser H, Naidu S. Leukodystrophies: pathogenesis, diagnosis, strategies, therapies, and future research directions. J Child Neurol 2003; 18 (9) 578-590
  • 4 Kohlschütter A, Bley A, Brockmann K , et al. Leukodystrophies and other genetic metabolic leukoencephalopathies in children and adults. Brain Dev 2010; 32 (2) 82-89
  • 5 Bonkowsky JL, Nelson C, Kingston JL, Filloux FM, Mundorff MB, Srivastava R. The burden of inherited leukodystrophies in children. Neurology 2010; 75 (8) 718-725
  • 6 Schiffmann R, van der Knaap MS. Invited article: an MRI-based approach to the diagnosis of white matter disorders. Neurology 2009; 72 (8) 750-759
  • 7 Nave KA. Myelination and support of axonal integrity by glia. Nature 2010; 468 (7321) 244-252
  • 8 Savas JN, Toyama BH, Xu T, Yates III JR, Hetzer MW. Extremely long-lived nuclear pore proteins in the rat brain. Science 2012; 335 (6071) 942
  • 9 Kang SH, Fukaya M, Yang JK, Rothstein JD, Bergles DE. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 2010; 68 (4) 668-681
  • 10 Baumann N, Pham-Dinh D. Biology of oligodendrocyte and myelin in the mammalian central nervous system. Physiol Rev 2001; 81 (2) 871-927
  • 11 Jahn O, Tenzer S, Werner HB. Myelin proteomics: molecular anatomy of an insulating sheath. Mol Neurobiol 2009; 40 (1) 55-72
  • 12 Aggarwal S, Yurlova L, Simons M. Central nervous system myelin: structure, synthesis and assembly. Trends Cell Biol 2011; 21 (10) 585-593
  • 13 White R, Krämer-Albers EM. Axon-glia interaction and membrane traffic in myelin formation. Front Cell Neurosci 2014; 7: 284
  • 14 de Castro F, Bribián A, Ortega MC. Regulation of oligodendrocyte precursor migration during development, in adulthood and in pathology. Cell Mol Life Sci 2013; 70 (22) 4355-4368
  • 15 Wake H, Lee PR, Fields RD. Control of local protein synthesis and initial events in myelination by action potentials. Science 2011; 333 (6049) 1647-1651
  • 16 Bartzokis G, Beckson M, Lu PH, Nuechterlein KH, Edwards N, Mintz J. Age-related changes in frontal and temporal lobe volumes in men: a magnetic resonance imaging study. Arch Gen Psychiatry 2001; 58 (5) 461-465
  • 17 van der Knaap MS, Pronk JC, Scheper GC. Vanishing white matter disease. Lancet Neurol 2006; 5 (5) 413-423
  • 18 Berger J, Forss-Petter S, Eichler FS. Pathophysiology of X-linked adrenoleukodystrophy. Biochimie 2014; 98: 135-142
  • 19 Vanderver A, Hussey H, Schmidt JL, Pastor W, Hoffman HJ. Relative incidence of inherited white matter disorders in childhood to acquired pediatric demyelinating disorders. Semin Pediatr Neurol 2012; 19 (4) 219-223
  • 20 van der Knaap MS, Breiter SN, Naidu S, Hart AA, Valk J. Defining and categorizing leukoencephalopathies of unknown origin: MR imaging approach. Radiology 1999; 213 (1) 121-133
  • 21 Pfeifenbring S, von Baumgarten L, Schüller U , et al. Biopsy findings of symptomatic cerebral X-linked adrenoleukodystrophy and histological differentiation from multiple sclerosis. Neuropathol Appl Neurobiol 2014; 40 (5) 658-661
  • 22 Baronica KB, Mlinac K, Ozretić D, Vladić A, Bognar SK. Arylsulfatase a gene polymorphisms in relapsing remitting multiple sclerosis: genotype-phenotype correlation and estimation of disease progression. Coll Antropol 2011; 35 (Suppl. 01) 11-16
  • 23 Eichler F, Van Haren K. Immune response in leukodystrophies. Pediatr Neurol 2007; 37 (4) 235-244
  • 24 Feenstra I, Vissers LE, Orsel M , et al. Genotype-phenotype mapping of chromosome 18q deletions by high-resolution array CGH: an update of the phenotypic map. Am J Med Genet A 2007; 143A (16) 1858-1867
