Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Oct 10.
Published in final edited form as: Glia. 2021 Apr 14;69(10):2309–2331. doi: 10.1002/glia.24008

Mechanisms of demyelination and neurodegeneration in globoid cell leukodystrophy

M Laura Feltri 1,2,3, Nadav I Weinstock 1,2, Jacob Favret 1,4, Narayan Dhimal 1,2, Lawrence Wrabetz 1,2,3, Daesung Shin 1,4
PMCID: PMC8502241  NIHMSID: NIHMS1745195  PMID: 33851745

Abstract

Globoid cell leukodystrophy (GLD), also known as Krabbe disease, is a lysosomal storage disorder causing extensive demyelination in the central and peripheral nervous systems. GLD is caused by loss-of-function mutations in the lysosomal hydrolase, galactosylceramidase (GALC), which catabolizes the myelin sphingolipid galactosylceramide. The pathophysiology of GLD is complex and reflects the expression of GALC in a number of glial and neural cell types in both the central and peripheral nervous systems (CNS and PNS), as well as leukocytes and kidney in the periphery. Over the years, GLD has garnered a wide range of scientific and medical interests, especially as a model system to study gene therapy and novel preclinical therapeutic approaches to treat the spontaneous murine model for GLD. Here, we review recent findings in the field of Krabbe disease, with particular emphasis on novel aspects of GALC physiology, GLD pathophysiology, and therapeutic strategies.

Keywords: demyelination, Krabbe disease, leukodystrophy, neurodegeneration

1 |. INTRODUCTION

Myelination is an evolutionary adaptation of vertebrates, which facilitates saltatory electrical conduction along axons and allows for more rapid neural transmission (Tasaki, 1939). Myelin exists in both the central and peripheral nervous systems (CNS and PNS, respectively) and is formed by the wrapping of oligodendrocyte or Schwann cell plasma membrane around axons. This process is exquisitely regulated and many different genetic defects can result in a hypomyelinating leukodystrophy (reviewed in Wolf, Ffrench-Constant, & van der Knaap, 2021). Compared to other membranes, myelin has a very high lipid content and an unusual composition, specifically enriched in glycosphingolipids (galactosylceramide and sulfatide; Chrast, Saher, Nave, & Verheijen, 2011). Myelin is an incredibly stable structure (M. E. Smith, 1968), likely influenced by its unique lipid and protein composition. In fact, evidence of intact myelin sheath with still detectable galactosylceramide was present in nerve specimens from remnants of a 5,000 year old ancient hominoid (Hess et al., 1998). Despite its stability, myelin is continuously remodeled to some degree postdevelopment (Lasiene, Matsui, Sawa, Wong, & Horner, 2009; Yeung et al., 2014; Young et al., 2013) and in response to injury and disease. The process of myelin degradation is complex and relies on a number of proteins and mechanisms in both myelinating cells and cells that do not make myelin. Of known importance for myelin turnover is the presence of the glycosphingolipid specific hydrolyzing enzyme, galactosylceramidase (GALC).

Globoid cell leukodystrophy (GLD, also Krabbe disease, “KD”) was first described by the Danish neurologist Knud Krabbe (Krabbe, 1916). GLD is caused by mutations in the GALC gene, that encodes a lysosomal enzyme catabolizing the myelin lipid component galactosylceramide (GalCer) (Figure 1). GALC deficiency causes rapid and severe demyelination, neurodegeneration, and neuroinflammation of both the CNS and PNS (Figures 2 and 3). The incidence of GLD is estimated at 1:100,000–1:250,000 (Barczykowski, Foss, Duffner, Yan, & Carter, 2012) and is classified either as infantile-onset (presentation within the first year of life, mean survival 24 months) or late-onset (presentation after the first year of life, mean survival 4–6 years after onset; Orsini, Escolar, Wasserstein, & Caggana, 1993).

FIGURE 1.

FIGURE 1

Galactosphingolipid biology in myelinating oligodendrocyte. The myelin sheath is a compact layer of lipid-rich membranes produced by oligodendrocytes in the central nervous system and by Schwann cells in the peripheral nervous system. Galactosylceramide (GalCer), a glycosphingolipid synthesized by ceramide synthase (CERS2) and ceramide galactosyl transferase (CGT), is a major component of myelin, present in the extracellular side of the membrane (so-called intraperiod line), see inset. GalCer can be further processed into sulfatides by addition of a sulfate group (red dots) by cerebroside sulfotransferase (CST). GalCer is catabolized into galactose (blue hexagons) and ceramide by galactosylceramidase and saposin A in the lysosome. Deficiency of these enzymes cause globoid cell leukodystrophy. CERS2, Ceramide synthase 2; CGT, ceramide galactosyl transferase; CST, cerebroside sulfotransferase; MBP, myelin basic protein; PLP, proteolipid protein

FIGURE 2.

FIGURE 2

Cell-specific pathophysiology in the CNS of globoid cell leukodystrophy. “GALC +” area depicts cells that express galactosylceramidase enzyme and are physiologically normal. “GALC Null” area contains cells that lack galactosylceramidase enzyme and represent globoid cell leukodystrophy (GLD). GALC Null oligodendrocytes produce psychosine from GalCer via acid ceramidase, undergo diffuse loss of myelin, and contribute to secondary axonal degeneration. Psychosine in neurons causes axonal degeneration. Microglia and globoid cells are unable to digest myelin (GalCer) efficiently, accumulate cytosolic crystals and become proinflammatory, further contributing to axonal degeneration and compromising remyelination. Astrocytes become activated, presumably in response to the surrounding pathology

FIGURE 3.

FIGURE 3

Demyelination, axonal degeneration and globoid cells in the nervous system of GLD-Krabbe patients. (a) Luxol-Fast-Blue, periodic acid schiff (PAS) staining of the cerebellar white matter from a GLD patient shows profound demyelination (absence of blue-stained myelin), neurodegeneration (low density of purple stained axons, arrows) and Globoid cells filled with PAS positive glycolipids storage (arrowhead). (b) Epon semithin sections of brachial plexus nerve from a globoid Leukodystrophy patient show very low density of myelinated fibers, myelinated fibers with onion bulbs (arrowhead) and many demyelinated fibers (double arrowheads), all indicative of chronic demyelination. Fiber density is also low, suggestive of chronic axonal degeneration. A globoid cell macrophage is indicated with an asterisk. Tissue (a) and image (b) courtesy of Dr. Julia Kofler and the NDRD brain and tissue bank at the University of Pittsburgh and Krabbe Partners for Research

The human GALC gene is mapped to chromosome 14q13 (Cannizzaro, Chen, Rafi, & Wenger, 1994) and more than 130 mutations have thus far been catalogued in the Human Gene Mutation Database (HGMD). At least 128 of those mutations have been reported to cause GLD (http://www.hgmd.org; Wenger, Rafi, Luzi, Datto, & Costantino-Ceccarini, 2000). The most common mutation in the European patient population is a large deletion spanning exons 11–17 (Rafi, Luzi, Chen, & Wenger, 1995), c.1161+6532_polyA+9kbdel (IVS10del30kb) that is always present with a c.550C>T (502C>T) polymorphism on the same allele, which eliminates the entire coding region for the 30-kDa enzyme subunit and about 15% of the coding region for the 50-kDa subunit. This mutation is absent in the Japanese population, which is another large GLD cohort. Instead, Japanese patients have a 12 bp deletion along with a 3 bp new insertion either c.683_694del12insCTC or c.2002A>C (635_646del12insCTC or 1954A>C; C. Xu, Sakai, Taniike, Inui, & Ozono, 2006), resulting in deletion of 5 amino acids and insertion of 2 amino acids that affects the formation of GALC quaternary structure (Tatsumi et al., 1995; Wenger, Rafi, & Luzi, 1997). In addition, a high prevalence of the infantile form was detected in two separate inbred communities in Israel. The Druze population commonly have a c.1796T>G (1748T>G) transversion in exon 15, while the Moslem Arab population have a c.1630G>A (1582G>A) transition in exon14 (Rafi, Luzi et al., 1996).

The infantile form of GLD accounts for almost 90% of the total cases of the patients (Orsini et al., 1993). Clinical features of the classical infantile form of GLD include progressive neurologic deterioration with decorticate posturing, peripheral neuropathy, blindness, deafness, autonomic instability, and death (Orsini et al., 1993). Initial symptoms of untreated children include irritability, feeding difficulties and poor head control. As the disease progresses, affected children experience loss of vision along with hyperactive reflexes and deterioration of motor and cognitive function (Suzuki, 2003). Children with GLD and/or other leukodystrophies are usually also prone to respiratory and urinary tract infections (Anderson et al., 2014) as well as a host of secondary complications. The most advanced stages of infantile GLD are very severe and progress to paralysis and severe neurologic decline (Escolar, West, Dallavecchia, Poe, & LaPoint, 2016).

While supportive care significantly improves the quality of life for GLD patients, hematopoietic stem cell therapy (HSCT) is the only approved disease-modifying treatment for GLD, and only asymptomatic patients benefit from this procedure.

Despite the relatively low incidence of KD, an extensive number of research studies have explored disease pathogenesis and therapeutic interventions in KD. This was facilitated by the long-term availability of spontaneously occurring disease models of KD among various species including dogs (Fletcher, Suzuki, & Martin, 1977; Hirth & Nielsen, 1967; Victoria, Rafi, & Wenger, 1996), cats (Johnson, 1970; Sigurdson, Basaraba, Mazzaferro, & Gould, 2002), sheep (Pritchard, Napthine, & Sinclair, 1980), and primates (Baskin et al., 1998; Luzi, Rafi, Victoria, Baskin, & Wenger, 1997). In particular, the twitcher mouse model, first characterized in 1980 by the Jackson laboratory, was one of the earliest leukodystrophy and LSD models discovered in mice (Duchen, Eicher, Jacobs, Scaravilli, & Teixeira, 1980). Accordingly, there have been nearly 100 studies using the twitcher mouse model, ranging in focuses from disease mechanism, gene therapy (GT), stem cell transplantation, and remyelination strategies. Despite the abundance of research, a number of underlying mechanistic questions remain unanswered, including the normal biology and function of GALC and the pathogenesis of demyelination resulting from its absence. Here, we review the current understanding of GALC, GLD pathogenesis, and therapeutic strategies, focusing on recent work that has provided a new understanding of this process.

2 |. PHYSIOLOGIC ROLE OF GALC

2.1 |. Galactosylceramide and myelin

The major physiologic substrate for GALC is the sphingolipid galactosylceramide (GalCer). Sphingolipids are a group of bioactive lipids that share a common sphingosine backbone. Sphingolipids act both as structural components and signaling molecules regulating cell death, proliferation, and cell differentiation. The structural makeup of sphingolipids includes a sphingolipid backbone, which can be modified with a fatty acid group to form ceramide (reviewed in Gault, Obeid, & Hannun, 2010). The two major pathways of sphingolipid metabolism include a de novo synthesis pathway, in which serine, palmityl-coA, and stearol-coA are enzymatically modified to generate ceramides. Alternatively, a sophisticated salvage pathway exists, in which sphingolipids can be extensively modified to form a number of different species with unique functions. Within the salvage pathway, there are six different endoplasmic reticulum (ER) enzymes that exist for ceramide synthesis, CerS 1–6, each of which generates ceramides of unique fatty acid lengths (Gault et al., 2010). The length of the fatty acid component dictates the biophysical properties and hydrophobicity of the sphingolipid. For example, myelin ceramides tend to be of very long fatty acid length (C22–C24) and are generated primarily by CerS2 (Imgrund et al., 2009). Ceramides are subsequently modified with various polar head groups including glucose or galactose moieties in variable sequences (gangliosides, globosides, and glycosphingolipids) or phosphocholine (sphingomyelin). Both sphingosine and ceramide can also be phosphorylated, to form sphingosine-1-phosphate (S1P) and ceramide-1-phosphate, important mediators of cell survival and death. Interestingly, a number of S1P analogues are popular pharmacologic agents for demyelinating diseases like Multiple Sclerosis, though their mechanism is thought to work primarily through immune modulation (Brinkmann et al., 2010). Notably, a recent article reports a that S1P receptor agonist improves demyelination in twitcher mice (Béchet, O’Sullivan, Yssel, Fagan, & Dev, 2020).

GalCer is a sphingolipid component of the outer leaflet of plasma membranes in many different cell types (Blanchette, Lin, Ratto, & Longo, 2006) and is synthetized by the galactosylation of ceramide by an ER enzyme, ceramide galactosyl transferase (CGT, [Sprong et al., 1998]). Like most sphingolipids, both hydroxylated and non-hydroxylated forms of GalCer exist, though their specific functions are not well understood. While GalCer may be expressed by many cell types, it is especially abundant in the nervous system where it represents 2% of grey matter and 12% of white matter, hence its historic terminology as “cerebroside” or “galactocerebroside” dating to 1874 (Fry, Weissbarth, Lehrer, & Bornstein, 1974; Thudichum, 1874). The abundance of GalCer in brain white matter is due to its enriched localization in myelin sheaths, created by oligodendrocyte membranes (or Schwann cells in the peripheral nervous system; Figure 1). Indeed, GalCer reaches up to 24% of total lipids in myelin sheaths (Garbay, Heape, Sargueil, & Cassagne, 2000; Norton & Cammer, 1984; O’Brien, 1965). In fact, the popular oligodendrocyte marker “O1” recognizes a GalCer antigen (Bansal, Warrington, Gard, Ranscht, & Pfeiffer, 1989; Sommer & Schachner, 1981). GalCer also serves as a substrate for synthesis of the sphingolipid sulfatide, which is another myelin-enriched lipid component (recognized by the “O4” oligodendrocyte marker). Of note, the myelin predominant species of both GalCer and sulfatide contain very long fatty acyl chains comprised of 22–24 carbons. This is in contrast to the composition of most cellular sphingolipids, which often span 16–18 carbons in length.

It is hypothesized that the function of myelin GalCer and sulfatide aid in membrane compaction and stability. These galactosphingolipids are localized in the extracellular surfaces of myelin, forming the intraperiod line in the wrapped myelin sheath (Boggs, Gao, & Hirahara, 2008; Figure 1). Knockout animals for CGT, which were unable to generate GalCer or sulfatide, had only minor defects in both CNS and PNS myelin formation and compaction, but a clinical phenotype possibly due to altered neurophysiological properties of myelin and defects in myelin maintenance (splitting; Bosio, Binczek, & Stoffel, 1996; Coetzee et al., 1996). The altered neurophysiological properties of CGT-null myelin are likely due to disruption of axonal paranodal domains (Dupree, Girault, & Popko, 1999). Interestingly, myelin of the CGT-null tissues was paradoxically enriched for glucosylceramide (GluCer), suggesting a compensatory presence of this lipid for myelin compaction, but not for myelin insulating function. Surprisingly, further investigation revealed an overall minor role of GluCer in myelin function. Independent oligodendrocyte deletion of UDP-glucose ceramide glucosyltransferase (Ugcg), thereby preventing the synthesis of glucosylceramide, did not cause a myelin defect (Saadat et al., 2010). Similarly, compound deletion of oligodendrocyte UGCG and global CGT strongly resembled the global CGT-null myelin, suggesting that the function of myelin galactosphingolipids is important and distinct from glucosphingolipids (Saadat et al., 2010). Interestingly, sulfatides, rather than GalCer, seems to be crucial for formation of paranodal domains and to prevent myelin splitting. Mice with genetic deletion of galactosyl 3′-sulfotransferase (CST), which prevents the generation of sulfatide while permitting the generation of GalCer, had similar paranodal anomalies (Honke et al., 2002). This highlights the unique functional roles in myelin of galactosylceramide and sulfatides from other sphingolipids and emphasizes the necessity for further investigation regarding myelin sphingolipid structure and function.

2.2 |. Lysosomal GALC

The myelin sphingolipid substrate GalCer is degraded by the lysosomal hydrolase GALC. Lysosomes are membrane bound organelles that serve as the major site of macromolecule catabolism in the cell. There are currently over 200 lysosomal proteins identified (Pu, Guardia, Keren-Kaplan, & Bonifacino, 2016), categorized generally as acid hydrolases involved in specific catabolic processes or lysosome membrane proteins involved in lysosome stability and regulation. Lysosomes are maintained at a pH of 4.5–5 by an ATP-dependent proton pump and contain >50 different hydrolytic enzymes (hydrolases) that catabolize a large range of macromolecules including glycosides, sulfates, phosphates, lipids, phospholipids, proteins, and nucleic acids.

The deficiency of a single lysosomal hydrolase will result in the inability to degrade the enzyme’s respective substrate, resulting in a specific lysosomal storage disorder (LSD; reviewed in Ferreira & Gahl, 2017). Nearly one third of the lysosomal hydrolases are involved in lipid metabolism (Lübke, Lobel, & Sleat, 2009), nine of which specifically degrade sphingolipids. The sphingolipid polar head groups are cleaved by one of eight different hydrolases, while acid ceramidase cleaves the final ceramide backbone into sphingosine. Additionally, there are multiple sphingolipid hydrolase coactivators that facilitate sphingolipid catabolism. Of particular interest are the prosaposin derived activators, saposins A–D, which are transcribed and cleaved from a common precursor. Each saposin seems to have the ability to activate the function of a specific sphingolipid hydrolase, emphasizing the nonoverlapping nature of these enzymes (Schuette, Pierstorff, Huettler, & Sandhoff, 2001).

Precursor GALC is synthesized in the ER as a 80-kDa precursor protein with three structural domains. Like many lysosomal hydrolyzing enzymes, it is cleaved in the lysosome into two subunits, a 50- and a 30-kDa subunit. The functional significance for the cleavage of GALC remains unknown. In vitro data suggests that the precursor form may be enzymatically active, though the pH requirements for GALC function imply that most GALC activity likely occur in the lysosome, where the cleaved forms predominate (reviewed in Shin, Feltri, & Wrabetz, 2016). Like other sphingolipid enzymes, GALC works in concert with one of the four saposins, specifically saposin A (SapA), thought to help shuttle the sphingolipid to the enzymatic active site. Recent crystal structure data revealed that GALC functions as a compound heterodimer comprised of two units of GALC and two units of SapA (Hill et al., 2018). Genetic knockout mice of SapA showed similar (albeit delayed) pathology to GALC knockout animals (Matsuda et al., 2001), emphasizing the congruent functions. A small number of patients with clinical and pathological features resembling GLD have been identified which include mutations in the SapA domain of PSAP rather than GALC (Spiegel et al., 2005).

2.3 |. Trafficking of GALC and cross-correction

Lysosomal hydrolases are modified in the trans Golgi network with mannose-6-phosphate (M6P) residues via GlcNAc phosphotransferase. M6P-tagged hydrolases are then properly sorted for lysosomal delivery via M6P receptor (MPR) binding in the Golgi (Coutinho, Prata, & Alves, 2012). In the low pH of the lysosome, the hydrolase-MPR complex dissociates. Hydrolases remain in the lysosomal matrix while unbound MPR is recycled to the Golgi apparatus. Some of the synthesized M6P-tagged enzymes is thought to escape the lysosome targeting and are secreted in the extracellular space where they can be taken up by cells using the same MPR, which are found (at reduced levels) on the plasma membrane. This process thereby provides a mechanism, termed cross-correction, in which neighboring cells can share lysosomal enzyme and is central to the rationale for many LSD therapies.

