Abstract
Age-associated neurodegenerative diseases such as amyotrophic lateral sclerosis (ALS), Parkinson's disease (PD) and Alzheimer's disease (AD) are an unmet health need, with significant economic and societal implications, and an ever-increasing prevalence. Membrane lipid rafts (MLRs) are specialised plasma membrane microdomains that provide a platform for intracellular trafficking and signal transduction, particularly within neurons. Dysregulation of MLRs leads to disruption of neurotrophic signalling and excessive apoptosis which mirrors the final common pathway for neuronal death in ALS, PD and AD. Sphingomyelinase (SMase) and phospholipase (PL) enzymes process components of MLRs and therefore play central roles in MLR homeostasis and in neurotrophic signalling. We review the literature linking SMase and PL enzymes to ALS, AD and PD with particular attention to attractive therapeutic targets, where functional manipulation has been successful in preclinical studies. We propose that dysfunction of these enzymes is upstream in the pathogenesis of neurodegenerative diseases and to support this we provide new evidence that ALS risk genes are enriched with genes involved in ceramide metabolism (P=0.019, OR = 2.54, Fisher exact test). Ceramide is a product of SMase action upon sphingomyelin within MLRs, and it also has a role as a second messenger in intracellular signalling pathways important for neuronal survival. Genetic risk is necessarily upstream in a late age of onset disease such as ALS. We propose that manipulation of MLR structure and function should be a focus of future translational research seeking to ameliorate neurodegenerative disorders.
Keywords: apoptosis, enzyme activity, genetics, membrane dynamics, neurodegeneration, neurotrophic factors
Introduction
Membrane lipid rafts (MLRs) are specialised plasma membrane microdomains that are integral to the regulation of intracellular trafficking and signal transduction. MLRs contain high concentrations of four lipid classes: cholesterol, gangliosides, phospholipids and sphingolipids of which sphingomyelin is a subtype. In the brain, MLRs are present in neurons, astrocytes and microglia [1]. MLRs exist in two forms: either as flask-like invaginations termed caveolae (‘little caves’) or in a planar non-caveolar form [2]. In neurons, MLRs exist exclusively in the planar form and preferentially accumulate on somatic and axonal membranes. Neuronal MLRs are crucial for diverse functionality including neuronal adhesion, neuritogenesis, growth cone advancement, synapse formation and synapse maintenance [3–5]. In particular, MLRs modulate cell surface receptors that are positioned within their substance [6]. Consequently, pro-survival and pro-growth neuronal signalling pathways depend on the unique composition and regulation of lipids within the MLR, so much so that the majority of neurodegenerative diseases are linked with alterations in the biophysical properties of these microdomains [7,8]. Structural remodelling of MLRs is regulated by lipid-modifying enzymes [9]; certain types of these enzymes have been linked to neurodegeneration [10].
Neurotrophins are polypeptides that activate signalling pathways to promote the survival, development and function of neurons. The four most researched neurotrophins in mammals include nerve growth factor (NGF), brain-derived neurotrophic factor (BDNF), neurotrophin-3 (NT-3) and neurotrophin-4 (NT-4). Each of these neurotrophins can bind specifically to a tyrosine kinase (Trk) receptor, and all can bind to the low affinity p75 receptor [11], thus activating a downstream signalling cascade to drive the expression of pro-survival, pro-growth or pro-apoptotic genes [12]. These receptors are localised within MLRs and thus MLRs provide an essential platform for neurotrophin signalling pathways.
MLR function is linked to neurodegenerative diseases
By definition neurodegenerative diseases are associated with ageing, and it is notable that ageing is associated with dysfunction directly and indirectly linked to MLRs. For example, age-associated loss of cholesterol from MLR leads to impaired pre-synaptic vesicle fusion and diminished neurotransmission [1,13]. Indeed ageing is associated with a generalised change in the composition of neuronal MLR including sphingomyelin [14] and gangliosides [15].
Amyotrophic lateral sclerosis (ALS) is an incurable age-of-onset neurodegenerative disorder affecting 1–2 per 100,000 people worldwide [16]. ALS is characterised by the progressive degeneration of upper and lower motor neurons within the motor cortex, brainstem and spinal cord, which leads to paralysis and death usually by respiratory failure within 2–5 years of symptom onset [17]. ALS is multifactorial with a number of described genetic and environmental risk factors. More than 50 potentially causative or disease-modifying genes have been associated with ALS; however, pathogenic variants in SOD1, C9ORF72, FUS and TARDBP are the most common [18]. Although phenotypically indistinguishable from one another, 10% of cases are familial, usually with an autosomal dominant inheritance pattern, whilst the remaining 90% are sporadic, defined as having no family history of the disease [17].
Neuronal cytoplasmic TDP-43-positive inclusions are the hallmark pathology of ALS which correlate with neuronal loss [19]. Disruption of MLRs is a feature of ALS pathophysiology [20,21] and is associated with impaired neurotrophic signalling [22]. Abnormal neurotrophic signalling is a feature of ALS [23,24], and in particular, deficient neurotrophic signalling associated with increased vulnerability to neuronal injury [25–27] is proposed as a mechanism underlying motor neuron toxicity. Neurotrophic signalling is an important regulator of apoptosis, and excessive neuronal apoptosis has also been observed in ALS [28].
Parkinson's disease (PD) is a debilitating neurodegenerative disorder of the motor system affecting 1–2 per 1000 people [29]. PD is characterised by the loss of dopaminergic neurons in the substantia nigra pars compacta [30]. The hallmark pathology of PD is the formation of α-synuclein aggregates within neurons in the form of Lewy bodies and Lewy neurites [31]. PD can affect multiple body systems, often as a consequence of autonomic nervous system dysfunction. Therefore, symptoms often include dysphagia, delayed gastric emptying and constipation [32]. Similar to ALS, alterations in the composition of MLRs have been reported in PD [33]. α-Synuclein has been shown to associate specifically with MLRs and this association is required for the normal synaptic localization of α-synuclein [34]. Excessive neuronal apoptosis has also been observed in PD [35].