  • 25 Horn MA, Retterstøl L, Abdelnoor M, Skjeldal OH, Tallaksen CM. Adrenoleukodystrophy in Norway: high rate of de novo mutations and age-dependent penetrance. Pediatr Neurol 2013; 48 (3) 212-219
  • 26 Ługowska A, Ponińska J, Krajewski P, Broda G, Płoski R. Population carrier rates of pathogenic ARSA gene mutations: is metachromatic leukodystrophy underdiagnosed?. PLoS ONE 2011; 6 (6) e20218
  • 27 Heim P, Claussen M, Hoffmann B , et al. Leukodystrophy incidence in Germany. Am J Med Genet 1997; 71 (4) 475-478
  • 28 Sereni C, Paturneau-Jouas M, Aubourg P, Baumann N, Feingold J. Adrenoleukodystrophy in France: an epidemiological study. Neuroepidemiology 1993; 12 (4) 229-233
  • 29 Kirk EP, Fletcher JM, Sharp P, Carey B, Poulos A. X-linked adrenoleukodystrophy: the Australasian experience. Am J Med Genet 1998; 76 (5) 420-423
  • 30 Takemoto Y, Suzuki Y, Tamakoshi A , et al. Epidemiology of X-linked adrenoleukodystrophy in Japan. J Hum Genet 2002; 47 (11) 590-593
  • 31 Steenweg ME, Vanderver A, Blaser S , et al. Magnetic resonance imaging pattern recognition in hypomyelinating disorders. Brain 2010; 133 (10) 2971-2982
  • 32 La Piana R, Tonduti D, Gordish Dressman H , et al. Brain magnetic resonance imaging (MRI) pattern recognition in Pol III-related leukodystrophies. J Child Neurol 2014; 29 (2) 214-220
  • 33 Yang E, Prabhu SP. Imaging manifestations of the leukodystrophies, inherited disorders of white matter. Radiol Clin North Am 2014; 52 (2) 279-319
  • 34 van der Knaap MS, Ramesh V, Schiffmann R , et al. Alexander disease: ventricular garlands and abnormalities of the medulla and spinal cord. Neurology 2006; 66 (4) 494-498
  • 35 Welker KM, Patton A. Assessment of normal myelination with magnetic resonance imaging. Semin Neurol 2012; 32 (1) 15-28
  • 36 van der Knaap MS, Valk J, Bakker CJ , et al. Myelination as an expression of the functional maturity of the brain. Dev Med Child Neurol 1991; 33 (10) 849-857
  • 37 Pujol J, López-Sala A, Sebastián-Gallés N , et al. Delayed myelination in children with developmental delay detected by volumetric MRI. Neuroimage 2004; 22 (2) 897-903
  • 38 Yang Y, Muzny DM, Reid JG , et al. Clinical whole-exome sequencing for the diagnosis of mendelian disorders. N Engl J Med 2013; 369 (16) 1502-1511
  • 39 Raymond GV, Jones RO, Moser AB. Newborn screening for adrenoleukodystrophy: implications for therapy. Mol Diagn Ther 2007; 11 (6) 381-384
  • 40 Orsini JJ, Martin MM, Showers AL , et al. Lysosomal storage disorder 4+1 multiplex assay for newborn screening using tandem mass spectrometry: application to a small-scale population study for five lysosomal storage disorders. Clin Chim Acta 2012; 413 (15-16) 1270-1273
  • 41 Duffner PK, Caggana M, Orsini JJ , et al. Newborn screening for Krabbe disease: the New York State model. Pediatr Neurol 2009; 40 (4) 245-252 , discussion 253–255
  • 42 Orsini JJ, Morrissey MA, Slavin LN , et al. Implementation of newborn screening for Krabbe disease: population study and cutoff determination. Clin Biochem 2009; 42 (9) 877-884
  • 43 Waggoner DJ, Tan CA. Expanding newborn screening for lysosomal disorders: opportunities and challenges. Dev Disabil Res Rev 2011; 17 (1) 9-14
  • 44 Lantos JD. Dangerous and expensive screening and treatment for rare childhood diseases: the case of Krabbe disease. Dev Disabil Res Rev 2011; 17 (1) 15-18
