GangliosidesandGangliosidoses:PrinciplesofMolecular … · 2013. 7. 16. · 2000). Gangliosides...

14
Disease Focus Editor’s Note: Disease Focus articles provide brief overviews of a neural disease or syndrome, emphasizing potential links to basic neural mechanisms. They are presented in the hope of helping researchers identify clinical implications of their research. For more information, see http://www.jneurosci.org/misc/ifa_minireviews.dtl. Gangliosides and Gangliosidoses: Principles of Molecular and Metabolic Pathogenesis Konrad Sandhoff 1 and Klaus Harzer 2 1 LIMES, Kekule ´-Institut, 53121 Bonn, Germany and 2 Neurometabolic Laboratory, Klinik fu ¨r Kinder- und Jugendmedizin, University of Tu ¨bingen, 72076 Tu ¨bingen, Germany Gangliosides are the main glycolipids of neuronal plasma membranes. Their surface patterns are generated by coordinated processes, involving biosynthetic pathways of the secretory compartments, catabolic steps of the endolysosomal system, and intracellular traffick- ing. Inherited defects in ganglioside biosynthesis causing fatal neurodegenerative diseases have been described so far almost exclusively in mouse models, whereas inherited defects in ganglioside catabolism causing various clinical forms of GM1- and GM2-gangliosidoses have long been known. For digestion, gangliosides are endocytosed and reach intra-endosomal vesicles. At the level of late endosomes, they are depleted of membrane-stabilizing lipids like cholesterol and enriched with bis(monoacylglycero)phosphate (BMP). Lysosomal catabolism is catalyzed at acidic pH values by cationic sphingolipid activator proteins (SAPs), presenting lipids to their respective hydrolases, electrostatically attracted to the negatively charged surface of the luminal BMP-rich vesicles. Various inherited defects of ganglioside hydrolases, e.g., of -galactosidase and -hexosaminidases, and of GM2-activator protein, cause infantile (with tetraparesis, dementia, blindness) and different protracted clinical forms of GM1- and GM2-gangliosidoses. Mutations yielding proteins with small residual catabolic activities in the lysosome give rise to juvenile and adult clinical forms with a wide range of clinical symptomatology. Apart from patients’ differences in their genetic background, clinical heterogeneity may be caused by rather diverse substrate specificities and functions of lysosomal hydrolases, multifunctional properties of SAPs, and the strong regulation of ganglioside catabolism by membrane lipids. Currently, there is no treatment available for neuronal ganglioside storage diseases. Therapeutic approaches in mouse models and patients with juvenile forms of gangliosidoses are discussed. Introduction 1 Gangliosides had been discovered by Ernst Klenk in the 1930s (Klenk, 1937, 1939) when he analyzed postmortem brain tissues of patients with Tay–Sachs disease, a fatal infantile form of amaurotic idiocy. At that time the clinical diagnosis “amaurotic idiocy” comprised an inher- ited disease characterized by high num- bers of lipid-laden cells in the nervous system and the visceral organs, mental re- tardation, and impaired vision or blind- ness. With the help of biochemical, enzymatic, molecular genetic, morpho- logical, and animal studies, the clinical picture of amaurotic idiocy has been re- solved in the last five decades into two ma- jor groups of diseases at the molecular level: lysosomal ganglioside storage dis- eases, comprising GM1-gangliosidoses and 4 forms of GM2-gangliosidoses, and 10 forms of neuronal ceroid lipofuscino- ses poorly characterized so far (Jalanko and Braulke, 2009). Gangliosides are typical components of neuronal plasma membranes. The first gan- glioside structure, that of ganglioside GM1 (Fig. 1), was elucidated by Kuhn and Wie- gand (1963). Six major and many minor ganglioside species have been identified in mammalian brains (Yu et al., 2011). Inher- ited defects in ganglioside metabolism can cause fatal neurodegenerative diseases. Best known so far are inherited defects in lyso- somal ganglioside catabolism, the GM1- and GM2-gangliosidoses. Up to now, no defects in ganglioside biosynthesis have been described in hu- mans with the exception of two reports on three patients with an infantile onset re- fractory epilepsy (Simpson et al., 2004; Fragaki et al., 2013), caused by a defi- ciency of ganglioside GM3-synthase. It is associated with psychomotor delay, blindness, deafness, and respiratory chain dysfunction. In addition, one patient with a genetically undisclosed basis of infantile Received Feb. 19, 2013; revised April 30, 2013; accepted May 9, 2013. This work was supported by the Deutsche Forschungsgemeinschaft. Dedicated on the occasion of her 60th birthday to Ingeborg Kra ¨geloh- Mann, professor of Neuropediatrics, Tu ¨bingen. Correspondence should be addressed to either of the following: Konrad Sandhoff, LIMES, Kekule ´-Institut, Gerhard-Domagk-Str. 1, 53121 Bonn, Germany, E-mail: [email protected]; or Klaus Harzer, Neurometabolic Laboratory, Klinik fu ¨r Kinder- und Jugendmedizin, University of Tu ¨bingen, 72076 Tu ¨bingen, Germany, E-mail: [email protected]. DOI:10.1523/JNEUROSCI.0822-13.2013 Copyright © 2013 the authors 0270-6474/13/3310195-14$15.00/0 The Journal of Neuroscience, June 19, 2013 33(25):10195–10208 • 10195

Transcript of GangliosidesandGangliosidoses:PrinciplesofMolecular … · 2013. 7. 16. · 2000). Gangliosides...

  • Disease Focus

    Editor’s Note: Disease Focus articles provide brief overviews of a neural disease or syndrome, emphasizing potential links to basicneural mechanisms. They are presented in the hope of helping researchers identify clinical implications of their research. For moreinformation, see http://www.jneurosci.org/misc/ifa_minireviews.dtl.

    Gangliosides and Gangliosidoses: Principles of Molecularand Metabolic Pathogenesis

    Konrad Sandhoff1 and Klaus Harzer21LIMES, Kekulé-Institut, 53121 Bonn, Germany and 2Neurometabolic Laboratory, Klinik für Kinder- und Jugendmedizin, University of Tübingen, 72076Tübingen, Germany

    Gangliosides are the main glycolipids of neuronal plasma membranes. Their surface patterns are generated by coordinated processes,involving biosynthetic pathways of the secretory compartments, catabolic steps of the endolysosomal system, and intracellular traffick-ing. Inherited defects in ganglioside biosynthesis causing fatal neurodegenerative diseases have been described so far almost exclusivelyin mouse models, whereas inherited defects in ganglioside catabolism causing various clinical forms of GM1- and GM2-gangliosidoseshave long been known. For digestion, gangliosides are endocytosed and reach intra-endosomal vesicles. At the level of late endosomes,they are depleted of membrane-stabilizing lipids like cholesterol and enriched with bis(monoacylglycero)phosphate (BMP). Lysosomalcatabolism is catalyzed at acidic pH values by cationic sphingolipid activator proteins (SAPs), presenting lipids to their respectivehydrolases, electrostatically attracted to the negatively charged surface of the luminal BMP-rich vesicles. Various inherited defects ofganglioside hydrolases, e.g., of �-galactosidase and �-hexosaminidases, and of GM2-activator protein, cause infantile (with tetraparesis,dementia, blindness) and different protracted clinical forms of GM1- and GM2-gangliosidoses. Mutations yielding proteins with smallresidual catabolic activities in the lysosome give rise to juvenile and adult clinical forms with a wide range of clinical symptomatology.Apart from patients’ differences in their genetic background, clinical heterogeneity may be caused by rather diverse substrate specificitiesand functions of lysosomal hydrolases, multifunctional properties of SAPs, and the strong regulation of ganglioside catabolism bymembrane lipids. Currently, there is no treatment available for neuronal ganglioside storage diseases. Therapeutic approaches in mousemodels and patients with juvenile forms of gangliosidoses are discussed.

    Introduction1Gangliosides had been discovered byErnst Klenk in the 1930s (Klenk, 1937,1939) when he analyzed postmortembrain tissues of patients with Tay–Sachsdisease, a fatal infantile form of amauroticidiocy. At that time the clinical diagnosis“amaurotic idiocy” comprised an inher-ited disease characterized by high num-

    bers of lipid-laden cells in the nervoussystem and the visceral organs, mental re-tardation, and impaired vision or blind-ness. With the help of biochemical,enzymatic, molecular genetic, morpho-logical, and animal studies, the clinicalpicture of amaurotic idiocy has been re-solved in the last five decades into two ma-jor groups of diseases at the molecularlevel: lysosomal ganglioside storage dis-eases, comprising GM1-gangliosidosesand 4 forms of GM2-gangliosidoses, and10 forms of neuronal ceroid lipofuscino-ses poorly characterized so far (Jalankoand Braulke, 2009).

    Gangliosides are typical components ofneuronal plasma membranes. The first gan-glioside structure, that of ganglioside GM1(Fig. 1), was elucidated by Kuhn and Wie-

    gand (1963). Six major and many minorganglioside species have been identified inmammalian brains (Yu et al., 2011). Inher-ited defects in ganglioside metabolism cancause fatal neurodegenerative diseases. Bestknown so far are inherited defects in lyso-somal ganglioside catabolism, the GM1-and GM2-gangliosidoses.

    Up to now, no defects in gangliosidebiosynthesis have been described in hu-mans with the exception of two reports onthree patients with an infantile onset re-fractory epilepsy (Simpson et al., 2004;Fragaki et al., 2013), caused by a defi-ciency of ganglioside GM3-synthase. Itis associated with psychomotor delay,blindness, deafness, and respiratory chaindysfunction. In addition, one patient witha genetically undisclosed basis of infantile

    Received Feb. 19, 2013; revised April 30, 2013; accepted May 9, 2013.This work was supported by the Deutsche Forschungsgemeinschaft.

    Dedicated on the occasion of her 60th birthday to Ingeborg Krägeloh-Mann, professor of Neuropediatrics, Tübingen.

    Correspondence should be addressed to either of the following: KonradSandhoff, LIMES, Kekulé-Institut, Gerhard-Domagk-Str. 1, 53121 Bonn,Germany, E-mail: [email protected]; or Klaus Harzer, NeurometabolicLaboratory, Klinik für Kinder- und Jugendmedizin, University of Tübingen,72076 Tübingen, Germany, E-mail: [email protected].

    DOI:10.1523/JNEUROSCI.0822-13.2013Copyright © 2013 the authors 0270-6474/13/3310195-14$15.00/0

    The Journal of Neuroscience, June 19, 2013 • 33(25):10195–10208 • 10195

    http://www.jneurosci.org/misc/ifa_minireviews.dtl

  • Figure 1. Pathway of lysosomal sphingolipid degradation. The eponyms of known metabolic diseases and those of SAPs (red) necessary for in vivo degradation are indicated. Hydrolases are givenin green. Heterogeneity in the lipid part of the sphingolipids is not indicated. Variant AB, Variant AB of GM2-gangliosidosis �deficiency of GM2-activator protein (GM2-AP)�; Sap, sphingolipidactivator protein (modified from Sandhoff and Kolter, 1995).

    10196 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

  • GM2-synthase deficiency, a gangliosideGM3 storage and loss of higher ganglio-sides has been described, passed away atage 3.5 months (Fishman et al., 1975).

    However, blocks in ganglioside bio-synthesis generated in gene-manipulatedmice cause a broad spectrum of patholog-ical phenotypes (Proia, 2003).

    Ganglioside biosynthesis and its defectsin genetically manipulated miceGlycolipids are expressed in a cell-type-and stage-specific manner, gangliosidesbeing the main glycolipids of neuronalsurfaces. Their surface patterns are gener-ated by coordinated processes, involvingbiosynthetic and catabolic steps and intra-cellular trafficking.

