<<

VU Research Portal

Comparing and contrasting disorders: a neuropathological approach to pathophysiology Bugiani, M.

2015

document version Publisher's PDF, also known as Version of record

Link to publication in VU Research Portal

citation for published version (APA) Bugiani, M. (2015). Comparing and contrasting white matter disorders: a neuropathological approach to pathophysiology.

General rights Copyright and moral rights for the publications made accessible in the public portal are retained by the authors and/or other copyright owners and it is a condition of accessing publications that users recognise and abide by the legal requirements associated with these rights.

• Users may download and print one copy of any publication from the public portal for the purpose of private study or research. • You may not further distribute the material or use it for any profit-making activity or commercial gain • You may freely distribute the URL identifying the publication in the public portal ?

Take down policy If you believe that this document breaches copyright please contact us providing details, and we will remove access to the work immediately and investigate your claim.

E-mail address: [email protected]

Download date: 02. Oct. 2021

Chapter 1 Introduction

Myelin, myelination and disorders: from development to pathology to repair

Genetic white matter disorders: definition, clinical features and diagnosis Genetic white matter disorders are inherited encephalopathies characterized by selective involvement of the white matter of the . Most are progressive. The first report of a presumably familial white matter disorder dates back to passage between 18th and 19th century, when Pelizaeus and Merzbacher separately described the familial occurrence of a chronic progressive ‘diffuse sclerosis’ (as opposed to the already recognized ‘’), an encephalopathy with lack of myelin and sclerotic hardening of the cerebral white matter (Pelizaeus, 1899; Merzbacher, 1910). In 1913 Schilder reported an adult patient with ´encephalitis periaxialis diffusa´, the sibling of whom presumably had the same disease (Schilder, 1913). Subsequent descriptions of similar cases prompted the coin of the term “” (leuko, white and dystrophy, lack of growth, thus degeneration) (Bielschowsky & Henneberg, 1928). Hence, in the original concept of the word, are hereditary, progressive diseases characterized by white matter degeneration. Disorders such as metachromatic leukodystrophy (MLD) and globoid cell leukodystrophy (GLD, also called ) owe their names to the staining reactions or unusual cell morphology seen on histopathological specimens. Analysis of stored substrates within white matter subsequently helped to unravel the underlying enzymatic defect in several instances. However, many genetic diseases that affect the white matter remain pathologically and pathogenetically poorly characterized with an unknown biochemical or molecular basis. Because many of these disorders demonstrate a failure in myelination (hypomyelination) rather than a loss of previously deposited myelin (demyelination), the term leukodystrophy sensu stricto has been considered too narrow, and recently juxtaposed by the broader term “leukoencephalopathy”. This latter term would also cover those hereditary white matter disorders in which myelin is normally deposited and no myelin degeneration occurs, and those that do not progress and may even improve over time. Noteworthy, many also designate genetic and acquired defects causing secondary myelin degeneration as leukoencephalopathies (Kaye, 2001; Kohlschütter & Eichler, 2011). The choice for one term or the other and the opinion on which disorders are included under these headings are strongly coloured by convention, and the two words are employed by many interchangeably.

A scientific definition should reflect the knowledge of its time. An important question is how we should define genetic white matter disorders at this time. The term leukodystrophy in its intentional meaning is not applicable to all genetic white matter disorders, because many are not progressive or characterized by loss of

myelin. This term has survived because of its popularity, but has lost its precision in the light of the current knowledge. Many use the term leukoencephalopathy to define all disorders that affect exclusively or predominantly the white matter of the . This choice is linguistically correct, but ignores the need to have terms to distinguish genetic from acquired disorders, and progressive from static conditions. Some destine the word leukodystrophy specifically to inherited white matter disorders. This choice may also be considered infelicitous, because it causes further confusion. The term would define both degenerative disorders, as in its original and still widely perceived meaning, and static conditions such as white matter disorders due to chromosomal abnormalities. Obviously, a consensus redefinition of these terms is urgently needed.

Genetic white matter disorders affect patients of all ages, from the prenatal period to senescence (Osterman et al., 2012). Incidence has been estimated to broadly range from 1:5,000 to 2:100,000 live births (Heim et al., 1997; Bonkowsky et al., 2010), but depending on definitions it is higher approaching 1:1000-2000 live births. As advances in imaging and molecular diagnostic techniques have enormously broadened the spectrum of these disorders, these numbers probably still represent an underestimation.

The clinical manifestations of white matter disorders are relatively heterogeneous and also depend on the age at onset (table 1). An insidious onset of symptom may complicate the diagnosis in the early disease stages, requiring a high index of suspicion and careful probing of the family history. Especially in infancy and early childhood, it can be difficult to distinguish whether a condition is static or truly progressive. With the exception of early infantile-onset cases, patients may exhibit normal early development before losing skills as the white matter disease evolves. A subtle cognitive decline may be the earliest sign preceding the loss of previously acquired motor skills. However, the onset may be or seem abrupt and triggered by traumas or infections, as in vanishing white matter (VWM), (AxD) and sometimes X-linked (X-ALD). The bilateral and symmetric involvement of the cerebral white matter manifests typically as a spastic quadriparesis with cerebellar . More focal clinical signs may also appear, including , , extrapyramidal movement disorders, optic atrophy or . Cognitive functions may remain relatively spared until late in the disease course. Although there is significant overlap in symptomatology, certain clinical signs can be clues to particular conditions. often coexists in Krabbe disease, MLD, Waardenburg-Hirschsprung’s syndrome and, in some instances, Pelizaeus-Merzbacher disease (PMD) and Pelizaeus-Merzbacher-

like diseases, and mitochondrial and peroxisomal disorders. is characteristic for megalencephalic leukoencephalopathy with subcortical cysts (MLC), and AxD, and some peroxisomal disorders and organic acidurias. Characteristic accompanying non-neurological findings such as bone, tooth or skin anomalies or adrenal insufficiency, are also valuable diagnostic clues. Notably, as stated above, not all genetic white matter disorders are progressive. For example, children with the mild form of leukoencephalopathy with thalamus and brainstem involvement and high lactate (LTBL) or with MLC due to autosomal dominant GLIALCAM improve clinically and radiologically over time.

The clinical manifestations in adults are broadly similar to those in children, reflecting the dysfunction of cerebral and/or spinal cord white matter (table 1) (Ahmed et al., 2014). Typically, adult patients present with progressive cognitive or neuropsychiatric difficulties, often associated with progressive lower limb or pseudobulbar signs. Cognitive dysfunction is generally characterized by impaired attention and memory, psychomotor slowing, defective visuospatial and executive skills, and personality changes typical of subcortical . Notably, cortical symptoms such as aphasia, neglect, or apraxia are usually lacking. As in children, the clinical presentation may be precipitated by trauma, infection, or endogenous or exogenous toxins. Patients who present acutely are commonly misdiagnosed, since a genetic disorder is often not considered in adults. Additionally, the clinical picture may be confounded by concurrent substance abuse, psychiatric disease, underlying systemic conditions or incorrect preceding diagnostic labels such as vascular or multiple sclerosis. In the late stages, adult patients as well as children may progress into a state of akinetic mutism, stupor, coma, and ultimately death.

Recognition of leukodystrophies has been revolutionized by magnetic resonance imaging (MRI) complemented by ancillary advanced imaging technologies. The high sensitivity of MRI allows distinction of hypomyelinating leukodystrophies from other pathologies and recognition of myelination delay at follow up. Additionally, MRI may reveal disease-specific patterns of signal abnormalities and characteristic features that strongly address the clinical diagnosis (van der Knaap et al., 1991 & 1999; Schiffmann & van der Knaap, 2009; Steenweg et al., 2010; van der Voorn et al., 2006) (fig. 1). Examples are the contrast enhancement in X-ALD and AxD, or the presence of cysts in MLC and RNASET2-deficient leukoencephalopathy. Calcifications suggest Aicardi-Goutières syndrome (AGS) or RNASET2-deficient leukoencephalopathy, while massive rarefaction of the cerebral white matter is virtually pathognomonic of VWM. Indeed, the validity of MRI pattern recognition in

Periventricular predominance Frontal Subcortical MLD, Krabbe, LBSL predominance predominance AxD, MLD, HDLS Canavan, L2-OHGA Thin Basal nuclei corpus involvement callosum HABC, Pol-III, fucosidosis Pol-III, HCC, fucosidosis, Salla Thalamic involvement Diffuse LTBL, LBSL, HEMS involvement VWM, Canavan, Posterior MLC, LBSL, predominance end-stage X-ALD, Krabbe

Frontal and/or Optic radiation temporal cysts involvement MLC, AGS (early-onset), Pol-III, HEMS RNASET2-deficient leukodystrophy Cerebellar involvement AxD, Krabbe, CTX, LBSL, Brainstem ADLD, MLC, Pol-III, HABC, involvement Salla, Cockayne, PMD, LBSL, AxD, ADLD, CTX PMD-like, HEMS HEMS, PMD-like

Figure 1. Regional distribution of white matter abnormalities and structures specifically involved in some genetic white matter disorders. Abbreviations are listed on page 7. Modified from Osterman et al., 2012.

defining white matter disorders is confirmed beyond doubt by the increasing number of novel entities identified using this approach (van der Knaap et al., 1999).

The diagnostic work-up of genetic white matter disorders is further aided by neurophysiological testing and functional investigation of organs other than the nervous system. When revealing symmetric involvement of long spinal tracts and peripheral , evoked potentials and conduction velocities may be helpful in differentiating leukodystrophies from acquired demyelinating disorders. Absence of multimodal evoked potentials central responses addresses towards hypomyelination syndromes and, in particular, PMD. Thorough ophthalmological evaluation may reveal a cataract, as in cerebrotendinous xanthomatosis, some

hypomyelination syndromes (hypomyelination with congenital cataract), and congenital variants of VWM. Abnormal endocrine findings may suggest X-ALD (adrenal cortical insufficiency), PolR-III defects (hypogonadotropic hypogonadism), AARS2-related leukoencephalopathy or VWM (premature ovarian failure).

The definite diagnosis of a genetic white matter disorder is achieved through specific laboratory and genetic tests (table 1). An omnicomprehensive laboratory protocol does not exist. Age at onset, pattern of neurological involvement and MRI findings dictate the diagnostic work-up and allow for a successful clinical diagnosis (Kohlschütter & Eichler, 2011). However, despite the advances in MRI diagnostics and explosion of discovery over the last decade, a definite etiological diagnosis is still not reached in up to 20% of the patients (Kohlschutter et al., 2010; Perlman & Mar, 2012).

Table 1. Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Inhibited transcription of Adult-onset autosomal Cerebellar, pyramidal, Cerebral WM and myelin genes, including dominant LMNB1 (AD) and autonomic middle cerebellar MG PLP1; altered Oct-1 leukodystrophy symptoms peduncles involvement nuclear localisation (oxidative stress) Neonatal form: , Extensive calcifications, TREX1 (AR, AD de spasticity, dystonia, cerebral hypoplasia, Pleiocytosis (CSF), - Aicardi-Goutières novo), RNASEH2A-C, Dysfunctional DNA marked developmental white matter signal interferon (CSF), MG syndrome SAMHD1, ADAR and repair delay and regression; abnormalities IFIH1 (AR) later-onset variants with milder phenotype Toxic aggregates of Megalencephaly, Diffuse white matter GFAP; defective psychomotor abnormalities with GFAP (AD, most often MG proteasome Alexander disease regression, ataxia and anterior predominance; de novo) degradation/auto- seizures; adults: bulbar in adults mainly brain phagy; glutamate- symptoms stem abnormalities mediated excitotoxicity Allan–Herndon–Dudley Severe retardation in Severely delayed Decreased T3 uptake in SLC16A2/MCT8 (XR) Thyroid function, MG syndrome males myelination and Cerebral and callosal Adult-onset atrophy, frontal white leukoencephalopathy Behavioral problems matter involvement, no with neuroaxonal CSF1R (AD) MG Poorly understood and dementia enhancement, possible spheroids and lacunar- or stroke-like pigmented glia lesions

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Diffuse subcortical Megalencephaly, signal abnormalities; N-acetylaspartate Poorly understood, Canavan disease ASPA (AR) , psychomotor involvement of thalami (urine), MG possibly regression and globi pallidi; osmoregulation defect increased NAA (MRS) Cerebral autosomal Dysregulated Notch dominant arteriopathy Vascular pattern, often signalling leading to with subcortical infarcts NOTCH3 (AD) Strokes, migraines involvement anterior MG progressive vascular and leuko- temporal lobes smooth muscle cell loss encephalopathy Disrupted cholesterol Cerebrotendinous Cerebellar white matter Cholestanol (plasma), homeostasis, CYP27A1 (AR) Xanthomas, cataracts xanthomatosis lesions, calcifications MG cholestanol-induced apoptosis Dwarfism, typical Impaired transcription- facies, peripheral coupled subpathway of neuropathy, cataract, Hypomyelination, DNA repair test nucleotide excision ERCC6 and ERCC8 Cockayne syndrome pigmentary retinopathy, putaminal calcifications, (fibroblasts), DNA repair; altered (AR) sensorineural hearing vermian atrophy MG redox balance; loss, lipoatrophy dysfunctional photosensitivity, mitochondria Ribosomal RNA Cystic leuko- Anterior temporal cysts, degradation defect with encephalopathy calcifications, RNASET2 (AR) Non-progressive course MG lysososmal storage; without congenital CMV-like myelin pathology poorly megalencephaly pattern understood

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Movement disorders, Altered protein and Folate receptor defect FOLR1 (AR) Hypomyelination Folates (CSF), MG epilepsy; treatable myelin composition Accumulation of fucose-containing glycolipids and oligosaccharides; Coarse facial features, -L-fucosidase activity Fucosidosis FUCA1 (AR) Hypomyelination developmentally- dysostosis multiplex (leukocytes), MG regulated loss; selective Purkinje cell apoptotic loss Impaired protein Giant axonal Curly hair, peripheral Features reminiscent of GAN (AR) MG degradation, myelin neuropathy type I neuropathy Alexander disease pathology poorly understood Posterior peri- Developmental ventricular; streaky Galactocerebrosidase Globoid cell regression, spasticity, tigroid appearance; Psychosine cytotoxicity GALC (AR), PSAP (AR) activity (leukocytes), leukodystrophy opisthotonus; late onset optic nerve swelling; to oligodendroglia MG milder involvement pyramidal tracts only (adults)

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Bioenergetic and oxidative stress Variable phenotype, (increased lipid Glycine AMT, GLDC, GCSH, from life-threatening White matter Glycine (plasma versus peroxidation, NMDAR- leukoencephalopathy LIAS, GLRX5 and neonatal to transient or vacuolation CSF), MG mediated gluthatione and variants BOLA3 (AR) late-onset forms reduction); myelin pathology poorly understood Hypomyelination, TUBB4A (mostly de Movement disorders, Hypomyelination with characteristic novo; rarely affected additionally atrophy of the basal anatomic pattern, MG Poorly understood sibs of somatic mosaic encephalopathy of ganglia and vermian and putaminal parent) variable severity atrophy Hypomyelination and cataracts and possibly FAM126A (AR) Hypomyelination MG Poorly understood congenital cataract peripheral neuropathy Reduced catalytic In childhood to early Leukoencephalopathy activity, protein adulthood cerebellar Brainstem and spinal with brainstem and MG expression or DARS2 (AR) ataxia and spasticity; cord involvement; spinal cord involvement dimerization; myelin usually no cognitive elevated lactate (MRS) and high lactate pathology poorly impairment understood Cerebroretinal Telomere DNA microangiopathy White matter cysts, replication defect, with calcifications and CTC1, NDP (AR) Retinopathy MG calcifications myelin pathology poorly cysts understood (Labrune’s disease)

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Leukoencephalopathy Optic atrophy; broad with metaphyseal XR knees and wrists, no Hypomyelination Bone x-ray Oligopenia chondrodysplasia contractures Leukoencephalopathy Pathologic fractures, Frontally accentuated Aberrant regulation of with polycystic TYROBP/DAP12 (AR), rapidly progressing atrophy, basal nuclei Bone x-ray; MG microglial phagocytosis, osteodysplasia TREM2 (AR) presenile dementia calcifications oligodendropathy (Nasu–Hakola disease)

Thalamic and Leukoencephalopathy mesencephalic with thalamus and EARS2 (AR) Later improvement involvement, MG Poorly understood brainstem involvement aplasia splenium and high lactate corpus callosum; elevated lactate (MRS)

Megalencephaly, slowly Megalencephalic progressive ataxia and Subcortical cysts leukoencephalopathy MCL1 (AR) or Ion-and-water spasticity, seizures; in temporal poles and MG with subcortical cysts GLIALCAM (AD, AR) homeostasis defect later improvement for frontoparietal regions the AD variant Accumulation of Behavioral changes, Diffuse white matter Arylsulfatase A activity within lipid Metachromatic ARSA (AR), PSAP pyramidal signs, ataxia, abnormalities; streaky (leukocytes), sulfatides membranes; leukodystrophy (AR), SUMF1 (AR) peripheral neuropathy tigroid appearance; (urine), MG cytotoxicity to sparing of U-fibers oligodendroglia

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings

Large heterogeneous Lactate (plasma, urine, Nuclear and Diffuse white matter oxidative stress, ATP group of disorders, CSF), respiratory chain Mitochondrial disorders mitochondrial DNA involvement, possibly reduction, frequently multi-organ activity (muscle, genes cavitations or cysts; Ca2+ overload symptoms fibroblasts), MG elevated lactate (MRS)

Abnormal posttranslational Typical craniofacial and processing and Oculodentodigital skeletal anomalies, GJA1 (AD, AR) Hypomyelination MG localisation; diminished dysplasia ocular anomalies, heart cell-cell coupling (heart arrhythmias and eye); myelin pathology poorly understood Large heterogeneous “Toxic” effect on White matter cystic Urinary organic Organic acidurias Different genes group of disorders, and degeration, oedema acids, MG possible myelin Involvement of left-right dysfunction, pyramidal connections and Ovarioleukodystrophy AARS2 MG Poorly understood tract involvement; descending tracts; ovarian failure cerebellar atrophy Peripheral neuropathy, Sensorineural hearing central SOX10 (AD, affected loss, pigmentary Dysregulated hypomyelination, sibs of germinal mosaic abnormalities, Hypomyelination MG developmental Waardenburg and parents) Hirschsprung disease, myelination Hirschsprung syndrome neurologic symptoms