  • 45 Nakamura K, Hattori K, Endo F. Newborn screening for lysosomal storage disorders. Am J Med Genet C Semin Med Genet 2011; 157C (1) 63-71
  • 46 Boelens JJ. Trends in haematopoietic cell transplantation for inborn errors of metabolism. J Inherit Metab Dis 2006; 29 (2-3) 413-420
  • 47 Sevin C, Aubourg P, Cartier N. Enzyme, cell and gene-based therapies for metachromatic leukodystrophy. J Inherit Metab Dis 2007; 30 (2) 175-183
  • 48 Orchard PJ, Tolar J. Transplant outcomes in leukodystrophies. Semin Hematol 2010; 47 (1) 70-78
  • 49 Escolar ML, Poe MD, Provenzale JM , et al. Transplantation of umbilical-cord blood in babies with infantile Krabbe's disease. N Engl J Med 2005; 352 (20) 2069-2081
  • 50 Martin PL, Carter SL, Kernan NA , et al. Results of the cord blood transplantation study (COBLT): outcomes of unrelated donor umbilical cord blood transplantation in pediatric patients with lysosomal and peroxisomal storage diseases. Biol Blood Marrow Transplant 2006; 12 (2) 184-194
  • 51 Valayannopoulos V. Enzyme replacement therapy and substrate reduction therapy in lysosomal storage disorders with neurological expression. Handb Clin Neurol 2013; 113: 1851-1857
  • 52 Stroobants S, Gerlach D, Matthes F , et al. Intracerebroventricular enzyme infusion corrects central nervous system pathology and dysfunction in a mouse model of metachromatic leukodystrophy. Hum Mol Genet 2011; 20 (14) 2760-2769
  • 53 Moser HW, Raymond GV, Lu SE , et al. Follow-up of 89 asymptomatic patients with adrenoleukodystrophy treated with Lorenzo's oil. Arch Neurol 2005; 62 (7) 1073-1080
  • 54 Aubourg P, Adamsbaum C, Lavallard-Rousseau MC , et al. A two-year trial of oleic and erucic acids (“Lorenzo's oil”) as treatment for adrenomyeloneuropathy. N Engl J Med 1993; 329 (11) 745-752
  • 55 Moser HW, Moser AB, Hollandsworth K, Brereton NH, Raymond GV. “Lorenzo's oil” therapy for X-linked adrenoleukodystrophy: rationale and current assessment of efficacy. J Mol Neurosci 2007; 33 (1) 105-113
  • 56 Berger J, Pujol A, Aubourg P, Forss-Petter S. Current and future pharmacological treatment strategies in X-linked adrenoleukodystrophy. Brain Pathol 2010; 20 (4) 845-856
  • 57 Van Haren K, Bonkowsky JL, Bernard G , et al , on behalf of the GLIA Consortium. Consensus statement on preventive and symptomatic care of leukodystrophy patients. Mol Genet Metab 2014; ; In press
  • 58 Cohen-Cymberknoh M, Shoseyov D, Kerem E. Managing cystic fibrosis: strategies that increase life expectancy and improve quality of life. Am J Respir Crit Care Med 2011; 183 (11) 1463-1471
  • 59 Brimley CJ, Lopez J, van Haren K , et al. National variation in costs and mortality for leukodystrophy patients in US children's hospitals. Pediatr Neurol 2013; 49 (3) 156-162 , e1
  • 60 Barrell C. Juvenile metachromatic leukodystrophy: understanding the disease and implications for nursing care. J Pediatr Oncol Nurs 2007; 24 (2) 64-69
  • 61 Holmes L, Cornes MJ, Foldi B, Miller F, Dabney K. Clinical epidemiologic characterization of orthopaedic and neurological manifestations in children with leukodystrophies. J Pediatr Orthop 2011; 31 (5) 587-593
  • 62 Nelson C, Mundorff MB, Korgenski EK, Brimley CJ, Srivastava R, Bonkowsky JL. Determinants of health care use in a population-based leukodystrophy cohort. J Pediatr 2013; 162 (3) 624-628 , e1
  • 63 Anderson HM, Wilkes J, Korgenski EK , et al. Preventable infections in children with leukodystrophy. Ann Clin Transl Neurol 2014; 1 (5) 370-374