    Their hydrophobic membrane an-chors, the ceramide residues, are formedin the endoplasmic reticulum (ER), glyco-sylation of ceramide takes place at theGolgi and trans-Golgi network (TGN),and their constitutive degradation mainlyin the endolysosomal compartment (Kolterand Sandhoff, 1999). After formation ofglucosylceramide (GlcCer) from cer-amide (Cer) at the cytosolic surface ofGolgi membranes and its flip to the lumi-nal surface of Golgi membranes, GlcCergets converted to lactosylceramide (Lac-Cer). At the luminal surfaces of Golgi andTGN membranes, LacCer is sialylated toform small precursor gangliosides (GM3,GD3, and GT3). They are then convertedby membrane-bound promiscuous glyco-syltransferases to complex gangliosidestructures, which reach cellular surfacesby vesicular transport of the secretorypathway. This concept of “combinatorialganglioside biosynthesis” (Kolter et al.,2002) allows the generation of cell-specific patterns on neuronal surfaces de-pending on the availability of precursormolecules, expression of biosynthetic en-zymes and their regulation. Generationand analysis of mutant mice with defectsin the pathways of ganglioside biosynthe-sis supported this model (Kolter et al.,2002; Sandhoff and Kolter, 2003). Mutantmice defective in b-series gangliosideswere viable and without apparent neuro-logical abnormalities (Kawai et al., 2001;Okada et al., 2002) and reach an almostnormal life span. Mice defective inGalNAc-transferase, missing the majorbrain gangliosides, were viable but suf-fered from instability of brain structureswith detachment of myelin sheets fromaxonal membranes (Schnaar, 2010) andmale infertility. A combined defect ofGM3-synthase and GM2-synthase causesan early fatal disease. Here, the absence of

    all a-series and b-series gangliosides triggersthe formation of 0-series gangliosides,which are not observed in the wild-typebrain. Their additional synthesis, however,cannot compensate the loss of all the majorneuronal ganglioside structures. Surprisingwas that the total ganglioside content in thebrains of the various mutant mice stayed al-most unchanged at the wild-type level(Sandhoff, 2012).

    The ubiquitous block of GlcCer syn-thesis, the precursor of the main glyco-sphingolipids (GSLs), is lethal in mice atembryonic day 6 (Yamashita et al., 1999).However, its cell-type-specific knock outin neurons (Jennemann et al., 2005),hepatocytes (Jennemann et al., 2010), orkeratinocytes (Jennemann et al., 2007)still allowed complete embryogenesis butwith the birth of fatally sick animals.Apparently, some complex glycolipids de-rived from GlcCer such as the stage-specific embryonic antigens (Kannagi etal., 1983; Eggens et al., 1989) involved incellular adhesion processes are needed atearly embryonic stages for developmentof a multicellular organism (Kolter et al.,2000).

    Gangliosides have been discovered asGSLs of neuronal membranes by lipidanalysis of Tay–Sachs disease (Klenk,1937, 1939), but their complex chemicalstructure was elucidated only later in the1960s (Kuhn and Wiegandt, 1963). Theirstorage in neuronal lysosomes paved theway for the analysis of their cellularmetabolism and lysosomal catabolism(Sandhoff et al., 1969; Sandhoff, 1977)

    Clinical phenotypes, correlations topathological and biochemicalphenotypes

    All types of gangliosidoses are caused byinherited defects in ganglioside catabo-lism (Fig. 1) and can manifest with severe,extremely variable clinical symptoms (Ta-ble 1) in almost any age group, althoughmost patients have an early infantile onsetand a late infantile or juvenile, fatal end oftheir disease, which is dominated by thecentral nervous involvement (Fig. 2A)(Lyon et al., 1996a). The cerebral degen-eration is marked by the lipid storagepresent in neurons with the ultrastruc-turally characteristic “membranous cy-toplasmic bodies,” which are secondary,“storage” lysosomes (Fig. 2B). These stor-age bodies distend the neurons (Fig. 2C),which eventually are lost, but the exactreason is unclear (for example, cell inac-tivity by ganglioside-induced impairmentof synaptic transmission is discussed).The peripheral and autonomous nerve

    systems are also involved, but with minorclinical relevance, though lysosomal stor-age bodies are demonstrable at places.

    In addition to the neuronal involve-ment, an extraneural involvement (Table1) is striking in infantile and partly presentin juvenile patients with GM1-gangli-osidosis; even an intrauterine manifesta-tion with hydrops fetalis is possible, and adysmorphic (gargoyle-like) face is regularin early infantile patients. Juvenile andadult GM1-gangliosidosis patients have apredominant or almost pure neurologicpicture (Suzuki et al., 2001). Extraneuralmanifestations are absent in GM2-gangliosidosis with the exception of vari-ant 0, also called Sandhoff disease, inwhich slight visceromegaly is frequent(Gravel et al., 2001) and some involve-ment of bones and even the heart can oc-cur (Venugopalan and Joshi, 2002).

    The clinical phenotypes in the gangli-osidoses can, though with some caveats,be correlated to the different types andnumbers of substrates, which are accumu-lated in these diseases. Examples of thesubstrates of the acid hydrolases whosedeficiencies define the diseases are ganglio-sides, asialogangliosides, other glycolipids,specific oligosaccharides and, in case of acid�-galactosidase, mucopolysaccharides. Inthe deficiencies, the patterns of accumulatedsubstrates can be explained in a simplifiedformat as follows. The main substratesof the acid �-galactosidase, which aredeficient in GM1-gangliosidosis, are GM1-ganglioside (Fig. 1), specific oligosaccha-rides, and keratan sulfate, so that this diseaseis marked not only by ganglioside storage(whose main pathologic correlate is neuro-nal degeneration) but also by oligosacchari-dosis and mucopolysaccharidosis, with theextraneuronal clinical involvement dueto the latter. The main substrate of the�-hexosaminidase A deficient in GM2-gangliosidosis variant B (also calledTay–Sachs disease) or suppressed in theGM2-activator-deficient variant AB, isGM2-ganglioside, so the disease is “only” aganglioside storing disorder. The main sub-strates of the �-hexosaminidases A and Bdeficient in GM2-gangliosidosis variant 0are GM2-ganglioside and oligosaccharides,so the disease includes gangliosidosis andoligosaccharidosis, with some visceral in-volvement due to the latter.

    Another correlation can, though withsome reasonable doubts, be assumed be-tween the time (as patients’ age) of onsetor manifestation, or the duration (chro-nicity) of the disease, and the rudimentarycatabolic functions of the defective (if notcompletely absent) acid hydrolases in the

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10197

  • patients. This “residual activity hypothe-sis” postulates higher residual functionsfor late starting/manifesting or chronicforms of gangliosidoses than for early (in-fantile) disease forms (see Protracted clin-ical forms of gangliosidoses and thethreshold theory, below). Although thelow residual, probably pathophysiologi-cally relevant activities of the defective hy-drolases cannot always reliably be assessedunder laboratory conditions in vitro, thathypothesis seemed to be acceptable formany patients who were studied enzymat-ically. As to those residual functions, it isinteresting that in late manifesting GM1-gangliosidosis the almost only substraterelevant for the biochemical phenotype isthe ganglioside accumulation, particu-larly in neurons, while for the breakdownof other substrates the residual functionsseem to be nearly sufficient.

    However, in a certain type of geneticacid �-galactosidase deficiency, also a suf-ficient catabolism of GM1-ganglioside ispossible, whereas the breakdown of othersubstrates is severely impaired, and the re-sulting disease is not gangliosidosis but mu-copolysaccharidosis IV (Morquio) type B(Suzuki et al., 2001). This indicates different

    substrate (e.g., the ganglioside/activatorprotein complex) binding sites, protein-folding-relevant sites, or subdomains in theenzyme, which are required for the catabo-lism of different substrates, and suggeststhat these sites are differentially or alterna-tively defective in different patients whohave then very variable diseases caused bytheir �-galactosidase defects. On the otherhand, a special GM1-ganglioside-storingdisease (associated with oligosaccharidestorage) results from an indirect deficiencyof acid �-galactosidase (and sialidase, and aspecific sulfatase) in galactosialidosis, whichis caused by the genetic deficiency of the so-called protective protein (identical to car-boxypeptidase A) (d’Azzo et al., 2001).When this protein is defective, the func-tional complex of the normal protein withacid �-galactosidase and sialidase (and thesulfatase) cannot be formed, and these en-zymes, though having a normal structure,are subject to rapid decay. The resultingclinical phenotype is very similar to that inclassic GM1-gangliosidosis (and to that inthe genetic sialidase defect, sialidosis). So anenzymatic diagnosis of GM1-gangliosidosishas always to be confirmed by demonstrat-

    ing the presence of normal sialidase activity,or by molecular gene analysis.

    Summary outline ofclinical symptomatologyThe main symptomatology of the gangli-osidoses is given as a partially simplifiedsummary in Table 1. Clinical detailsshould be found in the special literature(Lyon et al., 1996a, b; Gravel et al., 2001;Suzuki et al., 2001). Pathologists havefound that in gangliosidosis the brain issometimes slightly infiltrated with inflam-matory cells considered to be a responseto the dysfunction of the lipid-laden neu-rons and to the abundance of antigeni-cally acting gangliosides. Here followsome supplementary clinical remarks.Neuroimaging has no essential role in thecharacterization of gangliosidoses, but inlate disease stages cerebral and cerebellaratrophy can often be documented andslight T2-weighted hyperintensities mayappear earlier in the brainstem and whitematter (Wilken et al., 2008). Magneticresonance spectroscopy (N-acetyl aspar-tate, myoinositol, and other signals) maybe helpful in documenting neuronal lossand inflammation (Assadi et al., 2008).

    Table 1. The main clinical hallmarks and symptoms in gangliosidoses

    Disease Manifestation Regular symptoms/signs Facultative symptoms/signs Remarks

    GM1-gangliosidosis Infantilea,b Dysmorphic face, severe dysostosis, hepato-megaly, unable to sit, muscle hypotonia,later on spasticity, visual failure, foam cellsin bone marrow, oligosacchariduria,mucopolysacchariduria

    Cherry-red macular spotc, nystagmus,seizures, splenomegaly, joint contrac-tures, hematologic signs, vacuolatedblood lymphocytesd

    Rapid or 1–2 year downhill course withpsychomotor deterioration, generaldystrophy, feeding problems

    Juvenilea By 4 years or later: dysarthria, extrapyramidalsigns including dystonia

    Slight intellectual impairment, mild bonechanges, foam cells in bone marrow

    Some patients have also parts or attenu-ated forms of the symptoms listed forthe infantile disease, other patientsbelong to the chronic group

    Adulta As in juvenile disease, but with more insidiousonset

    Psychosis Disease is chronic rather than with onlyadult manifestation

    Chronic Combines late infantile, juvenile, andsometimes adult manifestations inwhich the above listed symptoms arepartly or gradually developed

    GM2-gangliosidosis, variants B,B1 and 0

    Infantilee Loss of head/trunk control, smiling, by about 6months, cherry-red macular spot c, highstartle response to noise, muscle hypotoniaby 1 year, epilepsy, tetraparesis with somespasticity, dementia, blindness

    Megalencephaly, visceral involvement andoligosacchariduria only in variant 0

    Downhill course mostly less rapid than inearly GM1-gangliosidosis

    Juvenile/subacute Onset by 3 to 6 years, dysarthria, loss ofspeech, spastic paraparesis, later onparaplegyf, pyramidal signsf, cerebellarataxia, mental deterioration

    Seizures, irritability, psychiatric signs,denervation muscle atrophy (EMG),areflexia, cherry red macular spotc

    Clinical variability even in siblingsg, aboveremark to juvenile GM1-gangliosidosishere also apt

    Adult Clumsiness, lower motor neuron disease,denervation muscle atrophy, cerebellarataxia

    Psychosis High clinical variability, oldest patient was76 years

    Chronic See remark to chronic GM1-gangliosidosisGM2-gangliosidosis, variant AB Most as in variants B, B1, 0 Cherry red macular spotc No adult patients knownaGrouping of GM1-gangliosidosis patients as type I, II, and III of the disease according to the age at clinical onset is questionable by the often ambiguous onset or insidious or chronic manifestation through all, except the early infantile, agegroups (Lyon et al, 1996b). bEarlier named “generalized gangliosidosis.” cSee Fig. 2D. dSee Fig. 2E. eEarlier named “infantile amaurotic idiocy”; relatively high prevalence of variant B (Tay–Sachs disease) in Ashkenazim. fSee Fig. 2A. gManypatients are Ashkenazim, but a genetic isolate of variant B1 is in Spain.

    10198 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

  • The prenatal and postnatal laboratory di-agnosis of gangliosidoses is usually madeenzyme biochemically (e.g., in blood sam-ples), and/or molecularly. Many aspectsof genotype–phenotype correlation inthese diseases were described in the specialliterature. Therapeutic strategies such asbone marrow transplantation (BMT),use of drugs with substrate reducing,chaperon-like and anti-inflammatory ef-fects, and intracerebral or intrathecal en-zyme replacement are under investigation(see Therapeutic approaches, below).

    Pathway of ganglioside degradationTopology of endocytosis and lysosomaldigestion of gangliosidesLysosomes are intracellular stomachs. Theydegrade macromolecules and release theircomponents as nutrients into the cytosol forsalvage pathways and energy metabolism.