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Infantile onset in PLP1 (XR) Symmetric confluent Poorly formed myelin; Pelizaeus-Merzbacher majority; , MG white matter signal unfolded protein disease impaired vision, ataxia, abnormalities response seizures Pelizaeus-Merzbacher- GJA12/GJC2/Cx47 Indistinguishable from Symmetric confluent MG Poorly understood like disease type I (AR) PMD abnormalities Congenital to early infantile onset, early Hypomyelination, Pelizaeus–Merzbacher- AIMP1 (AR) microcephaly, atrophy; N- MG Poorly understood like disease type II progressive course, acetylaspartate (MRS) kyphoscoliosis Pol III-related POLR3A, POLR3B Tooth anomalies, Hypomyelination MG Oligodendrocyte loss leukodystrophies (AR) retarded puberty Altered vesicular Clinically intermediate transport of N-acetyl- between severe aspartylglutamate, Sialic acid storage neonatal storage Free sialic acid (urine, reduced aspartate and SLC17A5 (AR) Hypomyelination disease disease and CSF), MG glutamate transport into the more benign Salla synaptic vesicles; disease myelin pathology poorly understood Skin, hair and nail ERCC2/XPD, Deregulation of thyroid Trichothiodystrophy abnormalities, Hypomyelination MG ERCC3/XPB hormone target genes photosensitivity

Table 1 (cont). Typical clinical, MRI, diagnostic and pathophysiological features of a selection of genetic white matter disorders

Mutated genes Distinctive clinical Distinctive MRI Disease Diagnostic tests Pathophysiology and inheritance features findings Rapid deterioration Progressive white following minor head matter cystic rarefaction MG Abnormal unfolded Vanishing white matter EIF2B1-5 (AR) trauma and febrile and cavitation protein response illness; premature

ovarian failure Cerebral: behavioral Cerebral: changes, motor predominantly Peroxisomal metabolic regression, acute posterior dysfunction; X-linked progression. periventricular, Very long chain fatty inflammation, lipo- ABCD1 (XR) Adrenoleukodystrophy Myeloneuropathy: contrast enhancement. acids (plasma); MG oxidative stress, chronic progressive Myeloneuropathy: deficient ALDP function spastic paraparesis corticospinal tract in microglia Adrenal: failure involvement Peroxisomal metabolic Very long chain fatty dysfunction, Diffuse or restricted Zellweger syndrome Typical facies, visual acids and other mitochondrial Different PEX genes white matter and Zellweger-like and hearing loss, liver peroxisomal dysfunction, oxidative (AR), ACOX1 (AR) involvement, disorders disease metabolites (plasma); stress, protein polymicrogyria MG misfolding, aberrant cell signalling, inflammation

AD, autosomal dominant; AR, autosomal recessive; XR, X-linked recessive; MG: molecular genetic analysis; CSF: cerebrospinal fluid; GFAP: glial fibrillary acidic protein; MRS: magnetic resonance spectroscopy; CMV: cytomegalovirus; NMDAR: N-methyl-D-aspartate receptor; ATP: adenosine triphosphate; ALDP: adrenoleukodystrophy protein (ABCD1).

Organization and structure of the brain white matter The white matter of the brain, so called because of its gleaming blanching aspect on macroscopic examination, is composed of myelinated , glia cells (myelinating oligodendrocytes and oligodendrocyte progenitor cells [OPCs], NG2- glia, and microglia) and blood vessels. The main component of white matter is the myelin sheath. Primordial myelin sheaths appeared in protostomes, lower bilateralia with centralized nervous system. Across evolution, the myelin sheath has evolved into an extended and modified plasma membrane wrapped around the axons that originates from and is part of a specific glia cell type, the oligodendrocyte (Geren & Raskind, 1953). Myelin is organized as multiple, spiral compacted layers (fig. 2). The non-compacted adaxonal (innermost) myelin layer is connected to the cell body through cytoplasmic channels containing , vesicles and outposts of endoplasmic reticulum (ER) and Golgi apparatus that are necessary for myelin maintenance and turnover (Perkins et al., 2008). Each oligodendrocyte can myelinate as many as 40 or more separate axons, but provides myelin for only a segment of any given . The periodic interruptions where axons are left uncovered are the nodes of Ranvier, highly specialized posts of ions interchange and critical to the proper functioning of both axons and myelin (Quarles et al., 2006; Arancibia-Carcamo & Attwell, 2014).

Myelin

Myelin acts as a high resistance electrical insulator that facilitates conduction (Waxman & Bangalore, 2004). In myelinated axons, the excitable axonal membrane is exposed to the extracellular space only at the nodes of Ranvier, where the process of myelination ensures clustering of voltage-gated sodium channels. Because of the high resistance of the myelin sheath, membrane excitement at a node of Ranvier results in outward flow of the locally generated circuit and depolarization of the axonal membrane at the next node. The low capacitance of the myelin sheath implies that little energy is needed to depolarize the remaining internodal membrane, resulting in an increased speed of circuit spreading. This mode of impulse propagation is known as saltatory conduction. In myelinated fibres conduction velocity is proportional to the axonal diameter. If axons of projecting pathways were not myelinated, the human spinal cord would need to be as large as a big tree trunk and the optic nerves0.75 meter in diameter

Figure 2. Myelin sheath ultrastructure. Transverse section of a myelinated axon from the optic nerve of an adult rat. The course of the flattened oligodendrocytic process beginning at the outer tongue (T) and ending at the internal mesaxon (Mi) can be traced. The major dens line (D) alternates with the paler intraperiod line (I). Note that where adjacent sheaths come together an intraperiod line is formed between them, so that an uninterrupted line is formed between them. The axon contains and . Reproduced from Peters & Vaughn, 1970.

in order to maintain equivalent conduction velocities. Therefore, myelin has the evolutionary advantage of facilitating conduction while preserving space and energy (Waxman & Bangalore, 2004; Nave & Werner, 2014).

Myelin plays a role in modulating the caliber and supporting the long-term structural integrity and viability of the axon it enwraps (Nave, 2010a & 2010b; Nave & Werner, 2014). Radial growth of axons is dependent on both myelination and organization and composition of the axonal cytoskeleton (mediated by the local accumulation and phosphorylation of neurofilaments), which is itself influenced by myelin cues (Brady et al., 1999; Rao et al., 2003, Sanchez et al., 2000). Axons in the brain of shiverer (Brady et al., 1999) and jimpy mice (Robain, 1977; Rosenfeld & Freidrich, 1983) respectively lacking the major CNS structural protein myelin basic protein (MBP) and mutant in the proteolipoprotein (PLP), are thinner than wild-type axons. Notably, some myelin constituents are not per se essential for

myelination, but are required for axonal integrity. Mice lacking 2’:3’-cyclic nucleotide 3’-phosphodisterase (CNP), another major protein concentrated in specific regions of the myelin sheaths associated with the oligodendrocytic cytoplasm, myelinate normally but later exhibit axonal swelling and (Lappe-Siefke et al., 2003; Edgar et al., 2009). In Plp1-null mice, oligodendrocytes assemble compact myelin that has only ultrastructural abnormalities and allows a normal motor development. However, secondary focal blockage of axonal transport ensues leading to axonal swellings and Wallerian degeneration of the distal axons (Griffiths et al., 1998; Rosenbluth et al., 2006).

It has been suggested that myelin also provides essential trophic support to axons (Nave, 2010a & 2010b; Hirrlinger & Nave, 2014). This hypothesis stems from different observations, including the similarity between axonal pathology in Plp1- null mice and mitochondrial disorders (Ferreirinha et al., 2004; Tarrade et al., 2006), the higher risk of degeneration of thinner compared to thicker axons (thinner axons are ensheathed by thinner myelin) (Griffiths et al., 1998; Edgar et al., 2009), the presence of ATPases along the entire internodal membrane (where the majority of axonal mitochondria are located, but the myelin sheath creates a diffusion barrier to free metabolic exchange) (McGrail et al., 1991; Young et al., 2008), and the apparent contradiction of relatively short half-lived glycolytic being conveyed by slow anterograd axonal transport (Brady & Lasek, 1981; Oblinger et al., 1988; Yuan et al., 1999). On these bases, it has been speculated that myelinating glia support energy generation in axons by delivering glycolysis products (lactate, pyruvate and its derivatives) for mitochondria in long fiber tracts (Nave, 2010b). Indeed, monocarboxylate transporters for glycolytic breakdown products exist on oligodendrocytes and axons, and neuronal activity can be maintained by lactate as only energy source. Supporting this model, in conditional Cox10 mutant mice, in which mitochondrial respiration is selectively inactivated in myelinating cells, oligodendrocytes survive by aerobic glycolysis and release lactate to the axonal compartment (Funfschilling et al., 2012). Oligodendrocyte-derived lactate is then rapidly metabolized within white matter tracts in vivo. These findings suggest metabolic coupling as a novel physiologic aspect of axon-glia interaction. In this perspective, reduced oligodendroglial trophic support could promote neurodegeneration in many conditions, including different genetic white matter disorders (Hirrlinger & Nave, 2014).

Oligodendrocytes

Developmental myelination, the process by which myelin is first deposited around axons, starts in the human spinal cord at 12 weeks gestation, and in telencephalon around the 14th week. Myelin can be detected in the basal nuclei starting from 20 weeks, in the pre- and postcentral gyrus and optic radiation at 35 weeks, and in the acoustic radiation at 40 weeks. It then proceeds in a caudal-to-rostral and dorsal- to-ventral fashion in the brain white matter during the first two years of life (fig. 3), continuing through the fourth decade in the intra-cortical connections (Yakovlev & Lecours, 1967; van der Knaap & Valk, 2005; Lebel et al., 2008). There is, however, increased evidence that myelination occurs throughout the entire life, either to replace lost oligodendrocytes and myelin or to myelinate previously unmyelinated axons (Bartzokis et al., 2012).

The generation of myelin involves a tightly regulated and finely tuned pathway of OPC specification, proliferation, migration and differentiation culminating, given the appropriate environmental cues, in myelination of appropriate receptive axons (fig. 4). This process is regulated by the interplay of intrinsic and extrinsic cues, these latter impacting on the development of the oligodendrocyte lineage by signalling through intracellular pathways on transcription factors (Emery, 2010; Mitew et al., 2014). Many of these regulatory mechanisms shape different transitions in oligodendrocyte development, and are also reactivated in the post-natal brain for repair purposes (Fancy et al., 2011). Of note, the vast knowledge on oligodendrocyte development and myelination derives from studies of transgenic mice either (constitutively or conditionally) lacking or overexpressing given oligodendrocyte-, myelin- or myelination-related molecules, raising the issue of inter-species differences (Bradl & Lassmann, 2010). The many more oligodendrocytes needed to myelinate a huge amount of axons in the large white matter of the and the much longer and complex developmental process support the idea that human oligodendrocytes may be generated for a longer time period and possibly from more progenitor sources than in the far smaller rodent brain. However, several lines of evidence indicate that a similar molecular machinery operates in humans as in rodents (Jakovcevski et al., 2009).

During development, OPCs appear in the forebrain in sequential waves. At early stages, they predominantly arise from ventral regions in the neural tube under the influence of extracellular signalling molecules as sonic hedgehog (SHH) (Orentas et al., 1999). The initial specification of the oligodendrocytic lineage is reliant on the basic helix-loop-helix (bHLH) transcription factor Olig2, the expression of which is induced (via Notch-1/Smoothened signalling) by SHH (Pringle et al., 1996; Lu et al., 2000). However, at later developmental stages ventrally-derived cells are

A B

C D

Figure 3. Development of myelin in the human brain. Parasagittal sections stained using the Weil method show development of myelin in the human brain at 28 (A) and 40 weeks gestational age (B), at 4 months (C) and at 2 years (D). Reproduced from Flechsig, 1920.

largely replaced by dorsally-derived OPCs (Vallstedt et al., 2005; Kessaris et al., 2006), which specify under the influence of the Wnt/- and bone morphogenic protein (BMP) pathways (Kasai et al., 2005; See & Grinspan, 2009). These OPCs give rise to the majority of adult oligodendrocytes in the mouse forebrain. Notably, SHH-independent pathways also participate to dorsal oligodendrocyte specification. The fibroblast growth factor (FGF)-2, for example, increases Olig2 expression and OPC specification via (MAPK, ERK1/ERK2- mediated) suppression of SMAD signalling, the main effector of dorsal BMP blockade of oligodendrocytes (Chandran et al., 2003; Bilican et al., 2008; Furusho et al., 2011). Other transcription factors at play during this phase contribute either to refine the dorso-ventral domains of the neural tube (NKX2.2, PAX6 [Vallstedt et al., 2005]) or to support oligodendrocyte specification (Ascl1/Mash1 [Parras et al., 2007; Sugimori et al., 2008; Nakatani et al., 2013], SOX10 [Wang et al., 2014]).

The spinal cord witnesses a similar sequence of molecular events, but OPC generation remains heavily skewed towards ventral origins.

Regulation of OPC proliferation and migration ensures that adequate OPC numbers reach the final site of myelination. OPC proliferation occurs not only during development, but also in the adult CNS and upon myelin loss, when developmental pathways are at least in part recapitulated (Kang et al., 2010; Clarke et al., 2012). Most extracellular signals promoting OPC proliferation also promote survival and inhibit differentiation (Wang et al., 2007; Cui et al., 2010). The most potent mitogen regulating total OPC numbers is platelet derived growth factor (PDGF)-A, signalling through the OPC receptor PDGFR (Noble et al., 1988; Richardson et al., 1988; Pringle et al., 1992). Other OPC mitogens are FGF-2 and the insulin growth factor (IGF)-1 that operate alone or synergically to promote OPC proliferation (Chandran et al., 2003; Furusho et al., 2011; Baron et al., 2000; Zeger et al., 2007; Frederick et al., 2007). Extracellular effectors regulating OPC migration include motogenic factors (stimulating OPC motility), adhesion and contact molecules (present in the extracellular matrix [ECM] and on cells here contained), and long-distance chemotactic cues. The main motogenic molecules are PDGF-AA and FGF-2 (Baron et al., 2000). The effects of these molecules may be locally modified by cues present in the ECM, both proteins (laminin, fibronectin, merosin, tenascin-C, anosmin-1) and cell adhesion molecules (polysialylated neuronal cell adhesion molecule [PSA-NCAM], ephrins, v1 integrins, claudin- 11/oligodendrocyte specific protein, neural/glial antigen 2 [NG2], and N-cadherin) (de Castro et al., 2013; Mitew et al., 2014). Chemoattractants and repellents guide longer distance OPC migration. Among these, PDGF-AA, FGF-2 and the secreted chemokine (C-X-C motif) ligand 12 (CXCL12) attract OPCs, while - secreted CXCL1 and semaphorins repel them (Noble et al., 1988; Bribián et al., 2006; Dziembowska et al., 2005; Tsai et al., 2002; Spassky et al., 2002; Taniguchi et al., 2009; Armendáriz et al., 2012). Importantly, the effects of chemotactic cues vary depending on the CNS region, the developmental stage and the specific receptors expressed by different OPC populations. Notably, axonal signals also regulate OPC proliferation and migration. Neuregulin-1 (NRG-1), for example, acts as (PI3K/Akt-mediated) proliferation signal as well as a (MAPK-driven) differentiation cue in presence of laminin-2, and has temporally different effects on OPC migration, chemo-attracting OPC during development but being unnecessary for post-natal OPC migration (Canoll et al., 1999; Fernandez et al., 2000; Flores et al., 2000: Colognato et al., 2002 & 2004; Ortega et al., 2012).

Once they have reached their final destination, OPCs can terminally differentiate into post-mitotic pre-myelinating oligodendrocytes. This is a key point in the myelination process. In mice, OPC terminal differentiation and myelination are almost co-occurring events, with pre-myelinating cells rapidly progressing to myelination or undergoing apoptosis (Barres et al., 1992; Trapp et al., 1997). By contrast, the developing human brain seems to harbour pre-myelinating oligodendrocytes for longer periods, before these cells finally start to myelinate (Back et al., 2002). Greater complexity of the human brain, including its larger size and longer development, and the existence of unique regions and functions, presumably account for the need of a greater potential and more complex regulation of oligodendrocyte differentiation and myelination.

The current model of oligodendrocyte terminal differentiation has been heavily influenced by the simple observation that, in absence of mitogens and serum, cultured OPCs spontaneously exit the cell cycle and differentiate in myelinating oligodendrocytes. This has suggested that much of the regulation of oligodendrocyte differentiation and myelination may be at the level of inhibiting this “default” pathway, with relief of inhibition leading to oligodendrocyte differentiation and myelination (Emery, 2010). Observations in vivo, however, show that the balance between OPC proliferation and differentiation is a tightly regulated process ensuring that oligodendrocyte lineage progression takes place in an orderly sequence and preventing differentiated patterns of gene expression from being induced prematurely or in the wrong cells (Hughes et al., 2013). Indeed, many of the factors participating in this process are inhibitory. Among these are the axonal signals Leucin-rich repeat and immunoglobulin domain-containing-1 (LINGO-1), Jagged-1 and Delta-1. These two latter signal via Notch-1 in OPCs and, through activation of the Hes1 and Hes5 transcription factors, hamper OPC differentiation by inhibiting pro-differentiation factors such as SOX10 (Jepson et al., 2012; Wang et al., 1998; Kondo & Raff, 2000; Liu et al., 2006). Interestingly, in presence of contactin/F3, Notch-1 signalling promotes rather than inhibits OPC differentiation (Hu et al., 2003). BMPs and the oligodendrocyte-specific G-protein coupled receptor 17 (GRP17) also act as negative regulators of oligodendrocyte differentiation and myelination. Their signalling is mediated by the bHLH molecules ID2 and ID4 that directly bind Olig1 and Olig2 inhibiting their function (Cheng et al., 2007; Chen et al., 2009; Samanta & Kessler, 2004). Promoters of differentiation include insulin growth factor-1 (IGF-1) and the triiodothyronine/thyroid hormone 3 (T3), the importance in vivo of which is supported by the hypomyelination phenotype of patients with congenital hypothyroidism (Carson et al., 1993;

SPECIFICATION DIFFERENTIATION, MATURATION & MYELINATION OPC pre-myelinating myelinating (PDGFR, NG2) oligodendrocyte oligodendrocyte (PLP, DM20, APC) Wnt /cat (MBP, MOG) Lingo1, PSA-NCAM

miR-219 PDGF miR-338

Olig2, Nkx2.2 YY1, Sox10 MRF NG2 Olig2 DAPI Jagged1 NF PLP DAPI HDAC NF MBP DAPI

Shh/Gli TH LIF astrocyte NRG1 laminins neurotransmitters ATP adenosine GFAP DAPI WHITE MATTER CORTEX

Nkx6.1 Olig2 Hes1, Hes5, Id2, Id4, Sox5, Sox6 miR-219, miR-338 Sox9 Sox10, Ascl1, Zfp191 Nkx2.2 Nkx2.2, Nkx6.2 Olig1 nuclear Tcf7I2 / Tcf4 active WNT Olig1 cytoplasmic, MRF, YY1

Rodriguez-Pena, 1997; Gupta et al., 1995; Jagannathan et al., 1998).