Alzheimer's disease (AD) is a progressive neurodegenerative disorder and the most common cause of dementia. There are currently 40–50 million people living with dementia worldwide, and this number is expected to triple by 2050 [36]. AD is characterised by two hallmark pathologies in the brain: (i) the formation of extracellular amyloid beta (Aβ) plaques and (ii) deposits of neurofibrillary tangles of hyperphosphorylated tau protein [37]. These neuropathological changes are accompanied by glial cell activation, release of pro-inflammatory mediators and neuronal death, leading to brain atrophy [38]. Much like ALS and PD, MLRs are strongly linked with AD pathophysiology. BACE1 and γ-secretase are two enzymes involved in the cleavage of the amyloid precursor protein (APP) to produce Aβ. Both of these enzymes are targeted to MLRs [39,40], which increases the efficiency of processing of APP bound directly to cholesterol molecules within each lipid raft [41]. As a result, MLR structure directly promotes the production of Aβ plaques [42]. Indeed, disruption of MLRs [43] and excessive neuronal apoptosis [44] have previously been observed in AD. Moreover, interventions that preserve or restore MLRs can mitigate AD-associated neurodegeneration [45].
For the purposes of this review, two classes of lipid-modifying enzymes important for MLR homeostasis will be discussed: phospholipases (PLs) and sphingomyelinases (SMases). We aim to summarise evidence linking the function of PLs and SMases to neurodegeneration via MLR maintenance. We will focus on the archetypal neurodegenerative diseases ALS, PD and AD. Homeostasis of other lipid subtypes within MLRs including gangliosides and cholesterol have been reviewed elsewhere [10,46]. We suggest that dysregulation of PL and SMase enzymes can cause disruption within MLRs leading to altered neurotrophin-receptor binding; these enzymes produce second messengers which act downstream of neurotrophin receptors to modify signal transduction within the neuron. Impaired neurotrophic signalling is a well-described mechanism in the context of neurodegeneration [12].
SMase dysregulation disrupts MLRs and impairs neurotrophic signalling
Sphingomyelin (SM) interacts with cholesterol to provide structural support for MLRs. Cholesterol is the ‘dynamic glue’ that holds MLRs together and most raft-associated proteins rely on cholesterol for their function [47]. SM expression is regulated by the sphingomyelin cycle where SM is hydrolysed to ceramide and phosphocholine by a small family of SMase enzymes; the reverse reaction is catalysed by SM synthase enzymes [48]. This mechanism is important for MLR homeostasis, but it also creates an environment to support efficient downstream neurotrophin signalling. SMases can be neutral (nSMase) or acidic (aSMase) depending on the optimal pH for their enzymatic activity [49]. Alterations in the activity of SMase enzymes operating within the sphingomyelin cycle have been frequently associated with neurodegeneration [50–52]. For example, inactivation of nSMase 2 correlates with the formation of TDP-43 aggregates in neuronal cells and exacerbates the disease phenotype in TDP-43 transgenic mice [53], and Aβ has been shown to directly activate SMase enzymes [54]. We argue that SMase dysregulation causes neurodegeneration via disruption of MLRs and a breakdown in the neurotrophic signalling cascade.
Ceramide is a product of SM metabolism that can act as a modulator of membrane structure or as a secondary messenger in intracellular signalling pathways [55] including signal transduction downstream of the p75 neurotrophin receptor [56–58]. Blocking ceramide production through inhibition of SMases can prevent NGF-induced apoptosis in hippocampal neurons after p75 activation i.e. downstream of receptor binding [59]. Ceramide also plays a key role in raft formation and growth [60,61] which has indirect consequences for activity of neurotrophin receptors including p75. Ceramide produced via activation of nSMases has been linked to neuronal apoptosis in SOD1-G93A ALS mice [62], and motor neurons over-expressing the ALS-associated SOD1-G93A protein are more susceptible to p75-induced apoptosis [63]. Given the complexity of this system, it is not surprising that the relationship between ceramide production and neurodegeneration is not linear: we have shown that ceramide can exacerbate neuronal toxicity. However, at low concentrations, ceramide can be neuroprotective and promote axonal development; for example, nSMase inhibition blocks the positive effects of the neurotrophic factor, NGF, on hippocampal neuron outgrowth [64].
Outside of p75 signalling, SMase activity has also been identified as a modulator of the other key neurotrophic signalling receptor subtype: Trk receptors. Binding of BDNF and NGF to Trk receptors promotes neuronal viability and is dependent on the presence of basal nSMase activity. Knockdown of nSMase in granule neurons and PC12 cells prevents neurotrophin-induced Akt phosphorylation [65]. Phosphorylation of Akt is an important rate-limiting step which protects cells from apoptosis and promotes cell survival [66].
nSMase enzymes have also been shown to influence the secretion of extracellular vesicles (EVs) from the plasma membrane [67,68]. Recently, a direct link between EVs and BDNF-dependent neurogenesis was described [69], which implicates nSMase enzymes in the regulation of neurotrophic signalling via modulation of EVs.
aSMase, encoded by the SMPD1 gene, is a key enzyme involved in sphingolipid metabolism and regulation of MLR assembly [70]. MLR scaffolding proteins have been shown to directly interact with p75 and Trk receptors [71], and amplify TrkB signalling [22]. Dysregulation of aSMase has been shown to cause destabilisation of MLRs [72], which could lead to the impairment of downstream neurotrophin signalling. Reduced aSMase enzymatic activity has been described in PD patients with SMPD1 mutations, and loss of aSMase expression correlates with increased α-synuclein levels in vitro [73]; indeed SMPD1 mutations are associated with increased risk for PD [51]. We have previously demonstrated that coding and non-coding mutations which reduce function of caveolin-1 disrupt MLRs and increase the risk for ALS [74].