    Macromolecules and membrane compo-nents reach the lysosomal compartment fordigestion by autophagy (Florey and Over-holtzer, 2012), phagocytosis (Delves et al.,2006; Florey and Overholtzer, 2012), and byendocytotic pathways (Kolter and Sandhoff,2005). Whereas water-soluble lysosomalhydrolases can attack water-soluble macro-molecules directly, the digestion of ganglio-sides, GSLs, and membranes needs a morecomplex cooperation between soluble hydro-lases, lipid binding and transfer proteins, andluminal intra-endolysosomal vesicles (Kolterand Sandhoff, 2005; Kolter and Sandhoff,2010). Due to the lipid phase problem, water-soluble glycosidases hardly attack amphiphilicGSLs as components of vesicular membranes,but need the help of membrane perturbingand lipid binding proteins, the sphingolipidactivator proteins (SAPs).

    Our view of the topology of lysosomalganglioside digestionFor their digestion, gangliosides of cellularsurfaces reach luminal intra-endolysosomalvesicles or intra-endosomal membranes(IMs) (Fig. 3; Burkhardt et al., 1997; Möbiuset al., 1999), which are generated during en-docytosis. Luminal vesicles are formed bysuccessive steps of vesicle budding and fis-sion controlled by the endosomal-sortingcomplex proteins (Wollert and Hurley,2010). The vesicles are prepared for lyso-somal digestion by a lipid-sorting processbeginning at the level of endosomes (Kolterand Sandhoff, 2005; Gallala et al., 2011).Cholesterol is sorted out by two sterol bind-ing proteins, NPC-2 and NPC-1 (Abdul-Hammed et al., 2010), and sphingomyelinis degraded by acid sphingomyelinase(ASM; V. Oninla, B. Breiden, K. Sandhoff,

    Figure 2. Clinical and morphological observations in gangliosidoses. A, Near end stage in a Spanish 13-year-old patient with GM2-gangliosidosis variant B1 (photograph courtesy of H.H. Goebel,Mainz). B, GM2-gangliosidosis variant 0 in a 23-week-old fetus; and part of a cortical neuron; early stage of membranous cytoplasmic bodies (two clusters marked by crosses), i.e., lipid storingsecondary lysosomes, �36,000 (courtesy W. Schlote, Tübingen). C, The large cells are neurons distended by lysosomal storage in the medulla oblongata of a 1.5-year-old patient with GM1-gangliosidosis. The small dark bodies are nuclei from increased numbers of glial cells (“reactive gliosis”). Cresyl violet stain �300 (courtesy of H.U. Benz, Tübingen). D, Cherry red macular spot in aninfantile patient with GM2-gangliosidosis variant B (Tay–Sachs disease). The dark red spot in the middle of the light central area is secondary to lipid storage in neuronal cells in this area; the storingcells have lost their processes that normally cover the fovea centralis. The fovea normally appears as yellow, but is changed to the red macular spot showing the color of the choroidea behind theretina. E, Lymphocytes with clusters of vacuoles (storage lysosomes) in blood smear of an early infantile patient with GM1-gangliosidosis, indicating the “generalized storage disorder.” May-Grünwald stain; �1500 (courtesy of H.U. Benz, Tübingen).

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10199

  • unpublished observations). Anionic bis-(monoacylglycero)phosphate (BMP) thatstimulates ganglioside catabolic steps sub-stantially is formed from phosphatidyl glyc-erol in the IMs (Gallala and Sandhoff, 2011;Gallala et al., 2011). Whereas the lysosomalperimeter membrane seems to be quite re-sistant against lysosomal degradation, IMsare digested with the help of lipid bindingproteins (SAPs) and hydrolytic enzymes.Perimeter membranes are protected bya thick glycocalyx enriched in polylac-tosamine structures, and by a high choles-terol level (Hay, 1989; Appelqvist et al.,2012), the chaperon HSP70 (Kirkegaard etal., 2010), and presumably also by a highlateral pressure attenuating the interactionwith GM2AP, a lipid binding protein essen-tial for ganglioside catabolism (Giehl et al.,1999).

    Biochemistry of ganglioside catabolismand its diseasesGanglioside catabolism proceeds in a stepwiseprocedure (Fig. 1) at the surface of luminal in-

    tralysosomal vesicle and membrane struc-tures. These are enriched with anionic BMP,which attracts polycationic proteins, glycosi-dases,andSAPs, totheganglioside-containingmembrane surfaces in the acidic intralyso-somal environment. Whereas GM1 hydrolyz-ing �-galactosidase and GM2 splitting HexAbind to these anionic surfaces, they do not at-tack the membrane bound ganglioside sub-strates intheabsenceofmembraneperturbinglipid binding and transfer proteins. The firstcaseofGM1storagewasdescribedin1963asaspecial form of infantile amaurotic idiocy(JatzkewitzandSandhoff,1963).Hydrolysisofvesicle-bound GM1 by lysosomal ganglioside�-galactosidase needs the presence ofGM2AP or saposin B (Sap-B; Wilkening etal., 2000). Anionic lipids like BMP in theGM1 carrying vesicles stimulate GM1 hy-drolysis efficiently. Inherited defects of theGM1 degrading �-galactosidase causeGM1-gangliosidoses (Okada and O’Brien,1968).

    The nonspecific and versatile GM1 de-grading �-galactosidase occurs as part of alysosomal multi-enzyme complex, contain-ing sialidase, cathepsin A (the so-called pro-tective protein) (d’Azzo et al., 2001), andN-acetylaminogalacto-6-sulfate sulfa-tase (Pshezhetsky and Ashmarina, 2001).Due to changes of the substrate specificityof variant patient enzymes, an inheriteddefect of GM1-�-galactosidase may alsolead to a major accumulation of galac-tose containing keratan sulfate and oligo-saccharides, a phenotype called Morquiosyndrome, type B (mucopolysaccharido-sis IV B) (Neufeld and Muenzer, 2001;Okumiya et al., 2003).

    Hydrolysis of GM2 is facilitated at an-ionic vesicular surfaces by the cooperationof two proteins, HexA (�,�) and themembrane perturbing and lipid bindingprotein GM2AP (Fig. 4) (Kolter andSandhoff, 2006). At acidic pH values, thelatter one is a protonated cationic amphi-philic protein and can bind to the anionic,

    Figure 3. Proposed topology of endocytosis and lysosomal lipid and membrane digestion (Gallala et al., 2011). “A section of the plasma membrane is internalized by way of coated pits orcaveolae. These membrane patches include GSLs (red) and receptors such as epidermal growth factor receptor (EGFR; blue). These vesicles fuse with the early endosomes, which mature to lateendosomes. Endosomal perimeter membranes form invaginations, controlled by ESCRT proteins (Wollert and Hurley, 2010), which bud off, forming intra-endosomal vesicles. Lipid sorting occurs atthis stage. The pH of the lumen is �5. At this pH, ASM is active and degrades sphingomyelin of the intra-endosomal vesicles to ceramide, whereas the perimeter membrane is protected against theaction of ASM by the glycocalyx facing the lumen. This decrease in sphingomyelin, coupled with the increase in ceramide, facilitates the binding of cholesterol to NPC2 and its transport to theperimeter membrane of the late endosome where it is transferred to NPC1 (Kwon et al., 2009). This protein mediates the export of cholesterol through the glycocalyx, eventually reaching cholesterolbinding proteins in the cytosol. Ultimately, late endosomes fuse with lysosomes. The GSLs are harbored in intralysosomal vesicles that face the lumen of the lysosome, and are degraded by hydrolaseswith the assistance of SAPs. The products of this degradation are exported to the cytosol or loaded on CD1b immunoreceptors and exported to the plasma membrane for antigen presentation.Gradients of pH in the lysosol, and intra-endolysosomal vesicle content of cholesterol (Chol), BMP, sphingomyelin (SM, hypothetical), and ceramide (Cer, hypothetical) are shown” (modified fromKolter and Sandhoff, 2010).

    10200 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

  • BMP-rich membrane surfaces, and lift aganglioside molecule (Fig. 4). A HexAbinding helix of the GM2AP facilitatesformation of the Michaelis–Menten com-plex for the degradation of GM2, the Tay–Sachs ganglioside, and glycolipid GA2.Inherited defects of either of the proteinsresult in fatal neurodegenerative diseases(Kolter and Sandhoff, 2006). Tay–Sachsdisease is caused by mutations in theHexA gene with loss of HexA (�,�) and S(�,�) activity. HexA mutations that stillallow the formation of the heterodimericHexA (�,�) and affect only its activityagainst anionic substrates result in B1variant of GM2-gangliosidosis (Kytzia etal., 1983; Kytzia and Sandhoff, 1985). Onthe other hand, inherited defects in HexBgene cause the loss of HexA (�,�) andHexB (�,�) activities in Sandhoff disease.Here, the combined deficiency of bothmajor hexosaminidase isoenzymes causes

    the additional storage of neutral glycolip-ids, especially of globoside in the visceralorgans, and of oligosaccharides. The raredefect of GM2AP results in AB-variant ofGM2-gangliosidoses (Sandhoff et al.,1989; Sandhoff and Kolter, 2003). Genet-ically manipulated knock-out mice areavailable as models for these diseases tostudy their course and pathogenesis, andto investigate therapeutic approaches.

    Protracted clinical forms ofgangliosidoses and the threshold theoryComplete or nearly complete loss of GM1or GM2 catabolic activities results in fatalinfantile forms of storage diseases. How-ever, allelic mutations, which produceproteins with some residual catabolic ac-tivities, give rise to protracted clinicalforms often described as late infantile, ju-venile, or chronic diseases (Sandhoff et al.,1989) with a wide range of clinical symp-

    tomatology. At the biochemical level, theclinical heterogeneity is paralleled by avariation of the extent and the pattern ofglycolipid accumulation and by differentlevels of residual catabolic activities de-tected in cultures of patients’ fibroblasts(Fig. 5). Catabolic activities were assayedin cell cultures using the physiological, ra-diolabeled lipid substrates instead of thewater-soluble synthetic substrates, whichserve widely for diagnostic purposes. Thewater-soluble substrates may well lead toerroneous data because of the lipid phaseproblem and the absent control byGM2AP (and other activators) of the ca-tabolism of physiological lipid substratessuch as GM2 ganglioside. According toFigure 5, GM2 cleavage by HexA was al-most absent in fibroblasts of infantile pa-tients, and significantly higher in those ofjuvenile and adult patients (Kytzia et al.,1984; Leinekugel et al., 1992). This obser-

    Figure 4. Mechanism of GM2-AP-Liftase. A, Model for the interaction of GM2-activator protein with luminal lysosomal membranes (IM) in the degradation of ganglioside GM2 (modified fromWendeler et al., 2004a). “GM2-AP interacts with the membrane by dipping the two exposed hydrophobic loops, V90 –W94 and V153–L163, into the apolar part of the membrane. Ganglioside GM2is recognized by specific sites at the rim of the cavity. In the open protein conformation, the large hydrophobic area reaching from the apolar phase of the membrane to the activator’s cavity lowersthe energy barrier for lipids leaving the membrane in an upward direction. After the ceramide tail has arrived inside the activator’s cavity, the inward-bending of the hydrophobic loop V153–L163is favored, and the conformation changes to the closed form (Wright et al., 2003). This folding in of the hydrophobic loop leaves a more polar patch close to the membrane. The activator is thenanchored only by the loop V90 –W94. It may now rotate slightly upward to expose all polar patches more fully to the solvent, and it may also leave the membrane and interact with the degradingenzyme. The photoaffinity label analogs C(n � 5,1)-TPD-GM2 were photo-incorporated specifically into the region V153–L163” (Sandhoff, 2012). B, BMP and GM2-AP stimulate the hydrolysis ofLUV-bound ganglioside GM2. The degradation of ganglioside GM2 inserted into LUVs doped with 0 mol % BMP (E), 5 mol % BMP(�), 10 mol % BMP (·), 20 mol % BMP (Œ), or 25 mol % BMP (f)was measured in the presence of increasing concentrations of GM2-AP (0 –2.2 �M) (Werth et al., 2001).

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10201

  • vation can well be modeled on the basis ofa greatly simplified kinetic model (Con-zelmann and Sandhoff, 1983, 1991;Leinekugel et al., 1992) presented in Fig-ure 5. A nice correlation was observed be-tween the predicted and the measuredlipid substrate turnover, and also betweenthe decrease of the ganglioside GM2 turn-over and the clinical course of the disease.Ten to 20% of normal GM2 cleaving ac-tivity appear already compatible withnormal life. Similar observations werereported for metachromatic leukodystro-phy (MLD), Gaucher, Sandhoff, andASM-deficient Niemann–Pick disease(Kudoh et al., 1983; Kytzia et al., 1984;Leinekugel et al., 1992; Graber et al.,1994).