Signalling via the Wnt/-catenin pathway has emerged as a key regulator of oligodendrocyte development, and exemplifies how the targets of signalling pathways may change depending on the differentiation stage and cellular context. Canonical Wnt signalling leads to nuclear localisation of -catenin, which recruits the transcription factor Tcf7l2/Tcf4 and inhibits terminal differentiation (Shimuzu et al., 2005; Fancy et al., 2009; Fu et al., 2012). Mice with elevated Wnt/-catenin signalling in the oligodendrocyte lineage show blocked oligodendrocyte differentiation and hypomyelination (Fancy et al., 2009). Repression of Wnt signalling is then necessary for oligodendrocyte differentiation. Decrease in - catenin levels allows for binding of Tcf7l2 to the chromatin-remodelling proteins histone deacetylase complexes 1 and 2 (HDAC1/2, see below) and to Olig2 (Ye et al., 2009). In agreement with a role of Tcf7l2 in chromatin remodelling during differentiation, Tcf7l2 expression is tightly regulated and occurs only in post-mitotic non-dividing pre-myelinating oligodendrocytes given that nuclear levels of -catenin are low (Fu et al., 2012). Tcf7l2 would therefore act as a molecular switch promoting or inhibiting oligodendrocyte differentiation depending on the available binding partners.

The identification of oligodendrocytic transcription factors required for terminal differentiation has been greatly aided by knockout animal models. These have revealed that Olig2, the main oligodendrocyte lineage marker, has stage-specific functions in oligodendrocyte development and that its target genes differ according to the differentiation stage. Olig2 is a key player in OPC specification (see above) and is also necessary for terminal differentiation of postnatal OPCs, but has no effects on myelination at later differentiation stages (Mei et al., 2013).

In OPCs, Olig2 induces chromatin remodelling by directing the histone acetylating

Figure 4. Regulation of oligodendrocyte development. After specification, proliferating (circular green arrow) oligodendrocyte progenitor cells (OPCs) undergo sequential steps of differentiation and maturation toward immature pre-myelinating and then myelinating cells. Markers identifying the maturation stage are specified. The process is highly controlled and regulated by a program composed by several intrinsic and extrinsic factors. Boxes represent oligodendrocyte-lineage transitions that are dependent on specific transcription factors (based on mouse knockout experiments). Coloured gradient bars represent predicted temporal expression patterns. The contribution of astrocytic and axonal signals is indicated. Abbreviations are listed on page 7 and found in the text.

molecule BRG1 to target genes critical for terminal differentiation (including SOX10) and myelination (including the myelin regulatory factor [Myrf]), therefore inducing their expression (Yu et al., 2013). The transcription factor Ascl1/Mash1 also supports oligodendrocyte terminal differentiation via promoting the expression of Nkx2.2 (Sugimori et al., 2008). Notably, in both Mash1 and Nkx2.2 knockout mice the total numbers of post-mitotic oligodendrocytes are reduced (Parras et al., 2007; Qi et al., 2001). Nkx2.2 must then be downregulated concurrent with the onset of myelination.

In addition to genetically programmed extracellular ligands, there is evidence that OPC terminal differentiation is also driven by the level of axonal activity. Release of adenosine by active axons activates purinergic receptors on OPCs and promotes differentiation. Axonal release of ATP stimulates adjacent astrocytes to secrete the pro-myelination cytokine leukaemia inhibitory factor (LIF), which in turn acts on OPCs counteracting the inhibitory effects of BMPs (Ishibashi et al., 2006).

The final stage of oligodendrocyte development is myelination. Myelination occurs in a very short time window in the lifetime of the individual oligodendrocyte, during which myelin sheaths are formed and the number of sheaths is determined (Watkins et al., 2008; Czopka et al., 2013). For this to take place, intrinsic and extrinsic regulators interact dynamically to control the balance between differentiation and myelination in a spatiotemporally specific manner, preventing transcriptional repressors of myelin genes from remaining activated after positive activators of myelin gene transcription are brought into play.

The simplest mechanism for determining whether an individual axon is myelinated would be the expression of inhibitory or permissive cues for myelination on the surface of the axon itself. This mechanism would also have the important benefit of allowing control of myelin at the subcellular level, explaining how individual axons are selected for myelination. Intriguingly, most axonal ligands influencing myelination are inhibitory, preventing myelination initiation or excessive myelination. Among these, Lingo1, PS-NCAM, and Jagged-1 inhibit oligodendrocyte process extension and initial contact with the axon (Decker et al., 2000; Fewou et al., 2007; Wang et al., 1998; Hu et al., 2003; Watkins et al., 2008; Mi et al., 2005). Conversely, axonal signals promoting myelination include laminin- 2 (binding oligodendrocyte 1-integrin receptors), netrin-1 (binding the DCC receptor) and axonal L1 ligand (binding oligodendroglial contactin). These pro- myelination signals converge in activating the non-receptor tyrosine kinase Fyn

eventually resulting in reorganisation of the cytoskeletal and microtubules, extension of oligodendrocyte processes and increased branch formation (Morisette & Carbonetto, 1995; Colognato et al., 2002; Hu et al., 2009; Liang et al., 2004; Bauer et al., 2009; Laursen et al., 2009; Rajasekharan et al., 2009). Fyn-mediated morphological remodelling occurs partly via activation of Rho GTPases by the non- receptor tyrosine kinase FAK (focal adhesion kinase) (Chun et al., 2003; Bacon et al., 2007; Kim et al., 2008; O’Meara et al., 2013). Interestingly, FAK regulates oligodendrocytic morphology in an opposite fashion depending on the composition of the ECM, inhibiting process expansion in the presence of fibronectin and promoting process maturation in the presence of laminin-2 (Lafrenaye & Fuss, 2010). Thus, FAK appears to be a key regulator of ECM-cytoskeletal responses.

Axonal signals are also required to establish adequate myelin thickness. Amongst these are laminin-2/1 integrin and NRG-1/ErbB (Lee et al., 2006; Barros et al., 2009; Taveggia et al., 2007; Brinkmann et al., 2008). Notably, NRG-1 expression is influenced by neuronal activity (Ziskin et al., 2007; Liu et al., 2011), confirming this as a major promoter of OPC proliferation, oligodendrogenesis, and myelin remodelling (Gibson et al., 2014). Other pathways downstream of signals regulating myelin thickness are the Raf/MEK/ERK pathway, targeted by FGF-2 and brain-derived neurotrophic factor (BDNF), and the PI3K/Akt/mTOR (mammalian target of rapamycin) pathway, the constitutive activation of which results in hypermyelination (Furusho et al., 2012; Xiao et al., 2010 & 2012; Ishii et al., 2013; Wong et al., 2013; Flores et al., 2008; Narayanan et al., 2009).

Numerous transcription factors have been identified that are involved in regulating myelination. Of note, some are also expressed at earlier differentiation stages, suggesting that their role in promoting differentiation and activity at myelin gene promoters must be subject to regulation of additional cues differentially expressed between OPCs and myelinating cells. Olig1, for example, is expressed throughout oligodendrocyte differentiation, but becomes necessary only at the onset of myelination when it interacts with SOX10 to promote the transcription of MBP (Xin et al., 2005; Li et al., 2007). Interestingly, oligodendrocyte maturation coincides with nucleo-cytoplasmic translocation of Olig1, which is a prerequisite to membrane expansion and maturation (Niu et al., 2012). Nucleo-cytoplasmic translocation is a known, although not fully explored mechanism of transcription factor inactivation. The mechanisms by which Olig1 promotes membrane maturation when excluded from the nucleus are still obscure, but may involve sequestration of inhibitory factors away from the nucleus or recruitment of other promoting factors in or to the cytoplasm. Besides acting at myelin gene promoters,

SOX10 directly interacts and induces the pro-myelination factor Myrf. Myrf is present only in post-mitotic oligodendrocytes during terminal differentiation, when it mediates the transition from pre-myelinating to myelin forming cells. Conditional inactivation of Myrf in oligodendrocytes causes differentiation to stall at an early pre-myelinating stage (Emery et al., 2009). Inducible conditional ablation of Myrf in mature oligodendrocytes causes demyelination even in adult animals, suggesting a fundamental role of Myrf also in myelin maintenance (Koenning et al., 2012). In agreement with this, Myrf binding to DNA is enriched close to genes for myelin components and enzymes and cytoskeletal proteins involved in the generation of the myelin sheath (Cahoy et al., 2008; Bujalka et al., 2013).

As the window of time available for the onset of myelination is very narrow (Czopka et al., 2013), oligodendrocytes possess a complex machinery to synthesize, sort, and traffic enormous amounts of membrane components in short time and in a coordinated manner (Quarles et al., 2006; Simons & Trotter, 2007). To ensure the proper lipid-rich composition of the myelin sheath, specific lipid-lipid and lipid- protein interactions must ensue. The myelin sheath has a unique lipid composition, as it is enriched in cholesterol, , and ethanolamine plasmalogens. The importance of maintaining the optimal ratio of individual lipid classes is illustrated by the adaptive plasticity of the process in genetically manipulated mice. Most are synthesized in the ER, possibly preassembled early in the secretory pathway, and are transported to the membrane via vesicular and non-vesicular mechanisms (Simons et al., 2000; Taylor et al., 2002; Krämer et al., 1997; White & Kramer-Albers, 2014). The major protein (PLP)/DM20 associates with lipids and is transported to myelin by vesicular transport through the biosynthetic pathway (Bradl & Lassmann, 2007; Simons et al., 2000). MBP, which is crucial for the proper assembly of myelin, is targeted by transport of its mRNA in the form of granules assembled in the cell body, transported along microtubules in the oligodendroglial processes, and then localized to the myelin membrane where it ensures myelin compaction (Ainger et al., 1997; Barbarese et al., 1999; Aggarwal et al., 2013; Müller et al., 2013). This alternative mechanism of local synthesis could have been developed to ensure that MBP exerts is adhesive action only at the appropriate site. In vitro studies suggest that both delivery systems may be at least in part under the control of neuronal signals (Trajkovic et al., 2006; Bradl & Lassmann, 2007).

Recent studies using in vivo time-lapse imaging of zebrafish embryos and quantitative analysis of electron micrographs from optic nerve serial sections have shown that radial and longitudinal expansion of the future myelin sheath occur

quite simultaneously (Snaidero et al., 2014). According to this model, the innermost layer of the sheath is laterally expanding and squeezes in between the preceding layer and the axon, an event called wrapping. Myelin compaction would then progress from the abaxonal layers towards the adaxonal inner tongue, allowing the growing sheath to expand underneath the previous layer (Snaidero et al., 2014; Nave & Werner, 2014).

Epigenetic regulation, including chromatin remodelling, DNA methylation and non- coding RNAs, has recently been recognized as a fundamental player contributing to oligodendrocyte development and myelination (Yu et al., 2010; Liu & Casaccia, 2010). One of the best characterized chromatin remodelling is nucleosomal histone modification by acetylation on lysine residues, which is dynamically mediated by histone acetyltransferases (HATs) and HDACs. HATs add acetyl groups and favour a transcriptionally competent chromatin structure, while HDACs remove acetyl groups resulting in chromatin compaction. Recent work, however, shows that HDACs can also participate in transcriptional activation of certain myelin genes (Liu et al., 2009). During oligodendrocyte development, HDAC activity is essential for OPC differentiation and myelin gene expression with temporally-specific effects: the activity is critical for OPC differentiation, but is apparently not required for myelin gene expression after myelination onset (Shen et al., 2005; Humphrey et al., 2008; Swiss et al., 2011). Actually, HDACs likely promote oligodendrocyte differentiation by inhibiting the expression of pathways and genes that otherwise block differentiation, including the Wnt/-catenin and Notch signalling pathways (see above) (Shakèd et al., 2008; Kim & Lassar, 2003; Ye et al., 2009; Kao et al., 1998; Yamaguchi et al., 2005). In line with this, HDAC mutants develop severe myelination deficits due to a block of oligodendrocyte differentiation (Jacob et al., 2011).

MicroRNAs are short non-coding RNAs processed from endogenous genomic loci that target mRNA species with complementary sequence to repress their translation and stability (Bartel, 2009) MicroRNAs function at multiple stages during oligodendrocyte development to direct and finely tune differentiation processes (Svaren, 2014; He et al., 2014). MicroRNAs expression is required for cell cycle exit and terminal differentiation. A possible negative regulator of proliferation is miR34a, which targets cyclin D1 and Notch1 (Bremer et al., 2010; Viader et al., 2011). MiR219 and miR338 target genes (Hes5, SOX6, PDGFR) that act to maintain OPCs in a proliferative undifferentiated state (Zhao et al., 2010; Dugas et

al., 2010). MiR219 may additionally target transcription factors potentially involved in neurogenesis (Dugas et al., 2010; Zhao et al., 2010). This suggests a dual role of microRNAs in regulating oligodendrocyte lineage progression and preventing expression of neuronal or other cell lineage genes. Finally, expression of microRNAs is required on a continual basis in mature oligodendrocyte for proper maintenance of myelin. MiR23 targets the nuclear envelope protein lamin b1 (Lin & Fu, 2009). LMNB1 gene duplications cause adult-onset autosomal dominant leukodystrophy, which is associated with myelin loss and repressed production of MBP, PLP and myelin oligodendrocyte glycoprotein (MOG) (Lin et al., 2014). MiR219 regulates lipid metabolism by targeting a fatty acid elongase that synthesizes saturated and polyunsaturated very long chain fatty acids incorporated into the myelin sheath (Shin et al., 2009).

NG2 glia

NG2 glia are the most recent member of the club of glial cells, identified by their expression of the chondroitin sulphate proteoglycan 4 (CSPG4) (Levine & Nishiyama, 1996). In the developing brain, NG2 glia are a highly proliferating population giving rise to OPCs and protoplasmic astrocytes in the gray matter, whereas their lineage appears restricted to OPCs in the white matter (Zhu et al., 2008 & 2011). In the adult brain, NG2 glia constitute the virtual totality of proliferating cells outside the neurogenic niches and are maintained in homeostatic balance under normal conditions (Hughes et al., 2013). They co-express PDGFR and Olig2 (like OPCs during development) and generate oligodendrocytes and further NG2 glia (Dimou et al., 2008; Ge et al., 2009; Simon et al., 2011; Nishiyama et al., 2009; Geha et al., 2010). They maintain however their regional intrinsic differences, with white matter NG2 glia having a much higher propensity to generate mature oligodendrocytes than their gray matter counterparts (Dimou et al., 2008; Kang et al., 2010; Vigano et al., 2013). The existence in the adult brain of NG2 glia not involved in oligodendrocyte generation has raised the hypothesis that this glial cell type has other functions beyond being OPCs. NG2 glia differ from other glia cell types by receiving synapses from neurons with clear ultrastructural specializations (Behrendt et al., 2013; de Biase et al., 2010; Sakry et al., 2011), a feature for which they are also referred to as “synantocytes”. Different observations suggest that this synaptic communication regulates NG2 glia proliferation and/or differentiation. In mice, sensory deprivation during development leads to NG2 glia proliferation, whereas physical activity in adult animals induces their cell cycle exit

and differentiation into mature oligodendrocytes (Dimou & Götz, 2012; Mangin et al., 2012; Simon et al., 2011). In line with this hypothesis, differentiation of NG2 glia coincides with lost of their synaptic input (Mangin & Gallo, 2011). Within this scenario, NG2 glia may contribute to activity-dependent regulation of myelination and of myelin remodelling in the adult brain. The discovery of proliferating NG2 glia also in the human brain (Geha et al., 2010) suggests that these events take place in our species as well.

Astrocytes

Astrocytes are the predominant cell population in the CNS. Once considered simple scaffolding elements, astrocytes are now recognized as an extremely heterogeneous cell type essential for CNS development and maintenance of CNS homeostasis (Freeman, 2010; Zhang & Barres, 2010; Sofroniew & Vinters, 2010; Verkhratsky & Parpura, 2010). During development, astrocytes progenitors promote synaptogenesis and neurite outgrowth and are involved in synapse stabilization and elimination (Molofsky et al., 2012; Barres, 2008). Postnatally, astrocytes preserve the integrity of the blood-brain and blood-cerebrospinal fluid barriers, control the extracellular ionic milieu, provide metabolic support to neurons, facilitate perivascular flow of cerebrospinal fluid, ensure proper synaptic transmission and plasticity, and are involved in cerebral blood flow regulation (Verkhratsky et al., 2012; Schummers et al, 2008; Sofroniew & Vinters, 2010; Verkhratsky & Parpura, 2010; Barres, 2008; Iliff & Nedergaard, 2013). Astrocytes also participate in regulating developmental myelination and myelin maintenance in the adult brain (Barnett & Linington, 2013; Lanciotti et al., 2013). The role of astrocytes as modulators of oligodendrocyte proliferation and differentiation postulated over 20 years ago based on experimental studies (Noble et al., 1988; Richardson et al., 1988) was definitively confirmed by the identification of human leukoencephalopathies linked to mutations in astrocyte-specific gene products such as the intermediate filament glial fibrillary acidic protein (GFAP, AxD; Brenner et al., 2001) and MLC1 (MLC; Leegwater et al., 2001).