While cross-correction of lysosomal enzyme is an important concept in the LSD field, the extent of its role in biology and therapy remains to be precisely defined. Most studies have focused on in vitro characterization of this process (Fratantoni, Hall, & Neufeld, 1969; Neufeld & Fratantoni, 1970), with an emphasis on supplying lysosomal enzyme to neighboring deficient cells. However, some evidence has emerged questioning the efficiency of cross-correction in vivo in GLD, Metachromatic leukodystrophy and other neurodegenerative LSDs (Y. Kondo, Wenger, Gallo, & Duncan, 2005; Matthes et al., 2015). More recent experiments using in vitro and in vivo GALC knockout strategies showed that GALC negative or mutant cells are not able to uptake GALC through the M6P receptor pathway as efficiently as wild type cells (Weinstock et al., 2020). Furthermore, the overall biologic significance of GALC cross-correction in physiologic conditions remains a mystery. A recent conditional mouse was specifically engineered to produce a Cre-dependent, LAMP1-GALC fusion protein that maintained GALC activity while being tethered in the membrane of lysosomes, thus preventing secretion and cross-correction (Mikulka et al., 2020). Interestingly, twitcher mice expressing the LAMP1-GALC fusion protein did not have any symptoms and survived normally. This suggests that there is no apparent function for GALC secretion in normal physiology. Therefore, the role of cross-correction in therapy and biology for GALC and other lysosomal hydrolases is a major question for the field moving forward.

2.4 |. Expression of GALC

The cellular expression of GALC seems to reflect the localization of its substrate GalCer, as it is expressed ubiquitously by all cell types of the nervous system though enriched in oligodendrocytes (Zhang et al., 2014). Interestingly, a growing body of evidence is emerging regarding the expression of GALC in nonmyelinating cells. For example, GALC is highly expressed by neurons in a number of studies (Dolcetta et al., 2004; Weinstock et al., 2020; Zhang et al., 2014). Similarly, recent data indicates that macrophages, which normally produce low levels of GALC, dramatically upregulate GALC expression when recruited to sites of demyelination (Weinstock, Shin, et al., 2020). This raises the possibility that GALC gene expression can be dynamically regulated when lysosomes are challenged by the accumulation of its substrate. GALC protein is also expressed in astrocytes and microglia (Weinstock, Kreher, et al., 2020), and in several non-nervous system tissues such as kidney.

In addition to variability in the cellular expression of GALC, the temporal expression of GALC is also of interest. Previous studies hinted that GALC rescue experiments in GALC deficient mice and humans were most efficacious when delivered as early as possible (Rafi, Rao, Luzi, Curtis, & Wenger, 2012). While early treatment may seem intuitive, the precise temporal requirements for GALC were not previously recognized. A recent study found that GALC expression peaks in murine brain tissues shortly after birth, approximately 4–6 days postnatally. Intriguingly, temporal deletion of GALC at or before this period correlated with the development of more severe disease and suggests a precise mechanism governing GALC expression in early development (Weinstock, Kreher, et al., 2020).

The transcriptional regulation of GALC has not yet been elucidated, but transgenic mouse studies using a BAC expressing the human GALC gene and its upstream regulatory sequence, was sufficient to prevent GLD pathology in twitcher mice (De Gasperi et al., 2004). More recently, a landmark study showed that a master lysosomal transcription factor, TFEB, recognizes CLEAR elements and is present in this promoter region of GALC (Sardiello et al., 2009). By this theory, lysosomal enzymes are co-transcribed and should be co-expressed among all cell types. Data regarding more selective mechanisms of expression of unique lysosomal enzymes is limited. Nonetheless, a strong body of work surrounding TFEB has now been characterized regarding lysosomal biogenesis in response to amino acid metabolism. For example, when amino acid pools in the lysosome are low, the master nutritional sensor mTOR, which docks at the lysosomal membrane, releases inhibition of TFEB to orchestrate signaling cascades that induce autophagy and lysosomal biogenesis (Settembre et al., 2012). This work contributed to a paradigm shift to redefine the lysosome from a “dead-end” metabolic organelle, to a dynamic metabolic signaling hub for the regulation of the cell function. Whether a similar homeostatic process occurs in response to sphingolipids and GalCer metabolism remains to be determined.

3 |. PATHOPHYSIOLOGY OF GLD

3.1 |. Psychosine toxicity

As GALC is a lysosomal enzyme, GLD belongs to the superfamily of LSDs, characterized by widespread and severe tissue damage, engorgement and misfunction of lysosomes. However, unlike other LSDs, the primary GALC substrate, GalCer, does not broadly accumulate in patient tissues, and paradoxically even decreases (Vanier & Svennerholm, 1974). While this change may reflect overall myelin loss, in which GalCer is highly concentrated, the biochemical explanation for why GalCer does not accumulate relative to other lipids has been a major question in the field since the initial characterization of KD. Instead, a minor substrate, psychosine, also known as galactosylsphingosine, accumulates. Psychosine is categorized as a lyso-sphingolipid, meaning it appears like a typical sphingolipid, in this case galactosylceramide, minus its acyl chain (Figure 2).

Psychosine is almost undetectable in normal tissues, is a potent toxin and is considered a major culprit for demyelination, loss of oligodendrocytes, and neuron degeneration in KD (“Psychosine hypothesis”; Miyatake & Suzuki, 1972). Psychosine is a known inducer of in vitro cell death (Tohyama, Matsuda, & Suzuki, 2001) in different cell types (Haq, Giri, Singh, & Singh, 2003; Jatana, Giri, & Singh, 2002; Tanaka & Webster, 1993), including those which express GALC (Formichi et al., 2007). Although the cytotoxicity of psychosine, in vitro, is well established, the actual mechanism has been difficult to elucidate and may be multifaceted. Descriptions of psychosine toxicity include inhibition of Protein Kinase C (PKC; Hannun & Bell, 1987, 1989), mitochondrial Cytochrome C oxidase (Igisu, Hamasaki, Ito, & Ou, 1988; Igisu & Nakamura, 1986; Ribbens, Moser, Hubbard, Bongarzone, & Maegawa, 2014), and caspase activation (Zaka & Wenger, 2004). Furthermore, psychosine has been shown to induce dispersion of subcellular organelles like the trans Golgi network and endosomal vesicles (Kanazawa, Takematsu, Yamamoto, Yamamoto, & Kozutsumi, 2008) and cause defects in peroxisomal (Khan, Haq, Giri, Singh, & Singh, 2005) and mitochondrial (Voccoli, Tonazzini, Signore, Caleo, & Cecchini, 2014) function. It was speculated that psychosine may exert its toxicity through a plasma membrane receptor, TDAG8, that would function as a “psychosine receptor” (Im, Heise, Nguyen, O’Dowd, & Lynch, 2001). However, a study using the enantiomer of psychosine, which in theory should prevent specific protein interactions, still had high toxicity (Hawkins-Salsbury et al., 2013). Recently, psychosine was proposed to alter the pH and degradative capacity of lysosomes, thus inducing broad lysosomal dysfunction (Folts, Scott-Hewitt, Pröschel, Mayer-Pröschel, & Noble, 2016).

Currently, the most widely accepted theory of psychosine toxicity is that psychosine (and other lysosphingolipids) can act as a detergent and disturb membrane microdomain organization of lipid rafts, thereby causing cell death (Hawkins-Salsbury et al., 2013; Spassieva & Bieberich, 2016; White et al., 2009). Psychosine can accumulate in lipid rafts of myelinating glia in brains and nerves very early in the disease process (White et al., 2009). In fact, psychosine can and does accumulate in a wide range of cell types in KD mice including blood cells and thymus cells (Chuang et al., 2013; Galbiati et al., 2007; Zhu, Lopez-Rosas, Qiu, Van Breemen, & Bongarzone, 2012). An emerging body of work also suggests that psychosine can accumulate in lipid raft organization of neurons and directly induce neuronal apoptosis (Castelvetri et al., 2011; Teixeira et al., 2014) or dysfunction (Castelvetri et al., 2011; White et al., 2009; White et al., 2011), and axonal degeneration due to abnormal neurofilament phosphorylation (Cantuti-Castelvetri et al., 2012), fast axonal transport (Cantuti Castelvetri et al., 2013), and neuromuscular junction function (Cantuti-Castelvetri et al., 2015). Psychosine has even been proposed to cause α-synuclein formation and neurodegeneration in Krabbe mice and patients (B. R. Smith et al., 2014). Therefore, the proposed mechanisms of psychosine toxicity are broad and much question still remains regarding the direct mechanism of psychosine-induced pathogenesis in KD.

3.2 |. The molecular origin of psychosine

Even more controversial than the biological effect of psychosine in KD has been the biochemical and cellular origin of psychosine synthesis. Although a catabolic pathway from the direct deacylation of GalCer was logical, postulated decades ago, and repeatedly investigated, no biochemical evidence to support formation of psychosine via GalCer deacylation were achieved (reviewed in Suzuki, 2016). This led to the belief that psychosine is synthesized anabolically through the addition of galactose to sphingosine (Cleland & Kennedy, 1960; Y. N. Lin & Radin, 1973). However, the question remained of why the undigested GalCer did not accumulate, except possibly in crystals found abundantly in myelinating cells and globoid cells throughout the nervous system (Duchen et al., 1980; Irino & Suzuki, 1990; Jacobs, Scaravilli, & De Aranda, 1982; Okeda, Suzuki, Horiguchi, & Fujii, 1979; Takahashi, Igisu, Suzuki, & Suzuki, 1983; Yajima, Fletcher, & Suzuki, 1977).

In 2019, genetic experiments in the laboratory of Dr. Mark Sands solved this mystery. Genetic deletion of the catabolic enzyme acid ceramidase (Asah1) in twitcher mice completely abolished psychosine production, proving that psychosine is generated by the deacylation of GalCer by acid ceramidase (Li et al., 2019). This study was also significant for showing the direct toxic effects of psychosine, in vivo, and the extent of its contribution to pathology. The implication from this study is that GalCer does not accumulate significantly in KD because it is readily degraded into psychosine. Furthermore, this biochemical discovery has potential for significant advances for patients with KD, especially if an acid ceramidase inhibitor aimed at preventing psychosine formation proves efficacious. One note of caution for inhibiting acid ceramidase for therapeutic purpose is that acid ceramidase deficiency causes Farber Disease, a severe LSD characterized by accumulation of ceramide. The phenotype of compound twitcher/Farber mice was similar to that of Farber mice alone and less severe than twitcher alone. These compound mice had longer life spans compared to twitcher mice, as well as reduced demyelination and fewer globoid cells, ultimately proving the “psychosine hypothesis”: psychosine is indeed responsible for much of the demyelination and axonal degeneration present in KD. However, given the confounding effect of acid ceramidase deficiency that caused a Farber Disease phenotype in the mice, it was not possible to determine if psychosine is sufficient to explain the entirety of twitcher pathology. In fact, several lines of evidence suggest that psychosine, even if clearly toxic and an important cause of pathology, is not sufficient to cause the entirety of the KD phenotype. For example, twitcher mice completely deficient for the synthetic GalCer enzyme (CGT) were unable to produce psychosine but instead had a neurodegenerative phenotype (Ezoe et al., 2000a). Alternatively, mice replicating the human KD mutation GALC E130K, termed twi-5J, had less psychosine than traditional twitcher mice, but had a more severe phenotype (Potter et al., 2013).

A second major historic question in the field surround the cellular origin of psychosine production. Studying twitcher mice for over 40 years has yielded many important tenants of KD pathology including characterization of the severe demyelination occurring in the CNS and PNS, the prominent role of neurodegeneration and axonal pathology and the contribution of globoid cells to neuroinflammation (Duchen et al., 1980). However, given the ubiquitous nature of the W339X Galc nonsense mutation in twitcher, question regarding cellular autonomy and the cellular origin of psychosine were challenging. The use of conditional mutagenesis in knock-out mice have been used for almost three decades (Gu, Zou, & Rajewsky, 1993) to elucidate cell autonomy in development and in many genetic diseases. However, the application of Cre-Lox technology to lysosomal storage diseases has generally been limited, due to the suspicion that cross-correction of GALC by nonrecombined cells would mask any phenotype caused by cell-specific GALC deletion.

Recently, two groups have generated independent approaches to floxed Galc animals to further pursue the question of cellular autonomy in KD pathogenesis. The first approach engineered a “non cross-correctable” Cre-dependent, LAMP1-GALC fusion protein expressed artificially by the Rosa26-promoter (Mikulka et al., 2020). Fusion GALC in these mice is tethered to the lysosomal membrane protein LAMP1. The second group followed a more traditional approach and engineered a GALC floxed allele allowing endogenous GALC expression, secretion, and thus cross-correction (Weinstock, Kreher, et al., 2020; Weinstock, Shin, et al., 2020) In both studies, Galc floxed animals were crossed to Mpz-Cre, which express Cre specifically in Schwann cells. The data generated from both studies showed definitively that Schwann cells produce psychosine autonomously, which is sufficient to generate a severe demyelinating neuropathy. The similarity of phenotypes of these two mutant mice, one impeding and one retaining the possibility of cross-correction, strongly suggests that cross-correction of GALC by unrecombined cells does not occur in vivo (Weinstock, Shin, et al., 2020). Further experiments in vitro showed that one reason for poor cross-correction is that GALC knockout or mutant cells are not able to uptake GALC through the M6P receptor pathway as efficiently as wild type cells. Importantly, despite equal amount of psychosine, the amount of demyelination and axonal degeneration was more severe in the global knockout than in the Schwann cell specific knockout, further indicating that psychosine does not account for the entirety of GLD pathology, and also that cells other than Schwann cells need GALC and contribute to overall disease.

3.3 |. Macrophages and globoid reaction

GLD was originally diagnosed pathologically by the presence of pathognomonic Periodic Acid-Schiff (PAS) stained “globoid cells” in brains of autopsy patients (Figure 3). These multinucleated cells have a traditional storage phenotype including distended cytoplasm, engorged lysosomes, unique lipid crystals (presumably GalCer) (Austin, 1963; Jacobs et al., 1982; Yunis & Lee, 1970) and partially digested myelin sheaths. Globoid cells accumulate in areas of highest demyelination in KD tissues (Figure 3; D’Agostino, Sayre, & Hayles, 1963), and these storage-laden macrophages follow the spatio-temporal pattern of normal myelination (Taniike & Suzuki, 1994). There has been some disagreement as to whether globoid cells comes from infiltrating monocyte-derived macrophages or innate microglia (Nicaise, Bongarzone, & Crocker, 2016). Frequent observations of PAS-positive globoid cells and macrophages surrounding blood vessels in KD brains suggested that globoid cells represent infiltrating monocyte-derived macrophages from the periphery. Similarly, there has been some question as to whether macrophages can be converted to globoid cells via galactosylceramide (Austin & Lehfeldt, 1965) or psychosine. In addition to questions about the mechanistic origin of globoid cells, a larger question regarding their role in pathogenesis and the significance in disease progression remained unanswered and largely speculative.

The recently generated floxed Galc mouse was able to shed light on some of these important questions. The most striking difference between nerves from global and Schwann cell conditional knockout mice was the presence of globoid cells in the former but not the latter, pointing to macrophages and innate immune cells as independently requiring GALC (Weinstock, Shin, et al., 2020). Indeed, in vivo and in vitro experiments confirmed that macrophages require GALC to assist in phagocytosis and digestion of myelin. Specifically, when GALC deficient macrophages are confronted with GalCer (as opposed to psychosine), they transform into globoid cells and express a molecular phenotype associated with impaired phagocytosis, oxidative stress and a pro-inflammatory phenotype. Furthermore, the presence of globoid cells seems to negatively impact the overall health of the nerve and contributes to additional inflammation, demyelination and axonal degeneration.

Interestingly, the well-documented benefit of HSCT in presymptomatic KD patients seems to also exert its effect by providing the nervous system with phagocytic competent cells, rather than by cross-correction of GALC (Weinstock, Shin, et al., 2020). Analogous conclusions were recently obtained in a postmortem analysis of brains of patients with metachromatic leukodystrophy, a lysosomal storage disease due loss of function of Arylsulfatase A (Wolf et al., 2020). These data are also in line with previous studies that showed that GALC haplo-insufficient mice had mild defects in remyelination following cuprizone-induced demyelination (Scott-Hewitt et al., 2017; Scott-Hewitt, Folts, & Noble, 2018). The association of GALC-deficiency and associated myelin-induced inflammation is particularly fascinating as GALC was recently discovered to be a possible risk factor in patients with Multiple Sclerosis (Sawcer et al., 2011).

The direct ability for GalCer to produce toxicity is also particularly interesting because the same GalCer inclusions found in globoid cells are also present in Schwann cells and oligodendrocytes. Therefore, GalCer may contribute to disease pathology via its redistribution from normal myelin to abnormal crystal inclusions. A number of recent studies have shown that crystal accumulation in cells can be toxic due to the formation of the NOD−, LRR−, and pyrin domain-containing protein 3 (NLRP3) inflammasome (Duewell et al., 2010; Rajamäki et al., 2010) or lysosome membrane permeabilization (LMP; Ono et al., 2018). Various types of crystals have been documented to converge in a similar pathological mechanism in a wide range of pathologies affecting the innate immune system (in particular macrophages) including gout (Martinon, Pétrilli, Mayor, Tardivel, & Tschopp, 2006), asbestos and silica lung disease (Dostert et al., 2008), atherosclerosis (Duewell et al., 2010), and even noncrystal molecules like amyloid-beta in Alzheimer’s disease (Halle et al., 2008). It should also be noted that myelin derived crystals were recently found to induce pathology in a demyelinating mouse model, though these crystals were proposed to be comprised of cholesterol (Cantuti-Castelvetri et al., 2018). The role of GalCer or crystal toxicity may, therefore, contribute to GLD pathology though much remains unknown.

3.4 |. Neurodegeneration

It has been suggested that neuronal pathology could be affected by neuron-autonomous GALC, independently from demyelination or neuroinflammation. Along this line, GALC is highly expressed in neurons (Dolcetta et al., 2004; Weinstock, Kreher, et al., 2020; Zhang et al., 2014) and neuronal pathology in GLD occurs before distinct demyelination in the twitcher and twi-5j mouse models (Cappello et al., 2016; Castelvetri et al., 2011; Potter et al., 2013). Neuronal swellings and varicosities were specifically documented in twitcher spinal cords at postnatal day 7, prior to overt CNS myelination (Castelvetri et al., 2011). Similarly, purified primary neurons from twitcher have abnormal neurites and axonal transport, attributed to some degree of autonomous psychosine production (Castelvetri et al., 2011). GALC has also been proposed to have a role in the maintenance of subventricular zone neurogenesis during the early postnatal period (Santambrogio et al., 2012). Interestingly, it was found that neuronal pathology was still present when twitcher mice were crossed to knockout mice for ceramide galactosyltransferase (Ezoe et al., 2000a), the enzyme that synthesizes galactosylceramide (and therefore upstream of psychosine formation). This suggests that neuronal GALC may function on a substrate and in a role unrelated to that of myelin and GalCer turnover. Nonetheless defining the autonomous role of GALC in neurons has been technically challenging using conventional global knockout or mutant models.