    Of course, additional factors inducedby the mutation and the storage process in

    the disease will also contribute to the clin-ical course and disease pathogenesis, e.g.,misregulation of proteins and intracellu-lar trafficking, formation of lytic and toxiccompounds (Nilsson et al., 1982; Igisuand Suzuki, 1984), depletion of precursorpools, alterations of composition andfunction of membranes, mislocalizedstorage compounds like GM1, which acti-vate ER-chaperons and trigger neuronalapoptosis (Tessitore et al., 2004), and al-tered regulation of endolysosomal pro-teins and enzymes by an increased load oflipids (see below). Also the formation ofmeganeurites and an increase in synapticspines may well disturb the connectivityin the nervous system (Purpura and Su-zuki, 1976).

    A genetic mutation and loss of a cata-bolic activity may not only result in accu-

    mulation of nondegradable substrates butalso affect the transcription rate of manyother genes. Mutations in the HexB geneand the subsequent loss of HexA andHexB activity in genetically manipulatedmice trigger a significant downregulationof �300 transcripts and a significant up-regulation of another �250 transcripts in-cluding genes of the immune system(Myerowitz et al., 2002).

    Genetically manipulated mice are notalways faithful copies of the human dis-ease. In contrast to Tay–Sachs patients,HexA knock-out mice reach an almostnormal life span and hardly get sick. Thereason seems to be a small difference inthe substrate specificity of murine and hu-man sialidase. Whereas human sialidase isalmost inactive, murine sialidase has a mi-nor but significant cleaving activity on the

    Figure 5. Residual catabolic activity correlates with clinical forms of GM2 gangliosidoses (modified from Sandhoff, 2012). A, Steady-state substrate concentration as a function of enzymeconcentration and activity (Conzelmann and Sandhoff, 1983). The model underlying this theoretical calculation assumes influx of the substrate into the lysosomal compartment at a constant rate(vi) and its subsequent utilization by the catabolic enzyme. Blue line� [S]eq steady-state substrate concentration;

    ………… � theoretical threshold of enzyme activity; ——-� critical thresholdvalue, taking limited solubility of substrate into account; red line � turnover rate of substrate (flux rate). B, To experimentally verify the basic assumptions for this model, studies were performedin cell culture (Leinekugel et al., 1992). The radiolabeled substrate ganglioside GM2 was added to cultures of skin fibroblasts with different activities of �-hexosaminidase A and its uptake andturnover measured. The correlation between residual enzyme activity and the turnover rate of the substrate was essentially as predicted: degradation rate of ganglioside GM2 increased steeply withresidual activity, to reach the control level at a residual activity of �10 –15% of normal. All cells with an activity above this critical threshold had a normal turnover. Comparison of the results of thesefeeding studies with the clinical status of the donor of each cell line basically confirmed our notions but also revealed the limitations of the cell culture approach.

    10202 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

  • storage compound ganglioside GM2 andthereby opens a catabolic bypass (Sand-hoff and Jatzkewitz, 1967; Sango et al.,1995). Mutant mice develop only a minorstorage process in some neurons, e.g., inthe pyramidal cells of the cortex (Taniikeet al., 1995). Apparently, the catabolic ac-tivity of the bypass (sequential action ofmurine sialidase to convert GM2 to GA2,and hex B to convert GA2 to LacCer) issufficient in almost all neurons of the ce-rebral cortex except in the murine pyra-midal cells and in a few others. These cellsprobably have a rather high biosyntheticrate for gangliosides, thereby slightly ex-ceeding their maximal catabolic capacity(Vmax of the catabolic system; Fig. 5) incase of hex A knocked out. Further patho-genetic cascades activated in these dis-eases, altered lipid trafficking, alteredautophagy, and induction of inflamma-tion, have been reviewed (Vitner et al.,2010; Xu et al., 2010).

    Many proteins involved in gangliosidemetabolism are nonspecificThe inherited defect of an endolysosomalprotein usually affects only one step inganglioside and sphingolipid catabolism.However, a more careful investigation ofthe proteins revealed that many of themhave rather promiscuous functions and,therefore, have a broader effect on metab-olism than originally expected. For glyco-sidases this has been known for a longtime, and it was no surprise to find outthat GM1-�-galactosidase hydrolyzesmany other �-galactosides in addition toganglioside GM1 and GA1 (Kolter andSandhoff, 2006). Mutations in the�-galactosidase gene can even change theenzyme’s substrate specificity and affectthe catabolism of GSLs and glycosamino-glycans differently.

    It was also no surprise to realize thathuman �-hexosaminidases have broadand partially overlapping substrate speci-ficities. Again some mutations in theHexA gene may also change substratespecificity of the patients’ HexA. Thosethat disrupt enzyme activity only againstanionic substrates (like GM2) and notagainst neutral substrates (like globoside)cause B1 variant of GM2-gangliosidosis(Kytzia et al., 1983), whereas those thataffect HexA activity against all substratesincluding neutral substrates cause Tay–Sachs disease (variant B of GM2-gangliosidosis). Active �-hexosaminidasesare the homodimers HexB (�,�) and HexS(�,�), and the heterodimer HexA (�,�).Each of them carries two active sites at thedomain interfaces (Maier et al., 2003; Le-

    mieux et al., 2006). Therefore, mutationsaffecting the dimer formation produce inac-tive monomeric subunits causing infantilediseases. The broad and overlapping speci-ficity of murine hexosaminidases A, B, and Sis highlighted by mice lacking both subunitsof �-hexosaminidase. They store ganglio-sides and glycosaminoglycans and displayphenotypic, pathologic, and biochemicalfeatures of mucopolysaccharidoses, notseen in Tay–Sachs and Sandhoff disease(Sango et al., 1996).

    The first activator protein described,the sulfatide activator protein, is nowcalled Sap-B. It was discovered as an es-sential cofactor for the lysosomal catabo-lism of sulfatides (Mehl and Jatzkewitz,1964). Its deficiency causes a late infantileform of MLD with a pronounced sulfatidestorage (Mehl and Jatzkewitz, 1964; Ste-vens et al., 1981). Sap-B combines withsulfatides, and the formed stoichiometricsoluble complex then forms a Michaelis–Menten complex with arylsulfatase A(Mehl and Jatzkewitz, 1964; Mraz et al.,1976). Though this suggested a high lipidbinding specificity of Sap-B, we nowknow that the human Sap-B is not specificfor sulfatide binding, but rather a promis-cuous lipid binding protein (Gärtner etal., 1983; Sun et al., 2008). It even stimu-lates the hydrolysis of bacterial lipids bybacterial hydrolases (Li et al., 1988). Alsothe other saposins have broad functions(Doering et al., 1999) and are rather un-specific lipid binding proteins, like theGM2AP (Wendeler et al., 2004b). Even theglycosyltransferases involved in the biosyn-thesis of complex gangliosides at Golgimembranes are rather versatile. There areonly one GalNAc-, one Gal-, and two sialyl-transferases available in Golgi membranesto convert the precursors LacCer, GM3,GD3, and GT3 into the different complexgangliosides of the 0-, a-, b-, and c-series(Kolter et al., 2002).

    Membrane lipids are regulators ofganglioside catabolismPromiscuous functions of endolysosomalproteins and enzymes make it difficult tounderstand the precise molecular pathol-ogy of the diseases caused by a defect ofany of these proteins. The complexity isfurther increased by the strong lipid de-pendency of many endolysosomal pro-teins, given that the lipid composition ofmany organelle membranes is mostly un-known, especially in patients’ tissues. Invitro experiments have shown that cata-bolic steps at surfaces of substrate carryingliposomes—simulating luminal endoly-sosomal vesicles—not only need low pH

    values, enzymes, and SAPs for significantdegradation rates, but also a sufficientconcentration of anionic lipids such asBMP in the liposomal membranes.

    Both, GM2AP and Sap-B, allow that�-galactosidase hydrolyzes GM1. There-fore, neither a defect of GM2AP nor ofSap-B causes a GM1 accumulation, sincethe other one left efficiently facilitates thereaction. However, the reaction is ratherslow in neutral liposomes and is stimu-lated 2- to 9-fold in the presence of ananionic phospholipid such as BMP, phos-phatidylinositol, dolicholphosphate, andphosphatidylserine (Wilkening et al.,2000). Even at acidic, lysosomal pH val-ues, these phospholipids convey negativecharges to the vesicular surfaces, which at-tract and bind the protonated and posi-tively charged hydrolases and SAPs tocatalyze the hydrolytic reaction. This viewalso applies to the degradation of lipo-somal ganglioside GM2 by HexA in thepresence of GM2AP. GM2AP is essentialfor the enzymatic hydrolysis of ganglio-side GM2, even in the presence of highBMP concentrations. Its absence causesthe fatal neurodegenerative AB variant ofGM2-gangliosidosis in vivo. Still, the reac-tion is stimulated by anionic BMP �100-fold (Werth et al., 2001) (Fig. 4B). Alsodownstream reactions (Fig. 1) are stimu-lated by anionic membrane lipids. At con-centrations of anionic phospholipids �10mol% in the liposomal membranes, thecleavage of GlcCer by �-glucosidase(Wilkening et al., 1998) and that of cer-amide by acid ceramidase (Linke et al.,2001) proceeds even in the absence of aSAP.

    Therapeutic approachesCurrently, there is no treatment availablefor patients with ganglioside storage dis-eases (van Gelder et al., 2012). A main ob-stacle is the blood– brain barrier, whichprevents the passage of therapeutic en-zymes and proteins into the brain. For theprevention of substrate storage, substrateinflux into the lysosomal compartmentmust not exceed this compartment’s cat-abolic capacity (see Protracted clinicalforms of gangliosidoses and the thresholdtheory, above; Conzelmann and Sand-hoff, 1983, 1991; Leinekugel et al., 1992).This means that therapeutic methodsshould either increase the catabolic rateabove the threshold or decrease the influxrate of substrates below the maximumcatabolic capacity of the affected cells, e.g.,by reducing the cellular substrate biosyn-thetic rate (Fig. 6). Therapeutic strategies

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10203

  • in use such as enzyme replacement therapy(ERT) for adult Gaucher disease and thoseunder study in animal models such as BMTyielded so far no substantial and long-lasting improvements in mouse modelswith GM1- and GM2-gangliosidosis. How-ever, intracerebroventricular administra-tion of a modified recombinant HexB(Matsuoka et al., 2011) and a highly phos-phomannosylated recombinant humanHexA reduced the level of storage com-pounds (Tsuji et al., 2011) in the HexA- andHexB-deficient mouse brain efficiently.

    Additional approaches using lowmolecular compounds penetrating theblood– brain barrier have been evaluatedin murine models of gangliosidoses, butso far without substantial success. Theyinclude the use of chemical chaperons,substrate analogs, or active site inhibitors(Matsuda et al., 2003; Tropak et al., 2004)to stabilize variant patient hydrolases dur-ing their biosynthesis and folding, andthereby enhance their hydrolytic activityas introduced by Fan et al. (1999).

    Another approach is the substrate reduc-tion therapy. It uses inhibitors of the biosyn-

    thetic pathway, such as iminosugars of thenojirimycin type (Butters et al., 2005; Fanturet al., 2012; Suzuki et al., 2012) and reducesthe influx of substrates into the lysosomes(Kolter and Sandhoff, 1999). However,treatment of two Tay–Sachs patients withN-butyldeoxynojirimycin (Miglustat) didnot arrest their clinical deterioration (Bembiet al., 2006), whereas a stable neurologicalpicture was reported for two patients withSandhoff disease after Miglustat treatment(Tallaksen and Berg, 2009; Wortmann et al.,2009).

    However, reducing the ganglioside bio-synthesis with inhibitors of the ceramideglucosyltransferase, N-butyldeoxynojirimy-cin or N-butyldeoxygalactonojirimycin, ledto an increased survival of Sandhoff mice upto 2 months (Andersson et al., 2004; Jeyaku-mar et al., 2004).

    In a clinical Phase I/II trial, treatment oflate onset GM2-gangliosidosis patients witha presumed chaperon of �-hexosamini-dases, pyrimethamine, for a 4 month periodenhanced their leukocyte HexA levels, butclinical efficacy could not be assessed(Clarke et al., 2011).

    A combination of different methods forpatients with significant residual enzymaticactivity, e.g., substrate reduction togetherwith enzyme enhancement therapy, non-steroidal anti-inflammatory drugs, and an-tioxidants is expected to yield better results.

    Gene therapyTo circumvent the blood– brain barrier,functional copies of the mutant gene weredirectly inserted into murine brain cellswith the help of retroviral and lentiviralvectors (Biffi and Naldini, 2005). Thefunctional enzyme should be stably over-expressed in the transfected mutant cellsand secreted, thereby cross-correcting ad-jacent cells (Arfi et al., 2005; Martino et al.,2005).