Phospholipase dysregulation disrupts MLRs and impairs neurotrophic signalling
Phospholipases (PLs) catalyse the hydrolysis and cleavage of phospholipids [75] and therefore play a central role in plasma membrane structure and homeostasis which includes MLRs. Phospholipids form the membrane permeability barrier, but they also regulate membrane protein function and serve as second messengers in signal transduction pathways [76]. PLs consist of four large families (PLA, PLB, PLC and PLD), categorised by the position of the cleavage site on the phospholipid backbone [77]. As for SMases, PL activity is also an important source of secondary messengers and lipid signalling molecules, including free fatty acids, lysophospholipids, inositol 1,4,5-triphosphate (IP3), diacylglycerol (DAG) and phosphatidic acid (PA), which play an important role in neurotrophic signalling [78–80]. For example, altered expression of the second messenger atypical protein kinase C (aPKC), which is activated by phosphatidic acid that is generated by PLD [81], has been implicated in ALS [82].
Multiple studies have discovered that altered PL activity is associated with neurodegeneration [83–87] often via an effect on neurotrophic signalling. PLC-γ has been shown to be directly involved in regulating neurotrophic signalling, with roles in synaptic plasticity, neurite outgrowth, neurotransmission and neuronal excitability [88]. Similar to SMases, this activity is dependent upon p75 receptor signalling within MLRs. siRNA-mediated inhibition of PLC-γ activity abolishes p75 up-regulation stimulated by NGF [89]. Moreover, phosphatidylcholine-specific PLC (PC-PLC) induces apoptosis in response to glutamate toxicity in HT22 cells [90], which links MLR dysfunction and neurotrophic signalling to excitotoxicity. This is significant because excitotoxicity is proposed to be a central mechanism for ALS pathogenesis [91].
PL activity has been linked to other mechanisms associated with neurodegeneration although the link with MLRs is currently less clear. For example, studies in synaptic endings of rats have also shown that both PC-PLC and PLD1 promote increased activation of the lipid messenger, DAG, in response to iron-induced oxidative injury, highlighting a role for these lipid modifying enzymes in oxidative stress signalling [92,93]. cPLA2 signalling, via the activation of arachidonic acid (AA), mediates neuronal cell death in response to beta-amyloid peptide [94]. Finally, overexpression of wild-type α-synuclein in IMR-32 cells can cause inhibition of PLD1 signalling, leading to neurofilament loss and a reduction in cell viability [95].
Based on the biochemical, molecular and cellular evidence we have presented, we propose that SMase and PL enzymes play a central role in lipid biology and MLR homeostasis in particular, with consequences for neurotrophic signalling and neurodegeneration. A summary of the pathophysiological mechanisms linked to enzymatic dysregulation of SMases and PLs is provided in Figure 1.
SMase and PL enzymes are promising therapeutic targets
Given their prominent role in apoptosis and neurotrophic signalling pathways, SMase and PL enzymes have been explored as potential therapeutic targets for ALS, PD and AD. Despite promising results, the selective targeting of SMase and PL enzymes is still in the preclinical stage. We have already described the complexity of this system demonstrating that effective neuroprotection may require up-regulation or down-regulation depending on specific biological context. However, given that overactivation of SMase and PL enzymes are most often associated with neurodegeneration, therapeutic targeting tends to focus on down-regulation of enzyme activity (Table 1). The attraction of targeting MLRs rather than neurotrophic signalling directly is that physiological control is maintained which should reduce the potential for excessive signalling leading to uncontrolled growth and potentially carcinogenesis.
Table 1. SMase and PL enzymes are potential therapeutic targets in neurodegenerative disease.
Enzyme | Disorder | Disease associated change | Therapeutic target/Intervention |
---|---|---|---|
Phospholipases | |||
cPLA2 | ALS | Increased mRNA expression in familial ALS patients and in ALS mice [107] | COX-2 inhibition reduced cPLA2 immunoreactivity and preserves motor neurons in the spinal cord of SOD1-G93A transgenic mice [107] |
AD | Increased activation in neurons [97] | Genetic ablation of cPLA2 prevents cognitive decline and preserves neuronal integrity in AD mice [108] | |
PD | Increased expression in microglia [109] | cPLA2 -/- mice are more resistant to PD-associated neurotoxicity [110,111] | |
cPLA2α | ALS | Increased protein levels in sporadic ALS patients and in ALS mice [84,104] | CNS-specific reduction of cPLA2α reverses AD and ALS disease progression [104,112] |
AD | Increased immunoreactivity of cPLA2α and its transcript [113–115] | ||
PLCD1 | ALS | Increased gene expression of PLCD1 and increased protein levels of PLCδ1 in ALS mouse model [100] | Genetic ablation of PLCD1 improves survival in ALS mice [100] |
Sphingomyelinases | |||
aSMase | AD | Increased activity [50,97,116] | Genetic inhibition of aSMase ameliorates autophagic dysfunction in AD mice [50,99]. |
nSMase | AD | Increased activity [97] | Inhibition of nSMase decreases apoptosis and Aβ-induced cytotoxicity [97] |
Enzyme expression changes, alterations in activity, associated mechanisms, associated neurodegenerative disorder(s), potential as therapeutic target, and references are shown; AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; PD, Parkinson's disease.
Sphingomyelinases
In theory, aSMase inhibitors could be used to prevent ceramide-driven apoptosis in neurodegenerative disease. The therapeutic potential of SMase inhibitors has not been investigated in pre-clinical models of ALS and PD. Although, indirect alteration of SMase function through modulation of sphingosine-1-phosphate, a known inhibitor of SMase enzymes [96], is currently undergoing phase II clinical trials for the treatment of ALS (Fingolimod (FTY720); NCT01786174). Conversely, SMase inhibition has been actively explored in AD [70]. SMase inhibition reduces the production of Aβ [54,97], and SMase inhibitors such as GW4896 and 3-O-methylsphingomyelin have been proposed as potential AD therapies [98], but translation to therapeutic trials has been limited by failure to achieve efficient target engagement [70]. Functional inhibitors of aSMase enzymes (FIASMAs) are a large group of compounds licensed for medical use which could have significant potential in neurodegenerative disease [99].