In vitro and in vivo studies have shown that astrocytes are a major source of many regulatory signals that influence OPC survival, oligodendrocyte differentiation/maturation and myelination (IGF-1, LIF, BDNF, Lingo-1, BMPs, CXCL10, CXCL12, CXCL1, PDGF, FGF-2) (Barnett & Linington, 2013). Astrocytes secrete ECM components (tenascin C, hyaluronan, fibronectin, laminin and members of the metalloproteinase family) and are implicated in ECM remodelling,

which may affect OPC proliferation/differentiation and myelination (Barnett & Linington, 2013; Harlow & Macklin, 2014). Simplified culture systems show that astrocytes also couple neuronal activity with myelinogenesis via ATP-, LIF- dependent mechanisms and possibly promote rapid myelin wrapping by material contribution of packaged lipids (Ishibashi et al., 2006; Sorensen et al., 2008; Watkins et al., 2008).

Astrocytes contribute to myelin maintenance by orchestrating the control of ion- water homeostasis at the nodes of Ranvier and preventing intramyelinic edema (Benfenati & Ferroni, 2010; Rash, 2010). When action potentials are transmitted through the white matter, depolarization of myelinated axons is associated with influx of sodium at the nodes of Ranvier and compensatory efflux of potassium at the paranodal regions covered by myelin. These fluctuations of ions are accompanied by osmotically driven shifts in water that require immediate compensation to allow further impulse transmission and prevent cellular swelling. Excessive osmotic water and potassium are siphoned away across the paranodal myelin into astrocytes via gap junctions. Long distance disposal of water and ions occurs via dispersion through the panglial syncytium, a network of astrocytes, oligodendrocytes and ependymal cells also interconnected by gap junctions. The crucial role of astrocytes in maintaining myelin integrity by potassium siphoning and gap junction communication is shown by white matter vacuolization and myelin degeneration in mice lacking gap junctions that form heterotopic interactions between oligodendrocytes and astrocytes (Tress et al., 2011 & 2012; Lutz et al., 2009; Kleopa et al., 2010).

The essential roles of astrocytes in maintaining CNS physiology have challenged the neurocentric view of brain functions. It has been suggested that failure of glial function results in development of many (if not all) neurological diseases (Verkhratsky & Parpura, 2010; Verkhratsky et al., 2012 & 2014). Given the important astrocytic contribution to the regulation of myelin deposition and maintenance, it is highly likely that astrocytes also participate in the pathogenesis of genetic white matter disorders other than AxD and MLC.

Axons

Axons and the ensheathing glia interact bidirectionally and throughout life. This interaction is essential for both partners. Axonal degeneration leads to loss of myelin and lack of myelin leads to axonal degeneration (Ellison et al., 2004; Bjartmar et al., 1999).

Although oligodendrocytes differentiate well in the absence of axons and can wrap even chemically fixed axons or nanofibers in vitro (Rosenberg et al., 2008; Lee et al., 2012), axonal signals participate in regulating oligodendroglial lineage progression and myelination in vivo (Zuchero & Barres, 2010). As illustrated above, axonal NRG-1 influences OPC proliferation, migration and differentiation (Canoll et al., 1999; Fernandez et al., 2000; Flores et al., 2000: Colognato et al., 2002 & 2004; Ortega et al., 2012), while LINGO-1, Jagged-1 and Delta-1 prevent differentiated patterns of gene expression from being induced prematurely in OPCs (Mi et al., 2005; Jepson et al., 2012; Wang et al., 1998). Axonal ligands like PS- NCAM, laminin-2 and netrin-1 control myelination initiation and mediate the influence on myelination of ECM components (Decker et al., 2000; Fewou et al., 2007; Morisette & Carbonetto, 1995; Rajasekharan et al., 2009). Neurons regulate membrane trafficking in oligodendrocytes and coordinate the transport of myelin components like PLP to the plasma membrane, thereby ensuring delivery at the appropriate developmental time (Trajkovic et al., 2006; Kippert et al., 2007). Axonal signals as laminin-2/1 integrin and NRG-1/ErbB are also required to establish adequate myelin thickness (Lee et al., 2006; Barros et al., 2009; Taveggia et al., 2007; Brinkmann et al., 2008).

In addition, neuronal activity may influence myelination (Demerens et al., 1996). Blockade of activity in the developing rat optic nerve by tetrodoxin or axotomy decreases OPC proliferation (Barres & Raff, 1993) and pathological or non- physiological electrical activity enhances OPC proliferation and differentiation or the rate of myelin development (Li et al., 2010; Goldsberry et al., 2011). Neuronal activity may also promote adaptive myelination after development. Optogenetic stimulation of mouse cortical neurons results in OPC proliferation, differentiation and myelin remodelling with accompanying changes in behavioural function (Gibson et al., 2014). The same may occur in the adult human brain, as suggested by studies on the effects of training and learning (possibly including learning a foreign language and a new job as adults, author’s note) showing structural white matter changes within the relevant neural circuit (Roberts et al., 2013; Scholz et al., 2009; Sampaio-Baptista et al., 2013; Schlegel et al., 2012; Wang & Yang, 2014).

Intriguingly, recent in vitro studies revealed two distinct modes of myelination by oligodendrocytes, one independent of neuronal activity and the other dependent on neuronal action potentials. NRG1 switches oligodendrocytes between these two myelination programmes by making oligodendrocyte lineage cells more sensitive to glutamate released from active axons and consequently increasing myelination. Such interaction of NRG1 and glutamatergic signalling may also provide a

mechanism by which myelination is focussed on the axons of active neurons (Lundgaard et al., 2013).

Given the above, an intriguing possibility is that abnormal neuronal activity in genetic diseases affecting the human cortex may as well impact on myelination. Indeed, myelination is perturbed in infantile-onset lysosomal neuronal storage disorders as gangliosidoses and neuronal ceroid lipofuscinosis (NCL), and in the white matter underlying the dysplastic cortex in Zellweger syndrome. Cln8-deficient mice modelling NCL show delayed myelination and increased OPC numbers, suggesting a defect in OPC maturation (Kuronen et al., 2012). Analysis of brain tissue from patients with GM1 and GM2 reveals failed myelin development and paucity of oligodendrocyte lineage cells, which may be compatible with a defect in OPC proliferation or survival (Haberland et al., 1973; van der Voorn et al., 2004).

Pathology and pathologic classification of genetic white matter disorders

The first pathologic classification of white matter disorders dates back to 1956, when Charles Poser and Ludo van Bogaert proposed a histologic systematization of the heterogeneous group of ‘diffuse cerebral scleroses’, chronic progressive diseases mainly affecting children characterized by diffuse myelin loss that until then had been grouped under the clinical umbrella of ‘Schilder’s disease’ (Poser & van Bogaert, 1956; Zeman & Whieldon, 1961). Poser introduced the concept of dysmyelination to differentiate those conditions in which normally constituted myelin is destroyed (myelinoclastic diseases) from those in which myelin does not form properly (Poser, 1957). Incorporating etiologic and prognostic implications with histologic features, Poser and van Bogaert proposed that diffuse cerebral scleroses fall into one of three categories: the myelinoclastic (demyelinating) group, closely related to multiple sclerosis; the dysmyelinating group of the leukodystrophies, probably related to some inherited enzymatic defect; and the leukoencephalitides, possibly of viral origin. Subsequently, the concept of dysmyelination was extended to include also those diseases in which myelin formation is delayed or arrested, as well as those in which the maintenance of already formed myelin appears to be disturbed (Poser 1961, 1968 & 1978).

The pathologic classification currently in use has added one category and separates primary diseases of myelin into three major types: demyelinative, dysmyelinative, and myelinolytic. Quoting James M. Powers (contribution in Naidu, 1999): “The prototypic demyelinative disease is multiple sclerosis (MS), included in

this category because of its inflammatory / immune-mediated destruction of biochemically normal myelin. These are dynamic lesions in which the acute or early lesions are characterized by glial (astrocytic and microglial) activation and perivascular astrocytic cuffs, followed by myelin-oligodendrocyte loss with macrophage infiltration and phagocytosis of myelin, followed by perivascular cuffs of macrophages filled with myelin debris and few lymphocytes, leading finally to a chronic fibrillary astrocytic scar with variable axonal loss. Leukodystrophies, primarily X-ALD, MLD and GLD are included in the dysmyelinative category and share an inherited (genetic) defect in myelin or myelin-forming cells. They generally do not manifest a true inflammatory response, and the myelin destruction appears to be owing to a biochemical abnormality in myelin (X-ALD, in part, MLD) or a molecular defect in the myelin-forming cells (GLD), particularly the oligodendrocyte. Hypomyelinative types, typified by PMD, are also included in this category. Myelinolytic diseases are distinct from the others because their lesions display a spongy or vacuolar myelinopathy owing to intralamellar edema. They may be genetic, such as Canavan’s disease (and MLC, author’s note), or acquired. Their separation has practical implications in that these lesions appear to be less detrimental to myelin, at least in the early stages, and more correctable or reversible. ALD and Canavan’s disease bridge the demyelinative-dysmyelinative and dysmyelinative-myelinolytic category, respectively.”

This classification has the major value of categorizing white matter disorders according to the main mechanism of white matter injury and recognizing the possibility that different pathomechanisms may contribute to determine a single disease. However, the choice of terms could be questioned, also at the light of the new insights into the pathogenesis of some diseases. The word ‘dysmyelination’ by itself implies some kind of aberration in the myelination process, which is considered intrinsic to oligodendrocytes and/or myelin. While the term still applies to very early onset diseases interfering de facto with developmental myelin deposition, it is more difficult to employ in case of adult onset variant of the same conditions. Krabbe disease is one such example. Infantile Krabbe disease typically displays marked paucity of myelin throughout the entire brain. In adult-onset Krabbe disease, by contrast, white matter degeneration appears limited to the corticospinal tracts. The term ‘myelinolytic’ is also questionable. Etymologically (Gr. , meaning breakdown or decomposition, as in cytolysis or hydrolysis), it implies dissolution or destruction of the white matter. However, no white matter destruction takes place in some of the disorders included in this category, which are rather characterized by intramyelinic edema and vacuolization. Again, a

classification should reflect the knowledge of its time. A new classification of genetic white matter disorders based on different criteria than the ones exposed above is perhaps also needed. The contribution of cell types other than oligodendrocytes driving myelin dysfunction or degeneration, including astrocytes and neurons, could also be considered.

Mechanisms of white matter injury

As stated above, the current pathologic classification reflects the main pathogenetic mechanisms that target the brain white matter. Different pathomechanisms may occur in the same condition either simultaneously or in sequence. The best characterized pathomechanism is probably demyelination, which results from direct insults targeting oligodendrocytes and myelin. This kind of injury is often referred to as primary demyelination to distinguish it from secondary demyelination (or Wallerian degeneration), in which myelin degenerates as a consequence of primary axonal loss (Ellison et al., 2004). It should be noted, however, that this distinction may be very difficult to achieve in demyelinating as well as dysmyelinating conditions. In both instances, loss or lack of myelin may coincide temporally with early axonal degeneration, raising the question of which cell type is truly primarily affected. This is the case in multiple sclerosis, but also VWM and some infantile onset primary neuronal disorders as gangliosidoses and NCL.

Depending on the developmental stage and pathomechanism involved, three main early patterns of tissue injury can be differentiated that eventually result in myelin loss (Bradl & Lassmann, 2010; Simons et al., 2014; Fern et al., 2014): (1) When oligodendrocytes and myelin are simultaneously targeted by the pathogenic insult, complete loss of myelin sheaths occurs while oligodendrocytes may either rapidly succumb by necrosis or transiently survive to then be cleared by apoptosis. This process characterizes dysmyelinating storage disorders. (2) Primary injuries involving the oligodendrocytes and not directly affecting the myelin sheaths, including some metabolic disturbances, result in incomplete loss of myelin. This pattern of tissue damage results from loss of individual oligodendrocytes with their respective myelin sheath. (3) Energy deficiency to the white matter leads to ‘dying back oligodendrogliopathy’, in which the damage progresses centripetally along the oligodendrocytic processes to last reach the cell body inducing caspase- independent apoptotic like death. This injury pattern implies an initial selective loss of distally enriched proteins (CNP and myelin-associated glycoprotein [MAG]) that

can be monitored with immunohistochemistry. Of note, the myelin (and axons) surrounding larger blood vessels may be remarkably spared.

Hypomyelination may result from deficiencies in or misfolding of key myelin proteins. The general pathomechanisms underlying this pattern of white matter injury are still poorly understood. The prototypic hypomyelinating leukoencephalopathy is PMD due to mutations in the PLP1 gene. PLP1 misfolding mutations result in oligodendrocytic loss due to ER stress-induced apoptosis, while duplications lead to oligodendrocyte arrested maturation, dysfunction and loss due to protein accumulation into the late endosomal/lysosomal compartment (Torii et al., 2014). Notably, PLP losses (null mutations) do not significantly affect oligodendrocytic numbers or myelination, but rather cause late-onset axonal degeneration (see above) (Griffiths et al., 1998; Rosenbluth et al., 2006). Arrested oligodendrocyte maturation also underlies hypomyelination due to hypothyroidism (see above) (Ibarrola & Rodriguez-Pena, 1997; Gupta et al., 1995; Jagannathan et al., 1998). The pathogenetic mechanisms leading to hypomyelination in other genetic disorders are largely unknown, also due to the rarity of the single conditions and scarcity of human tissue. However, it is conceivable that a reduction in the numbers of myelinating oligodendrocytes, either by cell loss, arrested maturation or both, may be a final common pathogenetic pathway leading to hypomyelination.

The so-called myelinolytic disorders are characterized by intramyelinic fluid accumulation with lamellar splitting along the intraperiod line (extracellular compartment) of the myelin sheath. It is conceivable that a general pathomechanism underlies myelinolysis, but lack of human tissue for many conditions prevents confirmation of this hypothesis. In Canavan disease, it has been suggested that intramyelinic edema results from abnormal water accumulation due to an osmotic-hydrostatic defect. Such defect has been hypothetically ascribed to astrocytes (Baslow & Guilfoyle, 2009 & 2013). Indeed, other genetically defined myelinolytic diseases are due to mutations in either astrocyte-specific gene products (MLC) or in gap junctions acting as water channels across the myelin sheaths. In these conditions, intramyelinic edema could therefore be a general manifestation of failed potassium siphoning and lost ion-water homeostasis. Whether this also applies to other genetic disease with ‘toxic’ intramyelinic edema such as disorders of organic acids, amino acids and urea cycle metabolism is not known.

Pathology of genetic white matter disorders

The pathologic characterization of genetic white matter disorders is largely limited to post-mortem material (Powers & De Vivo, 2002). On macroscopic examination (fig. 5), many conditions display similar macroscopic features that reflect the longstanding, diffuse involvement of the white matter. Typically, the brain is small and the white matter is atrophic with thinning of the corpus callosum and optic nerves and secondary dilation of the . The white matter is stiff and discoloured (hence the first designation of diffuse cerebral sclerosis) and the U- fibers are often spared. The cortex and deep gray structures are generally better preserved or even apparently normal. A predominant involvement of frontal or posterior brain areas is typical of AxD and X-ALD, respectively.

Notable exceptions to the general gross appearance of white matter disorders are the cavitating leukoencephalopathies. In VWM, the cerebral white matter varies from gelatinous to cystic to frankly cavitary to totally disappeared, but the corpus callosum, internal capsule and anterior commissure are generally relatively preserved. In infantile AxD, the affected frontal white matter often shows early cystic degeneration. Cysts can also be expected in the subcortical white matter especially of the temporal lobe in MLC and RNASET2-deficient leukodystrophy, and deeper in the hemispheric white matter in some mitochondrial leukoencephalopathies (NDUFS1, SDHAF1 and APOPT1 mutations). The white matter is grayish to indistinguishable from the gray matter in hypomyelinating disorders. Lacunae or larger infarcts can be appreciated in angiopathic white matter disorders, including cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy (CADASIL), AGS variants and some mitochondrial diseases as mitochondrial encephalopathy with lactic acidosis and stroke-like episodes (MELAS).

At light microscopy examination, the typifying features of genetic white matter disorders are myelin pallor with reduced myelin staining and eventually oligodendrocytic loss, relative sparing of axons and reactive to fibrillary astrocytosis. Spongy or vacuolated myelin due to intramyelinic edema (myelin splitting occurring at the intraperiod line) especially affecting the subcortical white matter is characteristic of myelinolytic leukoencephalopathies like Canavan disease and MLC. Macrophages, either scattered throughout the white matter or clustered around venules, are seen in many demyelinative and dysmyelinative conditions (see below), but are typically rare to absent in hypomyelinating and myelinolytic diseases. Reactive (isomorphic or anisomorphic) is prominent in

A B

Figure 5. Many genetic white matter disorders share common macroscopic features. Sagittal interhemispheric plane (A, Krabbe disease) and coronal cut (B, MLD) show confluent to diffuse loss or lack of white matter, sparing of the subcortical U-fibers (arrowheads), ventriculomegaly and thinning of the corpus callosum (arrow).

hypomyelinating disorders and mitochondrial diseases, but meagre in VWM and myelinolytic conditions. Astrocytes may display typical morphological changes, including the numerous Rosenthal fibres of AxD (and Labrune disease), the blunt coarse cells of VWM and the abundant Alzheimer type II astrocytes of Canavan disease. With the exception of X-ALD, inflammatory cells other than macrophages are generally insignificant in dysmyelinative and hypomyelinating conditions compared to demyelinating diseases as MS.