The new Galc conditional knockout mouse identified a key developmental period that requires GALC prior to myelination; exposing a critical period of vulnerability to GALC ablation between postnatal day 4–6 in mice (Weinstock, Kreher, et al., 2020). Early Galc deletion before postnatal day 4, results in a worse Krabbe phenotype, higher psychosine in the hindbrain, and a shorter life-span of the mice. Neurons in the hindbrain have more axonal degeneration when GALC is deleted early, in line with defective neurogenesis of the hindbrain found in a zebrafish model of GLD (Zizioli et al., 2014). During this vulnerable period, GALC is expressed in all brain cells, with the highest expression in neurons. Furthermore, neuronal GALC is involved in the development and maturation of immature T-box-brain-1 (TBR1) positive brainstem neurons, suggesting that brainstem development is affected by intrinsic neuronal GALC (Weinstock, Kreher, et al., 2020). This developmental role of GALC may correlate with the observed benefit of early therapeutic inventions for twitcher and Krabbe patients. Therefore, augmenting GALC levels before this defined period could possibly increase the efficacy of GLD therapy.

The mechanisms underlying the unique biological role of GALC in neurons remains unknown. Lysosomes are essential for formation of synaptic structures including dendritic spines (Vukoja et al., 2018). In neurons, lysosomes are present in dendrites and traffic bidirectionally, regulating dendritic spine number in an activity-dependent manner (Goo et al., 2017). Lysosomes control the excitatory synaptic signal strength by modulating the fate of the α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptors and sorting them for degradation or recycling (Fernández-Monreal, Brown, Royo, & Esteban, 2012). In hippocampal neurons, activity-dependent release of lysosomal Cathepsin B regulates the structural plasticity of dendritic spines by triggering MMP-9 activation and extracellular matrix remodeling (Padamsey et al., 2017). Therefore, it is possible that the aberrant synapse, caused by GALC-deficient lysosomes, may impact neuronal activity and function, eventually causing neuronal death, and abnormality in brain development (Hensch, 2005; Marín, 2016). In addition, if brainstem neurons do not fully mature, they could be vulnerable to accumulated psychosine, less responsive to dynamic signals, and eventually eliminated (Pfisterer & Khodosevich, 2017; Weinstock, Kreher, et al., 2020). Further investigation is required to understand the specific cellular events which require GALC for neural development and if similar mechanisms are present in other LSDs.

An additional theory relevant for neuronal pathogenesis in GLD is in the metabolism of a minor and lesser-studied substrate of GALC, lactosylceramide (Wenger, Sattler, & Hiatt, 1974). Lactosylceramide is synthesized by β-1,4-galactosyltransferase 6 in astrocytes during chronic CNS inflammation, but has been documented to contribute to neurodegeneration (Mayo et al., 2014), perhaps related to its regulation function of TNF-alpha induced astrocytosis (Pannu, Singh, & Singh, 2005). In the experimental autoimmune encephalomyelitis (EAE) model, sphingolipid lactosylceramide stimulates astrocytes (Mayo et al., 2014) and recruits microglia and infiltrating macrophages. In the canine GLD model, lactosylceramide was highly elevated in the cerebrospinal fluid and further increased with age, in addition to psychosine and GalCer (Corado et al., 2020), indicating a possibility of the contribution of accumulated lactosylceramide to GLD pathology.

4 |. SECONDARY PERTURBATIONS FROM GALC DYSFUNCTION

4.1 |. Lysosome accumulation and dysfunction

In most LSDs, a number of common pathologic molecular cascades occur at the cellular level as substrates accumulate leading to impaired lysosomal function. Lysosomes have multiple roles in cellular homeostasis including the regulation of endocytosis and autophagy via nuclear signal transduction as well as plasma membrane repair and exocytosis. All of these aspects of lysosomal biology are not surprisingly susceptible to impairments in LSDs. For example, under physiologic conditions there must be a sufficient number and volume of lysosomes to meet the metabolic requirements of the cell. The number of lysosomes varies from 50 to 1,000 per cell (H. Xu & Ren, 2015), and occupies 1–15% of cell volume (Marzella, Ahlberg, & Glaumann, 1982). These factors change dynamically in accordance with cellular demands (such as starvation) both transcriptionally and post-transcriptionally (Eriksson, Wäster, & Öllinger, 2020; Settembre et al., 2011). Instead, the cellular space occupied by lysosomes in LSDs increases drastically. Similarly, the pH of the lysosomal lumen is normally carefully maintained via the vacuolar ATPase (Platt, Boland, & van der Spoel, 2012). Substrate accumulation in LSDs can influence the regulation of lysosomal pH (Folts et al., 2016; Holopainen, Saarikoski, Kinnunen, & Järvelä, 2001). For example, lysosomes in the oligodendrocytes of GALC-deficient twitcher are enlarged and have higher pH. Psychosine has also been shown to disrupt lysosomal function by antagonizing the lysosomal vascular-type H + -ATPase (Tapper & Sundler, 1995), suggesting a change in lysosomal pH due to psychosine accumulation. Therefore, it has been suggested that re-acidification of lysosomal pH may ameliorate Krabbe pathogenesis (Folts et al., 2016).

There is also a growing body of evidence that suggests that distended lysosomes in LSDs, which often contain copious substrate, are structurally sensitive and prone to membrane rupture (reviewed in Wang, Gómez-Sintes, & Boya, 2018). This process is often referred to as LMP, and can potentially lead to cell death. If the lysosomes rupture, acidic hydrolases, including cathepsins, are released to the cytoplasm and have the potential to degrade cellular components. Their hydrolysis activity seems to continue to work to some degree in cytoplasmic pH, resulting in apoptosis or necrosis depending on the degree of permeabilization (Boya & Kroemer, 2008). LMP seems to be a common pathophysiologic mechanism in LSDs, though its specific role in GLD has yet to be explored.

In addition to perturbed lysosomal function, the LSDs can also cause dysfunction in the broader endosome-lysosome network. Substrate delivery to the lysosome occurs via well-coordinated steps of cargo formation and delivery. These include cargo recognition, autophagosome and endosome formation, fusion with the lysosome and subsequent recycling of proteins and lipids. Psychosine exposure to OLs caused abnormalities in endo-lysosomal transport, as proven by a decreased endocytic import. Furthermore, once the larger endosome trafficking system is perturbed, secondary accumulation of additional, biochemically unrelated, metabolites can also occur (Walkley & Vanier, 2009). In turn, this compounding effect can affect the integrity of other sub-organelles.

In addition, lysosome-mediated exocytosis regulates the proper targeting of proteins residing in vesicles, the plasma membrane, or extracellular space. Therefore, dysfunctional lysosomes can mis-target those proteins to the wrong place or the ER which could activate the unfolded protein response (UPR) and oxidative stress, which have been suggested as a key mechanism of Krabbe pathogenesis (Irahara-Miyana et al., 2018). Furthermore, toxic effect of misfolded GALC protein itself due to mutations and associated polymorphisms can generate similar ER or oxidative stresses (Irahara-Miyana et al., 2018; Shin et al., 2016), which may trigger neuroinflammation by attracting and activating immune cells (Formichi et al., 2007; Santambrogio et al., 2012). Finally, it is conceivable that dysregulated autophagy is involved in the inflammatory response. Indeed, autophagy modulates inflammation for normal host-pathogen defense, by controlling adaptive immunity through regulation of antigen presentation.

4.2 |. Autophagic dysfunction

Autophagy is a specialized “self-eating” cellular function, which eliminates unnecessary or damaged cellular components, providing nutrient and metabolic homeostasis. (Ravikumar et al., 2010). There are three types of autophagic pathways: macroautophagy, chaperone-mediated (CMA), and microautophagy. All three types use different routes to reach lysosomes for the degradation of autophagy-targeted materials. Macroautophagy (commonly referred to simply as “autophagy”) sequesters materials into double-membrane-layer bound autophagosomes and then fuses with lysosomes to form autophagolysosomes. The lipidated microtubule-associated protein light chain 3 (LC3-II) is involved in all parts of the autophagy process. p62 is another indicator of autophagic flux, because it interacts with autophagic substrates and delivers them to autophagosomes for degradation (Rusten & Stenmark, 2010). Autophagosomes are dispersed along microtubules in the cytosol to reach to and fuse with lysosomes, forming autolysosomes (Kast & Dominguez, 2017) where all of the cargo in autophagy is degraded and/or recycled for energy and membrane components. CMA is a selective autophagic pathway that only targets proteins having a KEFERQ motif or a similar sequence for degradation (Kaushik et al., 2011). Heat shock protein 70 recognizes and binds to the proteins destined for CMA, and docks on lysosomes via interaction with LAMP-2A, allowing lysosomal engulfment (Venugopal et al., 2009). Microautophagy is mediated by direct pinocytosis of cytosolic materials into lysosomes (Sahu et al., 2011).

Due to the intimate relationship of autophagy with lysosomal pathways, defects in autophagy are often secondary to the abnormally high accumulation of various lysosomal metabolites in most LSDs (Lieberman et al., 2012). In LSDs, autophagosome-lysosome fusion is impaired, which results in limited capacity for autophagic flux (Kollmann et al., 2012). Ultimately, the autophagosome is slowly processed by lysosomes, which results in the accumulation of autophagic substrates, LC3-II positive vacuoles, as well as ubiquitinated and aggregate-prone molecules in the cytoplasm, including p62 and alpha-synuclein (Ravikumar, Duden, & Rubinsztein, 2002; Teil et al., 2020). Since autophagy also has a role in the maintenance of mitochondria quality by selectively degrading dysfunctional mitochondria via mitophagy (Kim, Rodriguez-Enriquez, & Lemasters, 2007), impaired autophagic flux can lead to the increase of defective mitochondria. Similarly, lysosomes themselves have a quality control mechanism that relies on autophagy, termed lysophagy (Papadopoulos, Kravic, & Meyer, 2020). Therefore, lysosomal dysfunction, which can impair autophagic machinery, may further limit the normal cellular ability to repair damaged lysosomes via lysophagy.

Increased levels of LC3-positive autophagosomes have been observed in GALC-deficient oligodendrocytes (Ribbens et al., 2014). Abnormally large p62 cytoplasmic aggregates are also found in the brain and sciatic nerve of twitcher (Del Grosso et al., 2019). Co-accumulation of p62/LC3-II aggregates are mostly present in white matter regions undergoing demyelination and neuroinflammation (D. S. Lin et al., 2020). It is thought that psychosine impairs autophagic flux by affecting the formation steps of autophagosomes and autophagolysosomes (Del Grosso et al., 2016). Not surprisingly, there is also data from GLD tissues that show dysfunction in a complimentary protein degradative system known as the ubiquitin-proteasome system (D. S. Lin et al., 2020). Similarly, GALC deficient macrophages exposed to GalCer expressed markers of the closely related integrated stress response including CHOP, ATF4, and GADD34 (Weinstock, Shin, et al., 2020). Overall, it appears that GALC deficiency causes a broad cellular consequence in pathways involving metabolic regulation including autophagy and other protein quality control mechanisms. Suboptimal functioning of these important pathways may even have theoretical consequences in GALC haplo-insufficiency. For example, a strong association has now been developed between GBA1 haplo-insufficiency and the risk for neurodegenerative disorders like Parkinson’s disease (reviewed in Migdalska-Richards & Schapira, 2016). The link to neurodegenerative diseases for GALC, and other lysosomal hydrolases, is therefore of growing significance (Robak et al., 2017).

4.3 |. Progressive inflammation

High levels of inflammation in both the CNS and PNS is a prominent feature of GLD, documented by widespread microgliosis, CNS macrophage infiltration and expression of proinflammatory chemokines (LeVine, Wetzel, & Eilert, 1994). While microgliosis and globoid cell infiltration is clearly an important aspect of GLD pathology, there remains some question regarding the degree to which this process drives pathogenesis. There exists a longstanding correlation between globoid cell accumulation and areas of overt demyelination (Taniike & Suzuki, 1994), which was conventionally interpreted to signify that globoid cells form in areas of demyelination and myelin debris. However, some studies suggest that neuroinflammation precedes overt myelin loss (Potter et al., 2013;Santambrogio et al., 2012; Snook et al., 2014). For example, the twi-5j mouse model harboring Galc-E130K mutation had robust neuroinflammation in the absence of demyelination, showing an active gliosis reaction as early as 2 weeks postnatally, before demyelination. In addition, proinflammatory cytokines such as CCL2, CCL3, CCL5, CXCL1, CXCL10, IL1b, IL6, TNF-alpha, and toll-like receptor 2 (TLR2) are upregulated in presymptomatic twitcher brains starting postnatal 2 weeks old and increased exponentially with disease progression (Santambrogio et al., 2012; Snook et al., 2014). Similarly, astrogliosis also precedes overt demyelination and coincides with macrophage infiltration, arguing that astrocytes have a compounding role in the disease progression of GLD (Ijichi et al., 2013), possibly via over-expression of prostaglandin D signaling (Mohri et al., 2006).

Due to the assumption that proinflammatory conditions drive GLD pathogenesis, a number of attempts have used anti-inflammatory therapeutic approaches in twitcher mice. For example, the anti-inflammatory agent ibudilast was shown to modestly improve GLD by delaying symptom progression and demyelination (Kagitani-Shimono et al., 2005). Similarly, the S1P agonist fingolimod (FTY720), which acts by preventing lymphocyte egression from lymph nodes, was found to attenuate psychosine-induced glial cell death and demyelination both in vitro and ex-vivo models of twitcher mice (Béchet et al., 2020; O’Sullivan & Dev, 2015). Nonetheless the overall efficacy of anti-inflammatory agents alone on preventing GLD pathogenesis has been disappointing. Similarly, genetic crosses of twitcher mice with TNF-alpha R1 knockout mice did not significantly alter disease progression (Pedchenko, Bronshteyn, & LeVine, 2000) and crossing twitcher mice with MHC-II knockout mice had no effect on survival (Matsushima et al., 1994). These data further contributed to the question regarding the significance of neuroinflammation in GLD.

Furthermore, despite the evidence for a proinflammatory state being a driving force for GLD pathogenesis, there exists some evidence that macrophages may even be protective in GLD. When the twitcher mouse was crossed to a macrophage-deficient osteopetrotic background, which harbor mutations in Csf-1, the mice had a more severe phenotype, suggesting that macrophages play a protective role (Y. Kondo, Adams, Vanier, & Duncan, 2011). Our recent study showed that macrophages that express GALC are, in fact, neuroprotective and limit the overt demyelination, axonal degeneration, and clinical decline seen in global Galc knockout tissues (Weinstock, Shin, et al., 2020). We suspect that this effect is mediated by neuroinflammation elicited from myelin substrates in Galc deficient macrophages (i.e., globoid cells). This data is in line with a study in which Galc deficient oligodendrocytes were transplanted into the demyelinating shiverer mice, which are normally unable to myelinate effectively. Shiverer mice with transplanted Galc deficient oligodendrocytes were able to differentiate and myelinate normally (Y. Kondo et al., 2005), suggesting that the demyelinating phenotype of GLD is strongly influenced by non-oligodendrocyte conditions. Thus, the contrasting observations regarding a protective versus deleterious effects of innate immunity in GLD can be reconciled by the view that GALC positive macrophages are beneficial for GLD, while GALC deficient macrophages aggravate the disease.

5 |. THERAPIES FOR GLD

Due to the authentic nature of the twitcher model of GLD, a stunning number of therapeutic strategies have been tested on twitcher mice (reviewed in Mikulka & Sands, 2016). These include bone marrow transplantation (BMT) or HSCT (Ichioka, Kishimoto, Brennan, Santos, & Yeager, 1987; Yeager, Brennan, Tiffany, Moser, & Santos, 1984), neural and mesenchymal stem cell transplantation (Neri et al., 2011; Ripoll et al., 2011; Strazza et al., 2009; Taylor & Snyder, 1997), substrate reduction therapy (LeVine, Pedchenko, Bronshteyn, & Pinson, 2000), antioxidant therapy (Pannuzzo et al., 2010), pharmacological chaperone therapy (Berardi et al., 2014), enzyme replacement therapy (ERT; Lee et al., 2005), and gene therapy (GT) (Lattanzi et al., 2010; Lee et al., 2007; D. Lin et al., 2005; Meng, Shen, Watabe, Ohashi, & Eto, 2005; Rafi et al., 2012, 2015; Rafi, Luzi, & Wenger, 2021; Shen, Watabe, Ohashi, & Eto, 2001). Additionally, a large number of combination therapies have been tested (Galbiati et al., 2009; Karumuthil-Melethil et al., 2016; D. Lin et al., 2007; Rafi et al., 2015; Rafi, Luzi, & Wenger, 2020; Reddy et al., 2011; Ricca et al., 2015).

6 |. ENZYME REPLACEMENT THERAPY

The basis for enzyme replacement therapy (ERT) for lysosomal diseases is cross-correction, the phenomenon by which lysosomal enzymes (including GALC), are secreted from enzyme-producing cells and taken up by enzyme-deficient cells (Sands & Davidson, 2006), or supplied exogenously by therapeutic intervention. ERT has been successfully used in a number of LSDs including Gaucher’s disease (Schiffmann et al., 1997), Fabry’s disease (Germain, 2002), MPS-I (Pasqualim et al., 2015), and others, though peripheral disease seems easier to treat than neurological defects. This phenomenon is thought to highlight the difficult of delivering large proteins across the blood brain barrier (Thomas & Kermode, 2019). The molecular basis for ERT is that secreted lysosomal hydrolases are tagged with mannose-6-phosphate residues and taken up by M6P receptors (Coutinho et al., 2012). Human GALC has four potential N-glycosylation sites (Deane et al., 2011), though which site is required for activity is currently unknown.

Cross-correction of GALC was shown to occur, in vitro, in GLD fibroblasts from conditioned media and was inhibited by M6P (Nagano et al., 1998). This same process also occurs, in vitro, when using conditioned-media from fibroblasts transduced with lentiviral GALC cDNA (Rafi, Fugaro, et al., 1996). GALC-deficient oligodendrocytes were also able to take up exogenous GALC enzyme (Luddi et al., 2001), in vitro. An argument for evidence of in vivo cross correction of GALC to oligodendrocytes comes from a study that showed that twitcher oligodendrocytes transplanted into shiverer mice (myelin-deficient) were able to myelinate properly (Y. Kondo et al., 2005). While the authors concluded that twitcher oligodendrocytes received GALC from their environment, it is also possible that GLD pathogenesis relies on the role of GALC in cells other than oligodendrocytes, like microglia (discussed in Nicaise et al., 2016), macrophages, neurons and astrocytes, as suggested by recent data using cell-specific Galc (Weinstock, Shin, et al., 2020).

In general, GALC ERT has been disappointing in GLD and only seemed to improve survival in twitcher mice by a few days (Lee et al., 2005). Some speculated that this problem may have been: (a) due to poor ability of the recombinant enzyme to cross the blood–brain barrier (BBB) or (b) due to an immune response produced against the recombinant enzyme (Won, Singh, & Singh, 2016). The immune response was particularly pertinent because the twitcher mouse is a full null mouse and therefore may recognize injected recombinant GALC as foreign. To overcome this issue, one study generated a humanized mouse model of GLD using a knock-in of a human GLD mutation in the Galc locus (Matthes et al., 2015). While intracerebral injection of recombinant human GALC did not trigger an immune reaction and bypassed the BBB, there remained no improvement to the phenotype of the GLD mice. The conclusion, was that cross-correction of in vivo GALC to oligodendrocytes was not efficient (Matthes et al., 2015). In line with this data, a GT study showed that immunostaining for recombinant GALC was always restricted to transduced cells and was not able to cross-correct neighboring cells (Meng et al., 2005). Most recently, our study showed that in vitro GALC deficient cells, including both primary mouse Schwann cells and human patient fibroblasts, were less capable of receiving M6P-tagged GALC than their wildtype counterparts (Weinstock, Shin, et al., 2020). We also showed poor in vivo cross correction of GALC in Schwann cells.