Phospholipases
Knockdown of phospholipase C delta 1 (PLCD1), which encodes PLCδ1, in SOD1-G93A ALS mice delays symptom onset and prolongs survival [100]. This study also assessed nuclear shrinkage, a morphological change that occurs during apoptosis [100–102]. They found that motor neurons of PLCδ1−/−SOD1-G93A mice exhibited a reduction in nuclear shrinkage which would be consistent with an effect on neurotrophic signalling [100].
Cyclooxygenase-2 (COX-2) inhibitors have been shown to improve survival in the SOD1-G93A ALS mouse model [103] and to reduce expression of cytosolic PLA2 (cPLA2). Similarly, an antisense oligonucleotide against cPLA2α administered to SOD1-G93A mice ameliorates disease-associated elevation in levels of cPL2Aα protein, delays symptom onset and prolongs survival while preventing motor neuron loss [104].
Genes involved in MLR homeostasis are associated with neurodegeneration
We have recently developed a novel machine learning model called RefMap to increase the power of gene discovery by integrating motor neuron functional genomics with ALS genetics [105]. We examined the 690 candidate genes identified by RefMap to see which are linked with MLR homeostasis and SMase/PL function in particular. Twenty-two genes that met the filtering criteria include: ABCA1, ABCA2, ACER2, ANXA1, CERS5, GBA2, GRAMD1B, LPAR1, NAPEPLD, NTRK2, PLAA, PLPP6, S1PR3, SCARB1, SIGMAR1, SLC44A1, SPTAN1, TESK1, TMEM8B, TOR1A, TOR1B, TPD52. Information regarding their functional role in MLR homeostasis along with previous associations with neurodegenerative disease is provided in Table 2. We tested for enrichment of GO pathways associated with SMase, PL and MLR function within RefMap ALS genes and discovered that ‘ceramide metabolic process’ (GO:0006672) genes are significantly enriched within RefMap ALS genes (P=0.019, OR = 2.54, Fisher exact test). As a background we assumed the total set of genes expressed in iPSC-derived motor neurons (TPM>1, n=19,516) [89]. Overall, this suggests that dysfunction of MLR homeostasis and ceramide metabolism in particular may be upstream in the pathogenesis of ALS.
Table 2. MLR-associated ALS genes identified using RefMap.
RefMap ALS gene | Functional role in MLR homeostasis | Associated neurodegenerative disease | References |
---|---|---|---|
ABCA1 | Reduces formation of lipid raft domains | ALS | [117] |
ABCA2 | Regulates cholesterol and ceramide homeostasis | ALS, AD | [118] |
ACER2 | Catalyses the hydrolysis of ceramides to generate sphingosine | ALS | [119] |
ANXA1 | Facilitates MLR clustering; inhibitor of phospholipase A2 | ALS | [120] |
CERS5 | Synthesises c-16 ceramide, which is a pro-apoptotic ceramide | ALS | [121] |
GBA2 | Catalyses the conversion of glucosylceramide to free glucose and ceramide | ALS, HSP | [122] |
GRAMD1B | Regulates membrane cholesterol homeostasis | ALS | [123] |
LPAR1 | G-protein coupled receptor that localises to MLR | ALS, PD, AD | [124,125] |
NAPEPLD | A phospholipase D that cleaves N-acylphosphatidylethanolamines (NAPEs). NAPEs are involved in the consolidation of MLR structure | ALS, PD | [126,127] |
NTRK2 | Encodes neurotrophic tyrosine kinase receptor type 2, enriched within MLRs | ALS, AD | [128] |
PLAA | Phospholipase A2-activating protein present within MLRs; phospholipase A2 hydrolyses membrane phospholipids | ALS | [129] |
PLPP6 | Phospholipid phosphatase, involved in cholesterol synthesis | ALS | [130] |
S1PR3 | Sphingosine-1-phosphate receptor 3; sphingosine 1-phosphate (S1P) is a bioactive phospholipid growth factor that recruits proteins to MLRs | ALS | [131] |
SCARB1 | Localises to MLR and caveolin-1, involved in cholesterol homeostasis | ALS, AD | [132] |
SIGMAR1 | ER-resident protein with roles in MLR homeostasis | ALS, AD | [133–136] |
SLC44A1 | Encodes choline transporter-like protein 1, involved in phospholipid synthesis and MLR homeostasis | ALS | [137] |
SPTAN1 | Spectrin alpha chain, non-erythrocytic 1, associates with MLRs | ALS, HSP, PD, AD, ataxia | [138–140] |
TESK1 | Interacts with Spred1 which localizes in lipid raft/caveolae and inhibits ERK activation in collaboration with caveolin-1 | ALS | [141] |
TMEM8B | May have phospholipase A2 activity | ALS | [142] |
TOR1A / TOR1B | Encodes member of torsin protein family: Torsins are MLR-associated proteins important for lipid metabolism | ALS | [143] |
TPD52 | Interacts with the lipid raft protein MAL2 | ALS | [144] |
RefMap ALS gene ID, role within MLR homeostasis, associated neurodegenerative disorder(s) and references are shown; AD, Alzheimer's disease; ALS, amyotrophic lateral sclerosis; HSP, hereditary spastic paraplegia; PD, Parkinson's disease.