Careful survey of macrophages and of the myelin debris they contain is especially useful in dysmyelinating disorders (fig. 6). In this category of diseases, myelin is initially formed and then degenerates because it is biochemically abnormal (X-ALD, MLD), following the storage of toxic metabolites in oligodendrocytes (Krabbe disease), or for still unknown reasons (sudanophilic leukodystrophies in general). Breakdown of normal myelin yields galactolipids (sulfatide and cerebroside) and cholesterol; the former are rapidly degraded and are virtually not appreciable, while cholesterol is esterified by phagocytic cells where it persists for longer times and can be visualized with lipid stains as the oil red O. In lysosomal leukodystrophies galactolipids accumulate because of an enzymatic defect and become visible on

carbohydrate stains. Cerebroside, accumulating in Krabbe disease, is positive with the periodic acid Shiff (PAS) stain; sulfatide, stored in MLD, is PAS-positive and also metachromatic due to its anionic sulphate groups. In this, recognition of lipid components of the myelin debris helps both in addressing towards an underlying biochemical defect and in dating the duration of the lesions. The protein composition of the myelin debris also aids in determining the duration and degree of activity of the lesions, and is especially useful in demyelinating conditions.

Proteins that are contained in small amounts in the myelin sheath such as MOG or MAG are cleared much faster than proteins that are more abundant like PLP or MBP. Additionally, macrophages and their content may display characteristic morphologic features. Macrophages are typically striated and vacuolated in X-ALD, granular in MLD and pigmented in adult-onset leukoencephalopathy with neuroaxonal spheroids and pigmented glia. Multinucleated giant macrophages (globoid cells) are pathognomonic of Krabbe disease.

White matter lesions in microangiopathic leukoencephalopathies have no specific features and reflect the acute or chronic ischemia of the tissue. However, they may progress to white matter macrocystic degeneration, as in Labrune’s disease, AGS and MELAS. Perivascular mineralization to large calcifications are seen in AGS, leukoencephalopathy with calcifications and cysts, Cockayne syndrome and cerebrotendinous xanthomatosis. Involvement of the cortex especially the depth of the gyri and of selected neuronal populations may be seen in leukoencephalopathies due to energy defect, including microangiopathic conditions and mitochondrial diseases. Malformations of cortical development to frank cortical dysplasia may be encountered in some mitochondrial leukoencephalopathies (pyruvate dehydrogenase deficiency) and in peroxisomal biogenesis defects (Zellweger syndrome); the exact relation between cortical changes and white matter pathology in these cases is still undefined. Associated atrophy of the cerebellum and striatum suggests hypomyelination with atrophy of the basal ganglia and cerebellum (HABC).

Pathologic examination of biopsy specimens is usually not performed in white matter disorders, apart from selected cases of rapidly progressive demyelination that cannot be diagnostically resolved on MRI grounds.

A B C

D E F

G H I

Figure 6. Common and different microscopic features of genetic white matter disorders. Reduced myelin stain with relative sparing of the U-fibers (A, MBP, Krabbe disease), relative preservation of axons (B, Bodian, AxD), reactive (C, GFAP, MLD) and macrophages containing myelin debris (arrow in D, Kluver-PAS, X-ALD) are common to many conditions. Other features are distinctive, including metachromasia of storage material (E, Toluidine blue, MLD), multinucleated (globoid) cells (F, H&E, Krabbe disease), infiltration of inflammatory cells (G, H&E, X-ALD), Rosenthal fibers (H, H&E, AxD) and spongy myelin degeneration (I, Kluver, Canavan disease). Abbreviations are listed on page 7.

White matter potential for repair: remyelination and tissue remodelling

Remyelination is the process by which myelin sheaths are restored to demyelinated axons, re-enabling saltatory conduction and trophic support to axons

and resolving functional deficits (Franklin & ffrench-Constant, 2008). CNS remyelination is thus a truly regenerative process that, under permissive conditions, may be extremely effective. Efficient remyelination occurs after acute focal myelin loss, especially in younger individuals. In absence of remyelination, axonal degeneration ensues.

The remyelination process largely recapitulates developmental myelination (recapitulation hypothesis of remyelination) (Franklin & Hinks, 1999; Fancy et al., 2011). As developmental myelination, remyelination involves the same key steps of OPC activation, proliferation, migration and terminal differentiation, interaction with unmyelinated axons, expression of myelin genes and synthesis, wrapping and compaction of myelin. However, notable differences between remyelination and developmental myelination exist. The strict correlation between myelin sheath thickness and length and axon diameter typical of developmental myelination is lost during remyelination, which typically results in thinner and shorter sheath segments that are roughly constant independent of the axon diameter (Blakemore, 1974; Ludwin & Maitland, 1984). This is reflected by an increase of the G-ratio, i.e. the ratio between axonal diameter and myelinated fiber diameter. The mechanisms underlying this discrepancy remain unclear. Experimental work suggests that many axonal signals and signalling pathways controlling myelin parameters in developmental myelination (including NRG1, Notch and PI3K/Akt/mTOR) may be dispensable during remyelination (Brinkmann et al., 2008; Stidworthy et al., 2004; Harrington et al., 2010).

Remyelination involves the generation of new myelinating oligodendrocytes that are mostly derived from local OPCs (Prayoonwiwat & Rodriguez, 1993; Sim et al., 2002; ffrench-Constant & Raff, 1986; Dawson et al., 2003; Sim et al., 2006; Zawadzka et al, 2010). Adult OPCs differ from their perinatal counterparts in many aspects, including cell cycle time, motility rate, growth factors responsiveness and antigenic markers (Wolswijk & Noble, 1989; Fancy et al., 2011). Noteworthy, CNS remyelination may also be mediated by Schwann cells, the myelin forming cells of the peripheral nervous system (Itoyama et al., 1983; Dusart et al., 1992; Duncan & Hoffmann, 1997). Schwann cells involvement in CNS remyelination has been observed in animal models and in human demyelinating diseases characterized by a lack of reactive astrogliosis (Blakemore, 1975; Zawadzka et al, 2010). Transgenic fate mapping studies showed that the majority of these cells are actually derived from OPCs (Crang et al., 2004; Zawadzka et al., 2010), revealing an unanticipated differentiation potential of these CNS precursors in the adult brain.

The extrinsic and intrinsic signals regulating remyelination largely overlap with those involved in developmental myelination. OPC activation following injury involves the upregulation of many genes encoding transcription factors associated with oligodendrocyte generation during development (Olig2, Nkx2.2) (Fancy et al., 2004; Sim et al., 2002; Watanabe et al., 2004; Talbott et al., 2005; Shen et al., 2008). Both during development and regeneration PDFG and IGF-1 act as major OPC mitogens, whereas FGF-2 regulates the balance between OPC recruitment and differentiation (Woodruff et al., 2004; Fruttiger et al., 1999; Carson et al., 1993; Mason et al., 2003; Murtie et al., 2005; Zhou et al., 2006; Armstrong et al., 2002). Lingo-1, Notch and the canonical Wnt pathway play a negative regulatory role in remyelination similar to that in myelination (Mi et al., 2005 & 2009; Seifert et al., 2007; Stidworthy et al., 2004; Fancy et al., 2009). The transcription factor Olig1 plays key roles in early OPC development and in OPC differentiation during remyelination (Xin et al., 2005; Arnett et al., 2004).

The efficiency of remyelination declines with age due to age-associated changes in both the extrinsic environmental signals to which OPCs are exposed in remyelinating lesions and intrinsic determinants of OPC behaviour (Franklin & ffrench-Constant, 2008; Crawford et al., 2013). For example, overexpression of mitogens as PDGF in aged animals increases OPC recruitment to demyelinated areas, but does not improve remyelination (Woodruff et al., 2004). This suggests that OPC differentiation into myelin-forming cells is a major limiting step associated with ageing. Remyelination may also fail when myelin loss occurs in the context of a maladaptive immune response, as in chronic lesions of multiple sclerosis. In these conditions, the environment may contain elements that are inhibiting or lack elements that would promote remyelination (Franklin & ffrench-Constant, 2008; Crawford et al., 2013). Indeed, as developmental myelination, remyelination relies on the tight interaction of a multitude of extrinsic and intrinsic signals that regulate the behaviour of the oligodendrocyte lineage through the different stages of the process. The efficiency of remyelination may therefore depend on the presence or absence of regulatory factors, but also on the coordinated timing at which these factors come into play. If any of the stages of remyelination is delayed or dysregulated, oligodendrocytes may miss the critical time window in which the signalling environment is promotive to successful remyelination (Franklin, 2002). Arresting the remyelination process leaves the axons chronically deprived of the trophic support of myelin and prone to degeneration. In case of a disease also featuring early axonal damage, failure to remyelinate naked axons may further exacerbate the extent of axon degeneration.

Several studies have investigated the role of astrocytes in remyelination (Lundgaard et al., 2014). Again, these have been mostly focussed on multiple sclerosis and its models. Transplantation of astrocytes into demyelinated lesions may enhance remyelination by endogenous OPCs (Franklin et al., 1991). Similar to myelination in development, astrocytic signals also regulate remyelination (Barnett & Linington, 2012). However, astrocytic secreted factors have both promoting and inhibiting roles in relation to remyelination (Nair et al., 2008). Astrocytes have recently been implicated in the regulation of oligodendrocytic iron metabolism, which may be vital for oligodendrocytes also in the context of remyelination (Schulz et al., 2012). Gap junction coupling between astrocytes and oligodendrocytes is necessary for myelination and myelin maintenance (Odermatt et al., 2003; Orthmann-Murphy et al., 2007; Tress et al., 2012), The observation that expression of the oligodendrocyte connexin (Cx)47 and astrocyte Cx43 is downregulated in chronic demyelinated multiple sclerosis lesions suggests that oligodendrocyte- astrocyte gap junctions are also needed for effective remyelination (Markoullis et al., 2012). Upon injury, astrocytes become reactive and participate in tissue remodelling by increasing ECM protein and chondroitin sulphate proteoglycan production and deposition. In the case of severe and/or chronic injury, this contributes to the formation of a ‘glial scar’. Following chronic inflammation-induced demyelination, ECM remodelling also coincides with inhibition of OPC differentiation, oligodendrocyte process outgrowth and remyelination (Back et al., 2005; Harlow & Macklin, 2014; Pendleton et al., 2013). The balance between opposing ‘pathologic’ effects of astrogliosis and normal ‘physiologic’ functions of astrocytes is one major factor determining if astrocytes hamper or sustain remyelination in demyelinating diseases. In this respect, inhibition of astrogliosis has been suggested as a potential strategy to allow remyelination to occur.

In agreement with their role in developmental myelination and myelin remodelling in the adult brain, axons and neuronal activity can also influence remyelination. As illustrated above, oligodendrocytes possess two distinct myelination programmes, one independent of neuronal activity and the other dependent on neuronal action potentials via NRG and glutamatergic signalling on oligodendrocytic NMDA receptors (Lundgaard et al., 2013). In vivo mouse experiments show that remyelination of axons in demyelinated lesions is NMDA receptor dependent (Lundgaard et al., 2013). This suggests enhancement of NRG and NMDAR dependent remyelination as a therapeutic approach for promoting recovery after demyelination.

Remyelination in genetic leukoencephalopathies is much less studied. The endogenous potential for repair is likely to be limited in those diseases that result from intrinsic abnormalities of the oligodendrocyte leading to dysmyelination or hypomyelination. Nonetheless, a regenerative attempt could take place, and fail. Translating knowledge on remyelination (and remyelination failure) from multiple sclerosis and its models to genetic leukoencephalopathies is challenging and probably not entirely valid. Disease-specific mechanisms underlying myelin loss and possibly influencing repair potential must be taken into account. In multiple sclerosis, for example, the immune response not only is responsible for the inflammatory attack on myelin, but also has a beneficial permissive role in creating the conditions for remyelination and repair to take place (Franklin & Kotter, 2008). A similar scenario could theoretically be envisaged for inflammatory demyelinating leukodystrophies like X-ALD, but not for VWM where inflammation is typically lacking. Availability only of human tissue obtained at end-stage disease together with absence of relevant animal models (see below) are also major limitations to the recognition of the regenerative potential of genetic white matter disorders.

In vitro and in vivo models of myelination and genetic white matter disorders

The development of a method to grow OPCs from mixed glial cultures (McCarthy & de Vellis, 1980) has allowed the possibility to study OPC differentiation and maturation into myelin-forming cells, in ‘spontaneous’ conditions as well as after genetic manipulation or upon administration of biomolecules. Human and rodent OPCs can be isolated with shake-off techniques or by magnetic-activated cell sorting, and used for studies on cell viability and functionality, cell migration and cell differentiation (Barateiro & Fernandes, 2014; Dincman et al., 2012). However, this in vitro model does not allow communication between OPCs and neurons, which is essential for proper myelination. Oligodendrocyte-neuron co-cultures permit studying the molecular basis of myelination and axoglial signalling without interference of other glial cells (Laursen et al., 2009). Myelinating culture systems employing OPCs, neurons and astrocytes allow investigation of the astrocytic contribution to myelination and oligodendrocyte-neuron communication (Sorensen et al., 2008). In these culture systems, participating cell types can be selectively genetically manipulated. A major disadvantage of all in vitro models is the loss of the tissue architecture and cell-cell and cell-matrix interactions present in the brain. Ex vivo organotypic slice culture models overcome these issues. Organotypic slices from the cerebellum are usually employed to investigate changes in

myelination (Jaeger et al., 1988; Notterpeck et al., 1993). The choice of this brain structure is based on the fact that the cerebellar white matter is easily recognizable and contains high numbers of oligodendrocyte lineage cells (Zhang et al., 2011). Moreover, as the cerebellum myelinates postnatally, slice cultures of neonatal animals reproduce reliably the in vivo myelination process (Suzuki et al., 2012). Organotypic slices from non-myelinating animals as the shiverer mouse can be efficiently myelinated by ‘transplanted’ wild-type OPCs (Bin et al., 2012). Ex vivo slices from mice models of genetic white matter disorders have also been employed to address the issue of specific pathogenetic mechanisms, including oxidative damage in X-ALD (Fourcade et al., 2008).

In vitro and ex vivo culture models however all lack the temporal and spatial signalling existing in vivo, as well as any interaction with the immune system and blood circulation. Only in vivo models allow investigation of brain development and pathology within a complete organism. Among these, zebrafish has oligodendrocyte lineage cells and myelin homologous to mammals and presents the advantages of developing very rapidly and being transparent as embryos, simplifying in vivo imaging of myelin and myelination (Buckley et al., 2008). However, several aspects of zebrafish nervous system organization, including a different myelin composition compared to mammals, dictate circumspection when translating data inter-species. Said this, zebrafish has emerged as a promising genetic model system for the study of myelination and node of Ranvier formation (Pogoda et al., 2006) and as attractive option for addressing selected pathomechanisms of genetic white matter disorders, including Krabbe disease, MLC and RNASET2-related leukoencephalopathy (Zizioli et al., 2014; Sirisi et al., 2014; Haud et al., 2011).

Of all the other in vivo animal models, the most used is the mouse. The fundamental mechanisms underlying oligodendrocyte development and myelination are closely conserved between mouse and human, making this a suitable model system for studies on myelination and myelin pathology.

Mouse models with perturbed growth factor pathways and/or targeted ablation of receptors provided detailed insight into the molecular regulation of induction, proliferation and differentiation of OPCs (Rowitch, 2004). These studies revealed the crucial role of PDGF and FGF in OPC development (Richardson et al., 1988; Bansal et al., 1996; Fortin et al., 2005). Manipulation of transcription factors as OLIG2, NKX2-2 and SOX10 has elucidated the molecular machinery underlying cell specification and maturation (Bansal et al., 1996; Li & Richardson, 2008;

Rowitch, 2004). Spontaneously occurring and genetically engineered mouse models abnormally expressing major myelin proteins or lipids shed light on myelin composition and the mechanisms of myelin sheath formation. The shiverer and jimpy mutants, for example, allowed the identification of MBP and PLP as crucial components of the myelin sheath and enlightened their roles in myelin wrapping and compaction (Lunn et al., 1995). Targeted knockout of enzymes responsible for myelin lipid synthesis revealed a role for lipids in the stabilization of newly formed myelin (Coetzee et al., 1996; Kassmann & Nave, 2008). Immune-mediated, toxic, viral and genetic models of demyelination are used to understand the manifold aspects of MS and investigate the mechanisms of remyelination (Franklin & ffrench-Constant, 2008; Ransohoff, 2012). More recently, myelin mutants have also been employed as hosts to assess the myelination potential of isolated neural cells. Transplantation of rodent and human glial progenitors into non-myelinating animals as shiverer demonstrated these cells’ ability to migrate throughout the host brain and form myelin, in some instances in amounts sufficient to reverse the host phenotype (Ben-Hur & Goldman, 2008; Low et al., 2009; Windrem et al., 2008).

By contrast, generation of genetically manipulated mouse models of hereditary degenerative white matter disorders has often been disappointing. Mouse models of X-ALD, MLD, AGS, VWM and AxD, for instance, show biochemical or otherwise pathologic signs of disease, but have intact myelin and also lack the expected clinical phenotype (Lu et al., 1997; Hess et al., 1996; Behrendt & Roers, 2014; Geva et al., 2010; Hagemann et al., 2006). The absence of myelin pathology in these models is a major limit to their use in studies on pathophysiology and treatment. Greater quantity and complexity of the human brain with its unique regions and functions probably contributes to their limited success. Expansion of the cerebral hemispheric white matter is an evolutionary achievement of primates and humans. Mice have barely any white matter in the telencephalon outside the corpus callosum. Processes as oligodendrocyte development and myelination proceed at different pace in mice and humans and comparisons are neither easy, nor linear (Jakovcevski et al., 2009). Mice have a life span of approximately 2 years, only a fraction of the normal human life span. Also, maturation and ageing progress in the two species at totally different rates (Demetrius, 2005; Flurkey et al., 2007). Overall, mice appear to model primate oligodendrocyte and myelin developmental mechanisms much better than more complex degenerative myelin diseases. Generation of valid mouse models recapitulating the human disease is difficult. When achieved, study of these models in parallel with human brain tissue

should provide the better chance to obtain data that are both pathophysiologically powerful and trustfully translatable across species.