    Intracerebral injection of an adenovi-ral vector encoding the �-subunit ofHexA increased HexA activity to nearlynormal levels in HexB-deficient mice(Bourgoin et al., 2003). Also, the intracra-nial inoculation of recombinant adeno-associated viral (AAV) vectors encodingboth, human �- and � Hex-subunit genes,resulted in an almost generalized correc-

    Figure 6. Intracellular metabolic flux of sphingolipids. vi � the influx rate of the substrate into the lysosome; for further information see the text. Modified from Kolter and Sandhoff, 1999).

    10204 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

  • tion of the catabolic lysosomal defect inthe same mice (Cachón-González et al.,2006). Animals survived more than a yearwith delayed onset of the disease, reducedganglioside storage and inflammation,preserved motor function, and rescue ofacute neurodegenerative illness (Sargeantet al., 2012).

    AAV-mediated neonatal gene deliveryby intracerebroventricular injection seemsto be an effective approach to treat GM1-gangliosidoses. It resulted in a widespreadappearance of the missing �-galactosidaseactivity in the murine brain and a normal-ization of all GSL levels (Broekman et al.,2007; Baek et al., 2010).

    Though these results are promising, itwill take many more investigations andprogress to develop a real gene therapy forGSL and sphingolipid storage diseases.

    Conclusions and outlookTo identify the molecular basis of amau-rotic idiocy of the lipid accumulating type,it was important to isolate the storagecompounds and clarify their chemicalstructures. This allowed a search for thedefective catabolic steps to identify the en-zymes and lipid binding proteins in-volved, their genomic structures, and themutations causing the different gangli-osidoses. The fact that the main storagecompounds (gangliosides GM1 andGM2) are preferentially synthesized inneuronal cells is crucial for the start ofneurodegenerative diseases and compli-cates all therapeutic approaches. So far,efficient ERT is not feasible, since theblood– brain barrier prevents cerebral up-take of therapeutic proteins. Also geneticapproaches that appeared to be promisingin mouse models are still not reliable(Cachón-González et al., 2006).

    There is no simple model to under-stand the complex pathogenesis of mono-genetic gangliosidoses. The primary lipidstorage affects the endolysosomal systemof neurons and obviously does not triggeran effective feedback inhibition of the stor-age compounds’ biosynthesis. However, thestorage may well impair the turnover ofmembranes and macromolecules, affectingneuronal metabolism and function.

    Degradation of gangliosides requiresthe interaction of rather unspecific hydro-lases and lipid binding proteins at surfacesof intraluminal vesicles and membranestructures. These complex interactions,including the substantial dependency ofmany catabolic steps on lipid binding co-factors (SAPs) and the lipid compositionof the intraluminal, lysosomal mem-branes, seem to prevent an exact and

    quantitative understanding of the degra-dative sphingolipid pathways.

    We also do not know to what extentthe lipid load of the affected lysosomesdisturbs their primary functions as cellu-lar stomachs (Tamboli et al., 2011) to de-grade macromolecules and release theproducts into the cytosol as nutrientsfor biosynthetic pathways and energy me-tabolism. One should also keep in mindthat the cellular uptake of essential factors,for example, the uptake of vitamin B12involving several proteins, and that ofFe 2� via transferrin and the transferrinreceptor, may well be impaired by thelipid load causing a traffic jam in the en-dolysosomal system (Jeyakumar et al.,2009). The storage of GSLs in gangli-osidoses also triggers side effects such asthe accumulation of hydrophobic mate-rial including phospholipids and hydro-phobic protein fragments in the storagegranules (“membranous cytoplasmicbodies”) (Samuels et al., 1963), but alsotransmembrane spans of mitochondrialATP synthase and amyloid-�-peptide(Keilani et al., 2012). As in multiple sclero-sis, functionally impaired neurons giverise to an immune response, and areprone to digestion by microglia and in-vaded macrophages (Wada et al., 2000;Wekerle, 2005).

    Although we have learned a lot aboutthe function of gangliosides, their bio-chemistry, enzymology, and cell biology,we are still far away from a real under-standing of the molecular and cellularpathogenesis of ganglioside storage dis-eases and the development of therapeuticapproaches. However, the successful dis-section of the clinical picture of amauroticidiocy into 5 different ganglioside storagediseases involving 4 different genes, and agroup of 10 so far not extensively analyzedneuronal ceroidlipofuscinoses, may en-courage the search for similar clinical en-tities of monogenetic diseases but withcausative diversity, among the high num-ber of endemic inherited diseases, the mo-lecular basis of which still has to beelucidated.

    ReferencesAbdul-Hammed M, Breiden B, Adebayo MA, Ba-

    balola JO, Schwarzmann G, Sandhoff K(2010) Role of endosomal membrane lipidsand NPC2 in cholesterol transfer and mem-brane fusion. J Lipid Res 51:1747–1760.CrossRef Medline

    Andersson U, Smith D, Jeyakumar M, Butters TD,Borja MC, Dwek RA, Platt FM (2004) Im-proved outcome of N-butyldeoxygalactonojiri-mycin-mediated substrate reduction therapy ina mouse model of Sandhoff disease. NeurobiolDis 16:506–515. CrossRef Medline

    Appelqvist H, Sandin L, Björnström K, Saftig P,Garner B, Ollinger K, Kågedal K (2012) Sen-sitivity to lysosome-dependent cell death is di-rectly regulated by lysosomal cholesterolcontent. PLoS One 7:e50262. CrossRefMedline

    Arfi A, Bourgoin C, Basso L, Emiliani C, TanciniB, Chigorno V, Li YT, Orlacchio A, Poenaru L,Sonnino S, Caillaud C (2005) Bicistroniclentiviral vector corrects beta-hexosaminidasedeficiency in transduced and cross-correctedhuman Sandhoff fibroblasts. Neurobiol Dis20:583–593. CrossRef Medline

    Assadi M, Baseman S, Janson C, Wang DJ, Bila-niuk L, Leone P (2008) Serial 1H-MRS inGM2 gangliosidoses. Eur J Pediatr 167:347–352. CrossRef Medline

    Baek RC, Broekman ML, Leroy SG, Tierney LA,Sandberg MA, d’Azzo A, Seyfried TN, Sena-Esteves M (2010) AAV-mediated gene deliv-ery in adult GM1-gangliosidosis mice correctslysosomal storage in CNS and improves sur-vival. PLoS One 5:e13468. CrossRef Medline

    Bembi B, Marchetti F, Guerci VI, Ciana G, Ad-dobbati R, Grasso D, Barone R, Cariati R,Fernandez-Guillen L, Butters T, Pittis MG(2006) Substrate reduction therapy in the in-fantile form of Tay-Sachs disease. Neurology66:278 –280. CrossRef Medline

    Biffi A, Naldini L (2005) Gene therapy of storagedisorders by retroviral and lentiviral vectors.Hum Gene Ther 16:1133–1142. CrossRefMedline

    Bourgoin C, Emiliani C, Kremer EJ, Gelot A, Tan-cini B, Gravel RA, Drugan C, Orlacchio A,Poenaru L, Caillaud C (2003) Widespreaddistribution of beta-hexosaminidase activityin the brain of a Sandhoff mouse model aftercoinjection of adenoviral vector and manni-tol. Gene Ther 10:1841–1849. CrossRefMedline

    Broekman ML, Baek RC, Comer LA, Fernandez JL,Seyfried TN, Sena-Esteves M (2007) Completecorrection of enzymatic deficiency and neuro-chemistry in the GM1-gangliosidosis mousebrain by neonatal adeno-associated virus-mediated gene delivery. Mol Ther 15:30–37.CrossRef Medline

    Burkhardt JK, Hüttler S, Klein A, Möbius W,Habermann A, Griffiths G, Sandhoff K(1997) Accumulation of sphingolipids inSAP-precursor (prosaposin)-deficient fibro-blasts occurs as intralysosomal membranestructures and can be completely reversed bytreatment with human SAP-precursor. EurJ Cell Biol 73:10 –18. Medline

    Butters TD, Dwek RA, Platt FM (2005) Iminosugar inhibitors for treating the lysosomal glyco-sphingolipidoses. Glycobiology 15:43R–52R.CrossRef Medline

    Cachón-González MB, Wang SZ, Lynch A,Ziegler R, Cheng SH, Cox TM (2006) Effec-tive gene therapy in an authentic model ofTay-Sachs-related diseases. Proc Natl Acad SciU S A 103:10373–10378. CrossRef Medline

    Clarke JT, Mahuran DJ, Sathe S, Kolodny EH,Rigat BA, Raiman JA, Tropak MB (2011) Anopen-label Phase I/II clinical trial of pyrim-ethamine for the treatment of patientsaffected with chronic GM2 gangliosidosis(Tay-Sachs or Sandhoff variants). Mol GenetMetab 102:6 –12. CrossRef Medline

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10205

    http://dx.doi.org/10.1194/jlr.M003822http://www.ncbi.nlm.nih.gov/pubmed/20179319http://dx.doi.org/10.1016/j.nbd.2004.04.012http://www.ncbi.nlm.nih.gov/pubmed/15262262http://dx.doi.org/10.1371/journal.pone.0050262http://www.ncbi.nlm.nih.gov/pubmed/23166840http://dx.doi.org/10.1016/j.nbd.2005.04.017http://www.ncbi.nlm.nih.gov/pubmed/15953731http://dx.doi.org/10.1007/s00431-007-0469-0http://www.ncbi.nlm.nih.gov/pubmed/17387512http://dx.doi.org/10.1371/journal.pone.0013468http://www.ncbi.nlm.nih.gov/pubmed/20976108http://dx.doi.org/10.1212/01.wnl.0000194225.78917.dehttp://www.ncbi.nlm.nih.gov/pubmed/16434676http://dx.doi.org/10.1089/hum.2005.16.1133http://www.ncbi.nlm.nih.gov/pubmed/16218774http://dx.doi.org/10.1038/sj.gt.3302081http://www.ncbi.nlm.nih.gov/pubmed/12960974http://dx.doi.org/10.1038/sj.mt.6300004http://www.ncbi.nlm.nih.gov/pubmed/17164772http://www.ncbi.nlm.nih.gov/pubmed/9174667http://dx.doi.org/10.1093/glycob/cwi076http://www.ncbi.nlm.nih.gov/pubmed/15901676http://dx.doi.org/10.1073/pnas.0603765103http://www.ncbi.nlm.nih.gov/pubmed/16801539http://dx.doi.org/10.1016/j.ymgme.2010.09.004http://www.ncbi.nlm.nih.gov/pubmed/20926324

  • Conzelmann E, Sandhoff K (1983) Partial en-zyme deficiencies: residual activities and thedevelopment of neurological disorders. DevNeurosci 6:58 –71. CrossRef Medline

    Conzelmann E, Sandhoff K (1991) Biochemicalbasis of late-onset neurolipidoses. Dev Neuro-sci 13:197–204. CrossRef Medline

    d’Azzo A, Andria G, Strisciulio P, Galjaard H(2001) Galactosialidosis. In: The metabolicand molecular bases of inherited diseases, Ed 8(Scriver CR, Beaudet AL, Sly WS, Valle D,eds), pp 3811–3826. New York: McGraw-Hill.

    Delves P, Martin S, Burton D, Roit I (2006)Roit’s essential immunology, Ed 11. Malden,MA: Blackwell Publishing.