Despite these limitations, a number of promising approaches have been directed toward improving GALC delivery and uptake by twitcher neural tissues. For example, GALC has recently been encapsulated into poly-(lactide-co-glycolide) nanoparticles, functionalized with brain targeting peptides (Del Grosso et al., 2019). While this study showed promising in vivo GALC delivery to brains of twitcher mice, functional and clinical studies were not explored. A second group recently developed chimeric GALC enzymes engineered to express unique peptide domains hypothesized to improve secretion, uptake, and transport across the BBB (Ricca, Cascino, Morena, Martino, & Gritti, 2020). In particular, GALC fusion proteins to the signal peptide of iduronate-2-sulfatase (IDS) or the low-density lipoprotein receptor (LDLr)-binding domain (from ApoE II) were generated and produced by LV-transduced cells. Cells producing IDS-GALC had improved secretion of enzyme, compared to GALC-only producing cells, and similar or improved cross-correction of in vitro twitcher cells. Furthermore, the ApoE-GALC approach has the additional opportunity to leverage increase LDLr protein expression (and therefore increased cross-correction capabilities) via statin pharmacotherapy (Ricca et al., 2020). In summary, while recent works have shed light on the limitations of GALC cross-correction and the delivery of ERT across the BBB, exciting novel approaches continue to be developed to facilitate enzyme replacement in GLD and other LSDs.

7 |. HEMATOPOIETIC STEM CELL THERAPY

The first example of HSCT for LSDs was published in the Lancet in 1981 and showed reversal of clinical features of Hunter’s disease following transplantation (Hobbs et al., 1981). In the wake of this marquis paper, a number of groups were trying to test this paradigm in vivo, which presented challenges as some LSD mouse models were not severe enough to test survival (Hong, Sutherland, Matas, & Najarian, 1979), while others that were severe showed no improvement in survival (Miyawaki, Mitsuoka, Sakiyama, & Kitagawa, 1982). In 1984, a landmark study by Yeager and Santos was published that showed that HSCT delivered to irradiated twitcher mice could remarkably extend average survival from 40 to 80 days (Yeager et al., 1984). A follow up study showed that GALC activity was normalized in HSCT treated mice (Ichioka et al., 1987), and that psychosine production was drastically delayed in the CNS of mice ultimately reaching 30–35% of untreated levels at the time of death (Ichioka et al., 1987). PNS psychosine was also initially reduced, although it did accumulate to untreated levels by the time of death (Ichioka et al., 1987).

A repeat study, using donor mice genetically identified with a unique MHC isoform, confirmed the pronounced survival and psychosine reduction in HSC-treated twitcher mice (Hoogerbrugge et al., 1988). Furthermore, some donor derived bone marrow cells were found in twitcher brains, which could explain the increased GALC activity in the brain. Strikingly, the number of globoid cells continued to decrease in brains of grafted mice, while the number of “foamy macrophages” continued to increase. These foamy macrophages lacked the typical GalCer inclusions of globoid cells and were thought to reflect GALC-expressing donor macrophages involved in myelin turnover (Hoogerbrugge et al., 1988). A more in-depth analysis of these treated mice (A. Kondo et al., 1988; Suzuki & Suzuki, 1990) showed that there were areas of remyelination found throughout the CNS and PNS, which were attributed to the presence of GALC-expressing donor macrophages. While the mechanisms of HSCT was originally considered to occur via cross-correction, recent studies suggest that the beneficial properties may instead be from restored phagocytic function of donor macrophages, which retain GALC function (Weinstock, Shin, et al., 2020; Wolf et al., 2020).

Interestingly, the disease process in BMT-treated twitcher mice continued to progress in the CNS and PNS despite measurable improvement in the enzymatic activity after BMT. In fact, at the ultrastructural level, GalCer inclusions were still evident in oligodendrocytes and Schwann cells throughout survival of mice. Overall, it was suggestive that “the metabolic defect in the twitcher mouse still remained long after BMT” (A. Kondo et al., 1988). This data further argues that the therapeutic benefit of HSCT is likely through restoring macrophage function rather than supplying GALC to myelinating glia. Additional studies showed that BMT was effective not only in reducing globoid cells, but also in decreasing widespread markers of inflammation including TNFα (Wu et al., 2001). Along these lines, a more recent study showed that BMT was even more effective at inducing anti-inflammatory effects than the direct action of anti-inflammatory drugs (Luzi et al., 2009).

Initial human clinical trials for HSCT in GLD were first published in 1998 (four Late-Onset GLD, LOGLD, and one presymptomatic Early Infantile GLD, EIGLD; Krivit et al., 1998). Preliminary findings found that HSCT was efficacious as the four LOGLD patients had a reversal in CNS deterioration, as measured by decreased CSF protein and improved myelination by MRI, while the EIGLD patient had delayed symptom development. A 2005 study then showed that HSCT was particular helpful if delivered to presymptomatic EIGLD patients but was not efficacious in postsymptomatic EIGLD patients (Escolar et al., 2005). This landmark study emphasized the urgency of secure presymptomatic diagnosis for EIGLD and launched a host of clinical research including the implementation of newborn screening among several states as well novel studies to help better diagnose GLD. A follow up study of 18 EIGLD patients that received HSCT found that though most patients do significantly better than the natural history of EIGLD, all patients are abnormal and continue to develop disease progression both in the CNS and PNS (Wright, Poe, DeRenzo, Haldal, & Escolar, 2017).

8 |. GENE THERAPY

The twitcher model is one of oldest mouse models used for GT and has been tested with a number of viral delivery systems. Viral GT contains two components: the vehicle, which is comprised of a viral vector that is emptied of viral genes, and the genetic cargo, which in this case is GALC cDNA (often species matched to reflect the host species). The viral vehicles used for GT on twitcher mice have been broad and include adenovirus, lentivirus, and adeno-associated virus (AAV; Lee et al., 2007; D. Lin et al., 2005; Meng et al., 2005; Rafi et al., 2012, 2015; Shen et al., 2001). More recent approaches have used AAVrh10 (Rafi et al., 2012) or AAV9 (Marshall et al., 2018; Pan et al., 2019) serotypes, thought to have ideal transduction in brain and increased tropism for glia, without genome integration and minimal risk of inducing malignancy. It should be noted that lentiviruses, which integrate within the genome, are also of use particularly in the case of ex vivo transduction of hematopoietic stem cells, which have been very successful in the clinic (Aiuti et al., 2013; Biffi et al., 2013; Sessa et al., 2016).

Although viral GT should theoretically be curative, the efficacy in vivo has had major limitations. twitcher studies using viral-delivered GT tend to only modestly improve survival, typically by approximately 1.5–2-fold. Therefore, many approaches have been made to improve delivery and transducing ability. For example, some studies use tail-vein injections (Rafi et al., 2012), while others use intrathecal or intracerebral injections (D. Lin et al., 2005) while still others use multiple sites of injection (Rafi et al., 2012). Dose of virus is also very important, as a recent article had far better success in the twitcher mouse by using significantly higher doses of AAV9 (Marshall et al., 2018). It should also be noted, that AAV vectors have particularly strong tropism for neurons (Asokan, Schaffer, & Samulski, 2012) compared to glia, and so neurons need to provide a significant supply of GALC for other cell types via cross-correction (Mikulka & Sands, 2016). Therefore, discussions on improving cross-correction of GALC are relevant not just for ERT or HSCT, but also for viral GT. In addition, the oncogenic potential of AAV-vector should be clearly addressed before considering human trials (Li et al., 2021).

9 |. SUBSTRATE REDUCTION THERAPY

The idea of a pharmacological inhibitor of psychosine synthesis is of major interest in GLD. For example, the medication l-cycloserine has been shown to moderately reduce psychosine accumulation and GLD-associated toxicity in twitcher mice (LeVine et al., 2000). This medication, though toxic in its own right, has been found to be beneficial when used in conjunction with other forms of therapy (Ricca et al., 2015). Current groups are working to develop high-throughput screens to develop novel agents to inhibit psychosine production (Ribbens et al., 2014). Of note, genetic models of CGT/GALC double knockout mice found that reducing GalCer/Psychosine production was beneficial to some degree (Ezoe et al., 2000b), though total ablation of CGT was not curative (Ezoe et al., 2000a). Novel enzymatic targets for substrate reduction inhibitors now include the enzyme acid ceramidase, which could prevent the formation of psychosine via the degradative route (Li et al., 2019). For example, the chemotherapeutic agent carmofur, which directly inhibits acid ceramidase activity, reduced psychosine production and modestly extended life span of twitcher mice that were haplo-insufficient for Asah1 (Li et al., 2019).

10 |. COMBINATION THERAPY

Due to the partial effect of many different modalities of therapies, a number of groups have approached the concept of combining different modalities of therapy (reviewed in Ricca & Gritti, 2016). The effect of multimodal therapy has had profound improvements of survival on both the twitcher mouse model of GLD, as well as on the dog model of GLD. In particular, the use of HSCT has been particular efficacious in synergizing with viral-directed GT. The synergistic benefit of combining HSCT and GT has been replicated by many different labs with different viral vectors and regiments of delivery (Hawkins-Salsbury et al., 2015; D. Lin et al., 2007; Qin et al., 2012; Rafi et al., 2020; Reddy et al., 2011).

Work in uncovering the mechanism behind this synergy revealed that HSCT seems to have a profound immunomodulatory benefit that is not replicated by GT (Reddy et al., 2011). This effect seemed to be independent from the cross-corrective ability of circulating stem cells. This concept was highlighted by the fact that other stem cell therapies, like neural stem cell therapy (NSCT) or embryonic stem cell derived OLs, which should be able to cross-correct, were not efficacious in twitcher mice (Kuai et al., 2015; Ricca et al., 2015). Instead, when NSCT was given with HSCT, there was once again robust synergy similar to GT + HSCT, emphasizing the unique benefit of HSCT (Ricca et al., 2015). Taken together, these data are in agreement with the idea that HSCT may work independently of its ability to increase the amount of GALC activity in tissues. Similarly, the efficacy of ICV Mesenchymal stem cells in twitcher mice seemed to have an immunomodulatory effect, though not all cytokine levels were returned to normal (Ripoll et al., 2011).

Additional forms of “combination therapy” include the concept of augmenting HSCT precursor cells ex-vivo prior to transplantation. This strategy is particularly appealing for metabolic disease as patient-treated HSCs can be genetically corrected by (CRISPR-Cas editing) or transduced to overexpress the missing enzyme by lentiviral GT. The rationale for this approach is two-fold: (a) by inducing the patient’s own HSCs to produce the missing lysosomal enzyme the severe risk of allogenic transplantation would be avoided and (b) it would theoretically be possible to produce HSCT-derived cells that produce supra-physiologic levels of GALC that are even more efficient at correcting metabolic defects. This approach has remarkable effects in ARSA deficiency of metachromatic leukodystrophy patients (Biffi et al., 2013; Sessa et al., 2016). In light of recent data that shows limitations of cross-correction from HSCT in MLD patients (Wolf et al., 2020), it would be intriguing to evaluate if overexpressed GT-HSCT has improved cross-correctable capabilities. This approach would, in theory, be particularly appealing for GLD, where functional impairment of GALC deficient hematopoietic stem cells (Visigalli et al., 2010) has been described. Unfortunately, GALC overexpression in HSCs seems to be toxic, perhaps related to changes in the bioactive sphingolipids ceramide, sphingosine and sphinosine-1-phosphate (Costantino-Ceccarini et al., 1999). Novel alternative approaches have therefore considered modulating GLD HSCs using for miRNA GT that would increase GALC expression (Gentner et al., 2010).

11 |. CLOSING REMARKS

For over 50 years, physicians and scientists have made significant advances in understanding the molecular mechanisms of GLD, ranging from proposing the psychosine hypothesis (reviewed in Suzuki, 1998) to cloning GALC (Chen, Rafi, de Gala, & Wenger, 1993). Translational advances have also emerged as HSCT is the standard of care for presymptomatic patients with GLD (Escolar et al., 2005) and may be positively augmented by co-administration with viral GT (D. Lin et al., 2007). Nonetheless, the past year has marked a number of significant discoveries that seem to shed light on key unsolved questions in GLD, including the biochemical formation of psychosine from acid ceramidase (Li et al., 2019), the cellular origin of psychosine from myelinating glia (Mikulka et al., 2020; Weinstock, Shin, et al., 2020), the developmental role of GALC in neurons (Weinstock, Kreher, et al., 2020), and the beneficial effect of GALC expressing macrophages on remyelination and neuroprotection (Weinstock, Shin, et al., 2020). Many of these discoveries have been advanced with the development of novel tools, including genetic mouse models.

Despite these exciting discoveries, a number of important questions remain: Can acid ceramidase inhibitors be modulated to reduce toxicity, while remaining beneficial in patients with GLD? Are mechanisms of oligodendrocytes/microglia analogous to Schwann cells and macrophages? Do astrocytes (or neurons) of the CNS play an important part? Can cross correction be improved in GALC-deficient cells? Does the amount of enzyme available influence cross correction? Why does GALC secretion and transfer occur in physiology? Are the defects of cross correction reflective of other LSDs? If psychosine is generated by ASAH1 in lysosome, are the concentrations higher there? If so, does toxicity start at lysosome? Do the crystals evident by electron microscopy in GLD tissues contribute to pathology? What exactly are they composed of and how are they formed? For some time, researchers have been trying to optimize cell and tissue culture strategies to best represent normal and disease physiology, and one such research group has recently proposed the use of human induced pluripotent stem cells (hiPSCs) in developing 2D and 3D co-cultures to foster neuron-glia functional and dysfunctional interactions in order to study and define cell-specific role in neurodevelopment, neuroinflammation, and progression of the disease (Luciani, Gritti, & Meneghini, 2020). Such approaches might prove beneficial and fitting to answer some of the questions mentioned above.

The advances in GLD also have an impact on broader diseases. A recent discovery linked the association of GBA1 carrier mutants, responsible for the LSD Gaucher’s disease, with increased risk for Parkinson’s disease. It appears that mutations in GALC and other lysosomal enzymes have similar risk associations for neurodegenerative disease (Chang et al., 2017) and multiple sclerosis (Sawcer et al., 2011). These fascinating observations have, in turn, led to further unanswered questions in the field: how does GALC haploinsuffiency contribute to more common disorders of adulthood? Is there a unifying mechanism connecting lysosomal hydrolase deficiency and neurodegeneration? Can LSD treatments be beneficial in LSD carriers who develop neurodegenerative disease? While answers to these questions are likely to lead to new hypotheses, they emphasize the exciting nature of recent advances in the field and the broader application to common neurologic disorders.

ACKNOWLEDGMENTS

The authors thank past and current members of the Feltri, Wrabetz, and Shin laboratories for support and stimulating discussions. Work in the Feltri an Wrabetz laboratories was headstarted by the Empire State Development Corporation, Krabbe Disease Grants U446 and W753 to Lawrence Wrabetz, and the Hunter’s Hope Foundation; and it is currently supported by grants R01 NS111715 from the National Institute of Neurological Diseases and Stroke and 2018-023I4 from the European Leukodystrophy Association. Work in the Shin laboratory is supported by grant R01 NS 112327 from the National Institute of Neurological Diseases and Stroke. The authors thank the NDRD brain and tissue bank at the University of Pittsburgh, Krabbe Partners for Research and all the families for the gift of human tissues.

Funding information

Association Européenne contre les Leucodystrophies, Grant/Award Number: 2018-023I4; National Institute of Neurological Disorders and Stroke, Grant/Award Numbers: R01 NS 112327, R01 NS111715; Empire State Development Corporation, Krabbe Disease, Grant/Award Numbers: U446, W753; Hunter Hope Foundation