Aims

Oligodendrocytes, astrocytes, microglia, and neurons interact intimately. Consequently, the cellular pathologies of genetic white matter disorders are very complex. Indeed, the identification of novel disease genes, which has accelerated rapidly in recent years, has facilitated -often unexpected- insights into the biology and the interactions within the brain white matter. Genetic white matter disorders vary considerably regarding the affected genes and the pathophysiology. As illustrated in Table 1, only a subset of the mutated genes encodes myelin proteins. Many genetic leukoencephalopathies are caused by mutations that do not affect ‘classical’ myelin gene products, or even affect proteins considered to be expressed specifically in cells other than oligodendrocytes. AxD and MLC are due to mutations in the astrocytes-specific gene products GFAP, and MLC1 and GlialCAM (Brenner et al., 2001; Leegwater et al., 2001; Lopez-Hernandez et al., 2011). Hypomyelination and congenital cataract is caused by mutations in FAM126A encoding the primarily neuronal protein hyccin (Zara et al., 2006; Gazzero et al., 2012). Hereditary diffuse leukoencephalopathy with spheroids is caused by mutations affecting the gene encoding the colony stimulating factor 1 receptor (CSF1R), considered exclusively expressed in microglia (Rademakers et al., 2011; Erblich et al., 2011). Importantly, such prior knowledge may infer an unjustified bias in the search for the pathomechanism. For example, astrocytes are considered the primary site of pathology in AxD. Here, white matter degeneration coincides with the appearance of Rosenthal fibers, astrocytic aggregates of the mutant intermediate filament protein GFAP (Mignot et al., 2004; Sawaishi et al., 2009). However, whether astrocytic Rosenthal fibers indeed contribute to the white matter degeneration has not been satisfactorily shown. Overviewing the evolution of understanding of white matter physiology and pathology, it is clear that it is important to revise old concepts and definitions and that an unbiased approach is necessary to achieve further, truly new insights.

The aims of this thesis are to ascertain 1) if a cellular pathology perspective can be applied to the neuropathological evaluation of genetic white matter disorders; 2) if this approach can help categorize the major neuropathological findings in individual diseases; 3) if this approach can identify common pathomechanisms driving white matter dysfunction and degeneration; 4) if this information may be used to assess

the white matter repair potential; and 5) if this approach may contribute to a novel classification of genetic white matter disorders, which recapitulates all the above information.

To do this, we will compare and contrast the neuropathology of selected genetic white matter disorders of different aetiology, including VWM (chapter 2), MLC and ClC-2 related disease (chapter 3), a hypomyelination syndrome (chapter 4), and leukoencephalopathies due to vascular changes (chapter 5). Some of the diseases described are novel conditions. When available, the hypotheses formulated for human diseases will be verified and further investigated on their animal models using a combined evolutionary-developmental-pathological approach. A general discussion follows (chapter 6).

References

Aggarwal S et al. Myelin membrane assembly is driven by a phase transition of myelin basic proteins into a cohesive protein meshwork. PLOS Biol 2013; 11:e1001577.

Ahmed R et al. A practical approach to diagnosing adult onset leukodystrophies. J Neurol Neurosurg Psychiatry 2014;85:770-781.

Ainger K et al., Transport and localisation elements in myelin basic protein mRNA. J Cell Biol 1997;138:1077-1087.

Arancibia-Carcamo IL & Attwell D. The node of Ranvier in CNS pathology. Acta Neuropathol 2014;128:161-175.

Armendáriz BG et al. Expression of Semaphorin 4F in neurons and brain oligodendrocytes and the regulation of oligodendrocyte precursor migration in the optic nerve. Mol Cell Neurosci 2012;49:54-67.

Armstrong RC et al. Absence of fibroblast growth factor 2 promotes oligodendroglial repopulation of demyelinated white matter. J Neurosci 2002;22:8574-8585.

Arnett HA et al. TNFalpha promotes proliferation of oligodendrocyte progenitors and remyelination. Nat Neurosci 2001;4:1116-1122.

Back SA et al. Arrested oligodendrocyte lineage progression during human cerebral white matter development: dissociation between the timing of progenitor differentiation and myelinogenesis. J Neuropathol Exp Neurol 2002;61:197-211.

Back SA et al. Hyaluronan accumulates in demyelinated lesions and inhibits oligodendrocyte progenitor maturation. Nature Med 2005;11:966-972.

Bacon C et al. N-WASP regulates extension of filopodia and processes by oligodendrocyte progenitors, oligodendrocytes, and Schwann cells - implications for axon ensheathment at myelination. Glia 2007;55:844-858. Bansal R et al. Regulation of FGF receptors in the oligodendrocyte lineage. Mol Cell Neurosci 1996;7:263-275.

Barateiro A & Fernandes A. Temporal oligodendrocyte lineage progression: in vitro models of proliferation, differentiation and myelination. Biochim Biophys Acta 2014;1843:1917-1929.

Barbarese E et al. RNA on the road to myelin. J Neurocytol 1999;28:263-270.

Barnett SC & Linington C. Myelination: do astrocytes play a role? Neuroscientist 2013;19:442-450.

Baron W et al. PDGF and FGF-2 signaling in oligodendrocyte progenitor cells: regulation of proliferation and differentiation by multiple intracellular signaling pathways. Mol Cell Neurosci 2000;15:314-329.

Barres BA et al. Cell death and control of cell survival in the oligodendrocyte lineage. Cell 1992;70:31- 46.

Barres BA & Raff MC. Proliferation of oligodendrocyte precursor cells depends on electrical activity in axons. Nature 1993;361:258–260.

Barres BA. The mystery and magic of glia: a perspective on their roles in health and disease. Neuron 2008;60:430-40.

Barros CS et al. b1 Integrins are required for normal CNS myelination and promote AKT-dependent myelin outgrowth. Development 2009;136:2717-2724.

Bartel DP. MicroRNAs: target recognition and regulatory functions. Cell 2009;136:215-33.

Bartzokis G et al. Multimodal magnetic resonance imaging assessment of white matter aging trajectories over the lifespan of healthy individuals. Biol Psychiatry 2012;72:1026-1034.

Baslow MH & Guilfoyle DN. Are astrocytes the missing link between lack of brain activity and the spongiform leukodystrophy in Canavan disease? Neurochem Res 2009;34:1523-1534.

Baslow MH & Guilfoyle DN. Canavan disease, a rare early-onset human spongiform leukodystrophy: insights into its genesis and possible clinical interventions. Biochimie 2013;95:946-956.

Bauer NG et al. Role of the oligodendroglial cytoskeleton in differentiation and myelination. Glia 2009;57:1691-1705.

Behrendt G et al. Dynamic changes in myelin aberrations and oligodendrocyte generation in chronic amyloidosis in mice and men. Glia 2013;61:273-286.

Behrendt R & Roers A. Mouse models for Aicardi-Goutieres syndrome provide clues to the molecular pathogenesis of systemic autoimmunity. Clin Exp Immunol 2014;175:9-16.

Benfenati V & Ferroni S. Water transport between CNS compartments: functional and molecular interactions between aquaporins and ion channels. Neuroscience 2010;168:926-940.

Ben-Hur T & Goldman SA. Prospects of cell therapy for disorders of myelin. Ann N Y Acad Sci 2008;1142:218-249.

Bielschowsky M & Henneberg R. Ueber familiäre diffuse Sklerose (Leukodystrophia cerebri progressiva hereditaria). J Psychol Neurol 1928;36:131-181.

Bilican B et al. Induction of Olig2+ precursors by FGF involves BMP signalling blockade at the Smad level. PLoS One 2008;3:e2863.

Bin JM et al. Oligodendrocyte precursor cell transplantation into organotypic cerebellar shiverer slices: a model to study myelination and myelin maintenance. PLoS One 2012;7:e41237.

Bjartmar C et al. Axonal pathology in myelin disorders. J Neurocytol 1999;28:383-395.

Blakemore WF. Pattern of remyelination in the CNS. Nature 1974;249:577-578.

Blakemore WF. Remyelination by Schwann cells of axons demyelinated by intraspinal injection of 6- aminonicotinamide in the rat. J Neurocytol 1975;4:745-757.

Bonkowsky JL et al. The burden of inherited leukodystrophies in children. 2010;75:718–725.

Bradl M & Lassmann H. Oligodendrocytes: biology and pathology. Acta Neuropathol 2010;119:37-53.

Brady ST & Lasek RJ. Nerve-specific enolase and creatine phosphokinase in axonal transport: soluble proteins and the axoplasmic matrix. Cell 1981;23:515-523.

Brady ST et al. Formation of compact myelin is required for maturation of the axonal cytoskeleton. J Neurosci 1999;19:7278-7288.

Bremer J et al. Ablation of Dicer from murine Schwann cells increases their proliferation while blocking myelination. PLoS One 2010;5:e12450.

Brenner M et al. Mutations in GFAP, encoding glial fibrillary acidic protein, are associated with Alexander disease. Nat Genet 2001;27:117-120.

Bribián A et al. Anosmin-1 modulates the FGF-2-dependent migration of oligodendrocyte precursors in the developing optic nerve. Mol Cell Neurosci 2006;33:2-14.

Brinkmann BG et al. Neuregulin-1/ErbB signaling serves distinct functions in myelination of the peripheral and central nervous system. Neuron 2008;59:581-595.

Buckley CE et al. Zebrafish myelination: a transparent model for remyelination? Dis Model Mech 2008;1:221-227.

Bujalka H et al. MYRF is a membraneassociated transcription factor that autoproteolytically cleaves to directly activate myelin genes. PLoS Biol 2013;11:e1001625.

Cahoy JD et al. A transcriptome database for astrocytes, neurons, and oligodendrocytes: a new resource for understanding brain development and function. J Neurosci 2008;28:264-278.

Canoll PD et al. GGF/neuregulin induces a phenotypic reversion of oligodendrocytes. Mol Cell Neurosci 1999;13:79-94.

Carson MJ et al. Insulin-like growth factor I increases brain growth and central nervous system myelination in transgenic mice. Neuron 1993;10:729-740.

Chandran S et al. FGF-dependent generation of oligodendrocytes by a hedgehog-independent pathway. Development 2003;130:6599-6609.

Chen Y et al. The oligodendrocyte-specific G protein-coupled receptor GPR17 is a cell-intrinsic timer of myelination. Nat Neurosci 2009;12:1398-1406.

Cheng X et al. Bone morphogenetic protein signaling and olig1/2 interact to regulate the differentiation and maturation of adult oligodendrocyte precursor cells. Stem Cells 2007;25:3204-3214.

Chun SJ et al. Integrin-linked kinase is required for laminin-2-induced oligodendrocyte cell spreading and CNS myelination. J Cell Biol 2003;163:397-408.

Clarke LE et al. Properties and fate of oligodendrocyte progenitor cells in the corpus callosum, motor cortex, and piriform cortex of the mouse. J Neurosci 2012;32:8173-8185.

Coetzee T et al. Myelination in the absence of galactocerebroside and sulfatide: normal structure with abnormal function and regional instability. Cell 1996;86:209-219.

Colognato H et al. CNS integrins switch growth factor signalling to promote target dependent survival. Nat Cell Biol 2002;4:833-841.

Colognato H et al. Integrins direct Src family kinases to regulate distinct phases of oligodendrocyte development. J Cell Biol 2004;167:365-375.

Crang AJ et al. The remyelinating potential and in vitro differentiation of MOG-expressing oligodendrocyte precursors isolated from the adult rat CNS. Eur J Neurosci 2004;20:1445-1460.

Cui QL et al. Response of human oligodendrocyte progenitors to growth factors and axon signals. J Neuropathol Exp Neurol 2010;69:930-944.

Czopka T et al. Individual oligodendrocytes have only a few hours in which to generate new myelin sheaths in vivo. Dev Cell 2013;25:599-609.

Dawson MRL et al. NG2-expressing glial progenitor cells: an abundant and widespread population of cycling cells in the adult rat CNS. Mol Cell Neurosci 2003;24:476-488.

De Biase LM et al. Excitability and synaptic communication within the oligodendrocyte lineage. J Neurosci 2010;30:3600-3611. de Castro F et al. Regulation of oligodendrocyte precursor migration during development, in adulthood and in pathology. Cell Mol Life Sci 2013;70:4355-4368.

Decker L et al. Oligodendrocyte precursor migration and differentiation: combined effects of PSA residues, growth factors, and substrates. Mol Cell Neurosci 2000;16:422-439.

Demerens C et al. Induction of myelination in the central nervous system by electrical activity. Proc Natl Acad Sci USA 1996;93:9887-9892.

Demetrius L. Of mice and men. When it comes to study ageing and the means of slow it down, mice are not just small humans. EMBO Rep 2005;6:S39-44.

Dimou L & Gotz M. Shaping barrels: activity moves NG2+ glia. Nature Neurosci 2012;15:1176-1178.

Dimou L et al. Progeny of Olig2-expressing progenitors in the gray and white matter of the adult mouse cerebral cortex. J Neurosci 2008;28:10434-10442.

Dincman TA et al. Isolation of cortical mouse oligodendrocyte precursor cells. J Neurosci Methods 2012;209:219-226.

Dugas JC et al. Dicer1 and miR-219 are required for normal oligodendrocyte differentiation and myelination. Neuron 2010;65:597-611.

Duncan ID & Hoffman RL. Schwann cell invasion of the central nervous system of the myelin mutants. J Anat 1997;190:35-49.

Dusart I et al. Demyelination, and remyelination by Schwann cells and oligodendrocytes after kainate- induced neuronal depletion in the central nervous system. Neuroscience 1992;51:137-148.

Dziembowska M et al. A role for CXCR4 signaling in survival and migration of neural and oligodendrocyte precursors. Glia 2005;50:258-269.

Edgar J M et al. Early ultrastructural defects of axons and axon-glia junctions in mice lacking expression of Cnp1. Glia 2009;57:1815-1824.

Ellison D et al (eds). Neuropathology. A reference text of CNS pathology. 2nd ed. Edinburgh: Mosby Elsevier, 2004.

Emery B et al. Myelin gene regulatory factor is a critical transcriptional regulator required for CNS myelination. Cell 2009;138:172-185.

Emery B. Regulation of oligodendrocyte differentiation and myelination. Science 2010;330:779-782.

Erblich B et al. Absence of colony stimulation factor-1 receptor results in loss of microglia, disrupted brain development and olfactory deficits. PLoS One 2011;6:e26317.

Fancy SPJ et al. Increased expression of Nkx2.2 and Olig2 identifies reactive oligodendrocyte progenitor cells responding to demyelination in the adult CNS. Mol Cell Neurosci 2004;27:247-254.

Fancy SP et al. Dysregulation of the Wnt pathway inhibits timely myelination and remyelination in the mammalian CNS. Genes Dev 2009;23:1571-1585.

Fancy SPJ et al. Myelin regeneration: a recapitulation of development? Ann Rev Neurosci 2011;34:21- 43.

Fern RF et al. White matter injury: ischemic and nonischemic. Glia 2014;62:1780-1789.

Fernandez PA et al. Evidence that axon-derived neuregulin promotes oligodendrocyte survival in the developing rat optic nerve. Neuron 2000;28:81-90.

Ferreirinha F et al. Axonal degeneration in paraplegin-deficient mice is associated with abnormal mitochondria and impairment of axonal transport. J Clin Invest 2004;113:231-242.

Fewou SN et al. Down-regulation of polysialic acid is required for efficient myelin formation. J Biol Chem 2007;282:16700-16711. ffrench-Constant C & Raff MC. Proliferating bipotential glial progenitor cells in adult rat optic nerve. Nature 1986;319:499-502.

Flechsig P. Anatomie des menschlichen Gehirns und Rückenmarks auf myelogenetischer Grundlage. Leipzig, 1920.

Flores AI et al. Akt-mediated survival of oligodendrocytes induced by neuregulins. J Neurosci 2000;20:7622-7630.

Flores AI et al. Constitutively active Akt induces enhanced myelination in the CNS. J Neurosci 2008;28:7174-7183.

Flurkey K et al. The mouse in ageing research. In Fox JG et al. (eds). The mouse in biomedical research. 2nd ed. Burlington: Elsevier, 2007.

Fourcade S et al. Early oxidative damage underlying neurodegeneration in X-adrenoleukodystrophy. Hum Mol Genet 2008;17:1762-1773.

Fortin D et al. Distinct fibroblast growth factor (FGF)/FGF receptor signaling pairs initiate diverse cellular responses in the oligodendrocyte lineage. J Neurosci 2005;25:7470-7479.

Franklin RJM et al. Transplanted type-1 astrocytes facilitate repair of demyelinating lesions by host oligodendrocytes in adult rat spinal cord. J Neurocytol 1991;20:420-430.

Franklin RJM. Why does remyelination fail in multiple sclerosis? Nat Rev Neurosci 2002;3:705-714.

Franklin RJM & Hinks GL. Understanding CNS remyelination - clues from developmental and regeneration biology. J Neurosci Res 1999; 58:207-213.

Franklin RJM & ffrench-Constant C. Remyelination in the CNS: from biology to therapy. Nat Rev Neurosci 2008;9:839-855.

Franklin RJ & Kotter MR. The biology of CNS remyelination: the key to therapeutic advances. J Neurol 2008;255:19-25.

Frederick TJ et al. Synergistic induction of cyclin D1 in oligodendrocyte progenitor cells by IGF-I and FGF-2 requires differential stimulation of multiple signaling pathways. Glia 2007;55:1011-1022.

Freeman MR. Specification and morphogenesis of astrocytes. Science 2010;330:774-778.

Fruttiger M et al. Defective oligodendrocyte development and severe hypomyelination in PDGF-A knockout mice. Development 1999;126:457-467.

Fu H et al. Tcf7l2 is tightly controlled during myelin formation. Cell Mol Neurobiol 2012;32:345-352.

Funfschilling U et al. Glycolytic oligodendrocytes maintain myelin and long-term axonal integrity. Nature 2012;485:517-521.

Furusho M et al. Fibroblast growth factor signaling is required for the generation of oligodendrocyte progenitors from the embryonic forebrain. J Neurosci 2011;31:5055-5066.