    Doering T, Holleran WM, Potratz A, Vielhaber G,Elias PM, Suzuki K, Sandhoff K (1999) Sph-ingolipid activator proteins are required forepidermal permeability barrier formation.J Biol Chem 274:11038 –11045. CrossRefMedline

    Eggens I, Fenderson B, Toyokuni T, Dean B,Stroud M, Hakomori S (1989) Specific in-teraction between Lex and Lex determinants.A possible basis for cell recognition in preim-plantation embryos and in embryonal carci-noma cells. J Biol Chem 264:9476 –9484.Medline

    Fan JQ, Ishii S, Asano N, Suzuki Y (1999) Ac-celerated transport and maturation of lyso-somal alpha-galactosidase A in Fabrylymphoblasts by an enzyme inhibitor. NatMed 5:112–115. CrossRef Medline

    Fantur KM, Wrodnigg TM, Stütz AE, Pabst BM,Paschke E (2012) Fluorous iminoalditols actas effective pharmacological chaperonesagainst gene products from GLB(1) allelescausing GM1-gangliosidosis and Morquio Bdisease. J Inherit Metab Dis 35:495–503.CrossRef Medline

    Fishman PH, Max SR, Tallman JF, Brady RO, Ma-claren NK, Cornblath M (1975) DeficientGanglioside Biosynthesis: a novel human sph-ingolipidosis. Science 187:68 –70. CrossRefMedline

    Florey O, Overholtzer M (2012) Autophagy pro-teins in macroendocytic engulfment. TrendsCell Biol 22:374–380. CrossRef Medline

    Fragaki K, Ait-El-Mkadem S, Chaussenot A, GireC, Mengual R, Bonesso L, Bénéteau M, RicciJE, Desquiret-Dumas V, Procaccio V, Rötig A,Paquis-Flucklinger V (2013) Refractory epi-lepsy and mitochondrial dysfunction due toGM3 synthase deficiency. Eur J Hum Genet21:528 –534. CrossRef Medline

    Gallala HD, Sandhoff K (2011) Biological func-tion of the cellular lipid BMP—BMP as a keyactivator for cholesterol sorting and mem-brane digestion. Neurochem Res 36:1594 –1600. CrossRef Medline

    Gallala HD, Breiden B, Sandhoff K (2011)Regulation of the NPC2 protein-mediatedcholesterol trafficking by membrane lipids.J Neurochem 116:702–707. CrossRefMedline

    Gärtner S, Conzelmann E, Sandhoff K (1983) Ac-tivator protein for the degradation of globotri-aosylceramide by human alpha-galactosidase.J Biol Chem 258:12378–12385. Medline

    Giehl A, Lemm T, Bartelsen O, Sandhoff K, BlumeA (1999) Interaction of the GM2-activatorprotein with phospholipid-ganglioside bilayer

    membranes and with monolayers at the air-water interface. Eur J Biochem 261:650 – 658.Medline

    Graber D, Salvayre R, Levade T (1994) Accuratedifferentiation of neuronopathic and non-neuronopathic forms of Niemann-Pick dis-ease by evaluation of the effective residuallysosomal sphingomyelinase activity in intactcells. J Neurochem 63:1060 –1068. Medline

    Gravel R, Kaback MM, Proia RL, Sandhoff K, Su-zuki K, Suzuki K (2001) The GM2 gangli-osidoses. In: The metabolic and molecularbases of inherited diseases, Ed 8 (Scriver CR,Beaudet AL, Sly WS, Valle D, eds), pp 3827–3876. New York: McGraw-Hill.

    Hay ED (1989) Extracellular matrix, cell skele-tons, and embryonic development. Am J MedGenet 34:14 –29. CrossRef Medline

    Igisu H, Suzuki K (1984) Progressive accumula-tion of toxic metabolite in a genetic leukodys-trophy. Science 224:753–755. CrossRefMedline

    Jalanko A, Braulke T (2009) Neuronal ceroid li-pofuscinoses. Biochim Biophys Acta 1793:697–709. CrossRef Medline

    Jatzkewitz H, Sandhoff K (1963) On a biochem-ically special form of infantile amaurotic idi-ocy. Biochim Biophys Acta 70:354 –356.CrossRef Medline

    Jennemann R, Sandhoff R, Wang S, Kiss E,Gretz N, Zuliani C, Martin-Villalba A, JägerR, Schorle H, Kenzelmann M, BonrouhiM, Wiegandt H, Gröne HJ (2005) Cell-specific deletion of glucosylceramide syn-thase in brain leads to severe neural defectsafter birth. Proc Natl Acad Sci U S A 102:12459 –12464. CrossRef Medline

    Jennemann R, Sandhoff R, Langbein L, Kaden S, Ro-thermel U, Gallala H, Sandhoff K, Wiegandt H,Gröne HJ (2007) Integrity and barrier func-tion of the epidermis critically depend onglucosylceramide synthesis. J Biol Chem 282:3083–3094. Medline

    Jennemann R, Rothermel U, Wang S, Sandhoff R,Kaden S, Out R, van Berkel TJ, Aerts JM,Ghauharali K, Sticht C, Gröne HJ (2010)Hepatic glycosphingolipid deficiency andliver function in mice. Hepatology 51:1799 –1809. CrossRef Medline

    Jeyakumar M, Smith DA, Williams IM, Borja MC,Neville DC, Butters TD, Dwek RA, Platt FM(2004) NSAIDs increase survival in theSandhoff disease mouse: synergy withN-butyldeoxynojirimycin. Ann Neurol 56:642– 649. CrossRef Medline

    Jeyakumar M, Williams I, Smith D, Cox TM, PlattFM (2009) Critical role of iron in the patho-genesis of the murine gangliosidoses. Neuro-biol Dis 34:406 – 416. CrossRef Medline

    Kannagi R, Cochran NA, Ishigami F, Hakomori S,Andrews PW, Knowles BB, Solter D (1983)Stage-specific embryonic antigens (SSEA-3and -4) are epitopes of a unique globo-seriesganglioside isolated from human teratocarci-noma cells. EMBO J 2:2355–2361. Medline

    Kawai H, Allende ML, Wada R, Kono M, Sango K,Deng C, Miyakawa T, Crawley JN, Werth N,Bierfreund U, Sandhoff K, Proia RL (2001)Mice expressing only monosialogangliosideGM3 exhibit lethal audiogenic seizures. J BiolChem 276:6885– 6888. CrossRef Medline

    Keilani S, Lun Y, Stevens AC, Williams HN, Sjoberg

    ER, Khanna R, Valenzano KJ, Checler F, Bux-baum JD, Yanagisawa K, Lockhart DJ, WustmanBA, Gandy S (2012) Lysosomal dysfunction ina mouse model of Sandhoff disease leads to ac-cumulation of ganglioside-bound amyloid-betapeptide. J Neurosci 32:5223–5236. CrossRefMedline

    Kirkegaard T, Roth AG, Petersen NH, MahalkaAK, Olsen OD, Moilanen I, Zylicz A, KnudsenJ, Sandhoff K, Arenz C, Kinnunen PK, Ny-landsted J, Jäättelä M (2010) Hsp70 stabi-lizes lysosomes and reverts Niemann-Pickdisease-associated lysosomal pathology. Na-ture 463:549 –553. CrossRef Medline

    Klenk E (1937) Die Fettstoffe des Gehirnsbei Amaurotischer Idiotie und Niemann-Pick’scher Krankheit. Ber Ges Physiol96:659 – 660.

    Klenk E (1939) Niemann-Pick’sche Krankheitund Amaurotische Idiotie. Hoppe-Seyler’s ZPhysiol Chem 262:128 –143. CrossRef

    Kolter T, Sandhoff K (1999) Sphingolipids—their metabolic pathways and the pathobio-chemistry of neurodegenerative diseases.Angew Chem Int Ed Engl 38:1532–1568.CrossRef

    Kolter T, Sandhoff K (2005) Principles of lyso-somal membrane digestion: stimulation ofsphingolipid degradation by sphingolipid ac-tivator proteins and anionic lysosomal lipids.Annu Rev Cell Dev Biol 21:81–103. CrossRefMedline

    Kolter T, Sandhoff K (2006) Sphingolipid me-tabolism diseases. Biochim Biophys Acta1758:2057–2079. CrossRef Medline

    Kolter T, Sandhoff K (2010) Lysosomal degra-dation of membrane lipids. FEBS Lett 584:1700 –1712. CrossRef Medline

    Kolter T, Magin TM, Sandhoff K (2000)Biomolecule function: no reliable predictionfrom cell culture. Traffic 1:803– 804. CrossRefMedline

    Kolter T, Proia RL, Sandhoff K (2002) Combi-natorial ganglioside biosynthesis. J Biol Chem277:25859 –25862. CrossRef Medline

    Kudoh T, Velkoff MA, Wenger DA (1983) Up-take and metabolism of radioactively labeledsphingomyelin in cultured skin fibroblastsfrom controls and patients with Niemann-Pick disease and other lysosomal storage dis-eases. Biochim Biophys Acta 754:82–92.CrossRef Medline

    Kuhn R, Wiegandt H (1963) Die Konstitutionder Ganglio-N-tetraose und des GangliosidsGI. Chem Ber 96:866 – 880. CrossRef

    Kwon HJ, Abi-Mosleh L, Wang ML, DeisenhoferJ, Goldstein JL, Brown MS, Infante RE (2009)Structure of N-terminal domain of NPC1 re-veals distinct subdomains for binding andtransfer of cholesterol. Cell 137:1213–1224.CrossRef Medline

    Kytzia HJ, Sandhoff K (1985) Evidence fortwo different active sites on human beta-hexosaminidase A. Interaction of GM2 activatorprotein with beta-hexosaminidase A. J BiolChem 260:7568–7572. Medline

    Kytzia HJ, Hinrichs U, Maire I, Suzuki K, SandhoffK (1983) Variant of GM2-gangliosidosis withhexosaminidase A having a severely changedsubstrate specificity. EMBO J 2:1201–1205.Medline

    Kytzia HJ, Hinrichs U, Sandhoff K (1984) Diag-

    10206 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

    http://dx.doi.org/10.1159/000112332http://www.ncbi.nlm.nih.gov/pubmed/6421563http://dx.doi.org/10.1159/000112160http://www.ncbi.nlm.nih.gov/pubmed/1817024http://dx.doi.org/10.1074/jbc.274.16.11038http://www.ncbi.nlm.nih.gov/pubmed/10196186http://www.ncbi.nlm.nih.gov/pubmed/2470757http://dx.doi.org/10.1038/4801http://www.ncbi.nlm.nih.gov/pubmed/9883849http://dx.doi.org/10.1007/s10545-011-9409-2http://www.ncbi.nlm.nih.gov/pubmed/22033734http://dx.doi.org/10.1126/science.803227http://www.ncbi.nlm.nih.gov/pubmed/803227http://dx.doi.org/10.1016/j.tcb.2012.04.005http://www.ncbi.nlm.nih.gov/pubmed/22608991http://dx.doi.org/10.1038/ejhg.2012.202http://www.ncbi.nlm.nih.gov/pubmed/22990144http://dx.doi.org/10.1007/s11064-010-0337-6http://www.ncbi.nlm.nih.gov/pubmed/21136156http://dx.doi.org/10.1111/j.1471-4159.2010.07014.xhttp://www.ncbi.nlm.nih.gov/pubmed/21214551http://www.ncbi.nlm.nih.gov/pubmed/6313655http://www.ncbi.nlm.nih.gov/pubmed/10215880http://www.ncbi.nlm.nih.gov/pubmed/8051547http://dx.doi.org/10.1002/ajmg.1320340107http://www.ncbi.nlm.nih.gov/pubmed/2683777http://dx.doi.org/10.1126/science.6719111http://www.ncbi.nlm.nih.gov/pubmed/6719111http://dx.doi.org/10.1016/j.bbamcr.2008.11.004http://www.ncbi.nlm.nih.gov/pubmed/19084560http://dx.doi.org/10.1016/0006-3002(63)90764-9http://www.ncbi.nlm.nih.gov/pubmed/13957544http://dx.doi.org/10.1073/pnas.0500893102http://www.ncbi.nlm.nih.gov/pubmed/16109770http://www.ncbi.nlm.nih.gov/pubmed/17145749http://dx.doi.org/10.1002/hep.23545http://www.ncbi.nlm.nih.gov/pubmed/20432257http://dx.doi.org/10.1002/ana.20242http://www.ncbi.nlm.nih.gov/pubmed/15505823http://dx.doi.org/10.1016/j.nbd.2009.01.015http://www.ncbi.nlm.nih.gov/pubmed/19449457http://www.ncbi.nlm.nih.gov/pubmed/6141938http://dx.doi.org/10.1074/jbc.C000847200http://www.ncbi.nlm.nih.gov/pubmed/11133999http://dx.doi.org/10.1523/JNEUROSCI.4860-11.2012http://www.ncbi.nlm.nih.gov/pubmed/22496568http://dx.doi.org/10.1038/nature08710http://www.ncbi.nlm.nih.gov/pubmed/20111001http://dx.doi.org/10.1515/bchm2.1939.262.3-5.128http://dx.doi.org/10.1002/(SICI)1521-3773(19990601)38:113.0.CO%3B2-Uhttp://dx.doi.org/10.1146/annurev.cellbio.21.122303.120013http://www.ncbi.nlm.nih.gov/pubmed/16212488http://dx.doi.org/10.1016/j.bbamem.2006.05.027http://www.ncbi.nlm.nih.gov/pubmed/16854371http://dx.doi.org/10.1016/j.febslet.2009.10.021http://www.ncbi.nlm.nih.gov/pubmed/19836391http://dx.doi.org/10.1034/j.1600-0854.2000.011007.xhttp://www.ncbi.nlm.nih.gov/pubmed/11208070http://dx.doi.org/10.1074/jbc.R200001200http://www.ncbi.nlm.nih.gov/pubmed/12011101http://dx.doi.org/10.1016/0005-2760(83)90084-Xhttp://www.ncbi.nlm.nih.gov/pubmed/6626569http://dx.doi.org/10.1002/cber.19630960329http://dx.doi.org/10.1016/j.cell.2009.03.049http://www.ncbi.nlm.nih.gov/pubmed/19563754http://www.ncbi.nlm.nih.gov/pubmed/3158659http://www.ncbi.nlm.nih.gov/pubmed/6226523