DATA AVAILABILITY STATEMENT

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

REFERENCES

  1. Aiuti A, Biasco L, Scaramuzza S, Ferrua F, Cicalese MP, Baricordi C, … Naldini L (2013). Lentiviral hematopoietic stem cell gene therapy in patients with Wiskott-Aldrich syndrome. Science, 341(6148), 1233151. 10.1126/science.1233151 [DOI] [PMC free article] [PubMed] [Google Scholar]
  2. Anderson HM, Wilkes J, Korgenski EK, Pulsipher MA, Blaschke AJ, Hersh AL, … Bonkowsky JL (2014). Preventable infections in children with leukodystrophy. Annals of Clinical Translational Neurology, 1(5), 370–374. 10.1002/acn3.61 [DOI] [PMC free article] [PubMed] [Google Scholar]
  3. Asokan A, Schaffer DV, & Samulski RJ (2012). The AAV vector toolkit: Poised at the clinical crossroads. Molecular Therapy, 20(4), 699–708. 10.1038/mt.2011.287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  4. Austin JH (1963). Studies in Globoid (Krabbe) Leukodystrophy. II. Controlled thin-layer chromatographic studies of globoid body fractions in seven patients. Journal of Neurochemistry, 10, 921–930. 10.1111/j.1471-4159.1963.tb11919.x [DOI] [PubMed] [Google Scholar]
  5. Austin JH, & Lehfeldt D (1965). Studies in globoid (Krabbe) leukodystrophy. 3. Significance of experimentally-produced globoid-like elements in rat White matter and spleen. Journal of Neuropathology and Experimental Neurology, 24, 265–289. [PubMed] [Google Scholar]
  6. Bansal R, Warrington AE, Gard AL, Ranscht B, & Pfeiffer SE (1989). Multiple and novel specificities of monoclonal antibodies O1, O4, and R-mAb used in the analysis of oligodendrocyte development. Journal of Neuroscience Research, 24(4), 548–557. 10.1002/jnr.490240413 [DOI] [PubMed] [Google Scholar]
  7. Barczykowski AL, Foss AH, Duffner PK, Yan L, & Carter RL (2012). Death rates in the U.S. due to Krabbe disease and related leukodystrophy and lysosomal storage diseases. American Journal of Medical Genetics Part A, 158a(11), 2835–2842. 10.1002/ajmg.a.35624 [DOI] [PubMed] [Google Scholar]
  8. Baskin GB, Ratterree M, Davison BB, Falkenstein KP, Clarke MR, England JD, … Wenger DA (1998). Genetic galactocerebrosidase deficiency (globoid cell leukodystrophy, Krabbe disease) in rhesus monkeys (Macaca mulatta). Laboratory Animal Science, 48(5), 476–482. [PubMed] [Google Scholar]
  9. Béchet S, O’Sullivan SA, Yssel J, Fagan SG, & Dev KK (2020). Fingolimod rescues demyelination in a mouse model of Krabbe’s disease. The Journal of Neuroscience, 40(15), 3104–3118. 10.1523/jneurosci.2346-19.2020 [DOI] [PMC free article] [PubMed] [Google Scholar]
  10. Berardi AS, Pannuzzo G, Graziano A, Costantino-Ceccarini E, Piomboni P, & Luddi A (2014). Pharmacological chaperones increase residual β-galactocerebrosidase activity in fibroblasts from Krabbe patients. Molecular Genetics and Metabolism, 112(4), 294–301. 10.1016/j.ymgme.2014.05.009 [DOI] [PubMed] [Google Scholar]
  11. Biffi A, Montini E, Lorioli L, Cesani M, Fumagalli F, Plati T, … Naldini L (2013). Lentiviral hematopoietic stem cell gene therapy benefits metachromatic leukodystrophy. Science, 341(6148), 1233158. 10.1126/science.1233158 [DOI] [PubMed] [Google Scholar]
  12. Blanchette CD, Lin WC, Ratto TV, & Longo ML (2006). Galactosylceramide domain microstructure: impact of cholesterol and nucleation/growth conditions. Biophysical Journal, 90(12), 4466–4478. 10.1529/biophysj.105.072744 [DOI] [PMC free article] [PubMed] [Google Scholar]
  13. Boggs JM, Gao W, & Hirahara Y (2008). Myelin glycosphingolipids, galactosylceramide and sulfatide, participate in carbohydrate-carbohydrate interactions between apposed membranes and may form glycosynapses between oligodendrocyte and/or myelin membranes. Biochimica et Biophysica Acta, 1780(3), 445–455. 10.1016/j.bbagen.2007.10.015 [DOI] [PubMed] [Google Scholar]
  14. Bosio A, Binczek E, & Stoffel W (1996). Functional breakdown of the lipid bilayer of the myelin membrane in central and peripheral nervous system by disrupted galactocerebroside synthesis. Proceedings of the National Academy of Sciences of the United States of America, 93(23), 13280–13285. 10.1073/pnas.93.23.13280 [DOI] [PMC free article] [PubMed] [Google Scholar]
  15. Boya P, & Kroemer G (2008). Lysosomal membrane permeabilization in cell death. Oncogene, 27(50), 6434–6451. 10.1038/onc.2008.310 [DOI] [PubMed] [Google Scholar]
  16. Brinkmann V, Billich A, Baumruker T, Heining P, Schmouder R, Francis G, … Burtin P (2010). Fingolimod (FTY720): Discovery and development of an oral drug to treat multiple sclerosis. Nature Reviews. Drug Discovery, 9(11), 883–897. 10.1038/nrd3248 [DOI] [PubMed] [Google Scholar]
  17. Cannizzaro LA, Chen YQ, Rafi MA, & Wenger DA (1994). Regional mapping of the human galactocerebrosidase gene (GALC) to 14q31 by in situ hybridization. Cytogenetics and Cell Genetics, 66(4), 244–245. 10.1159/000133703 [DOI] [PubMed] [Google Scholar]
  18. Cantuti Castelvetri L, Givogri MI, Hebert A, Smith B, Song Y, Kaminska A, … Bongarzone ER (2013). The sphingolipid psychosine inhibits fast axonal transport in Krabbe disease by activation of GSK3β and deregulation of molecular motors. The Journal of Neuroscience, 33(24), 10048–10056. 10.1523/jneurosci.0217-13.2013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  19. Cantuti-Castelvetri L, Fitzner D, Bosch-Queralt M, Weil MT, Su M, Sen P, … Simons M (2018). Defective cholesterol clearance limits remyelination in the aged central nervous system. Science, 359(6376), 684–688. 10.1126/science.aan4183 [DOI] [PubMed] [Google Scholar]
  20. Cantuti-Castelvetri L, Maravilla E, Marshall M, Tamayo T, D’Auria L, Monge J, … Bongarzone ER (2015). Mechanism of neuromuscular dysfunction in Krabbe disease. The Journal of Neuroscience, 35(4), 1606–1616. 10.1523/jneurosci.2431-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  21. Cantuti-Castelvetri L, Zhu H, Givogri MI, Chidavaenzi RL, Lopez-Rosas A, & Bongarzone ER (2012). Psychosine induces the dephosphorylation of neurofilaments by deregulation of PP1 and PP2A phosphatases. Neurobiology of Disease, 46(2), 325–335. 10.1016/j.nbd.2012.01.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  22. Cappello V, Marchetti L, Parlanti P, Landi S, Tonazzini I, Cecchini M, … Gemmi M (2016). Ultrastructural characterization of the lower motor system in a mouse model of Krabbe disease. Scientific Reports, 6 (1), 1. 10.1038/s41598-016-0001-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  23. Castelvetri LC, Givogri MI, Zhu H, Smith B, Lopez-Rosas A, Qiu X, … Bongarzone ER (2011). Axonopathy is a compounding factor in the pathogenesis of Krabbe disease. Acta Neuropathologica, 122(1), 35–48. 10.1007/s00401-011-0814-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  24. Chang D, Nalls MA, Hallgrímsdóttir IB, Hunkapiller J, van der Brug M, Cai F, … Graham RR (2017). A meta-analysis of genome-wide association studies identifies 17 new Parkinson’s disease risk loci. Nature Genetics, 49(10), 1511–1516. 10.1038/ng.3955 [DOI] [PMC free article] [PubMed] [Google Scholar]
  25. Chen YQ, Rafi MA, de Gala G, & Wenger DA (1993). Cloning and expression of cDNA encoding human galactocerebrosidase, the enzyme deficient in globoid cell leukodystrophy. Human Molecular Genetics, 2(11), 1841–1845. 10.1093/hmg/2.11.1841 [DOI] [PubMed] [Google Scholar]
  26. Chrast R, Saher G, Nave KA, & Verheijen MH (2011). Lipid metabolism in myelinating glial cells: Lessons from human inherited disorders and mouse models. Journal of Lipid Research, 52(3), 419–434. 10.1194/jlr.R009761 [DOI] [PMC free article] [PubMed] [Google Scholar]
  27. Chuang WL, Pacheco J, Zhang XK, Martin MM, Biski CK, Keutzer JM, … Orsini JJ Jr. (2013). Determination of psychosine concentration in dried blood spots from newborns that were identified via newborn screening to be at risk for Krabbe disease. Clinica Chimica Acta, 419, 73–76. 10.1016/j.cca.2013.01.017 [DOI] [PubMed] [Google Scholar]
  28. Cleland WW, & Kennedy EP (1960). The enzymatic synthesis of psychosine. The Journal of Biological Chemistry, 235, 45–51. [PubMed] [Google Scholar]
  29. Coetzee T, Fujita N, Dupree J, Shi R, Blight A, Suzuki K, … Popko B (1996). Myelination in the absence of galactocerebroside and sulfatide: Normal structure with abnormal function and regional instability. Cell, 86(2), 209–219. 10.1016/s0092-8674(00)80093-8 [DOI] [PubMed] [Google Scholar]
  30. Corado CR, Pinkstaff J, Jiang X, Galban EM, Fisher SJ, Scholler O, … Bradbury AM (2020). Cerebrospinal fluid and serum glycosphingolipid biomarkers in canine globoid cell leukodystrophy (Krabbe disease). Molecular and Cellular Neurosciences, 102, 103451. 10.1016/j.mcn.2019.103451 [DOI] [PMC free article] [PubMed] [Google Scholar]
  31. Costantino-Ceccarini E, Luddi A, Volterrani M, Strazza M, Rafi MA, & Wenger DA (1999). Transduction of cultured oligodendrocytes from normal and twitcher mice by a retroviral vector containing human galactocerebrosidase (GALC) cDNA. Neurochemical Research, 24(2), 287–293. 10.1023/a:1022574323784 [DOI] [PubMed] [Google Scholar]
  32. Coutinho MF, Prata MJ, & Alves S (2012). Mannose-6-phosphate pathway: A review on its role in lysosomal function and dysfunction. Molecular Genetics and Metabolism, 105(4), 542–550. 10.1016/j.ymgme.2011.12.012 [DOI] [PubMed] [Google Scholar]
  33. D’Agostino AN, Sayre GP, & Hayles AB (1963). Krabbe’s disease. Globoid cell type of leukodystrophy. Archives of Neurology, 8, 82–96. 10.1001/archneur.1963.00460010098012 [DOI] [PubMed] [Google Scholar]
  34. De Gasperi R, Friedrich VL, Perez GM, Senturk E, Wen PH, Kelley K, … Gama Sosa MA (2004). Transgenic rescue of Krabbe disease in the twitcher mouse. Gene Therapy, 11(15), 1188–1194. 10.1038/sj.gt.3302282 [DOI] [PubMed] [Google Scholar]
  35. Deane JE, Graham SC, Kim NN, Stein PE, McNair R, Cachón-González MB, … Read RJ (2011). Insights into Krabbe disease from structures of galactocerebrosidase. Proceedings of the National Academy of Sciences of the United States of America, 108(37), 15169–15173. 10.1073/pnas.1105639108 [DOI] [PMC free article] [PubMed] [Google Scholar]
  36. Del Grosso A, Angella L, Tonazzini I, Moscardini A, Giordano N, Caleo M, … Cecchini M (2019). Dysregulated autophagy as a new aspect of the molecular pathogenesis of Krabbe disease. Neurobiology of Disease, 129, 195–207. 10.1016/j.nbd.2019.05.011 [DOI] [PubMed] [Google Scholar]
  37. Del Grosso A, Antonini S, Angella L, Tonazzini I, Signore G, & Cecchini M (2016). Lithium improves cell viability in psychosine-treated MO3.13 human oligodendrocyte cell line via autophagy activation. Journal of Neuroscience Research, 94(11), 1246–1260. 10.1002/jnr.23910 [DOI] [PubMed] [Google Scholar]
  38. Dolcetta D, Perani L, Givogri MI, Galbiati F, Orlacchio A, Martino S, … Bongarzone E (2004). Analysis of galactocerebrosidase activity in the mouse brain by a new histological staining method. Journal of Neuroscience Research, 77(3), 462–464. 10.1002/jnr.20169 [DOI] [PubMed] [Google Scholar]
  39. Dostert C, Pétrilli V, Van Bruggen R, Steele C, Mossman BT, & Tschopp J (2008). Innate immune activation through Nalp3 inflammasome sensing of asbestos and silica. Science, 320(5876), 674–677. 10.1126/science.1156995 [DOI] [PMC free article] [PubMed] [Google Scholar]
  40. Duchen LW, Eicher EM, Jacobs JM, Scaravilli F, & Teixeira F (1980). Hereditary leucodystrophy in the mouse: The new mutant twitcher. Brain, 103(3), 695–710. 10.1093/brain/103.3.695 [DOI] [PubMed] [Google Scholar]
  41. Duewell P, Kono H, Rayner KJ, Sirois CM, Vladimer G, Bauernfeind FG, … Latz E (2010). NLRP3 inflammasomes are required for atherogenesis and activated by cholesterol crystals. Nature, 464(7293), 1357–1361. 10.1038/nature08938 [DOI] [PMC free article] [PubMed] [Google Scholar]
  42. Dupree JL, Girault JA, & Popko B (1999). Axo-glial interactions regulate the localization of axonal paranodal proteins. The Journal of Cell Biology, 147(6), 1145–1152. 10.1083/jcb.147.6.1145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  43. Eriksson I, Wäster P, & Öllinger K (2020). Restoration of lysosomal function after damage is accompanied by recycling of lysosomal membrane proteins. Cell Death & Disease, 11(5), 370. 10.1038/s41419-020-2527-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  44. Escolar ML, Poe MD, Provenzale JM, Richards KC, Allison J, Wood S, … Kurtzberg J (2005). Transplantation of umbilical-cord blood in babies with infantile Krabbe’s disease. The New England Journal of Medicine, 352(20), 2069–2081. 10.1056/NEJMoa042604 [DOI] [PubMed] [Google Scholar]
  45. Escolar ML, West T, Dallavecchia A, Poe MD, & LaPoint K (2016). Clinical management of Krabbe disease. Journal of Neuroscience Research, 94(11), 1118–1125. 10.1002/jnr.23891 [DOI] [PubMed] [Google Scholar]
  46. Ezoe T, Vanier MT, Oya Y, Popko B, Tohyama J, Matsuda J, … Suzuki K (2000a). Biochemistry and neuropathology of mice doubly deficient in synthesis and degradation of galactosylceramide. Journal of Neuroscience Research, 59(2), 170–178. [DOI] [PubMed] [Google Scholar]
  47. Ezoe T, Vanier MT, Oya Y, Popko B, Tohyama J, Matsuda J, … Suzuki K (2000b). Twitcher mice with only a single active galactosylceramide synthase gene exhibit clearly detectable but therapeutically minor phenotypic improvements. Journal of Neuroscience Research, 59(2), 179–187. [DOI] [PubMed] [Google Scholar]
  48. Fernández-Monreal M, Brown TC, Royo M, & Esteban JA (2012). The balance between receptor recycling and trafficking toward lysosomes determines synaptic strength during long-term depression. The Journal of Neuroscience, 32(38), 13200–13205. 10.1523/jneurosci.0061-12.2012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  49. Ferreira CR, & Gahl WA (2017). Lysosomal storage diseases. Translational Science of Rare Diseases, 2(1–2), 1–71. 10.3233/trd-160005 [DOI] [PMC free article] [PubMed] [Google Scholar]
  50. Fletcher TF, Suzuki K, & Martin FB (1977). Galactocerebrosidase activity in canine globoid leukodystrophy. Neurology, 27(8), 758–766. 10.1212/wnl.27.8.758 [DOI] [PubMed] [Google Scholar]
  51. Folts CJ, Scott-Hewitt N, Pröschel C, Mayer-Pröschel M, & Noble M (2016). Lysosomal re-acidification prevents lysosphingolipid-induced lysosomal impairment and cellular toxicity. PLoS Biology, 14(12), e1002583. 10.1371/journal.pbio.1002583 [DOI] [PMC free article] [PubMed] [Google Scholar]
  52. Formichi P, Radi E, Battisti C, Pasqui A, Pompella G, Lazzerini PE, … Federico A (2007). Psychosine-induced apoptosis and cytokine activation in immune peripheral cells of Krabbe patients. Journal of Cellular Physiology, 212(3), 737–743. 10.1002/jcp.21070 [DOI] [PubMed] [Google Scholar]
  53. Fratantoni JC, Hall CW, & Neufeld EF (1969). The defect in Hurler and Hunter syndromes. II. Deficiency of specific factors involved in mucopolysaccharide degradation. Proceedings of the National Academy of Sciences of the United States of America, 64(1), 360–366. 10.1073/pnas.64.1.360 [DOI] [PMC free article] [PubMed] [Google Scholar]
  54. Fry JM, Weissbarth S, Lehrer GM, & Bornstein MB (1974). Cerebroside antibody inhibits sulfatide synthesis and myelination and demyelinates in cord tissue cultures. Science, 183(4124), 540–542. 10.1126/science.183.4124.540 [DOI] [PubMed] [Google Scholar]
  55. Galbiati F, Basso V, Cantuti L, Givogri MI, Lopez-Rosas A, Perez N, … Bongarzone ER (2007). Autonomic denervation of lymphoid organs leads to epigenetic immune atrophy in a mouse model of Krabbe disease. The Journal of Neuroscience, 27(50), 13730–13738. 10.1523/jneurosci.3379-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
  56. Galbiati F, Givogri MI, Cantuti L, Rosas AL, Cao H, van Breemen R, & Bongarzone ER (2009). Combined hematopoietic and lentiviral gene-transfer therapies in newborn Twitcher mice reveal contemporaneous neurodegeneration and demyelination in Krabbe disease. Journal of Neuroscience Research, 87(8), 1748–1759. 10.1002/jnr.22006 [DOI] [PubMed] [Google Scholar]
  57. Garbay B, Heape AM, Sargueil F, & Cassagne C (2000). Myelin synthesis in the peripheral nervous system. Progress in Neurobiology, 61(3), 267–304. 10.1016/s0301-0082(99)00049-0 [DOI] [PubMed] [Google Scholar]
  58. Gault CR, Obeid LM, & Hannun YA (2010). An overview of sphingolipid metabolism: From synthesis to breakdown. Advances in Experimental Medicine and Biology, 688, 1–23. 10.1007/978-1-4419-6741-1_1 [DOI] [PMC free article] [PubMed] [Google Scholar]
  59. Gentner B, Visigalli I, Hiramatsu H, Lechman E, Ungari S, Giustacchini A, … Naldini L (2010). Identification of hematopoietic stem cell-specific miRNAs enables gene therapy of globoid cell leukodystrophy. Science Translational Medicine, 2(58), 58ra84. 10.1126/scitranslmed.3001522 [DOI] [PubMed] [Google Scholar]
  60. Germain DP (2002). Fabry disease: Recent advances in enzyme replacement therapy. Expert Opinion on Investigational Drugs, 11(10), 1467–1476. 10.1517/13543784.11.10.1467 [DOI] [PubMed] [Google Scholar]
  61. Goo MS, Sancho L, Slepak N, Boassa D, Deerinck TJ, Ellisman MH, … Patrick GN (2017). Activity-dependent trafficking of lysosomes in dendrites and dendritic spines. The Journal of Cell Biology, 216(8), 2499–2513. 10.1083/jcb.201704068 [DOI] [PMC free article] [PubMed] [Google Scholar]