Furusho M et al. Fibroblast growth factor receptor signaling in oligodendrocytes regulates myelin sheath thickness. J Neurosci 2012;32:6631-6641.

Gazzero E et al. Hyccin, the molecule mutated in the leukodystrophy hypomyelination and congenital cataract (HCC), is a neuronal protein. PLoS One 2012;7:e32180.

Ge WP et al. Dividing glial cells maintain differentiated properties including complex morphology and functional synapses. Proc Natl Acad Sci U S A 2009;106:328-333.

Geha S et al. NG2+/Olig2+ cells are the major cycle-related cell population of the adult human normal brain. Brain Pathol 2010;20:399-411.

Geren BB & Raskind J. Development of the fine structure of the myelin sheath in sciatic nerves of chick embryos. Proc Natl Acad Sci U S A 1953;39:880-884.

Geva M et al. A mouse model for eukaryotic translation initiation factor 2B-leucodystrophy reveals abnormal development of brain white matter. Brain 2010;133:2448-2461.

Gibson EM et al. Neuronal activity promotes oligodendrogenesis and adaptive myelination in the mammalian brain. Science 2014;344(6183):1252304.

Goldsberry G et al. Accelerated myelination with motor system involvement in a neonate with immediate postnatal onset of seizures and hemimegalencephaly. Epilepsy Behav 2011;22:391-394.

Griffiths I et al. Axonal swellings and degeneration in mice lacking the major proteolipid of myelin. Science 1998;280:1610-1613.

Gupta RK et al. Brain metabolite changes on in vivo proton magnetic resonance spectroscopy in children with congenital hypothyroidism. J Pediatr 1995;126:389-392.

Haberland C et al. The white matter in GM2 gangliosidosis. A comparative histopathological and biochemical study. Acta Neuropathol 1973;24:43-55.

Hagemann TL et al. Alexander disease-associated glial fibrillary acidic protein mutations in mice induce rosenthal fiber formation and a white matter stress response. J Neurosci 2006;26:11162-11173.

Harlow DE & Macklin WB. Inhibitors of myelination: ECM changes, CSPGs and PTPs. Exp Neurol 2014;251:39-46.

Harrington EP et al. Oligodendrocyte PTEN required for myelin and axonal integrity not remyelination. Ann Neurol 2010;68:703-16.

Haud N et al. rnaset2 mutant zebrafish model familial cystic leukoencephalopathy and reveal a role for RNase T2 in degrading ribosomal RNA. Proc Natl Acad Sci U S A 2011;108:1099-1103.

He X et al. Unwrapping myelination by MicroRNAs. Neuroscientist 2012;18:45-55.

Heim P et al. Leukodystrophy incidence in Germany. Am J Med Genet 1997;71:475–1478.

Hess B et al. Phenotype of arylsulfatase A-deficient mice: relationship to human metachromatic leukodystrophy. Proc Natl Acad Sci U S A 1996;93:14821-14826.

Hirrlinger J & Nave KA. Adapting brain metabolism to myelination and long-range signal transduction. Glia 2014;62:1749-1761.

Hu Q-D et al. F3/contactin acts as a functional ligand for Notch during oligodendrocyte maturation. Cell 2003;115:163-175.

Hu J et al. Effects of extracellular matrix molecules on the growth properties of oligodendrocyte progenitor cells in vitro. J Neurosci Res 2009;87:2854-2862.

Hughes EG et al. Oligodendrocyte progenitors balance growth with self-repulsion to achieve homeostasis in the adult brain. Nat Neurosci 2013;16:10.

Humphrey GW et al. Complementary roles for histone deacetylases 1, 2, and 3 in differentiation of pluripotent stem cells. Differentiation 2008;76:348-356.

Ibarrola N & Rodriguez-Pena A. Hypothyroidism coordinately and transiently affects myelin protein gene expression in most rat brain regions during postnatal development. Brain Res 1997;752:285-293.

Iliff JJ & Nedergaard M. Is there a cerebral lymphatic system? Stroke 2013;44:S93-95.

Ishibashi T et al. Astrocytes promote myelination in response to electrical impulses. Neuron 2006;49:823-832.

Ishii A et al. Sustained activation of ERK1/2 MAPK in oligodendrocytes and Schwann cells enhances myelin growth and stimulates oligodendrocyte progenitor expansion. J Neurosci 2013;33:175-186.

Itoyama Y et al. Schwann cell remyelination of demyelinated axons in spinal cord multiple sclerosis lesions. Ann Neurol 1983;14:339-346.

Jacob C et al. How histone deacetylases control myelination. Mol Neurobiol 2011;44:303-312.

Jaeger CB et al. Cytology and organization of rat cerebellar organ cultures. Neuroscience 1988;26:509- 538.

Jagannathan NR et al. Reversal of abnormalities of myelination by thyroxine therapy in congenital hypothyroidism: localized in vivo proton magnetic resonance spectroscopy (MRS) study. Brain Res Dev Brain Res 1998;109:179-186.

Jakovcevski I et al. Oligodendrocyte development and the onset of myelination in the human fetal brain. Front Neuroanat 2009;3:5.

Jepson S et al. LINGO-1, a transmembrane signaling protein, inhibits oligodendrocyte differentiation and myelination through intercellular self-interactions. J Biol Chem 2012;287:22184-22195.

Kang SH et al. NG2+ CNS glial progenitors remain committed to the oligodendrocyte lineage in postnatal life and following neurodegeneration. Neuron 2010;68:668-681.

Kao HY et al. A histone deacetylase corepressor complex regulates the Notch signal transduction pathway. Genes Dev 1998;12:2269-2277.

Kasai M et al. Wnt signaling regulates the sequential onset of neurogenesis and gliogenesis via induction of BMPs. Genes Cells 2005;10:777-83.

Kassmann CM & Nave KA. Oligodendroglial impact on axonal function and survival - a hypothesis. Curr Opin Neurol 2008;21:235-241.

Kaye EM. Update on genetic disorders affecting white matter. Pediatr Neurol 2001;24:11–24.

Kessaris N et al. Competing waves of oligodendrocytes in the forebrain and postnatal elimination of an embryonic lineage. Nat Neurosci 2006; 9:173-179.

Kim DW & Lassar AB. Smad-dependent recruitment of a histone deacetylase/Sin3A complex modulates the bone morphogenetic protein-dependent transcriptional repressor activity of Nkx3.2. Mol Cell Biol 2003;23:8704-8717.

Kim H et al. Notch-regulated oligodendrocyte specification from radial glia in the spinal cord of zebrafish embryos. Dev Dyn 2008;237:2081-2089.

Kippert A et al. Rho regulates membrane transport in the endocytic pathway to control plasma membrane specialization in oligodendroglial cells. J Neurosci 2007;27:3560-3570.

Kleopa KA et al. Gap junction disorders of myelinating cells. Rev Neurosci 2010;21:397-419.

Koenning M et al. Myelin gene regulatory factor is required for maintenance of myelin and mature oligodendrocyte identity in the adult CNS. J Neurosci 2012;32:12528-12542.

Kohlschütter A & Eichler F. Childhood leukodystrophies: a clinical perspective. Expert Rev Neurother 2011;11:1485-1496.

Kohlschütter A et al. Leukodystrophies and other genetic metabolic leukoencephalopathies in children and adults. Brain Dev 2010;32:82-89.

Kondo T & Raff M. Basic helix–loop–helix proteins and the timing of oligodendrocyte differentiation. Development 2000;127:2989-2998.

Krämer EM et al. Oligodendrocytes direct glycosyl phosphatidylinositol-anchored proteins to the myelin sheath in -rich complexes. J Biol Chem 1997;272:8937-8945.

Kuronen M et al. Galactolipid deficiency in the early pathogenesis of neuronal ceroid lipofuscinosis model Cln8mnd: implications to delayed myelination and oligodendrocyte maturation. Neuropathol Appl Neurobiol 2012;38:471-486.

Lafrenaye AD & Fuss B. Focal adhesion kinase can play unique and opposing roles in regulating the morphology of differentiating oligodendrocytes. J Neurochem 2010;115:269-282.

Lanciotti A et al. Astrocytes: emerging stars in leukodystrophy pathogenesis. Transl Neurosci 2013;4: doi:10.2478/s13380-013-0118-1.

Lappe-Siefke C et al. Disruption of Cnp1 uncouples oligodendroglial functions in axonal support and myelination. Nature Genet 2003;33:366–374.

Laursen LS et al. An integrin-contactin complex regulates CNS myelination by differential Fyn phosphorylation. J Neurosci 2009;29:9174-9185.

Lebel C et al. Microstructural maturation of the human brain from childhood to adulthood. Neuroimage 2008;40:1044-1055.

Lee KK et al. Dominant-negative beta1 integrin mice have region-specific myelin defects accompanied by alterations in MAPK activity. Glia 2006;53:836-844.

Lee S et al. A culture system to study oligodendrocyte myelination processes using engineered nanofibers. Nat Methods 2012;9:917-922.

Leegwater PA et al. Mutations in MLC1 (KIAA0027), encoding a putative membrane protein, cause megalencephalic leukoencephalopathy with subcortical cysts. Am J Hum Genet 2001;68:831-838.

Levine JM & Nishiyama A. The NG2 chondroitin sulphate proetoglycan: a multifunctional proteoglycan associated with immature cells. Perspect Dev Neurobiol 1996;3:245-259.

Li H et al. Olig1 and Sox10 interact synergistically to drive myelin basic protein transcription in oligodendrocytes. J Neurosci 2007;27:14375-14382.

Li H & Richardson W. The evolution of Olig genes and their roles in myelination. Neuron Glia Biol 2008;4:129-135.

Li Q et al. Electrical stimulation of the medullary pyramid promotes proliferation and differentiation of oligodendrocyte progenitor cells in the corticospinal tract of the adult rat. Neurosci Lett 2010;479:128- 133.

Liang X et al. Signaling from integrins to Fyn to Rho family GTPases regulates morphologic differentiation of oligodendrocytes. J Neurosci 2004;24:7140-7149.

Lin ST & Fu YH. miR-23 regulation of lamin B1 is crucial for oligodendrocyte development and myelination. Dis Model Mech 2009;2:178-88.

Lin ST et al. Regulation of myelination in the central nervous system by nuclear lamin B1 and non- coding RNAs. Transl Neurosci 2014;3: doi: 10.1186/2047-9158-3-4.

Liu A et al. A molecular insight of Hes5- dependent inhibition of myelin gene expression: old partners and new players. EMBO J 2006;25:4833-4842.

Liu H et al. Histone deacetylase 11 regulates oligodendrocyte-specific gene expression and cell development in OL-1 oligodendroglia cells. Glia 2009;57:1-12.

Liu X et al. Specific regulation of NRG1 isoform expression by neuronal activity. J Neurosci 2011;31:8491-8501.

Lopez-Hernandez T et al. Mutant GlialCAM causes megalencephalic leukoencephalopathy with subcortical cysts, benign familial macrocephaly, and macrocephaly with retardation and autism. Am J Hum Genet 2011;88:422-432.

Low HP et al. Embryonic stem cell rescue of tremor and ataxia in myelin-deficient shiverer mice. J Neurol Sci 2009;276:133-137.

Lu JF et al. A mouse model for X-linked adrenoleukodystrophy. Proc Natl Acad Sci U S A 1997;94:9366-9371.

Lu QR et al. Sonic hedgehog – regulated oligodendrocyte lineage genes encoding bHLH proteins in the mammalian central nervous system. Neuron 2000;25:317-329.

Ludwin SK & Maitland M. Long-term remyelination fails to reconstitute normal thickness of central myelin sheaths. J Neurol Sci 1984;64:193-198.

Lundgaard I et al. Neuregulin and BDNF induce a switch to NMDA receptor-dependent myelination by oligodendrocytes. PLos Biol 2013;11:e1001743.

Lundgaard I et al. White matter astrocytes in health and disease. Neuroscience 2014;276:161-173.

Lunn KF et al. Myelin mutants: new models and new observations. Microsc Res Tech 1995;32:183-203.

Lutz SE et al. Deletion of astrocyte connexins 43 and 30 leads to a dysmyelinating phenotype and hippocampal CA1 vacuolation. J Neurosci 2009;29:7743-7752.

Mangin JM & Gallo V. The curious case of NG2 cells: transient trend or game changer? ASN Neuro 2011;3:e00052.

Mangin JM et al. Experience-dependent regulation of NG2 progenitors in the developing barrel cortex. Nat Neurosci 2012;15:1192-1194.

Markoullis K et al. Gap junction pathology in multiple sclerosis lesions and normal-appearing white matter. Acta Neuropathol 2012;123:873-886.

Mason JL et al. Insulin-like growth factor (IGF) signalling through type 1 IGF receptor plays an important role in remyelination. J Neurosci 2003;23:7710-7718.

McCarthy KD & de Vellis J. Preparation of separate astroglial and oligodendroglial cell cultures from rat cerebral tissue. J Cell Biol 1980;85:890-902.

McGrail K et al. Immunofluorescent localization of three Na, K-ATPase isozymes in the rat central nervous system: both neurons and glia can express more than one Na, K-ATPase. J Neurosci 1991;11:381-391.

Mei F et al. Stage-specific deletion of Olig2 conveys opposing functions on differentiation and maturation of oligodendrocytes. J Neurosci 2013;33:8454-8462.

Merzbacher L. Eine eigenartige familiärhereditäre Erkrankungsform (Aplasia axialis extra-corticalis congenita). Z Gesamte Neurol Psychiat 1910;3:1-138.

Mi S et al. LINGO-1 negatively regulates myelination by oligodendrocytes. Nat Neurosci 2005;8:745- 751.

Mi S et al. Promotion of central nervous system remyelination by induced differentiation of oligodendrocyte precursor cells. Ann Neurol 2009;65:304-315.

Mignot C et al. Alexander disease: putative mechanisms of an astrocytic encephalopathy. Cell Mol Life Sci 2004;61:369-385.

Mitew S et al. Mechanisms regulating the development of oligodendrocytes and central nervous system myelin. Neuroscience 2014;276:29-47.

Molofsky AV et al. Astrocytes and disease: a neurdevelopmental perspective. Genes Dev 2012;26:891- 907.

Morissette N & Carbonetto S. Laminin alpha 2 chain (M chain) is found within the pathway of avian and murine retinal projections. J Neurosci 1995;15:8067-8082.

Muller C et al. Making myelin basic protein: from mRNA transport to localized translation. Front Cell Neurosci 2013;7:169.

Murtie JC et al. PDGF and FGF2 pathways regulate distinct oligodendrocyte lineage responses in experimental demyelination with spontaneous remyelination. Neurobiol Dis 2005;19:171-182.

Nakatani H et al. Ascl1/Mash1 promotes brain oligodendrogenesis during myelination and remyelination. J Neurosci 2013;33:9752-9768.

Nair A et al. Astrocytes in multiple sclerosis: a product of their environment. Cell Mol Life Sci 2008;65:2702-2720.

Narayanan SP et al. Akt signals through the mammalian target of rapamycin pathway to regulate CNS myelination. J Neurosci 2009;29:6860-6870.

Nave KA. Myelination and support of axonal integrity by glia. Nature 2010a;468:244-252.

Nave KA. Myelination and the trophic support of long axons. Nat Rev Neurosci 2010b;11:275-283.

Naidu S. Clinical delineation of leukodystrophies. J Mol Neurosci 1999;12:185-192.

Nave KA & Werner HB. Myelination of the nervous system: mechanisms and functions. Annu Rev Cell Dev Biol 2014;30:503-533.

Nishiyama A et al. Polydendrocytes (NG2 cells): multifunctional cells with lineage plasticity. Nature Rev Neurosci 2009;10:9-22.

Niu J et al. Phosphorylated olig1 localizes to the cytosol of oligodendrocytes and promotes membrane expansion and maturation. Glia 2012;60:1427-1436.

Noble M et al. Platelet-derived growth factor promotes division and motility and inhibits premature differentiation of the oligodendrocyte/type-2 astrocyte progenitor cell. Nature 1988;333:560-562.

Notterpek LM et al. Myelination in cerebellar slice cultures: development of a system amenable to biochemical analysis. J Neurosci Res 1993;36:621-634.

Oblinger MM et al. Phosphofructokinase in the rat nervous system: regional differences in activity and characteristics of axonal transport. J Neurosci Res 1988:21:25-34.

Odermatt B et al. Connexin 47 (Cx47)-deficient mice with enhanced green fluorescent protein reporter gene reveal predominant oligodendrocytic expression of Cx47 and display vacuolized myelin in the CNS. J Neurosci 2003;23:4549-4559.

O’Meara RW et al. Integrin-linked kinase regulates process extension in oligodendrocytes via control of actin cytoskeletal dynamics. J Neurosci 2013;33:9781-9793.

Orentas DM et al. Sonic hedgehog signaling is required during the appearance of spinal cord oligodendrocyte precursors. Development 1999;126:2419-2429.

Ortega MC et al. Neuregulin-1/ErbB4 signaling controls the migration of oligodendrocyte precursor cells during development. Exp Neurol 2012;235:610-620.

Orthmann-Murphy JL et al. Loss-of-function GJA12/connexin47 mutations cause Pelizaeus– Merzbacher-like disease. Mol Cell Neurosci 2007;34:629-641.

Osterman B et al. Advances in the diagnosis of leukodystrophies. Future Neurol 2012;7:595-612.

Parras CM et al. The proneural gene Mash1 specifies an early population of telencephalic oligodendrocytes. J Neurosci 2007;27:4233-4242

Pelizaeus F. Über eine eigenartige familiäre Entwicklungshemmung vornehmlich auf motorischem Gebiete. Archiv Psych 1899;31:100-xx.

Pendleton JC et al. Chondroitin sulphate proteoglycans inhibit oligodendrocyte myelination through PTPσ. Exp Neurol 2013;247:113-121.

Perkins GA et al. Electron tomographic analysis of cytoskeletal cross-bridges in the paranodal region of the node of Ranvier in peripheral nerves. J Struct Biol 2008;161:469-480.

Perlman SJ & Mar S. Leukodystrophies. Adv Exp Med Biol 2012;724:154-171.