  • nosis of infantile and juvenile forms of GM2gangliosidosis variant 0. Residual activities to-ward natural and different synthetic sub-strates. Hum Genet 67:414 – 418. CrossRefMedline

    Leinekugel P, Michel S, Conzelmann E, SandhoffK (1992) Quantitative correlation betweenthe residual activity of beta-hexosaminidase Aand arylsulfatase A and the severity of the re-sulting lysosomal storage disease. Hum Genet88:513–523. Medline

    Lemieux MJ, Mark BL, Cherney MM, Withers SG,Mahuran DJ, James MN (2006) Crystallo-graphic structure of human beta-hexosamini-dase A: interpretation of Tay-Sachs mutationsand loss of GM2 ganglioside hydrolysis. J MolBiol 359:913–929. CrossRef Medline

    Li SC, Sonnino S, Tettamanti G, Li YT (1988)Characterization of a nonspecific activatorprotein for the enzymatic hydrolysis of glyco-lipids. J Biol Chem 263:6588 – 6591. Medline

    Linke T, Wilkening G, Sadeghlar F, Mozcall H, Ber-nardo K, Schuchman E, Sandhoff K (2001) In-terfacial regulation of acid ceramidase activity.Stimulation of ceramide degradation by lyso-somal lipids and sphingolipid activator proteins.J Biol Chem 276:5760–5768. CrossRef Medline

    Lyon G, Adams RD, Kolodny EH (1996a) Early in-fantile progressive metabolic encephalopathies:clinical problems and diagnostic considerations.In: Neurology of hereditary metabolic diseasesof children, Ed 2 (Lyon G, Adams RD, KolodnyEH, eds), pp 45–123. New York: McGraw-Hill.

    Lyon G, Adams RD, Kolodny EH (1996b)Childhood and adolescent hereditary meta-bolic disorders. In: Neurology of hereditarymetabolic diseases of children Ed 2 (Lyon G,Adams RD, Kolodny EH, eds), pp 177–281.New York: McGraw-Hill.

    Maier T, Strater N, Schuette CG, Klingenstein R,Sandhoff K, Saenger W (2003) The X-ray crys-tal structure of human beta-hexosaminidase Bprovides new insights into Sandhoff disease.J Mol Biol 328:669–681. CrossRef Medline

    Martino S, Marconi P, Tancini B, Dolcetta D, DeAngelis MG, Montanucci P, Bregola G, Sand-hoff K, Bordignon C, Emiliani C, ManservigiR, Orlacchio A (2005) A direct gene transferstrategy via brain internal capsule reverses thebiochemical defect in Tay-Sachs disease. HumMol Genet 14:2113–2123. CrossRef Medline

    Matsuda J, Suzuki O, Oshima A, Yamamoto Y,Noguchi A, Takimoto K, Itoh M, Matsuzaki Y,Yasuda Y, Ogawa S, Sakata Y, Nanba E, HigakiK, Ogawa Y, Tominaga L, Ohno K, Iwasaki H,Watanabe H, Brady RO, Suzuki Y (2003)Chemical chaperone therapy for brain pathol-ogy in G(M1)-gangliosidosis. Proc Natl AcadSci U S A 100:15912–15917. CrossRefMedline

    Matsuoka K, Tamura T, Tsuji D, Dohzono Y, Kita-kaze K, Ohno K, Saito S, Sakuraba H, Itoh K(2011) Therapeutic potential of intracerebro-ventricular replacement of modified humanbeta-hexosaminidase B for GM2 gangliosidosis.Mol Ther 19:1017–1024. CrossRef Medline

    Mehl E, Jatzkewitz H (1964) Eine Cerebrosidsulfa-tase aus Schweineniere. Hoppe-Seylers ZPhysiol Chem 339:260–276. CrossRef Medline

    Möbius W, Herzog V, Sandhoff K, Schwarzmann G(1999) Intracellular distribution of a biotin-labeled ganglioside, GM1, by immunoelectron

    microscopy after endocytosis in fibroblasts.J Histochem Cytochem 47:1005–1014. CrossRefMedline

    Mraz W, Fischer G, Jatzkewitz H (1976) Lowmolecular weight proteins in secondary lyso-somes as activators of different sphingolipidhydrolases. FEBS Lett 67:104 –109. CrossRefMedline

    Myerowitz R, Lawson D, Mizukami H, Mi Y, TifftCJ, Proia RL (2002) Molecular pathophysi-ology in Tay-Sachs and Sandhoff diseases asrevealed by gene expression profiling. HumMol Genet 11:1343–1350. CrossRef Medline

    Neufeld EF, Muenzer J (2001) The Mucopoly-saccharidoses. In: The metabolic and molecu-lar bases of inherited disease, Ed 8 (Scriver CR,Beaudet AL, Sly WS, Valle D, eds), pp 3421–3452. New York: McGraw-Hill.

    Nilsson O, Månsson JE, Håkansson G, Svenner-holm L (1982) The occurrence of psychosineand other glycolipids in spleen and liver fromthe three major types of Gaucher’s disease.Biochim Biophys Acta 712:453– 463. CrossRefMedline

    Okada M, Itoh Mi M, Haraguchi M, Okajima T,Inoue M, Oishi H, Matsuda Y, Iwamoto T,Kawano T, Fukumoto S, Miyazaki H, Furu-kawa K, Aizawa S, Furukawa K (2002)b-series Ganglioside deficiency exhibits nodefinite changes in the neurogenesis and thesensitivity to Fas-mediated apoptosis but im-pairs regeneration of the lesioned hypoglossalnerve. J Biol Chem 277:1633–1636. CrossRefMedline

    Okada S, O’Brien JS (1968) Generalized gangli-osidosis: beta-galactosidase deficiency. Sci-ence 160:1002–1004. CrossRef Medline

    Okumiya T, Sakuraba H, Kase R, Sugiura T(2003) Imbalanced substrate specificity ofmutant beta-galactosidase in patients withMorquio B disease. Mol Genet Metab 78:51–58. CrossRef Medline

    Proia RL (2003) Glycosphingolipid functions:insights from engineered mouse models. Phi-los Trans R Soc Lond B Biol Sci 358:879 – 883.CrossRef Medline

    Pshezhetsky AV, Ashmarina M (2001) Lyso-somal multienzyme complex: biochemistry,genetics, and molecular pathophysiology.Prog Nucleic Acid Res Mol Biol 69:81–114.CrossRef Medline

    Purpura DP, Suzuki K (1976) Distortion of neu-ronal geometry and formation of aberrantsynapses in neuronal storage disease. BrainRes 116:1–21. CrossRef Medline

    Samuels S, Korey SR, Gonatas J, Terry RD, WeissM (1963) Studies in Tay-Sachs disease. IV.Membranous cytoplasmic bodies. J Neuro-path Exp Neurol 22:81–97. CrossRef Medline

    Sandhoff K (1977) The biochemistry of sphin-golipid storage diseases. Angew Chem Int EdEngl 16:273–285. CrossRef Medline

    Sandhoff K (2012) My journey into the world ofsphingolipids and sphingolipidoses. Proc JpnAcad Ser B Phys Biol Sci 88:554 –582.CrossRef Medline

    Sandhoff K, Jatkewitz H (1967) A particle-bound sialyl lactosidoceramide splittingmammalian sialidase. Biochim Biophys Acta141:442– 444. CrossRef Medline

    Sandhoff K, Kolter T (1995) Glykolipide derZelloberfläche-Biochemie ihres Abbaus.

    Naturwissenschaften 82:403– 413. CrossRefMedline

    Sandhoff K, Kolter T (2003) Biosynthesis anddegradation of mammalian glycosphingolip-ids. Philos Trans R Soc Lond B Biol Sci 358:847– 861. CrossRef Medline

    Sandhoff K, Jatzkewitz H, Peters G (1969) Dieinfantile amaurotische Idiotie und verwandteFormen als Gangliosid-Speicherkrankheiten.Naturwissenschaften 56:356 –362. CrossRefMedline

    Sandhoff K, Conzelmann E, Neufeld EF, KabackMM, Suzuki K (1989) The GM2 gangli-osidosis. In: The metabolic basis of inheriteddisease. (Scriver CR, Beaudet AL, Sly WS,Valle D, eds), pp 1807–1839. New York:McGraw-Hill.

    Sango K, Yamanaka S, Hoffmann A, Okuda Y,Grinberg A, Westphal H, McDonald MP,Crawley JN, Sandhoff K, Suzuki K, Proia RL(1995) Mouse models of Tay-Sachs andSandhoff diseases differ in neurologic pheno-type and ganglioside metabolism. Nat Genet11:170 –176. CrossRef Medline

    Sango K, McDonald MP, Crawley JN, MackML, Tifft CJ, Skop E, Starr CM, HoffmannA, Sandhoff K, Suzuki K, Proia RL (1996)Mice lacking both subunits of lysosomal�-hexosaminidase display gangliosidosisand mucopolysaccharidosis. Nat Genet 14:348 –352. CrossRef Medline

    Sargeant TJ, Drage DJ, Wang S, Apostolakis AA,Cox TM, Cachón-González MB (2012)Characterization of inducible models of Tay-Sachs and related disease. PLoS Genet8:e1002943. CrossRef Medline

    Schnaar RL (2010) Brain gangliosides in axon-myelin stability and axon regeneration. FEBSLett 584:1741–1747. CrossRef Medline

    Simpson MA, Cross H, Proukakis C, Priestman DA,Neville DC, Reinkensmeier G, Wang H, Wiz-nitzer M, Gurtz K, Verganelaki A, Pryde A, Pat-ton MA, Dwek RA, Butters TD, Platt FM,Crosby AH (2004) Infantile-onset symptom-atic epilepsy syndrome caused by a homozygousloss-of-function mutation of GM3 synthase. NatGenet 36:1225–1229. CrossRef Medline

    Stevens RL, Fluharty AL, Kihara H, Kaback MM,Shapiro LJ, Marsh B, Sandhoff K, Fischer G(1981) Cerebroside sulfatase activator defi-ciency induced metachromatic leukodystro-phy. Am J Hum Genet 33:900 –906. Medline

    Sun Y, Witte DP, Ran H, Zamzow M, Barnes S,Cheng H, Han X, Williams MT, Skelton MR,Vorhees CV, Grabowski GA (2008) Neuro-logical deficits and glycosphingolipid accu-mulation in saposin B deficient mice. HumMol Genet 17:2345–2356. CrossRef Medline

    Suzuki Y, Oshima A, Nanba E (2001)�-Galactosidase deficiency (�-galactosidosis):GM1 gangliosidosis and Morquio B disease. In:The metabolic and molecular bases of inheriteddiseases, Ed 8 (Scriver CR, Beaudet AL, Sly WS,Valle D, eds), pp 3755–3809. New York:McGraw-Hill.