Conclusion and future directions
MLRs represent an organising centre for neurotrophic signalling in neurons. MLR disruption has already been described as an upstream cause of neurodegeneration [74]. Homeostasis of MLRs is tightly coupled to downstream signalling, particularly for Trk and p75 neurotrophin receptors which are positioned within the MLR. We have focused on SMases and PLs showing how activities of these enzymes are crucial modulators of neurotrophic signalling. Moreover, we have summarised evidence from the literature demonstrating that over activity of SMase and PL enzymes is linked to abnormal neurotrophic signalling and ultimately excessive apoptosis and neurodegeneration. A large number of studies provide specific examples linked to ALS, AD and PD.
An open question remains regarding the position of MLR dysfunction in the cascade of pathophysiology leading to neurodegenerative disease. It is possible that dysfunction of MLRs is simply a downstream consequence of neuronal toxicity. However, several arguments suggest that the role of SMases and PLs are more significant than that: firstly, these enzymes and their products have been linked to key mechanisms associated with these diseases including excitotoxicity, oxidative stress and protein aggregation. Second, manipulation of these enzymes can ameliorate the phenotype in key models of neurodegeneration. Finally, we show here evidence that proteins in these pathways are enriched with genetic mutations linked to ALS. Genetics are largely fixed at conception and so are, by definition, upstream in the cascade of pathogenesis leading to a late age of onset disease.
In this review we have focused on ALS, PD and AD, which are the most frequent neurodegenerative diseases. However, we acknowledge that this is far from an exhaustive list and although the evidence-base is strongest for these conditions, it is likely that other diseases are not represented only because key experiments have not yet been performed. In particular, FTD is on a spectrum with ALS and both diseases share key pathogenic mechanisms [106]; we suspect that much of the evidence we have presented for ALS would also apply to FTD.
We suggest that MLRs should be a key focus of translational research for neurodegenerative diseases. The potential is a means of manipulating neurotrophic signalling in a controlled and specific manner, which does not bypass physiological production of neurotrophins. Manipulation of SMases and PLs could improve pro-survival signalling efficiency by efficient organisation of neurotrophin receptors within the MLR, and simultaneously modulate second messenger transduction following receptor binding. The field awaits further validation using patient-derived cell models which are noticeably missing from the literature currently, to address this important opportunity.
Summary
Membrane lipid rafts (MLRs) are essential signalling platforms, and their dysregulation is strongly linked with neurodegenerative disease.
MLR homeostasis is regulated by sphingomyelinase (SMase) and phospholipase (PL) enzymes, and functional changes in these enzymes can cause impaired neurotrophic signalling and excessive apoptosis.
Genes involved in MLR homeostasis are enriched with genetic risk for amyotrophic lateral sclerosis (ALS).
SMase and PL enzymes are promising therapeutic targets for the treatment of neurodegenerative diseases.
Abbreviations
- AD
Alzheimer's disease
- ALS
amyotrophic lateral sclerosis
- DAG
diacylglycerol
- IP3
inositol trisphosphate
- JNK
c-Jun N-terminal kinase
- NAPE
N-acylphosphatidylethanolamine
- NGF
nerve growth factor
- NTR
neurotrophin receptor
- PD
Parkinson's disease
- PL
phospholipase
- PLC
phospholipase C
- PKC
protein kinase C
- MLR
membrane lipid raft
- SM
sphingomyelin
- SMase
sphingomyelinase
- TRK
tropomyosin-related kinase receptor
Competing Interests
The authors declare that there are no competing interests associated with the manuscript.
Funding
We acknowledge support from a Kingsland fellowship (to T.M.) and the NIHR Sheffield Biomedical Research Centre for Translational Neuroscience [grant number IS-BRC-1215-20017]. We also acknowledge support from the Wellcome Trust (to J.C.-K.) and NIHR [grant number NF-SI-0617-10077 (to P.J.S)]. Figure 1 was created with BioRender.com.
Author Contribution
All authors contributed equally to this work.
References
- 1.Egawa J., Pearn M.L., Lemkuil B.P., Patel P.M. and Head B.P. (2016) Membrane lipid rafts and neurobiology: age-related changes in membrane lipids and loss of neuronal function. J. Physiol. 594, 4565–4579 10.1113/JP270590 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 2.Pike L.J. (2003) Lipid rafts: bringing order to chaos. J. Lipid Res. 44, 655–667 10.1194/jlr.R200021-JLR200 [DOI] [PubMed] [Google Scholar]
- 3.Head B.P., Patel H.H. and Insel P.A. (2014) Interaction of membrane/lipid rafts with the cytoskeleton: impact on signaling and function: membrane/lipid rafts, mediators of cytoskeletal arrangement and cell signaling. Biochim. Biophys. Acta 1838, 532–545 10.1016/j.bbamem.2013.07.018 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4.Suzuki T., Zhang J., Miyazawa S., Liu Q., Farzan M.R. and Yao W.-D. (2011) Association of membrane rafts and postsynaptic density: proteomics, biochemical, and ultrastructural analyses. J. Neurochem. 119, 64–77 10.1111/j.1471-4159.2011.07404.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Whitehead S.N., Gangaraju S., Aylsworth A. and Hou S.T. (2012) Membrane raft disruption results in neuritic retraction prior to neuronal death in cortical neurons. Biosci. Trends 6, 183–191 10.5582/bst.2012.v6.4.183 [DOI] [PubMed] [Google Scholar]