Peters A & Vaughn JE. Morphology and development of the myelin sheath. In: Davison AN & Peters A Eds. Myelination. Charles c Thomas Publisher, Springfield, IL, 1970.

Pogoda HM et al. A genetic screen identifies genes essential for development of myelinated axons in zebrafish. Dev Biol 2006;298:118-131.

Poser CM. Leucodystrophy as an example of dysmyelinating disease. Proc Third Intern Cong Neuropath. Brussels, Editions "Acta Medica Belgica" 1957:106-111.

Poser CM. Leukodystrophy and the concept of dysmyelination. Arch Neurol 1961;4:323-332.

Poser CM. Diseases of the myelin sheath, in Minckler J (ed): Pathology of the Nervous System. New York, McGraw-Hill Book Co Inc, 1968.

Poser CM. Dysmyelination revisited. Arch Neurol 1978;35:401-408.

Poser CM & van Bogaert L. natural history and evolution of the concept of Schilder’s diffuse sclerosis. Acta Psychiat Scand 1956;31:285-331.

Powers JM & De Vivo D. Peroxisomal and mitochondrial disorders. In Graham DI & Lantos PL (eds). Greenfield’s neuropathology. 7th ed. London: Arnold, 2002.

Prayoonwiwat N & Rodriguez M. The potential for oligodendrocyte proliferation during . J Neuropathol Exp Neurol 1993;52:55-63.

Pringle NP et al. Determination of neuroepithelial cell fate: induction of the oligodendrocyte lineage by ventral midline cells and sonic hedgehog. Dev Biol 1996;177:30-42.

Qi Y et al. Control of oligodendrocyte differentiation by the Nkx2.2 homeodomain transcription factor. Development 2001;128:2723-2733.

Quarles RH et al. Myelin formation, structure and biochemistry. In: Basic neurochemistry: molecular, cellular and medical aspects (eds. G.J. Siegel, et al.), 7th Edition, Elsevier, pp. 51-71.

Rademakers R et al. Mutations in the colony stimulating factor 1 receptor (CSF1R) gene cause hereditary diffuse leukoencephalopathy with spheroids. Nat Genet 2011;44:200-205.

Rajasekharan S et al. Netrin 1 and Dcc regulate oligodendrocyte process branching and membrane extension via Fyn and RhoA. Development 2009;136:415-426.

Ransohoff RM. Animal models for multiple sclerosis: the good, the bad and the bottom line. Nat Neurosci 2012;15:1074-1077.

Rao MV et al. The middle molecular mass subunit carboxyl-terminal tail domains is essential for the radial growth and cytoskeletal architecture of axons but not for regulating neurofilament transport rate. J Cell Biol 2003;163:1021-1031.

Rash JE. Molecular disruptions of the panglial syncytium block potassium siphoning and axonal saltatory conduction: pertinence to neuromyelitis optica and other demyelinating diseases of the central nervous system. Neuroscience 2010;168:982-1008.

Richardson WD et al. A role for platelet-derived growth factor in normal gliogenesis in the central nervous system. Cell 1988;53:309-319.

Robain O. The jimpy mouse. Acta Zool Pathol Antwerp 1977;68:68-92.

Roberts RE et al. Individual differences in expert motor coordination associated with white matter microstructure in the cerebellum. Cereb Cortex 2013;23:2282-2292.

Rodriguez-Pena A et al. Neonatal hypothyroidism affects the timely expression of myelin associated glycoprotein in the rat brain. J Clin Invest 1993;91:812-818.

Rosenberg SS et al. The geometric and spatial constraints of the microenvironment induce oligodendrocyte differentiation. Proc Natl Acad Sci U S A 2008;105:14662-14667.

Rosenbluth J et al. Subtle myelin defects in PLPnull mice. Glia 2006;54:172-182.

Rosenfeld J & Friedrich VL Jr. Axonal swellings in jimpy mice: does lack of myelin cause neuronal abnormalities? Neuroscience 1983;10:959-966.

Rowitch DH. Glial specification in the vertebrate neural tube. Nat Rev Neurosci 2004;5:409-419.

Sakry D et al. Synapses between NG2 glia and neurons. J Anat 2011;219:2-7.

Samanta J & Kessler JA. Interactions between ID and OLIG proteins mediate the inhibitory effects of BMP4 on oligodendroglial differentiation. Development 2004;131:4131-4142.

Sampaio-Baptista C et al. Motor skill learning induces changes in white matter microstructure and myelination. J Neurosci 2013;33:19499-19503.

Sanchez I et al. Local control of neurofilament accumulation during radial growth of myelinating axons in vivo. Selective role of site-specific phosphorylation. J Cell Biol 2000;151:1013-1024.

Sawaishi Y. Review of Alexander disease: beyond the classical concept of leukodystrophy. Brain Dev 2009;31:493-498.

Schiffmann R & van der Knaap MS. An MRI-based approach to the diagnosis of white matter disorders. Neurology 2009;72:750-759.

Schilder P. Zur Frage der Encephalitis periaxialis diffusa (sogenannte diffuse Sklerose). Z Gesamte Neurol Psychiatr 1913;15:359-376.

Schlegel AA et al. White matter structure changes as adults learn a second language. J Cogn Neurosci 2012;24:1664-1670.

Scholz J et al. Training induces changes in white-matter architecture. Nat Neurosci 2009;12:1370-1371.

Schulz K et al. Iron efflux from astrocytes plays a role in remyelination. J Neurosci 2012;32:4841-4847.

Schummers J et al. Tuned responses of astrocytes and their influence on hemodynamic signals in the visual cortex. Science 2008;320:1638-1643.

See JM & Grinspan JB. Sending mixed signals: bone morphogenetic protein in myelination and demyelination. J Neuropathol Exp Neurol 2009;68:595-604.

Seifert T et al. Notch1 and its ligand Jagged1 are present in remyelination in a T-cell- and antibody- mediated model of inflammatory demyelination. Acta Neuropathol 2007;113:195-203.

Shimizu T et al. Wnt signaling controls the timing of oligodendrocyte development in the spinal cord. Dev Biol 2005;282:397-410.

Shin D et al. Dicer ablation in oligodendrocytes provokes neuronal impairment in mice. Ann Neurol 2009;66:843-57.

Shakèd M et al. Histone deacetylases control neurogenesis in embryonic brain by inhibition of BMP2/4 signaling. PLoS ONE 2008;3:e2668.

Shen S et al. Histone modifications affect timing of oligodendrocyte progenitor differentiation in the developing rat brain. J Cell Biol 2005;169:577-589.

Shen S et al. Age-dependent epigenetic control of differentiation inhibitors is critical for remyelination efficiency. Nat Neurosci 2008;11:1024-34.

Sim FJ et al. The age-related decrease in CNS remyelination efficiency is attributable to an impairment of both oligodendrocyte progenitor recruitment and differentiation. J Neurosci 2002;22:2451-2459.

Sim FJ et al. Complementary patterns of gene expression by human oligodendrocyte progenitors and their environment predict determinants of progenitor maintenance and differentiation. Ann Neurol 2006;59:763-779.

Simon C et al. Progenitors in the adult cerebral cortex: cell cycle properties and regulation by physiological stimuli and injury. Glia 2011;59:869-881.

Simons M et al. Assembly of myelin by association of proteolipid protein with cholesteroland galactosylceramide-rich membrane domains. J Cell Biol 2000;151:143-154.

Simons M & Trotter J. Wrapping it up: the cell biology of myelination. Curr Opin Neurobiol 2007;17:533- 540.

Simons M et al. A unified cell biological perspective on axon-myelin injury. J Cell Biol 2014;206:335- 345.

Sirisi S et al. Megalencephalic leukoencephalopathy with subcortical cysts protein 1 regulates glial surface localisation of GLIALCAM from fish to humans. Hum Mol Genet 2014;23:5069-5086.

Sofroniew MV & Vinters HV. Astrocytes: biology and pathology. Acta Neuropathol 2010;119:7-35.

Sorensen A et al. Astrocytes, but not olfactory ensheathing cells or Schwann cells, promote myelination of CNS axons in vitro. Glia 2008;56:750-763.

Spassky N et al. Directional guidance of oligodendroglial migration by class 3 semaphorins and netrin-1. J Neurosci 2002;22:5992-6004.

Steenweg ME et al. Magnetic resonance imaging pattern recognition in hypomyelinating disorders. Brain 2010;133:2971-2982.

Stidworthy MF et al. Notch1 and Jagged1 are expressed after CNS demyelination, but are not a major rate-determining factor during remyelination. Brain 2004;127:1928-1941.

Sugimori M et al. Ascl1 is required for oligodendrocyte development in the spinal cord. Development 2008;135:1271-1281.

Suzuki L et al. Organization of cerebral projections to identified cerebellar zones in the posterior cerebellum of the rat. J Neurosci 2012;32:10854-10869.

Svaren J. MicroRNA and transcriptional crosstalk in myelinating glia. Neurochem Int 2014;77:50-57.

Swiss VA et al. Identification of a gene regulatory network necessary for the initiation of oligodendrocyte differentiation. PLoS One 2011;6:e18088.

Talbott JF et al. Endogenous Nkx2.2+/Olig2+ oligodendrocyte precursor cells fail to remyelinate the demyelinated adult rat spinal cord in the absence of astrocytes. Exp Neurol 2005;192:11-24.

Taniguchi Y et al. Sema4D deficiency results in an increase in the number of oligodendrocytes in healthy and injured mouse . J Neurosci Res 2009;87:2833-2841.

Tarrade A et al. A of spastin is responsible for swellings and impairment of transport in a region of axon characterized by changes in composition. Hum Mol Genet 2006;15:3544- 3558.

Taveggia C et al. Type III neuregulin-1 promotes oligodendrocyte myelination. Glia 2007;56:284-293.

Taylor CM et al. Detergent-insoluble glycosphingolipid/cholesterol microdomains of the myelin membrane. J Neurochem 2002;81:993-1004.

Torii T et al. Pelizaeus-Merzbacher disease: cellular pathogenesis and pharmacologic therapy. Pediatr Int 2014;56:659-666.

Trajkovic K et al. Neuron to glia signaling triggers myelin membrane exocytosis from endosomal storage sites. J Cell Biol 2006;172:937-948.

Trapp BD et al. Differentiation and death of premyelinating oligodendrocytes in developing rodent brain. J Cell Biol 1997;137:459-468.

Tress O et al. Pathologic and phenotypic alterations in a mouse expressing a connexin47 missense mutation that causes Pelizaeus-Merzbacher-like disease in humans PLoS Genet 2011;7:e1002146.

Tress O et al. Panglial gap junctional communication is essential for maintenance of myelin in the CNS. J Neurosci 2012:32:7499-7518.

Tsai HH et al. The chemokine receptor CXCR2 controls positioning of oligodendrocyte precursors in developing spinal cord by arresting their migration. Cell 2002;110:373-383.

Vallstedt A et al. Multiple dorsoventral origins of oligodendrocyte generation in the spinal cord and hindbrain. Neuron 2005;45:55-67. van der Knaap MS et al. Pattern recognition in magnetic resonance imaging of white matter disorders in children and young adults. Neuroradiology 1991;33:478-493.

van der Knaap MS et al. Defining and categorizing leukoencephalopathies of unknown origin: MR imaging approach. 1999;213:121-133. van der Knaap MS & Valk J. Magnetic resonance of myelination and myelin disorders. 3rd ed. Berlin Heidelberg: Springer-Verlag, 2005. van der Voorn JP et al. The leukoencephalopathy of infantile GM1 gangliosidosis: oligodendrocytic loss and axonal dysfunction. Acta Neuropathol 2004;107:539-545. van der Voorn JP et al. Childhood white matter disorders: quantitative MR imaging and spectroscopy. Radiology 2006;241:510-517.

Verkhratsky A & Parpura V. Recent advances in (patho)physiology of astroglia. Acta Pharmacol Sinica 2010;31:1044-1054.

Verkhratsky A et al. Neurotransmitters and integration in neuronal-astroglial networks. Neurochem Res 2012;37:2326-2338.

Verkhratsky A et al. Neurological diseases as primary gliopathies: a reassessment of neurocentrism. ASN Neuro 2012;4: doi:10.1042/an20120010.

Verkhratsky A et al. Astrogliopathology: a central element of neuropsychiatric disease? Neuroscientist 2014;20:576-588.

Viader A et al. MicroRNAs modulate Schwann cell response to nerve injury by reinforcing transcriptional silencing of dedifferentiation-related genes. J Neurosci 2011;31:17358-17369.

Vigano F et al. Transplantation reveals regional differences in oligodendrocyte differentiation in the adult brain. Nat neurosci 2013;16:1370-1372.

Wang S et al. Notch receptor activation inhibits oligodendrocyte differentiation. Neuron 1998;21:63-75.

Wang S & Yang KM. White matter plasticity in adulthood. Neuroscience 2014;276:148-160.

Wang Z et al. Contrasting effects of mitogenic growth factors on myelination in neuron-oligodendrocyte co-cultures. Glia 2007;55:537-545.

Wang J et al. Transcription factor induction of human oligodendrocyte progenitor fate differentiation. Proc Natl Acad Sci U S A 2014;111:E2885-2894.

Watanabe M et al. Transient upregulation of Nkx2.2 expression in oligodendrocyte lineage cells during remyelination. Glia 2004;46:311-322.

Watkins TA et al. Distinct stages of myelination regulated by gamma-secretase and astrocytes in a rapidly myelinating CNS coculture system. Neuron 2008;60:555-569.

Waxman SG & Bangalore L. Electrophysiological consequences of myelination. In RA Lazzarini (ed.), Myelin biology and disorders. San Diego, CA: Elsevier Academic Press, 2004, pp. 117-141.

White R & Kramer-Albers EM. Axon-glia interaction and membrane trafficking in myelin formation. Front Cell Neurosci 2014;7:284.

Windrem MS et al. Neonatal chimerization with human glial progenitor cells can both remyelinate and rescue the otherwise lethally hypomyelinated shiverer Mouse. Cell Stem Cell 2008;2:553-565.

Wolswijk G & Noble M. Identification of an adult specific glial progenitor cell. Development 1989;105:387-400.

Wong AW et al. Oligodendroglial expression of TrkB independently regulates myelination and progenitor cell proliferation. J Neurosci 2013;33:4947-4957.

Woodruff RH et al. Platelet-derived growth factor regulates oligodendrocyte progenitor numbers in adult CNS and their response following CNS demyelination. Mol Cell Neurosci 2004;25:252-262.

Xiao J et al. Extracellular signal-regulated kinase 1/2 signaling promotes oligodendrocyte myelination in vitro. J Neurochem 2012;122:1167-1180.

Xiao J et al. Brain-derived neurotrophic factor promotes central nervous system myelination via a direct effect upon oligodendrocytes. Neurosignals 2010;18:186-202.

Xin M et al. Myelinogenesis and axonal recognition by oligodendrocytes in brain are uncoupled in Olig1- null mice. J Neurosci 2005;25:1354-1365.

Yakovlev PA & Lecours IR. The myelogenetic cycles of regional maturation of the brain. In: Regional development of the brain in early life (Minkowski A, editor). Oxford: Blackwell 1967.

Yamaguchi M et al. Histone deacetylase 1 regulates retinal neurogenesis in zebrafish by suppressing Wnt and Notch signaling pathways. Development 2005;132:3027-3043.

Ye P et al. Myelination is altered in insulin-like growth factor-I null mutant mice. J Neurosci 2002;22:6041-6051.

Ye F et al. HDAC1 and HDAC2 regulate oligodendrocyte differentiation by disrupting the beta-catenin- TCF interaction. Nat Neurosci 2009;12:829-838.

Young EA et al. Imaging correlates of decreased axonal Na+/K+ ATPase in chronic multiple sclerosis lesions. Ann Neurol 2008;63:428-435.

Yu Y et al. Olig2 targets chromatin remodelers to enhancers to initiate oligodendrocyte differentiation. Cell 2013;152:248-261.

Yuan A et al. Cotransport of glyceraldehyde-3-phosphate dehydrogenase and actin in axons of chicken motoneurons. Cell Mol Neurobiol 1999;19:733-744.

Zara F et al. Deficiency in hyccin, a newly identified membrane protein, causes hypomyelination and congenital cataract. Nat Genet 2006;38:1111-1113.

Zawadzka M et al. CNS-resident glial progenitor/stem cells produce Schwann cells as well as oligodendrocytes during repair of CNS demyelination. Cell Stem Cell 2010;6:578-590.

Zeger M et al. Insulin-like growth factor type 1 receptor signaling in the cells of oligodendrocyte lineage is required for normal in vivo oligodendrocyte development and myelination. Glia 2007;55:400-411.

Zeman W & Whieldon JA. Clinical considerations in “Schilder’s disease”. Am J Dis Chil 1962;104:635- 643.

Zhang H et al. Central nervous system remyelination in culture - a tool for multiple sclerosis research. Exp Neurol 2011;230:138-148.

Zhang Y & Barres BA. Astrocyte heterogeneity: an underappreciated topic in neurobiology. Curr Opin Neurobiol 2010;20:588-594.

Zhao X et al. MicroRNA-mediated control of oligodendrocyte differentiation. Neuron 2010;65:612-26.

Zhou YX et al. Retroviral lineage analysis of fibroblast growth factor receptor signaling in FGF2 inhibition of oligodendrocyte progenitor differentiation. Glia 2006;54:578-590.

Zhu X et al. NG2 cells generate both oligodendrocytes and gray matter astrocytes. Development 2008;135:145-157.

Zhu X et al. Age-dependent fate and lineage restriction of single NG2 cells. Development 2011;138:745- 753.

Ziskin JL et al. Vesicular release of glutamate from unmyelinated axons in white matter. Nat Neurosci 2007;10:321-330.

Zizioli D et al. Molecular cloning and knockdown of galactocerebrosidase in zebrafish: new insights into the pathogenesis of Krabbe disease. Biochim Biophys Acta 2014;1842:665-675.

Zuchero JB & Barres BA. Intrinsic and extrinsic control of oligodendrocyte development. Curr Opin Neurobiol 2013;23:914-920.