  62. Gu H, Zou YR, & Rajewsky K (1993). Independent control of immunoglobulin switch recombination at individual switch regions evidenced through Cre-loxP-mediated gene targeting. Cell, 73(6), 1155–1164. 10.1016/0092-8674(93)90644-6 [DOI] [PubMed] [Google Scholar]
  63. Halle A, Hornung V, Petzold GC, Stewart CR, Monks BG, Reinheckel T, … Golenbock DT (2008). The NALP3 inflammasome is involved in the innate immune response to amyloid-beta. Nature Immunology, 9(8), 857–865. 10.1038/ni.1636 [DOI] [PMC free article] [PubMed] [Google Scholar]
  64. Hannun YA, & Bell RM (1987). Lysosphingolipids inhibit protein kinase C: Implications for the sphingolipidoses. Science, 235(4789), 670–674. 10.1126/science.3101176 [DOI] [PubMed] [Google Scholar]
  65. Hannun YA, & Bell RM (1989). Regulation of protein kinase C by sphingosine and lysosphingolipids. Clinica Chimica Acta, 185(3), 333–345. 10.1016/0009-8981(89)90224-6 [DOI] [PubMed] [Google Scholar]
  66. Haq E, Giri S, Singh I, & Singh AK (2003). Molecular mechanism of psychosine-induced cell death in human oligodendrocyte cell line. Journal of Neurochemistry, 86(6), 1428–1440. 10.1046/j.1471-4159.2003.01941.x [DOI] [PubMed] [Google Scholar]
  67. Hawkins-Salsbury JA, Parameswar AR, Jiang X, Schlesinger PH, Bongarzone E, Ory DS, … Sands MS (2013). Psychosine, the cytotoxic sphingolipid that accumulates in globoid cell leukodystrophy, alters membrane architecture. Journal of Lipid Research, 54(12), 3303–3311. 10.1194/jlr.M039610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  68. Hawkins-Salsbury JA, Shea L, Jiang X, Hunter DA, Guzman AM, Reddy AS, … Sands MS (2015). Mechanism-based combination treatment dramatically increases therapeutic efficacy in murine globoid cell leukodystrophy. The Journal of Neuroscience, 35(16), 6495–6505. 10.1523/jneurosci.4199-14.2015 [DOI] [PMC free article] [PubMed] [Google Scholar]
  69. Hensch TK (2005). Critical period plasticity in local cortical circuits. Nature Reviews. Neuroscience, 6(11), 877–888. 10.1038/nrn1787 [DOI] [PubMed] [Google Scholar]
  70. Hess MW, Kirschning E, Pfaller K, Debbage PL, Hohenberg H, & Klima G (1998). 5000-year-old myelin: Uniquely intact in molecular configuration and fine structure. Current Biology, 8(15), R512–R513. 10.1016/s0960-9822(07)00334-x [DOI] [PubMed] [Google Scholar]
  71. Hill CH, Cook GM, Spratley SJ, Fawke S, Graham SC, & Deane JE (2018). The mechanism of glycosphingolipid degradation revealed by a GALC-SapA complex structure. Nature Communications, 9(1), 151. 10.1038/s41467-017-02361-y [DOI] [PMC free article] [PubMed] [Google Scholar]
  72. Hirth RS, & Nielsen SW (1967). A familial canine globoid cell leukodystrophy (“Krabbe Type”). The Journal of Small Animal Practice, 8(10), 569–575. 10.1111/j.1748-5827.1967.tb04499.x [DOI] [PubMed] [Google Scholar]
  73. Hobbs JR, Hugh-Jones K, Barrett AJ, Byrom N, Chambers D, Henry K, … Young EP (1981). Reversal of clinical features of Hurler’s disease and biochemical improvement after treatment by bone-marrow transplantation. Lancet, 2(8249), 709–712. 10.1016/s0140-6736(81)91046-1 [DOI] [PubMed] [Google Scholar]
  74. Holopainen JM, Saarikoski J, Kinnunen PK, & Järvelä I (2001). Elevated lysosomal pH in neuronal ceroid lipofuscinoses (NCLs). European Journal of Biochemistry, 268(22), 5851–5856. 10.1046/j.0014-2956.2001.02530.x [DOI] [PubMed] [Google Scholar]
  75. Hong C, Sutherland DE, Matas AJ, & Najarian JS (1979). Bone marrow transplantation for correction of enzyme deficiency disease. Transplantation Proceedings, 11(1), 498–503. [PubMed] [Google Scholar]
  76. Honke K, Hirahara Y, Dupree J, Suzuki K, Popko B, Fukushima K, … Taniguchi N (2002). Paranodal junction formation and spermatogenesis require sulfoglycolipids. Proceedings of the National Academy of Sciences of the United States of America, 99(7), 4227–4232. 10.1073/pnas.032068299 [DOI] [PMC free article] [PubMed] [Google Scholar]
  77. Hoogerbrugge PM, Suzuki K, Suzuki K, Poorthuis BJ, Kobayashi T, Wagemaker G, & van Bekkum DW (1988). Donor-derived cells in the central nervous system of twitcher mice after bone marrow transplantation. Science, 239(4843), 1035–1038. 10.1126/science.3278379 [DOI] [PubMed] [Google Scholar]
  78. Ichioka T, Kishimoto Y, Brennan S, Santos GW, & Yeager AM (1987). Hematopoietic cell transplantation in murine globoid cell leukodystrophy (the twitcher mouse): Effects on levels of galactosylceramidase, psychosine, and galactocerebrosides. Proceedings of the National Academy of Sciences of the United States of America, 84(12), 4259–4263. 10.1073/pnas.84.12.4259 [DOI] [PMC free article] [PubMed] [Google Scholar]
  79. Igisu H, Hamasaki N, Ito A, & Ou W (1988). Inhibition of cytochrome c oxidase and hemolysis caused by lysosphingolipids. Lipids, 23(4), 345–348. 10.1007/bf02537346 [DOI] [PubMed] [Google Scholar]
  80. Igisu H, & Nakamura M (1986). Inhibition of cytochrome c oxidase by psychosine (galactosylsphingosine). Biochemical and Biophysical Research Communications, 137(1), 323–327. 10.1016/0006-291x(86)91213-1 [DOI] [PubMed] [Google Scholar]
  81. Ijichi K, Brown GD, Moore CS, Lee JP, Winokur PN, Pagarigan R, … Crocker SJ (2013). MMP-3 mediates psychosine-induced globoid cell formation: Implications for leukodystrophy pathology. Glia, 61(5), 765–777. 10.1002/glia.22471 [DOI] [PMC free article] [PubMed] [Google Scholar]
  82. Im DS, Heise CE, Nguyen T, O’Dowd BF, & Lynch KR (2001). Identification of a molecular target of psychosine and its role in globoid cell formation. The Journal of Cell Biology, 153(2), 429–434. 10.1083/jcb.153.2.429 [DOI] [PMC free article] [PubMed] [Google Scholar]
  83. Imgrund S, Hartmann D, Farwanah H, Eckhardt M, Sandhoff R, Degen J, … Willecke K (2009). Adult ceramide synthase 2 (CERS2)-deficient mice exhibit myelin sheath defects, cerebellar degeneration, and hepatocarcinomas. The Journal of Biological Chemistry, 284(48), 33549–33560. 10.1074/jbc.M109.031971 [DOI] [PMC free article] [PubMed] [Google Scholar]
  84. Irahara-Miyana K, Otomo T, Kondo H, Hossain MA, Ozono K, & Sakai N (2018). Unfolded protein response is activated in Krabbe disease in a manner dependent on the mutation type. Journal of Human Genetics, 63(6), 699–706. 10.1038/s10038-018-0445-8 [DOI] [PubMed] [Google Scholar]
  85. Irino M, & Suzuki K (1990). Isolation and morphologic characterization in vitro of Schwann cells of the twitcher mouse. Laboratory Investigation, 62(6), 704–712. [PubMed] [Google Scholar]
  86. Jacobs JM, Scaravilli F, & De Aranda FT (1982). The pathogenesis of globoid cell leucodystrophy in peripheral nerve of the mouse mutant twitcher. Journal of the Neurological Sciences, 55(3), 285–304. 10.1016/0022-510x(82)90127-7 [DOI] [PubMed] [Google Scholar]
  87. Jatana M, Giri S, & Singh AK (2002). Apoptotic positive cells in Krabbe brain and induction of apoptosis in rat C6 glial cells by psychosine. Neuroscience Letters, 330(2), 183–187. 10.1016/s0304-3940(02)00655-9 [DOI] [PubMed] [Google Scholar]
  88. Johnson KH (1970). Globoid leukodystrophy in the cat. Journal of the American Veterinary Medical Association, 157(12), 2057–2064. [PubMed] [Google Scholar]
  89. Kagitani-Shimono K, Mohri I, Fujitani Y, Suzuki K, Ozono K, Urade Y, & Taniike M (2005). Anti-inflammatory therapy by ibudilast, a phosphodiesterase inhibitor, in demyelination of twitcher, a genetic demyelination model. Journal of Neuroinflammation, 2(1), 10. 10.1186/1742-2094-2-10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  90. Kanazawa T, Takematsu H, Yamamoto A, Yamamoto H, & Kozutsumi Y (2008). Wheat germ agglutinin stains dispersed post-golgi vesicles after treatment with the cytokinesis inhibitor psychosine. Journal of Cellular Physiology, 215(2), 517–525. 10.1002/jcp.21328 [DOI] [PubMed] [Google Scholar]
  91. Karumuthil-Melethil S, Marshall MS, Heindel C, Jakubauskas B, Bongarzone ER, & Gray SJ (2016). Intrathecal administration of AAV/GALC vectors in 10–11-day-old twitcher mice improves survival and is enhanced by bone marrow transplant. Journal of Neuroscience Research, 94(11), 1138–1151. 10.1002/jnr.23882 [DOI] [PMC free article] [PubMed] [Google Scholar]
  92. Kast DJ, & Dominguez R (2017). The cytoskeleton-autophagy connection. Current Biology, 27(8), R318–r326. 10.1016/j.cub.2017.02.061 [DOI] [PMC free article] [PubMed] [Google Scholar]
  93. Kaushik S, Bandyopadhyay U, Sridhar S, Kiffin R, Martinez-Vicente M, Kon M, … Cuervo AM (2011). Chaperone-mediated autophagy at a glance. Journal of Cell Science, 124(Pt 4), 495–499. 10.1242/jcs.073874 [DOI] [PMC free article] [PubMed] [Google Scholar]
  94. Khan M, Haq E, Giri S, Singh I, & Singh AK (2005). Peroxisomal participation in psychosine-mediated toxicity: Implications for Krabbe’s disease. Journal of Neuroscience Research, 80(6), 845–854. 10.1002/jnr.20529 [DOI] [PubMed] [Google Scholar]
  95. Kim I, Rodriguez-Enriquez S, & Lemasters JJ (2007). Selective degradation of mitochondria by mitophagy. Archives of Biochemistry and Biophysics, 462(2), 245–253. 10.1016/j.abb.2007.03.034 [DOI] [PMC free article] [PubMed] [Google Scholar]
  96. Kollmann K, Damme M, Markmann S, Morelle W, Schweizer M, Hermans-Borgmeyer I, … Braulke T (2012). Lysosomal dysfunction causes neurodegeneration in mucolipidosis II ‘knock-in’ mice. Brain, 135(Pt 9), 2661–2675. 10.1093/brain/aws209 [DOI] [PMC free article] [PubMed] [Google Scholar]
  97. Kondo A, Hoogerbrugge PM, Suzuki K, Poorthuis BJ, Van Bekkum DW, & Suzuki K (1988). Pathology of the peripheral nerve in the twitcher mouse following bone marrow transplantation. Brain Research, 460(1), 178–183. 10.1016/0006-8993(88)91220-6 [DOI] [PubMed] [Google Scholar]
  98. Kondo Y, Adams JM, Vanier MT, & Duncan ID (2011). Macrophages counteract demyelination in a mouse model of globoid cell leukodystrophy. The Journal of Neuroscience, 31(10), 3610–3624. 10.1523/jneurosci.6344-10.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  99. Kondo Y, Wenger DA, Gallo V, & Duncan ID (2005). Galactocerebrosidase-deficient oligodendrocytes maintain stable central myelin by exogenous replacement of the missing enzyme in mice. Proceedings of the National Academy of Sciences of the United States of America, 102(51), 18670–18675. 10.1073/pnas.0506473102 [DOI] [PMC free article] [PubMed] [Google Scholar]
  100. Krabbe K (1916). A new familial, infantile form of diffuse brain-sclerosis. Brain, 39(1–2), 74–114. [DOI] [PubMed] [Google Scholar]
  101. Krivit W, Shapiro EG, Peters C, Wagner JE, Cornu G, Kurtzberg J, … Lockman LA (1998). Hematopoietic stem-cell transplantation in globoid-cell leukodystrophy. The New England Journal of Medicine, 338(16), 1119–1126. 10.1056/nejm199804163381605 [DOI] [PubMed] [Google Scholar]
  102. Kuai XL, Ni RZ, Zhou GX, Mao ZB, Zhang JF, Yi N, … Wang ZW (2015). Transplantation of mouse embryonic stem cell-derived oligodendrocytes in the murine model of globoid cell leukodystrophy. Stem Cell Research & Therapy, 6(1), 30. 10.1186/s13287-015-0024-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
  103. Lasiene J, Matsui A, Sawa Y, Wong F, & Horner PJ (2009). Age-related myelin dynamics revealed by increased oligodendrogenesis and short internodes. Aging Cell, 8(2), 201–213. 10.1111/j.1474-9726.2009.00462.x [DOI] [PMC free article] [PubMed] [Google Scholar]
  104. Lattanzi A, Neri M, Maderna C, di Girolamo I, Martino S, Orlacchio A, … Gritti A (2010). Widespread enzymatic correction of CNS tissues by a single intracerebral injection of therapeutic lentiviral vector in leukodystrophy mouse models. Human Molecular Genetics, 19(11), 2208–2227. 10.1093/hmg/ddq099 [DOI] [PubMed] [Google Scholar]
  105. Lee WC, Courtenay A, Troendle FJ, Stallings-Mann ML, Dickey CA, DeLucia MW, … Eckman CB (2005). Enzyme replacement therapy results in substantial improvements in early clinical phenotype in a mouse model of globoid cell leukodystrophy. The FASEB Journal, 19(11), 1549–1551. 10.1096/fj.05-3826fje [DOI] [PubMed] [Google Scholar]
  106. Lee WC, Tsoi YK, Troendle FJ, DeLucia MW, Ahmed Z, Dicky CA, … Eckman CB (2007). Single-dose intracerebroventricular administration of galactocerebrosidase improves survival in a mouse model of globoid cell leukodystrophy. The FASEB Journal, 21(10), 2520–2527. 10.1096/fj.06-6169com [DOI] [PubMed] [Google Scholar]
  107. LeVine SM, Pedchenko TV, Bronshteyn IG, & Pinson DM (2000). L-cycloserine slows the clinical and pathological course in mice with globoid cell leukodystrophy (twitcher mice). Journal of Neuroscience Research, 60(2), 231–236. [DOI] [PubMed] [Google Scholar]
  108. LeVine SM, Wetzel DL, & Eilert AJ (1994). Neuropathology of twitcher mice: Examination by histochemistry, immunohisto-chemistry, lectin histochemistry and Fourier transform infrared microspectroscopy. International Journal of Developmental Neuroscience, 12(4), 275–288. 10.1016/0736-5748(94)90075-2 [DOI] [PubMed] [Google Scholar]
  109. Li Y, Miller CA, Shea LK, Jiang X, Guzman MA, Chandler RJ, … Sands MS (2021). Enhanced efficacy and increased long-term toxicity of CNS-directed, AAV-based combination therapy for Krabbe disease. Molecular Therapy, 29(2), 691–701. 10.1016/j.ymthe.2020.12.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  110. Li Y, Xu Y, Benitez BA, Nagree MS, Dearborn JT, Jiang X, … Sands MS (2019). Genetic ablation of acid ceramidase in Krabbe disease confirms the psychosine hypothesis and identifies a new therapeutic target. Proceedings of the National Academy of Sciences of the United States of America, 116(40), 20097–20103. 10.1073/pnas.1912108116 [DOI] [PMC free article] [PubMed] [Google Scholar]
  111. Lieberman AP, Puertollano R, Raben N, Slaugenhaupt S, Walkley SU, & Ballabio A (2012). Autophagy in lysosomal storage disorders. Autophagy, 8(5), 719–730. 10.4161/auto.19469 [DOI] [PMC free article] [PubMed] [Google Scholar]
  112. Lin D, Donsante A, Macauley S, Levy B, Vogler C, & Sands MS (2007). Central nervous system-directed AAV2/5-mediated gene therapy synergizes with bone marrow transplantation in the murine model of globoid-cell leukodystrophy. Molecular Therapy, 15(1), 44–52. 10.1038/sj.mt.6300026 [DOI] [PubMed] [Google Scholar]
  113. Lin D, Fantz CR, Levy B, Rafi MA, Vogler C, Wenger DA, & Sands MS (2005). AAV2/5 vector expressing galactocerebrosidase ameliorates CNS disease in the murine model of globoid-cell leukodystrophy more efficiently than AAV2. Molecular Therapy, 12(3), 422–430. 10.1016/j.ymthe.2005.04.019 [DOI] [PubMed] [Google Scholar]
  114. Lin DS, Ho CS, Huang YW, Wu TY, Lee TH, Huang ZD, … Chiang MF (2020). Impairment of proteasome and autophagy underlying the pathogenesis of Leukodystrophy. Cell, 9(5). 1124. 10.3390/cells9051124 [DOI] [PMC free article] [PubMed] [Google Scholar]
  115. Lin YN, & Radin NS (1973). Alternate pathways of cerebroside catabolism. Lipids, 8(12), 732–736. 10.1007/bf02531841 [DOI] [PubMed] [Google Scholar]
  116. Lübke T, Lobel P, & Sleat DE (2009). Proteomics of the lysosome. Biochimica et Biophysica Acta, 1793(4), 625–635. 10.1016/j.bbamcr.2008.09.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  117. Luciani M, Gritti A, & Meneghini V (2020). Human iPSC-based models for the development of therapeutics targeting neurodegenerative lysosomal storage diseases. Frontiers in Molecular Biosciences, 7, 224. 10.3389/fmolb.2020.00224 [DOI] [PMC free article] [PubMed] [Google Scholar]
  118. Luddi A, Volterrani M, Strazza M, Smorlesi A, Rafi MA, Datto J, … Costantino-Ceccarini E (2001). Retrovirus-mediated gene transfer and galactocerebrosidase uptake into twitcher glial cells results in appropriate localization and phenotype correction. Neurobiology of Disease, 8(4), 600–610. 10.1006/nbdi.2001.0407 [DOI] [PubMed] [Google Scholar]
  119. Luzi P, Abraham RM, Rafi MA, Curtis M, Hooper DC, & Wenger DA (2009). Effects of treatments on inflammatory and apoptotic markers in the CNS of mice with globoid cell leukodystrophy. Brain Research, 1300, 146–158. 10.1016/j.brainres.2009.09.017 [DOI] [PMC free article] [PubMed] [Google Scholar]
  120. Luzi P, Rafi MA, Victoria T, Baskin GB, & Wenger DA (1997). Characterization of the rhesus monkey galactocerebrosidase (GALC) cDNA and gene and identification of the mutation causing globoid cell leukodystrophy (Krabbe disease) in this primate. Genomics, 42(2), 319–324. 10.1006/geno.1997.4744 [DOI] [PubMed] [Google Scholar]
  121. Marín O (2016). Developmental timing and critical windows for the treatment of psychiatric disorders. Nature Medicine, 22(11), 1229–1238. 10.1038/nm.4225 [DOI] [PubMed] [Google Scholar]
  122. Marshall MS, Issa Y, Jakubauskas B, Stoskute M, Elackattu V, Marshall JN, … Bongarzone ER (2018). Long-term improvement of neurological signs and metabolic dysfunction in a mouse model of Krabbe’s disease after global gene therapy. Molecular Therapy, 26(3), 874–889. 10.1016/j.ymthe.2018.01.009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  123. Martinon F, Pétrilli V, Mayor A, Tardivel A, & Tschopp J (2006). Gout-associated uric acid crystals activate the NALP3 inflammasome. Nature, 440(7081), 237–241. 10.1038/nature04516 [DOI] [PubMed] [Google Scholar]