- 6.Jana A., Hogan E.L. and Pahan K. (2009) Ceramide and neurodegeneration: susceptibility of neurons and oligodendrocytes to cell damage and death. J. Neurol. Sci. 278, 5–15 10.1016/j.jns.2008.12.010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Allen J.A., Halverson-Tamboli R.A. and Rasenick M.M. (2007) Lipid raft microdomains and neurotransmitter signalling. Nat. Rev. Neurosci. 8, 128–140 10.1038/nrn2059 [DOI] [PubMed] [Google Scholar]
- 8.Hussain G., Wang J., Rasul A., Anwar H., Imran A., Qasim M.et al. (2019) Role of cholesterol and sphingolipids in brain development and neurological diseases. Lipids Health Dis. 18, 26 10.1186/s12944-019-0965-z [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Sezgin E., Levental I., Mayor S. and Eggeling C. (2017) The mystery of membrane organization: composition, regulation and roles of lipid rafts. Nat. Rev. Mol. Cell Biol. 18, 361–374 10.1038/nrm.2017.16 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Moll T., Shaw P.J. and Cooper-Knock J. (2019) Disrupted glycosylation of lipids and proteins is a cause of neurodegeneration. Brain. 143, 1332–1340 10.1093/brain/awz358 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Huang E.J. and Reichardt L.F. (2001) Neurotrophins: roles in neuronal development and function. Annu. Rev. Neurosci. 24, 677–736 10.1146/annurev.neuro.24.1.677 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Mitre M., Mariga A. and Chao M.V. (2017) Neurotrophin signalling: novel insights into mechanisms and pathophysiology. Clin. Sci. 131, 13–23 10.1042/CS20160044 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ledesma M.D., Martin M.G. and Dotti C.G. (2012) Lipid changes in the aged brain: effect on synaptic function and neuronal survival. Prog. Lipid Res. 51, 23–35 10.1016/j.plipres.2011.11.004 [DOI] [PubMed] [Google Scholar]
- 14.Trovò L., Van Veldhoven P.P., Martín M.G. and Dotti C.G. (2011) Sphingomyelin upregulation in mature neurons contributes to TrkB activity by Rac1 endocytosis. J. Cell Sci. 124, 1308–1315 10.1242/jcs.078766 [DOI] [PubMed] [Google Scholar]
- 15.Posse de Chaves E. and Sipione S. (2010) Sphingolipids and gangliosides of the nervous system in membrane function and dysfunction. FEBS Lett. 584, 1748–1759 10.1016/j.febslet.2009.12.010 [DOI] [PubMed] [Google Scholar]
- 16.Ingre C., Roos P.M., Piehl F., Kamel F. and Fang F. (2015) Risk factors for amyotrophic lateral sclerosis. Clin. Epidemiol. 7, 181–193 10.2147/CLEP.S37505 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Renton A.E., Chiò A. and Traynor B.J. (2014) State of play in amyotrophic lateral sclerosis genetics. Nat. Neurosci. 17, 17–23 10.1038/nn.3584 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Mejzini R., Flynn L.L., Pitout I.L., Fletcher S., Wilton S.D. and Akkari P.A. (2019) ALS genetics, mechanisms, and therapeutics: where are we now? Front. Neurosci. 13, 1310 10.3389/fnins.2019.01310 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Neumann M., Sampathu D.M., Kwong L.K., Truax A.C., Micsenyi M.C., Chou T.T.et al. (2006) Ubiquitinated TDP-43 in frontotemporal lobar degeneration and amyotrophic lateral sclerosis. Science 314, 130–133 10.1126/science.1134108 [DOI] [PubMed] [Google Scholar]
- 20.Antonini A., Caioli S., Saba L., Vindigni G., Biocca S., Canu N.et al. (2018) Membrane cholesterol depletion in cortical neurons highlights altered NMDA receptor functionality in a mouse model of amyotrophic lateral sclerosis. Biochim. Biophys. Acta Mol. Basis Dis. 1864, 509–519 10.1016/j.bbadis.2017.11.008 [DOI] [PubMed] [Google Scholar]
- 21.Zhai J., Ström A.-L., Kilty R., Venkatakrishnan P., White J., Everson W.V.et al. (2009) Proteomic characterization of lipid raft proteins in amyotrophic lateral sclerosis mouse spinal cord. FEBS J. 276, 3308–3323 10.1111/j.1742-4658.2009.07057.x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Sawada A., Wang S., Jian M., Leem J., Wackerbarth J., Egawa J.et al. (2019) Neuron‐targeted caveolin‐1 improves neuromuscular function and extends survival in SOD1 G93A mice. FASEB J. 33, 7545–7554 10.1096/fj.201802652RR [DOI] [PMC free article] [PubMed] [Google Scholar]
- 23.Mutoh T., Sobue G., Hamano T., Kuriyama M., Hirayama M., Yamamoto M.et al. (2000) Decreased phosphorylation levels of TrkB neurotrophin receptor in the spinal cords from patients with amyotrophic lateral sclerosis. Neurochem. Res. 25, 239–245 10.1023/A:1007575504321 [DOI] [PubMed] [Google Scholar]
- 24.Turner B.J., Murray S.S., Piccenna L.G., Lopes E.C., Kilpatrick T.J. and Cheema S.S. (2004) Effect of p75 neurotrophin receptor antagonist on disease progression in transgenic amyotrophic lateral sclerosis mice. J. Neurosci. Res. 78, 193–199 10.1002/jnr.20256 [DOI] [PubMed] [Google Scholar]
- 25.Bemelmans A.-P., Husson I., Jaquet M., Mallet J., Kosofsky B.E. and Gressens P. (2006) Lentiviral-mediated gene transfer of brain-derived neurotrophic factor is neuroprotective in a mouse model of neonatal excitotoxic challenge. J. Neurosci. Res. 83, 50–60 10.1002/jnr.20704 [DOI] [PubMed] [Google Scholar]