  • 64 Lu JF, Lawler AM, Watkins PA , et al. A mouse model for X-linked adrenoleukodystrophy. Proc Natl Acad Sci U S A 1997; 94 (17) 9366-9371
  • 65 Kobayashi T, Shinnoh N, Kondo A, Yamada T. Adrenoleukodystrophy protein-deficient mice represent abnormality of very long chain fatty acid metabolism. Biochem Biophys Res Commun 1997; 232 (3) 631-636
  • 66 Forss-Petter S, Werner H, Berger J , et al. Targeted inactivation of the X-linked adrenoleukodystrophy gene in mice. J Neurosci Res 1997; 50 (5) 829-843
  • 67 Morató L, Galino J, Ruiz M , et al. Pioglitazone halts axonal degeneration in a mouse model of X-linked adrenoleukodystrophy. Brain 2013; 136 (Pt 8) 2432-2443
  • 68 Min KT, Benzer S. Preventing neurodegeneration in the Drosophila mutant bubblegum. Science 1999; 284 (5422) 1985-1988
  • 69 Hagemann TL, Connor JX, Messing A. Alexander disease-associated glial fibrillary acidic protein mutations in mice induce Rosenthal fiber formation and a white matter stress response. J Neurosci 2006; 26 (43) 11162-11173
  • 70 Wang L, Colodner KJ, Feany MB. Protein misfolding and oxidative stress promote glial-mediated neurodegeneration in an Alexander disease model. J Neurosci 2011; 31 (8) 2868-2877
  • 71 Koç ON, Day J, Nieder M, Gerson SL, Lazarus HM, Krivit W. Allogeneic mesenchymal stem cell infusion for treatment of metachromatic leukodystrophy (MLD) and Hurler syndrome (MPS-IH). Bone Marrow Transplant 2002; 30 (4) 215-222
  • 72 Biffi A, Montini E, Lorioli L , et al. Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science 2013; 341 (6148) 1233158
  • 73 Cartier N, Hacein-Bey-Abina S, Bartholomae CC , et al. Hematopoietic stem cell gene therapy with a lentiviral vector in X-linked adrenoleukodystrophy. Science 2009; 326 (5954) 818-823
  • 74 Saher G, Rudolphi F, Corthals K , et al. Therapy of Pelizaeus-Merzbacher disease in mice by feeding a cholesterol-enriched diet. Nat Med 2012; 18 (7) 1130-1135
  • 75 Németh AH, Kwasniewska AC, Lise S , et al; UK Ataxia Consortium. Next generation sequencing for molecular diagnosis of neurological disorders using ataxias as a model. Brain 2013; 136 (Pt 10) 3106-3118
  • 76 Purnell SM, Bleyl SB, Bonkowsky JL. Clinical exome sequencing identifies a novel TUBB4A mutation in a child with static hypomyelinating leukodystrophy. Pediatr Neurol 2014; 50 (6) 608-611
  • 77 Jalal K, Carter R, Yan L, Barczykowski A, Duffner PK. Does galactocerebrosidase activity predict Krabbe phenotype?. Pediatr Neurol 2012; 47 (5) 324-329
  • 78 May T, Zusevics KL, Strong KA. On the ethics of clinical whole genome sequencing of children. Pediatrics 2013; 132 (2) 207-209
  • 79 Knoppers BM, Sénécal K, Borry P, Avard D. Whole-genome sequencing in newborn screening programs. Sci Transl Med 2014; 6 (229) cm2

Zoom Image
Fig. 1 Illustrative examples of magnetic resonance images of different leukodystrophies, and of leukodystrophy mimics. Images are axial slices. (A) T1 Image of a hypomyelinating leukodystrophy, TUBB4A. (B) T1 Image with contrast of a demyelinating leukodystrophy, X-linked adrenoleukodystrophy (X-ALD). (C) T2 Fluid-attenuated inversion-recovery (FLAIR) image of metachromatic leukodystrophy (MLD). (D) T2 FLAIR image of vanishing white matter disease (VWMD). (E) T2 FLAIR image of multiple sclerosis (MS). (F) T2 FLAIR image of acute disseminated encephalomyelitis (ADEM).
Zoom Image
Fig. 2 A proposed diagnostic algorithm for testing of leukodystrophies. CK, creatine kinase; MRI, magnetic resonance imaging; PMD, Pelizaeus-Merzbacher disease; VWMD, vanishing white matter disease.