  124. Marzella L, Ahlberg J, & Glaumann H (1982). Isolation of autophagic vacuoles from rat liver: Morphological and biochemical characterization. The Journal of Cell Biology, 93(1), 144–154. 10.1083/jcb.93.1.144 [DOI] [PMC free article] [PubMed] [Google Scholar]
  125. Matsuda J, Vanier MT, Saito Y, Tohyama J, Suzuki K, & Suzuki K (2001). A mutation in the saposin A domain of the sphingolipid activator protein (prosaposin) gene results in a late-onset, chronic form of globoid cell leukodystrophy in the mouse. Human Molecular Genetics, 10(11), 1191–1199. 10.1093/hmg/10.11.1191 [DOI] [PubMed] [Google Scholar]
  126. Matsushima GK, Taniike M, Glimcher LH, Grusby MJ, Frelinger JA, Suzuki K, & Ting JP (1994). Absence of MHC class II molecules reduces CNS demyelination, microglial/macrophage infiltration, and twitching in murine globoid cell leukodystrophy. Cell, 78(4), 645–656. 10.1016/0092-8674(94)90529-0 [DOI] [PubMed] [Google Scholar]
  127. Matthes F, Andersson C, Stein A, Eistrup C, Fogh J, Gieselmann V, … Matzner U (2015). Enzyme replacement therapy of a novel humanized mouse model of globoid cell leukodystrophy. Experimental Neurology, 271, 36–45. 10.1016/j.expneurol.2015.04.020 [DOI] [PubMed] [Google Scholar]
  128. Mayo L, Trauger SA, Blain M, Nadeau M, Patel B, Alvarez JI, … Quintana FJ (2014). Regulation of astrocyte activation by glycolipids drives chronic CNS inflammation. Nature Medicine, 20(10), 1147–1156. 10.1038/nm.3681 [DOI] [PMC free article] [PubMed] [Google Scholar]
  129. Meng XL, Shen JS, Watabe K, Ohashi T, & Eto Y (2005). GALC transduction leads to morphological improvement of the twitcher oligodendrocytes in vivo. Molecular Genetics and Metabolism, 84(4), 332–343. 10.1016/j.ymgme.2004.12.007 [DOI] [PubMed] [Google Scholar]
  130. Migdalska-Richards A, & Schapira AH (2016). The relationship between glucocerebrosidase mutations and Parkinson disease. Journal of Neurochemistry, 139(Suppl 1), 77–90. 10.1111/jnc.13385 [DOI] [PMC free article] [PubMed] [Google Scholar]
  131. Mikulka CR, Dearborn JT, Benitez BA, Strickland A, Liu L, Milbrandt J, & Sands MS (2020). Cell-autonomous expression of the acid hydrolase galactocerebrosidase. Proceedings of the National Academy of Sciences of the United States of America, 117(16), 9032–9041. 10.1073/pnas.1917675117 [DOI] [PMC free article] [PubMed] [Google Scholar]
  132. Mikulka CR, & Sands MS (2016). Treatment for Krabbe’s disease: Finding the combination. Journal of Neuroscience Research, 94(11), 1126–1137. 10.1002/jnr.23822 [DOI] [PMC free article] [PubMed] [Google Scholar]
  133. Miyatake T, & Suzuki K (1972). Globoid cell leukodystrophy: Additional deficiency of psychosine galactosidase. Biochemical and Biophysical Research Communications, 48(3), 539–543. 10.1016/0006-291x(72)90381-6 [DOI] [PubMed] [Google Scholar]
  134. Miyawaki S, Mitsuoka S, Sakiyama T, & Kitagawa T (1982). Sphingomyelinosis, a new mutation in the mouse: A model of Niemann-Pick disease in humans. The Journal of Heredity, 73(4), 257–263. 10.1093/oxfordjournals.jhered.a109635 [DOI] [PubMed] [Google Scholar]
  135. Mohri I, Taniike M, Taniguchi H, Kanekiyo T, Aritake K, Inui T, … Urade Y (2006). Prostaglandin D2-mediated microglia/astrocyte interaction enhances astrogliosis and demyelination in twitcher. The Journal of Neuroscience, 26(16), 4383–4393. 10.1523/jneurosci.4531-05.2006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  136. Nagano S, Yamada T, Shinnoh N, Furuya H, Taniwaki T, & Kira J (1998). Expression and processing of recombinant human galactosylceramidase. Clinica Chimica Acta, 276(1), 53–61. 10.1016/s0009-8981(98)00095-3 [DOI] [PubMed] [Google Scholar]
  137. Neri M, Ricca A, di Girolamo I, Alcala’-Franco B, Cavazzin C, Orlacchio A, … Gritti A (2011). Neural stem cell gene therapy ameliorates pathology and function in a mouse model of globoid cell leukodystrophy. Stem Cells, 29(10), 1559–1571. 10.1002/stem.701 [DOI] [PMC free article] [PubMed] [Google Scholar]
  138. Neufeld EF, & Fratantoni JC (1970). Inborn errors of mucopolysaccharide metabolism. Science, 169(3941), 141–146. 10.1126/science.169.3941.141 [DOI] [PubMed] [Google Scholar]
  139. Nicaise AM, Bongarzone ER, & Crocker SJ (2016). A microglial hypothesis of globoid cell leukodystrophy pathology. Journal of Neuroscience Research, 94(11), 1049–1061. 10.1002/jnr.23773 [DOI] [PMC free article] [PubMed] [Google Scholar]
  140. Norton WT, & Cammer W (1984). Isolation and characterization of myelin. In Myelin (pp. 147–195). Boston, MA: Springer. [Google Scholar]
  141. O’Brien JS (1965). Stability of the myelin membrane. Science, 147(3662), 1099–1107. 10.1126/science.147.3662.1099 [DOI] [PubMed] [Google Scholar]
  142. O’Sullivan C, & Dev KK (2015). Galactosylsphingosine (psychosine)-induced demyelination is attenuated by sphingosine 1-phosphate signalling. Journal of Cell Science, 128(21), 3878–3887. 10.1242/jcs.169342 [DOI] [PubMed] [Google Scholar]
  143. Okeda R, Suzuki Y, Horiguchi S, & Fujii T (1979). Fetal globoid cell leukodystrophy in one of twins. Acta Neuropathologica, 47(2), 151–154. 10.1007/bf00717039 [DOI] [PubMed] [Google Scholar]
  144. Ono H, Ohta R, Kawasaki Y, Niwa A, Takada H, Nakahata T, … Saito MK (2018). Lysosomal membrane permeabilization causes secretion of IL-1β in human vascular smooth muscle cells. Inflammation Research, 67(10), 879–889. 10.1007/s00011-018-1178-z [DOI] [PMC free article] [PubMed] [Google Scholar]
  145. Orsini JJ, Escolar ML, Wasserstein MP, & Caggana M (1993). Krabbe disease. In Adam MP, Ardinger HH, Pagon RA, Wallace SE, Bean LJH, Stephens K, & Amemiya A(Eds.), GeneReviews (®). Seattle, WA: University of Washington. [PubMed] [Google Scholar]
  146. Padamsey Z, McGuinness L, Bardo SJ, Reinhart M, Tong R, Hedegaard A, … Emptage NJ (2017). Activity-dependent exocytosis of lysosomes regulates the structural plasticity of dendritic spines. Neuron, 93(1), 132–146. 10.1016/j.neuron.2016.11.013 [DOI] [PMC free article] [PubMed] [Google Scholar]
  147. Pan X, Sands SA, Yue Y, Zhang K, LeVine SM, & Duan D (2019). An engineered galactosylceramidase construct improves AAV gene therapy for Krabbe disease in Twitcher mice. Human Gene Therapy, 30 (9), 1039–1051. 10.1089/hum.2019.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
  148. Pannu R, Singh AK, & Singh I (2005). A novel role of lactosylceramide in the regulation of tumor necrosis factor alpha-mediated proliferation of rat primary astrocytes. Implications for astrogliosis following neurotrauma. The Journal of Biological Chemistry, 280(14), 13742–13751. 10.1074/jbc.M411959200 [DOI] [PubMed] [Google Scholar]
  149. Pannuzzo G, Cardile V, Costantino-Ceccarini E, Alvares E, Mazzone D, & Perciavalle V (2010). A galactose-free diet enriched in soy isoflavones and antioxidants results in delayed onset of symptoms of Krabbe disease in twitcher mice. Molecular Genetics and Metabolism, 100(3), 234–240. 10.1016/j.ymgme.2010.03.021 [DOI] [PubMed] [Google Scholar]
  150. Papadopoulos C, Kravic B, & Meyer H (2020). Repair or lysophagy: Dealing with damaged lysosomes. Journal of Molecular Biology, 432(1), 231–239. 10.1016/j.jmb.2019.08.010 [DOI] [PubMed] [Google Scholar]
  151. Pasqualim G, Baldo G, de Carvalho TG, Tavares AM, Giugliani R, & Matte U (2015). Effects of enzyme replacement therapy started late in a murine model of mucopolysaccharidosis type I. PLoS One, 10(2), e0117271. 10.1371/journal.pone.0117271 [DOI] [PMC free article] [PubMed] [Google Scholar]
  152. Pedchenko TV, Bronshteyn IG, & LeVine SM (2000). TNF-receptor 1 deficiency fails to alter the clinical and pathological course in mice with globoid cell leukodystrophy (twitcher mice) but affords protection following LPS challenge. Journal of Neuroimmunology, 110(1–2), 186–194. 10.1016/s0165-5728(00)00345-3 [DOI] [PubMed] [Google Scholar]
  153. Pfisterer U, & Khodosevich K (2017). Neuronal survival in the brain: Neuron type-specific mechanisms. Cell Death & Disease, 8(3), e2643. 10.1038/cddis.2017.64 [DOI] [PMC free article] [PubMed] [Google Scholar]
  154. Platt FM, Boland B, & van der Spoel AC (2012). The cell biology of disease: Lysosomal storage disorders: The cellular impact of lysosomal dysfunction. The Journal of Cell Biology, 199(5), 723–734. 10.1083/jcb.201208152 [DOI] [PMC free article] [PubMed] [Google Scholar]
  155. Potter GB, Santos M, Davisson MT, Rowitch DH, Marks DL, Bongarzone ER, & Petryniak MA (2013). Missense mutation in mouse GALC mimics human gene defect and offers new insights into Krabbe disease. Human Molecular Genetics, 22(17), 3397–3414. 10.1093/hmg/ddt190 [DOI] [PMC free article] [PubMed] [Google Scholar]
  156. Pritchard DH, Napthine DV, & Sinclair AJ (1980). Globoid cell leucodystrophy in polled Dorset sheep. Veterinary Pathology, 17(4), 399–405. 10.1177/030098588001700402 [DOI] [PubMed] [Google Scholar]
  157. Pu J, Guardia CM, Keren-Kaplan T, & Bonifacino JS (2016). Mechanisms and functions of lysosome positioning. Journal of Cell Science, 129(23), 4329–4339. 10.1242/jcs.196287 [DOI] [PMC free article] [PubMed] [Google Scholar]
  158. Qin EY, Hawkins-Salsbury JA, Jiang X, Reddy AS, Farber NB, Ory DS, & Sands MS (2012). Bone marrow transplantation increases efficacy of central nervous system-directed enzyme replacement therapy in the murine model of globoid cell leukodystrophy. Molecular Genetics and Metabolism, 107(1–2), 186–196. 10.1016/j.ymgme.2012.05.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  159. Rafi M, Luzi P, & Wenger D (2021). Can early treatment of twitcher mice with high dose AAVrh10-GALC eliminate the need for BMT? [DOI] [PMC free article] [PubMed]
  160. Rafi MA, Fugaro J, Amini S, Luzi P, de Gala G, Victoria T, … Wenger DA (1996). Retroviral vector-mediated transfer of the galactocerebrosidase (GALC) cDNA leads to overexpression and transfer of GALC activity to neighboring cells. Biochemical and Molecular Medicine, 58(2), 142–150. 10.1006/bmme.1996.0042 [DOI] [PubMed] [Google Scholar]
  161. Rafi MA, Luzi P, Chen YQ, & Wenger DA (1995). A large deletion together with a point mutation in the GALC gene is a common mutant allele in patients with infantile Krabbe disease. Human Molecular Genetics, 4(8), 1285–1289. 10.1093/hmg/4.8.1285 [DOI] [PubMed] [Google Scholar]
  162. Rafi MA, Luzi P, & Wenger DA (2020). Conditions for combining gene therapy with bone marrow transplantation in murine Krabbe disease. Bioimpacts, 10(2), 105–115. 10.34172/bi.2020.13 [DOI] [PMC free article] [PubMed] [Google Scholar]
  163. Rafi MA, Luzi P, Zlotogora J, & Wenger DA (1996). Two different mutations are responsible for Krabbe disease in the Druze and Moslem Arab populations in Israel. Human Genetics, 97(3), 304–308. 10.1007/bf02185759 [DOI] [PubMed] [Google Scholar]
  164. Rafi MA, Rao HZ, Luzi P, Curtis MT, & Wenger DA (2012). Extended normal life after AAVrh10-mediated gene therapy in the mouse model of Krabbe disease. Molecular Therapy, 20(11), 2031–2042. 10.1038/mt.2012.153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  165. Rafi MA, Rao HZ, Luzi P, Luddi A, Curtis MT, & Wenger DA (2015). Intravenous injection of AAVrh10-GALC after the neonatal period in twitcher mice results in significant expression in the central and peripheral nervous systems and improvement of clinical features. Molecular Genetics and Metabolism, 114(3), 459–466. 10.1016/j.ymgme.2014.12.300 [DOI] [PubMed] [Google Scholar]
  166. Rajamäki K, Lappalainen J, Oörni K, Välimäki E, Matikainen S, Kovanen PT, & Eklund KK (2010). Cholesterol crystals activate the NLRP3 inflammasome in human macrophages: A novel link between cholesterol metabolism and inflammation. PLoS One, 5(7), e11765. 10.1371/journal.pone.0011765 [DOI] [PMC free article] [PubMed] [Google Scholar]
  167. Ravikumar B, Duden R, & Rubinsztein DC (2002). Aggregate-prone proteins with polyglutamine and polyalanine expansions are degraded by autophagy. Human Molecular Genetics, 11(9), 1107–1117. 10.1093/hmg/11.9.1107 [DOI] [PubMed] [Google Scholar]
  168. Ravikumar B, Sarkar S, Davies JE, Futter M, Garcia-Arencibia M, Green-Thompson ZW, … Rubinsztein DC (2010). Regulation of mammalian autophagy in physiology and pathophysiology. Physiological Reviews, 90(4), 1383–1435. 10.1152/physrev.00030.2009 [DOI] [PubMed] [Google Scholar]
  169. Reddy AS, Kim JH, Hawkins-Salsbury JA, Macauley SL, Tracy ET, Vogler CA, … Sands MS (2011). Bone marrow transplantation augments the effect of brain- and spinal cord-directed adeno-associated virus 2/5 gene therapy by altering inflammation in the murine model of globoid-cell leukodystrophy. The Journal of Neuroscience, 31(27), 9945–9957. 10.1523/jneurosci.1802-11.2011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  170. Ribbens JJ, Moser AB, Hubbard WC, Bongarzone ER, & Maegawa GH (2014). Characterization and application of a disease-cell model for a neurodegenerative lysosomal disease. Molecular Genetics and Metabolism, 111(2), 172–183. 10.1016/j.ymgme.2013.09.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  171. Ricca A, Cascino F, Morena F, Martino S, & Gritti A (2020). In vitro validation of chimeric β-galactosylceramidase enzymes with improved enzymatic activity and increased secretion. Frontiers in Molecular Biosciences, 7, 167. 10.3389/fmolb.2020.00167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  172. Ricca A, & Gritti A (2016). Perspective on innovative therapies for globoid cell leukodystrophy. Journal of Neuroscience Research, 94(11), 1304–1317. 10.1002/jnr.23752 [DOI] [PubMed] [Google Scholar]
  173. Ricca A, Rufo N, Ungari S, Morena F, Martino S, Kulik W, … Gritti A (2015). Combined gene/cell therapies provide long-term and pervasive rescue of multiple pathological symptoms in a murine model of globoid cell leukodystrophy. Human Molecular Genetics, 24(12), 3372–3389. 10.1093/hmg/ddv086 [DOI] [PMC free article] [PubMed] [Google Scholar]
  174. Ripoll CB, Flaat M, Klopf-Eiermann J, Fisher-Perkins JM, Trygg CB, Scruggs BA, … Bunnell BA (2011). Mesenchymal line-age stem cells have pronounced anti-inflammatory effects in the twitcher mouse model of Krabbe’s disease. Stem Cells, 29(1), 67–77. 10.1002/stem.555 [DOI] [PMC free article] [PubMed] [Google Scholar]
  175. Robak LA, Jansen IE, van Rooij J, Uitterlinden AG, Kraaij R, Jankovic J, … Shulman JM (2017). Excessive burden of lysosomal storage disorder gene variants in Parkinson’s disease. Brain, 140(12), 3191–3203. 10.1093/brain/awx285 [DOI] [PMC free article] [PubMed] [Google Scholar]
  176. Rusten TE, & Stenmark H (2010). p62, an autophagy hero or culprit? Nature Cell Biology, 12(3), 207–209. 10.1038/ncb0310-207 [DOI] [PubMed] [Google Scholar]
  177. Saadat L, Dupree JL, Kilkus J, Han X, Traka M, Proia RL, … Popko B (2010). Absence of oligodendroglial glucosylceramide synthesis does not result in CNS myelin abnormalities or alter the dysmyelinating phenotype of CGT-deficient mice. Glia, 58(4), 391–398. 10.1002/glia.20930 [DOI] [PMC free article] [PubMed] [Google Scholar]
  178. Sahu R, Kaushik S, Clement CC, Cannizzo ES, Scharf B, Follenzi A, … Santambrogio L (2011). Microautophagy of cytosolic proteins by late endosomes. Developmental Cell, 20(1), 131–139. 10.1016/j.devcel.2010.12.003 [DOI] [PMC free article] [PubMed] [Google Scholar]
  179. Sands MS, & Davidson BL (2006). Gene therapy for lysosomal storage diseases. Molecular Therapy, 13(5), 839–849. 10.1016/j.ymthe.2006.01.006 [DOI] [PubMed] [Google Scholar]
  180. Santambrogio S, Ricca A, Maderna C, Ieraci A, Aureli M, Sonnino S, … Gritti A (2012). The galactocerebrosidase enzyme contributes to maintain a functional neurogenic niche during early post-natal CNS development. Human Molecular Genetics, 21(21), 4732–4750. 10.1093/hmg/dds313 [DOI] [PubMed] [Google Scholar]
  181. Sardiello M, Palmieri M, di Ronza A, Medina DL, Valenza M, Gennarino VA, … Ballabio A (2009). A gene network regulating lysosomal biogenesis and function. Science, 325(5939), 473–477. 10.1126/science.1174447 [DOI] [PubMed] [Google Scholar]
  182. Sawcer S, Hellenthal G, Pirinen M, Spencer CC, Patsopoulos NA, Moutsianas L, … Compston A (2011). Genetic risk and a primary role for cell-mediated immune mechanisms in multiple sclerosis. Nature, 476(7359), 214–219. 10.1038/nature10251 [DOI] [PMC free article] [PubMed] [Google Scholar]
  183. Schiffmann R, Heyes MP, Aerts JM, Dambrosia JM, Patterson MC, DeGraba T, … Barton NW (1997). Prospective study of neurological responses to treatment with macrophage-targeted glucocerebrosidase in patients with type 3 Gaucher’s disease. Annals of Neurology, 42(4), 613–621. 10.1002/ana.410420412 [DOI] [PubMed] [Google Scholar]
  184. Schuette CG, Pierstorff B, Huettler S, & Sandhoff K (2001). Sphingolipid activator proteins: Proteins with complex functions in lipid degradation and skin biogenesis. Glycobiology, 11(6), 81r–90r. 10.1093/glycob/11.6.81r [DOI] [PubMed] [Google Scholar]