- 26.Ghavami S., Shojaei S., Yeganeh B., Ande S.R., Jangamreddy J.R., Mehrpour M.et al. (2014) Autophagy and apoptosis dysfunction in neurodegenerative disorders. Prog. Neurobiol. 112, 24–49 10.1016/j.pneurobio.2013.10.004 [DOI] [PubMed] [Google Scholar]
- 27.Kowiański P., Lietzau G., Czuba E., Waśkow M., Steliga A. and Moryś J. (2018) BDNF: a key factor with multipotent impact on brain signaling and synaptic plasticity. Cell. Mol. Neurobiol. 38, 579–593 10.1007/s10571-017-0510-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Shaw P.J. (2006) Molecular and cellular pathways of neurodegeneration in motor neurone disease. Neurosci. Neurologists 143–179 10.1142/9781860948961_0006 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Tysnes O.-B. and Storstein A. (2017) Epidemiology of Parkinson's disease. J. Neural Transm. 124, 901–905 10.1007/s00702-017-1686-y [DOI] [PubMed] [Google Scholar]
- 30.Goedert M., Spillantini M.G., Del Tredici K. and Braak H. (2013) 100 years of Lewy pathology. Nat. Rev. Neurol. 9, 13–24 10.1038/nrneurol.2012.242 [DOI] [PubMed] [Google Scholar]
- 31.Gómez-Benito M., Granado N., García-Sanz P., Michel A., Dumoulin M. and Moratalla R. (2020) Modeling Parkinson's disease with the alpha-synuclein protein. Front. Pharmacol. 11, 10.3389/fphar.2020.00356 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Cersosimo M.G. and Benarroch E.E. (2008) Neural control of the gastrointestinal tract: implications for Parkinson's disease. Mov. Disord. 23, 1065–1075 10.1002/mds.22051 [DOI] [PubMed] [Google Scholar]
- 33.Fabelo N., Martín V., Santpere G., Marín R., Torrent L., Ferrer I.et al. (2011) Severe alterations in lipid composition of frontal cortex lipid rafts from Parkinson's disease and incidental Parkinson's disease. Mol. Med. 17, 1107–1118 10.2119/molmed.2011.00119 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34.Fortin D.L., Troyer M.D., Nakamura K., Kubo S.-I., Anthony M.D. and Edwards R.H. (2004) Lipid rafts mediate the synaptic localization of α-synuclein. J. Neurosci. 24, 6715–6723 10.1523/JNEUROSCI.1594-04.2004 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Lev N., Melamed E. and Offen D. (2003) Apoptosis and Parkinson's disease. Prog. Neuropsychopharmacol. Biol. Psychiatry 27, 245–250 10.1016/S0278-5846(03)00019-8 [DOI] [PubMed] [Google Scholar]
- 36.Nichols E., Szoeke C.E.I., Vollset S.E., Abbasi N., Abd-Allah F., Abdela J.et al. (2019) Global, regional, and national burden of Alzheimer's disease and other dementias, 1990-2016: a systematic analysis for the Global Burden of Disease Study 2016. Lancet Neurol. 18, 88–106 10.1016/S1474-4422(18)30403-4 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Kang S., Lee Y.H. and Lee J.E. (2017) Metabolism-centric overview of the pathogenesis of Alzheimer's disease. Yonsei Med. J. 58, 479–488 10.3349/ymj.2017.58.3.479 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38.Shabir O., Moll T.A., Matuszyk M.M., Eyre B., Dake M.D., Berwick J.et al. (2020) Preclinical models of disease and multimorbidity with focus upon cardiovascular disease and dementia. Mech. Ageing Dev. 192, 111361 10.1016/j.mad.2020.111361 [DOI] [PubMed] [Google Scholar]
- 39.Hattori C., Asai M., Onishi H., Sasagawa N., Hashimoto Y., Saido T.C.et al. (2006) BACE1 interacts with lipid raft proteins. J. Neurosci. Res. 84, 912–917 10.1002/jnr.20981 [DOI] [PubMed] [Google Scholar]
- 40.Vetrivel K.S. and Thinakaran G. (2010) Membrane rafts in Alzheimer's disease beta-amyloid production. Biochim. Biophys. Acta 1801, 860–867 10.1016/j.bbalip.2010.03.007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41.Beel A.J., Sakakura M., Barrett P.J. and Sanders C.R. (2010) Direct binding of cholesterol to the amyloid precursor protein: an important interaction in lipid-Alzheimer's disease relationships? Biochim. Biophys. Acta 1801, 975–982 10.1016/j.bbalip.2010.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42.Rushworth J.V. and Hooper N.M. (2010) Lipid rafts: linking Alzheimer's amyloid-β production, aggregation, and toxicity at neuronal membranes. Int. J. Alzheimer's Dis. 2011, 603052 10.4061/2011/603052 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 43.Chadwick W., Brenneman R., Martin B. and Maudsley S. (2010) Complex and multidimensional lipid raft alterations in a murine model of Alzheimer's disease. Int. J. Alzheimers Dis. 2010, 604792 10.4061/2010/604792 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Niikura T., Tajima H. and Kita Y. (2006) Neuronal cell death in Alzheimer's disease and a neuroprotective factor, humanin. Curr. Neuropharmacol. 4, 139–147 10.2174/157015906776359577 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Wang S., Leem J.S., Podvin S., Hook V., Kleschevnikov N., Savchenko P.et al. (2021) Synapsin-caveolin-1 gene therapy preserves neuronal and synaptic morphology and prevents neurodegeneration in a mouse model of AD. Mol. Ther. Methods Clin. Dev. 21, 434–450 10.1016/j.omtm.2021.03.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Mesa-Herrera F., Taoro-González L., Valdés-Baizabal C., Diaz M. and Marín R. (2019) Lipid and lipid raft alteration in aging and neurodegenerative diseases: a window for the development of new biomarkers. Int. J. Mol. Sci. 20, 3810 10.3390/ijms20153810 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Simons K. and Ehehalt R. (2002) Cholesterol, lipid rafts, and disease. J. Clin. Invest. 110, 597–603 10.1172/JCI0216390 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 48.Adada M., Luberto C. and Canals D. (2016) Inhibitors of the sphingomyelin cycle: sphingomyelin synthases and sphingomyelinases. Chem. Phys. Lipids. 197, 45–59 10.1016/j.chemphyslip.2015.07.008 [DOI] [PubMed] [Google Scholar]