    Suzuki Y, Ichinomiya S, Kurosawa M, Matsuda J,Ogawa S, Iida M, Kubo T, Tabe M, Itoh M, Hi-gaki K, Nanba E, Ohno K (2012) Therapeuticchaperone effect of N-octyl 4-epi-beta-valienamine on murine G(M1)-gangliosidosis.Mol Genet Metab 106:92–98. CrossRef Medline

    Tallaksen CM, Berg JE (2009) Miglustat therapy

    Sandhoff and Harzer • Gangliosides and Gangliosidoses J. Neurosci., June 19, 2013 • 33(25):10195–10208 • 10207

    http://dx.doi.org/10.1007/BF00291402http://www.ncbi.nlm.nih.gov/pubmed/6436167http://www.ncbi.nlm.nih.gov/pubmed/1348043http://dx.doi.org/10.1016/j.jmb.2006.04.004http://www.ncbi.nlm.nih.gov/pubmed/16698036http://www.ncbi.nlm.nih.gov/pubmed/3360793http://dx.doi.org/10.1074/jbc.M006846200http://www.ncbi.nlm.nih.gov/pubmed/11104761http://dx.doi.org/10.1016/S0022-2836(03)00311-5http://www.ncbi.nlm.nih.gov/pubmed/12706724http://dx.doi.org/10.1093/hmg/ddi216http://www.ncbi.nlm.nih.gov/pubmed/15961412http://dx.doi.org/10.1073/pnas.2536657100http://www.ncbi.nlm.nih.gov/pubmed/14676316http://dx.doi.org/10.1038/mt.2011.27http://www.ncbi.nlm.nih.gov/pubmed/21487393http://dx.doi.org/10.1515/bchm2.1964.339.1.260http://www.ncbi.nlm.nih.gov/pubmed/5829234http://dx.doi.org/10.1177/002215549904700804http://www.ncbi.nlm.nih.gov/pubmed/10424884http://dx.doi.org/10.1016/0014-5793(76)80879-4http://www.ncbi.nlm.nih.gov/pubmed/8337http://dx.doi.org/10.1093/hmg/11.11.1343http://www.ncbi.nlm.nih.gov/pubmed/12019216http://dx.doi.org/10.1016/0005-2760(82)90272-7http://www.ncbi.nlm.nih.gov/pubmed/7126619http://dx.doi.org/10.1074/jbc.C100395200http://www.ncbi.nlm.nih.gov/pubmed/11682464http://dx.doi.org/10.1126/science.160.3831.1002http://www.ncbi.nlm.nih.gov/pubmed/5647842http://dx.doi.org/10.1016/S1096-7192(02)00199-3http://www.ncbi.nlm.nih.gov/pubmed/12559848http://dx.doi.org/10.1098/rstb.2003.1268http://www.ncbi.nlm.nih.gov/pubmed/12803921http://dx.doi.org/10.1016/S0079-6603(01)69045-7http://www.ncbi.nlm.nih.gov/pubmed/11550799http://dx.doi.org/10.1016/0006-8993(76)90245-6http://www.ncbi.nlm.nih.gov/pubmed/824017http://dx.doi.org/10.1097/00005072-196301000-00005http://www.ncbi.nlm.nih.gov/pubmed/13976171http://dx.doi.org/10.1002/anie.197702733http://www.ncbi.nlm.nih.gov/pubmed/406814http://dx.doi.org/10.2183/pjab.88.554http://www.ncbi.nlm.nih.gov/pubmed/23229750http://dx.doi.org/10.1016/0304-4165(67)90124-9http://www.ncbi.nlm.nih.gov/pubmed/6048332http://dx.doi.org/10.1007/BF01133673http://www.ncbi.nlm.nih.gov/pubmed/7477413http://dx.doi.org/10.1098/rstb.2003.1265http://www.ncbi.nlm.nih.gov/pubmed/12803917http://dx.doi.org/10.1007/BF00596925http://www.ncbi.nlm.nih.gov/pubmed/5360899http://dx.doi.org/10.1038/ng1095-170http://www.ncbi.nlm.nih.gov/pubmed/7550345http://dx.doi.org/10.1038/ng1196-348http://www.ncbi.nlm.nih.gov/pubmed/8896570http://dx.doi.org/10.1371/journal.pgen.1002943http://www.ncbi.nlm.nih.gov/pubmed/23028353http://dx.doi.org/10.1016/j.febslet.2009.10.011http://www.ncbi.nlm.nih.gov/pubmed/19822144http://dx.doi.org/10.1038/ng1460http://www.ncbi.nlm.nih.gov/pubmed/15502825http://www.ncbi.nlm.nih.gov/pubmed/6119902http://dx.doi.org/10.1093/hmg/ddn135http://www.ncbi.nlm.nih.gov/pubmed/18480170http://dx.doi.org/10.1016/j.ymgme.2012.02.012http://www.ncbi.nlm.nih.gov/pubmed/22436580

  • in juvenile Sandhoff disease. J Inherit MetabDis 32 [Suppl 1]:S289 –S293. CrossRefMedline

    Tamboli IY, Hampel H, Tien NT, Tolksdorf K,Breiden B, Mathews PM, Saftig P, Sandhoff K,Walter J (2011) Sphingolipid storage affectsautophagic metabolism of the amyloid pre-cursor protein and promotes A� generation.J Neurosci 31:1837–1849. CrossRef Medline

    Taniike M, Yamanaka S, Proia RL, Langaman C,Bone-Turrentine T, Suzuki K (1995) Neuro-pathology of mice with targeted disruption ofHexa gene, a model of Tay-Sachs disease. ActaNeuropathol 89:296 –304. CrossRef Medline

    Tessitore A, del P Martin M, Sano R, Ma Y, Mann L,Ingrassia A, Laywell ED, Steindler DA, Hender-shot LM, d’Azzo A (2004) GM1-ganglioside-mediated activation of the unfolded proteinresponse causes neuronal death in a neurode-generative gangliosidosis. Mol Cell 15:753–766.CrossRef Medline

    Tropak MB, Reid SP, Guiral M, Withers SG,Mahuran D (2004) Pharmacological en-hancement of beta-hexosaminidase activityin fibroblasts from adult Tay-Sachs andSandhoff Patients. J Biol Chem 279:13478 –13487. CrossRef Medline

    Tsuji D, Akeboshi H, Matsuoka K, Yasuoka H,Miyasaki E, Kasahara Y, Kawashima I, ChibaY, Jigami Y, Taki T, Sakuraba H, Itoh K(2011) Highly phosphomannosylated en-zyme replacement therapy for GM2 gangli-osidosis. Ann Neurol 69:691–701. CrossRefMedline

    van Gelder CM, Vollebregt AA, Plug I, van derPloeg AT, Reuser AJ (2012) Treatment op-tions for lysosomal storage disorders: devel-oping insights. Expert Opin Pharmacother 13:2281–2299. CrossRef Medline

    Venugopalan P, Joshi SN (2002) Cardiac in-volvement in infantile Sandhoff disease. J Pae-diatr Child Health 38:98 –100. CrossRefMedline

    Vitner EB, Platt FM, Futerman AH (2010) Com-mon and uncommon pathogenic cascades inlysosomal storage diseases. J Biol Chem 285:20423–20427. CrossRef Medline

    Wada R, Tifft CJ, Proia RL (2000) Microglial ac-tivation precedes acute neurodegeneration inSandhoff disease and is suppressed by bonemarrow transplantation. Proc Natl Acad SciU S A 97:10954 –10959. CrossRef Medline

    Wekerle H (2005) Immune pathogenesis ofmultiple sclerosis. Neurol Sci 26 [Suppl 1]:S1–S2. Medline

    Wendeler M, Hoernschemeyer J, Hoffmann D,Kolter T, Schwarzmann G, Sandhoff K(2004a) Photoaffinity labelling of the humanGM2-activator protein. Eur J Biochem 271:614 – 627. CrossRef Medline

    Wendeler M, Hoernschemeyer J, John M, WerthN, Schoeniger M, Lemm T, Hartmann R, Kes-sler H, Sandhoff K (2004b) Expression of theGM2-activator protein in the methylotrophicyeast Pichia pastoris, purification, isotopic la-beling, and biophysical characterization. Pro-tein Expr Purif 34:147–157. CrossRef Medline

    Werth N, Schuette CG, Wilkening G, Lemm T,Sandhoff K (2001) Degradation of membrane-bound ganglioside GM2 by beta -hexosamini-dase A. Stimulation by GM2 activator proteinand lysosomal lipids. J Biol Chem 276:12685–12690. CrossRef Medline

    Wilken B, Dechent P, Hanefeld F, Frahm J (2008)Proton MRS of a child with Sandhoff diseasereveals elevated brain hexosamine. Eur J Pae-diatr Neurol 12:56 – 60. CrossRef Medline

    Wilkening G, Linke T, Sandhoff K (1998) Lyso-

    somal degradation on vesicular membranesurfaces. Enhanced glucosylceramide degra-dation by lysosomal anionic lipids and activa-tors. J Biol Chem 273:30271–30278. CrossRefMedline

    Wilkening G, Linke T, Uhlhorn-Dierks G, Sand-hoff K (2000) Degradation of membrane-bound ganglioside GM1. Stimulation bybis(monoacylglycero)phosphate and the acti-vator proteins SAP-B and GM2-AP. J BiolChem 275:35814 –35819. CrossRef Medline

    Wollert T, Hurley JH (2010) Molecular mecha-nism of multivesicular body biogenesis byESCRT complexes. Nature 464:864 – 869.CrossRef Medline

    Wortmann SB, Lefeber DJ, Dekomien G, Wil-lemsen MA, Wevers RA, Morava E (2009)Substrate deprivation therapy in juvenileSandhoff disease. J Inherit Metab Dis 32[Suppl 1]:S307–S311. CrossRef Medline

    Wright CS, Zhao Q, Rastinejad F (2003) Struc-tural Analysis of Lipid Complexes of GM2-Activator Protein. J Mol Biol 331:951–964.CrossRef Medline

    Xu YH, Barnes S, Sun Y, Grabowski GA (2010)Multi-system disorders of glycosphingolipidand ganglioside metabolism. J Lipid Res 51:1643–1675. CrossRef Medline

    Yamashita T, Wada R, Sasaki T, Deng C, Bier-freund U, Sandhoff K, Proia RL (1999) A vi-tal role for glycosphingolipid synthesis duringdevelopment and differentiation. Proc NatlAcad Sci U S A 96:9142–9147. CrossRefMedline

    Yu RK, Tsai YT, Ariga T, Yanagisawa M (2011)Structures, biosynthesis, and functions ofgangliosides–an overview. J Oleo Sci 60:537–544. CrossRef Medline

    10208 • J. Neurosci., June 19, 2013 • 33(25):10195–10208 Sandhoff and Harzer • Gangliosides and Gangliosidoses

    http://dx.doi.org/10.1007/s10545-009-1224-7http://www.ncbi.nlm.nih.gov/pubmed/19898953http://dx.doi.org/10.1523/JNEUROSCI.2954-10.2011http://www.ncbi.nlm.nih.gov/pubmed/21289194http://dx.doi.org/10.1007/BF00309622http://www.ncbi.nlm.nih.gov/pubmed/7610760http://dx.doi.org/10.1016/j.molcel.2004.08.029http://www.ncbi.nlm.nih.gov/pubmed/15350219http://dx.doi.org/10.1074/jbc.M308523200http://www.ncbi.nlm.nih.gov/pubmed/14724290http://dx.doi.org/10.1002/ana.22262http://www.ncbi.nlm.nih.gov/pubmed/21520232http://dx.doi.org/10.1517/14656566.2012.729039http://www.ncbi.nlm.nih.gov/pubmed/23009070http://dx.doi.org/10.1046/j.1440-1754.2002.00765.xhttp://www.ncbi.nlm.nih.gov/pubmed/11869411http://dx.doi.org/10.1074/jbc.R110.134452http://www.ncbi.nlm.nih.gov/pubmed/20430897http://dx.doi.org/10.1073/pnas.97.20.10954http://www.ncbi.nlm.nih.gov/pubmed/11005868http://www.ncbi.nlm.nih.gov/pubmed/15883678http://dx.doi.org/10.1111/j.1432-1033.2003.03964.xhttp://www.ncbi.nlm.nih.gov/pubmed/14728689http://dx.doi.org/10.1016/j.pep.2003.11.010http://www.ncbi.nlm.nih.gov/pubmed/14766311http://dx.doi.org/10.1074/jbc.M007970200http://www.ncbi.nlm.nih.gov/pubmed/11278374http://dx.doi.org/10.1016/j.ejpn.2007.05.008http://www.ncbi.nlm.nih.gov/pubmed/17625939http://dx.doi.org/10.1074/jbc.273.46.30271http://www.ncbi.nlm.nih.gov/pubmed/9804787http://dx.doi.org/10.1074/jbc.M006568200http://www.ncbi.nlm.nih.gov/pubmed/10942779http://dx.doi.org/10.1038/nature08849http://www.ncbi.nlm.nih.gov/pubmed/20305637http://dx.doi.org/10.1007/s10545-009-1261-2http://www.ncbi.nlm.nih.gov/pubmed/19898952http://dx.doi.org/10.1016/S0022-2836(03)00794-0http://www.ncbi.nlm.nih.gov/pubmed/12909021http://dx.doi.org/10.1194/jlr.R003996http://www.ncbi.nlm.nih.gov/pubmed/20211931http://dx.doi.org/10.1073/pnas.96.16.9142http://www.ncbi.nlm.nih.gov/pubmed/10430909http://dx.doi.org/10.5650/jos.60.537http://www.ncbi.nlm.nih.gov/pubmed/21937853

    Gangliosides and Gangliosidoses: Principles of Molecular and Metabolic PathogenesisIntroductionSummary outline of clinical symptomatologyPathway of ganglioside degradationOur view of the topology of lysosomal ganglioside digestionProtracted clinical forms of gangliosidoses and the threshold theoryMany proteins involved in ganglioside metabolism are nonspecificMembrane lipids are regulators of ganglioside catabolismTherapeutic approaches

    Gene therapyConclusions and outlookReferences