  185. Scott-Hewitt NJ, Folts CJ, Hogestyn JM, Piester G, Mayer-Pröschel M, & Noble MD (2017). Heterozygote galactocerebrosidase (GALC) mutants have reduced remyelination and impaired myelin debris clearance following demyelinating injury. Human Molecular Genetics, 26(15), 2825–2837. 10.1093/hmg/ddx153 [DOI] [PMC free article] [PubMed] [Google Scholar]
  186. Scott-Hewitt NJ, Folts CJ, & Noble MD (2018). Heterozygous carriers of galactocerebrosidase mutations that cause Krabbe disease have impaired microglial function and defective repair of myelin damage. Neural Regeneration Research, 13(3), 393–401. 10.4103/1673-5374.228712 [DOI] [PMC free article] [PubMed] [Google Scholar]
  187. Sessa M, Lorioli L, Fumagalli F, Acquati S, Redaelli D, Baldoli C, … Biffi A (2016). Lentiviral haemopoietic stem-cell gene therapy in early-onset metachromatic leukodystrophy: An ad-hoc analysis of a non-randomised, open-label, phase 1/2 trial. Lancet, 388(10043), 476–487. 10.1016/s0140-6736(16)30374-9 [DOI] [PubMed] [Google Scholar]
  188. Settembre C, Di Malta C, Polito VA, Garcia Arencibia M, Vetrini F, Erdin S, … Ballabio A (2011). TFEB links autophagy to lysosomal biogenesis. Science, 332(6036), 1429–1433. 10.1126/science.1204592 [DOI] [PMC free article] [PubMed] [Google Scholar]
  189. Settembre C, Zoncu R, Medina DL, Vetrini F, Erdin S, Erdin S, … Ballabio A (2012). A lysosome-to-nucleus signalling mechanism senses and regulates the lysosome via mTOR and TFEB. The EMBO Journal, 31(5), 1095–1108. 10.1038/emboj.2012.32 [DOI] [PMC free article] [PubMed] [Google Scholar]
  190. Shen JS, Watabe K, Ohashi T, & Eto Y (2001). Intraventricular administration of recombinant adenovirus to neonatal twitcher mouse leads to clinicopathological improvements. Gene Therapy, 8(14), 1081–1087. 10.1038/sj.gt.3301495 [DOI] [PubMed] [Google Scholar]
  191. Shin D, Feltri ML, & Wrabetz L (2016). Altered trafficking and processing of GALC mutants correlates with globoid cell Leukodystrophy severity. The Journal of Neuroscience, 36(6), 1858–1870. 10.1523/jneurosci.3095-15.2016 [DOI] [PMC free article] [PubMed] [Google Scholar]
  192. Sigurdson CJ, Basaraba RJ, Mazzaferro EM, & Gould DH (2002). Globoid cell-like leukodystrophy in a domestic longhaired cat. Veterinary Pathology, 39(4), 494–496. 10.1354/vp.39-4-494 [DOI] [PubMed] [Google Scholar]
  193. Smith BR, Santos MB, Marshall MS, Cantuti-Castelvetri L, Lopez-Rosas A, Li G, … Bongarzone ER (2014). Neuronal inclusions of α-synuclein contribute to the pathogenesis of Krabbe disease. The Journal of Pathology, 232(5), 509–521. 10.1002/path.4328 [DOI] [PMC free article] [PubMed] [Google Scholar]
  194. Smith ME (1968). The turnover of myelin in the adult rat. Biochimica et Biophysica Acta, 164(2), 285–293. 10.1016/0005-2760(68)90154-9 [DOI] [PubMed] [Google Scholar]
  195. Snook ER, Fisher-Perkins JM, Sansing HA, Lee KM, Alvarez X, MacLean AG, … Bunnell BA (2014). Innate immune activation in the pathogenesis of a murine model of globoid cell leukodystrophy. The American Journal of Pathology, 184(2), 382–396. 10.1016/j.ajpath.2013.10.011 [DOI] [PMC free article] [PubMed] [Google Scholar]
  196. Sommer I, & Schachner M (1981). Monoclonal antibodies (O1 to O4) to oligodendrocyte cell surfaces: An immunocytological study in the central nervous system. Developmental Biology, 83(2), 311–327. [DOI] [PubMed] [Google Scholar]
  197. Spassieva S, & Bieberich E (2016). Lysosphingolipids and sphingolipidoses: Psychosine in Krabbe’s disease. Journal of Neuroscience Research, 94(11), 974–981. 10.1002/jnr.23888 [DOI] [PMC free article] [PubMed] [Google Scholar]
  198. Spiegel R, Bach G, Sury V, Mengistu G, Meidan B, Shalev S, … Zeigler M (2005). A mutation in the saposin A coding region of the prosaposin gene in an infant presenting as Krabbe disease: First report of saposin A deficiency in humans. Molecular Genetics and Metabolism, 84(2), 160–166. 10.1016/j.ymgme.2004.10.004 [DOI] [PubMed] [Google Scholar]
  199. Sprong H, Kruithof B, Leijendekker R, Slot JW, van Meer G, & van der Sluijs P (1998). UDP-galactose:Ceramide galactosyltransferase is a class I integral membrane protein of the endoplasmic reticulum. The Journal of Biological Chemistry, 273(40), 25880–25888. 10.1074/jbc.273.40.25880 [DOI] [PubMed] [Google Scholar]
  200. Strazza M, Luddi A, Carbone M, Rafi MA, Costantino-Ceccarini E, & Wenger DA (2009). Significant correction of pathology in brains of twitcher mice following injection of genetically modified mouse neural progenitor cells. Molecular Genetics and Metabolism, 97(1), 27–34. 10.1016/j.ymgme.2009.01.005 [DOI] [PubMed] [Google Scholar]
  201. Suzuki K (1998). Twenty five years of the “psychosine hypothesis”: A personal perspective of its history and present status. Neurochemical Research, 23(3), 251–259. 10.1023/a:1022436928925 [DOI] [PubMed] [Google Scholar]
  202. Suzuki K (2003). Globoid cell leukodystrophy (Krabbe’s disease): Update. Journal of Child Neurology, 18(9), 595–603. 10.1177/08830738030180090201 [DOI] [PubMed] [Google Scholar]
  203. Suzuki K (2016). My encounters with Krabbe disease: A personal recollection of a 40-year journey with young colleagues. Journal of Neuroscience Research, 94(11), 965–972. 10.1002/jnr.23735 [DOI] [PubMed] [Google Scholar]
  204. Suzuki K, & Suzuki K (1990). Myelin pathology in the twitcher mouse. Annals of the new York Academy of Sciences, 605, 313–324. 10.1111/j.1749-6632.1990.tb42405.x [DOI] [PubMed] [Google Scholar]
  205. Takahashi H, Igisu H, Suzuki K, & Suzuki K (1983). The twitcher mouse: An ultrastructural study on the oligodendroglia. Acta Neuropathologica, 59(3), 159–166. 10.1007/bf00703199 [DOI] [PubMed] [Google Scholar]
  206. Tanaka K, & Webster HD (1993). Effects of psychosine (galactosylsphingosine) on the survival and the fine structure of cultured Schwann cells. Journal of Neuropathology and Experimental Neurology, 52(5), 490–498. 10.1097/00005072-199309000-00007 [DOI] [PubMed] [Google Scholar]
  207. Taniike M, & Suzuki K (1994). Spacio-temporal progression of demyelination in twitcher mouse: With clinico-pathological correlation. Acta Neuropathologica, 88(3), 228–236. 10.1007/bf00293398 [DOI] [PubMed] [Google Scholar]
  208. Tapper H, & Sundler R (1995). Bafilomycin A1 inhibits lysosomal, phagosomal, and plasma membrane H(+)-ATPase and induces lysosomal enzyme secretion in macrophages. Journal of Cellular Physiology, 163(1), 137–144. 10.1002/jcp.1041630116 [DOI] [PubMed] [Google Scholar]
  209. Tasaki I (1939). The electro-saltatory transmission of the nerve impulse and the effect of narcosis upon the nerve fiber. American Journal of Physiology-Legacy Content, 127(2), 211–227. [Google Scholar]
  210. Tatsumi N, Inui K, Sakai N, Fukushima H, Nishimoto J, Yanagihara I, … Okada S (1995). Molecular defects in Krabbe disease. Human Molecular Genetics, 4(10), 1865–1868. 10.1093/hmg/4.10.1865 [DOI] [PubMed] [Google Scholar]
  211. Taylor RM, & Snyder EY (1997). Widespread engraftment of neural progenitor and stem-like cells throughout the mouse brain. Transplantation Proceedings, 29(1–2), 845–847. 10.1016/s0041-1345(96)00163-7 [DOI] [PubMed] [Google Scholar]
  212. Teil M, Arotcarena ML, Faggiani E, Laferriere F, Bezard E, & Dehay B (2020). Targeting α-synuclein for PD therapeutics: A pursuit on all fronts. Biomolecules, 10(3), 391. 10.3390/biom10030391 [DOI] [PMC free article] [PubMed] [Google Scholar]
  213. Teixeira CA, Miranda CO, Sousa VF, Santos TE, Malheiro AR, Solomon M, … Sousa MM (2014). Early axonal loss accompanied by impaired endocytosis, abnormal axonal transport, and decreased microtubule stability occur in the model of Krabbe’s disease. Neurobiology of Disease, 66, 92–103. 10.1016/j.nbd.2014.02.012 [DOI] [PMC free article] [PubMed] [Google Scholar]
  214. Thomas R, & Kermode AR (2019). Enzyme enhancement therapeutics for lysosomal storage diseases: Current status and perspective. Molecular Genetics and Metabolism, 126(2), 83–97. 10.1016/j.ymgme.2018.11.011 [DOI] [PubMed] [Google Scholar]
  215. Thudichum JLW (1874). Researches on the chemical constitution of the brain. [Google Scholar]
  216. Tohyama J, Matsuda J, & Suzuki K (2001). Psychosine is as potent an inducer of cell death as C6-ceramide in cultured fibroblasts and in MOCH-1 cells. Neurochemical Research, 26(6), 667–671. 10.1023/a:1010991420942 [DOI] [PubMed] [Google Scholar]
  217. Vanier MT, & Svennerholm L (1974). Chemical pathology of Krabbe’s disease. I. Lipid composition and fatty acid patterns of phosphoglycerides in brain. Acta Paediatrica Scandinavica, 63(4), 494–500. 10.1111/j.1651-2227.1974.tb04838.x [DOI] [PubMed] [Google Scholar]
  218. Venugopal B, Mesires NT, Kennedy JC, Curcio-Morelli C, Laplante JM, Dice JF, & Slaugenhaupt SA (2009). Chaperone-mediated autophagy is defective in mucolipidosis type IV. Journal of Cellular Physiology, 219(2), 344–353. 10.1002/jcp.21676 [DOI] [PubMed] [Google Scholar]
  219. Victoria T, Rafi MA, & Wenger DA (1996). Cloning of the canine GALC cDNA and identification of the mutation causing globoid cell leukodystrophy in West Highland White and cairn terriers. Genomics, 33(3), 457–462. 10.1006/geno.1996.0220 [DOI] [PubMed] [Google Scholar]
  220. Visigalli I, Ungari S, Martino S, Park H, Cesani M, Gentner B, … Biffi A (2010). The galactocerebrosidase enzyme contributes to the maintenance of a functional hematopoietic stem cell niche. Blood, 116 (11), 1857–1866. 10.1182/blood-2009-12-256461 [DOI] [PMC free article] [PubMed] [Google Scholar]
  221. Voccoli V, Tonazzini I, Signore G, Caleo M, & Cecchini M (2014). Role of extracellular calcium and mitochondrial oxygen species in psychosine-induced oligodendrocyte cell death. Cell Death & Disease, 5 (11), e1529. 10.1038/cddis.2014.483 [DOI] [PMC free article] [PubMed] [Google Scholar]
  222. Vukoja A, Rey U, Petzoldt AG, Ott C, Vollweiter D, Quentin C, … Haucke V (2018). Presynaptic biogenesis requires axonal transport of lysosome-related vesicles. Neuron, 99(6), 1216–1232. 10.1016/j.neuron.2018.08.004 [DOI] [PubMed] [Google Scholar]
  223. Walkley SU, & Vanier MT (2009). Secondary lipid accumulation in lysosomal disease. Biochimica et Biophysica Acta, 1793(4), 726–736. 10.1016/j.bbamcr.2008.11.014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  224. Wang F, Gómez-Sintes R, & Boya P (2018). Lysosomal membrane permeabilization and cell death. Traffic, 19(12), 918–931. 10.1111/tra.12613 [DOI] [PubMed] [Google Scholar]
  225. Weinstock NI, Kreher C, Favret J, Nguyen D, Bongarzone ER, Wrabetz L, … Shin D (2020). Brainstem development requires galactosylceramidase and is critical for pathogenesis in a model of Krabbe disease. Nature Communications, 11(1), 5356. 10.1038/s41467-020-19179-w [DOI] [PMC free article] [PubMed] [Google Scholar]
  226. Weinstock NI, Shin D, Dhimal N, Hong X, Irons EE, Silvestri NJ, … Feltri ML (2020). Macrophages expressing GALC improve peripheral Krabbe disease by a mechanism independent of cross-correction. Neuron, 107(1), 65–81. 10.1016/j.neuron.2020.03.031 [DOI] [PMC free article] [PubMed] [Google Scholar]
  227. Wenger DA, Rafi MA, & Luzi P (1997). Molecular genetics of Krabbe disease (globoid cell leukodystrophy): Diagnostic and clinical implications. Human Mutation, 10(4), 268–279. [DOI] [PubMed] [Google Scholar]
  228. Wenger DA, Rafi MA, Luzi P, Datto J, & Costantino-Ceccarini E (2000). Krabbe disease: Genetic aspects and progress toward therapy. Molecular Genetics and Metabolism, 70(1), 1–9. 10.1006/mgme.2000.2990 [DOI] [PubMed] [Google Scholar]
  229. Wenger DA, Sattler M, & Hiatt W (1974). Globoid cell leukodystrophy: Deficiency of lactosyl ceramide beta-galactosidase. Proceedings of the National Academy of Sciences of the United States of America, 71(3), 854–857. 10.1073/pnas.71.3.854 [DOI] [PMC free article] [PubMed] [Google Scholar]
  230. White AB, Galbiati F, Givogri MI, Lopez Rosas A, Qiu X, van Breemen R, & Bongarzone ER (2011). Persistence of psychosine in brain lipid rafts is a limiting factor in the therapeutic recovery of a mouse model for Krabbe disease. Journal of Neuroscience Research, 89(3), 352–364. 10.1002/jnr.22564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  231. White AB, Givogri MI, Lopez-Rosas A, Cao H, van Breemen R, Thinakaran G, & Bongarzone ER (2009). Psychosine accumulates in membrane microdomains in the brain of krabbe patients, disrupting the raft architecture. The Journal of Neuroscience, 29(19), 6068–6077. 10.1523/jneurosci.5597-08.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
  232. Wolf NI, Breur M, Plug B, Beerepoot S, Westerveld ASR, van Rappard DF, … Bugiani M (2020). Metachromatic leukodystrophy and transplantation: Remyelination, no cross-correction. Annals of Clinical Translational Neurology, 7(2), 169–180. 10.1002/acn3.50975 [DOI] [PMC free article] [PubMed] [Google Scholar]
  233. Wolf NI, Ffrench-Constant C, & van der Knaap MS (2021). Hypomyelinating leukodystrophies—Unravelling myelin biology. Nature Reviews. Neurology, 17(2), 88–103. 10.1038/s41582-020-00432-1 [DOI] [PubMed] [Google Scholar]
  234. Won JS, Singh AK, & Singh I (2016). Biochemical, cell biological, pathological, and therapeutic aspects of Krabbe’s disease. Journal of Neuroscience Research, 94(11), 990–1006. 10.1002/jnr.23873 [DOI] [PMC free article] [PubMed] [Google Scholar]
  235. Wright MD, Poe MD, DeRenzo A, Haldal S, & Escolar ML (2017). Developmental outcomes of cord blood transplantation for Krabbe disease: A 15-year study. Neurology, 89(13), 1365–1372. 10.1212/wnl.0000000000004418 [DOI] [PMC free article] [PubMed] [Google Scholar]
  236. Wu YP, McMahon EJ, Matsuda J, Suzuki K, Matsushima GK, & Suzuki K (2001). Expression of immune-related molecules is down-regulated in twitcher mice following bone marrow transplantation. Journal of Neuropathology and Experimental Neurology, 60(11), 1062–1074. 10.1093/jnen/60.11.1062 [DOI] [PubMed] [Google Scholar]
  237. Xu C, Sakai N, Taniike M, Inui K, & Ozono K (2006). Six novel mutations detected in the GALC gene in 17 Japanese patients with Krabbe disease, and new genotype-phenotype correlation. Journal of Human Genetics, 51(6), 548–554. 10.1007/s10038-006-0396-3 [DOI] [PubMed] [Google Scholar]
  238. Xu H, & Ren D (2015). Lysosomal physiology. Annual Review of Physiology, 77, 57–80. 10.1146/annurev-physiol-021014-071649 [DOI] [PMC free article] [PubMed] [Google Scholar]
  239. Yajima K, Fletcher TF, & Suzuki K (1977). Canine globoid cell leukodystrophy. Part 1. Further ultrastructural study of the typical lesion. Journal of the Neurological Sciences, 33(1–2), 179–197. 10.1016/0022-510x(77)90192-7 [DOI] [PubMed] [Google Scholar]
  240. Yeager AM, Brennan S, Tiffany C, Moser HW, & Santos GW (1984). Prolonged survival and remyelination after hematopoietic cell transplantation in the twitcher mouse. Science, 225(4666), 1052–1054. 10.1126/science.6382609 [DOI] [PubMed] [Google Scholar]
  241. Yeung MS, Zdunek S, Bergmann O, Bernard S, Salehpour M, Alkass K, … Frisén J (2014). Dynamics of oligodendrocyte generation and myelination in the human brain. Cell, 159(4), 766–774. 10.1016/j.cell.2014.10.011 [DOI] [PubMed] [Google Scholar]
  242. Young KM, Psachoulia K, Tripathi RB, Dunn SJ, Cossell L, Attwell D, … Richardson WD (2013). Oligodendrocyte dynamics in the healthy adult CNS: Evidence for myelin remodeling. Neuron, 77(5), 873–885. 10.1016/j.neuron.2013.01.006 [DOI] [PMC free article] [PubMed] [Google Scholar]
  243. Yunis EJ, & Lee RE (1970). Tubules of globoid leukodystrophy: A right-handed helix. Science, 169(3940), 64–66. 10.1126/science.169.3940.64 [DOI] [PubMed] [Google Scholar]
  244. Zaka M, & Wenger DA (2004). Psychosine-induced apoptosis in a mouse oligodendrocyte progenitor cell line is mediated by caspase activation. Neuroscience Letters, 358(3), 205–209. 10.1016/j.neulet.2003.12.126 [DOI] [PubMed] [Google Scholar]
  245. Zhang Y, Chen K, Sloan SA, Bennett ML, Scholze AR, O’Keeffe S, … Wu JQ (2014). An RNA-sequencing transcriptome and splicing database of glia, neurons, and vascular cells of the cerebral cortex. The Journal of Neuroscience, 34(36), 11929–11947. 10.1523/jneurosci.1860-14.2014 [DOI] [PMC free article] [PubMed] [Google Scholar]
  246. Zhu H, Lopez-Rosas A, Qiu X, Van Breemen RB, & Bongarzone ER (2012). Detection of the neurotoxin psychosine in samples of peripheral blood: Application in diagnostics and follow-up of Krabbe disease. Archives of Pathology & Laboratory Medicine, 136(7), 709–710. 10.5858/arpa.2011-0667-LE [DOI] [PMC free article] [PubMed] [Google Scholar]
  247. Zizioli D, Guarienti M, Tobia C, Gariano G, Borsani G, Bresciani R, … Presta M (2014). Molecular cloning and knockdown of galactocerebrosidase in zebrafish: New insights into the pathogenesis of Krabbe’s disease. Biochimica et Biophysica Acta, 1842(4), 665–675. 10.1016/j.bbadis.2014.01.008 [DOI] [PubMed] [Google Scholar]

Associated Data

This section collects any data citations, data availability statements, or supplementary materials included in this article.

Data Availability Statement

Data sharing is not applicable to this article as no new data were created or analyzed in this study.

RESOURCES