- 49.Goñi F.M. and Alonso A. (2002) Sphingomyelinases: enzymology and membrane activity. FEBS Lett. 531, 38–46 10.1016/S0014-5793(02)03482-8 [DOI] [PubMed] [Google Scholar]
- 50.Lee J.K., Jin H.K., Park M.H., Kim B.-R., Lee P.H., Nakauchi H.et al. (2014) Acid sphingomyelinase modulates the autophagic process by controlling lysosomal biogenesis in Alzheimer's disease. J. Exp. Med. 211, 1551–1570 10.1084/jem.20132451 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Gan-Or Z., Ozelius L.J., Bar-Shira A., Saunders-Pullman R., Mirelman A., Kornreich R.et al. (2013) The p.L302P mutation in the lysosomal enzyme gene SMPD1 is a risk factor for Parkinson disease. Neurology 80, 1606–1610 10.1212/WNL.0b013e31828f180e [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Ong W.-Y., Herr D.R., Farooqui T., Ling E.-A. and Farooqui A.A. (2015) Role of sphingomyelinases in neurological disorders. Expert Opin. Ther. Targets 19, 1725–1742 10.1517/14728222.2015.1071794 [DOI] [PubMed] [Google Scholar]
- 53.Iguchi Y., Eid L., Parent M., Soucy G., Bareil C., Riku Y.et al. (2016) Exosome secretion is a key pathway for clearance of pathological TDP-43. Brain 139, 3187–3201 10.1093/brain/aww237 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Grimm M.O.W., Grimm H.S., Pätzold A.J., Zinser E.G., Halonen R., Duering M.et al. (2005) Regulation of cholesterol and sphingomyelin metabolism by amyloid-β and presenilin. Nat. Cell Biol. 7, 1118–1123 10.1038/ncb1313 [DOI] [PubMed] [Google Scholar]
- 55.van Blitterswijk W.J., van der Luit A.H., Veldman R.J., Verheij M. and Borst J. (2003) Ceramide: second messenger or modulator of membrane structure and dynamics? Biochem. J. 369, 199–211 10.1042/bj20021528 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Pettus B.J., Chalfant C.E. and Hannun Y.A. (2002) Ceramide in apoptosis: an overview and current perspectives. Biochim. Biophys. Acta (BBA) - Mol. Cell Biol. Lipids 1585, 114–125 10.1016/S1388-1981(02)00331-1 [DOI] [PubMed] [Google Scholar]
- 57.Dobrowsky R.T., Jenkins G.M. and Hannun Y.A. (1995) Neurotrophins induce sphingomyelin hydrolysis: modulation by co-expression of p75NTR with Trk receptors (*). J. Biol. Chem. 270, 22135–22142 10.1074/jbc.270.38.22135 [DOI] [PubMed] [Google Scholar]
- 58.Dobrowsky R.T., Werner M.H., Castellino A.M., Chao M.V. and Hannun Y.A. (1994) Activation of the sphingomyelin cycle through the low-affinity neurotrophin receptor. Science 265, 1596–1599 10.1126/science.8079174 [DOI] [PubMed] [Google Scholar]
- 59.Brann A.B., Tcherpakov M. and Williams I.M. (2002) Nerve growth factor-induced p75-mediated death of cultured hippocampal neurons is age-dependent and transduced through ceramide generated by neutral sphingomyelinase. J. Biol. Chem. 277, 9812–9818 10.1074/jbc.M109862200 [DOI] [PubMed] [Google Scholar]
- 60.Zeidan Y.H. and Hannun Y.A. (2010) The acid sphingomyelinase/ceramide pathway: biomedical significance and mechanisms of regulation. Curr. Mol. Med. 10, 454–466 10.2174/156652410791608225 [DOI] [PubMed] [Google Scholar]
- 61.Gulbins E. and Grassmé H. (2002) Ceramide and cell death receptor clustering. Biochim. Biophys. Acta 1585, 139–145 10.1016/S1388-1981(02)00334-7 [DOI] [PubMed] [Google Scholar]
- 62.Cutler R.G., Pedersen W.A., Camandola S., Rothstein J.D. and Mattson M.P. (2002) Evidence that accumulation of ceramides and cholesterol esters mediates oxidative stress-induced death of motor neurons in amyotrophic lateral sclerosis. Ann. Neurol. 52, 448–457 10.1002/ana.10312 [DOI] [PubMed] [Google Scholar]
- 63.Pehar M., Vargas M.R., Robinson K.M., Cassina P., Diaz-Amarilla P.J., Hagen T.M.et al. (2007) Mitochondrial superoxide production and nuclear factor erythroid 2-related factor 2 activation in p75 neurotrophin receptor-induced motor neuron apoptosis. J. Neurosci. 27, 7777–7785 10.1523/JNEUROSCI.0823-07.2007 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64.Brann A.B., Scott R., Neuberger Y., Abulafia D., Boldin S., Fainzilber M.et al. (1999) Ceramide signaling downstream of the p75 neurotrophin receptor mediates the effects of nerve growth factor on outgrowth of cultured hippocampal neurons. J. Neurosci. 19, 8199–8206 10.1523/JNEUROSCI.19-19-08199.1999 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Candalija A., Cubí R., Ortega A., Aguilera J. and Gil C. (2014) Trk receptors need neutral sphingomyelinase activity to promote cell viability. FEBS Lett. 588, 167–174 10.1016/j.febslet.2013.11.032 [DOI] [PubMed] [Google Scholar]
- 66.Manning B.D. and Toker A. (2017) AKT/PKB signaling: navigating the network. Cell 169, 381–405 10.1016/j.cell.2017.04.001 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 67.Menck K., Sönmezer C., Worst T.S., Schulz M., Dihazi G.H., Streit F.et al. (2017) Neutral sphingomyelinases control extracellular vesicles budding from the plasma membrane. J. Extracell. Vesicles 6, 1378056 10.1080/20013078.2017.1378056 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Skotland T., Sagini K., Sandvig K. and Llorente A. (2020) An emerging focus on lipids in extracellular vesicles. Adv. Drug Deliv. Rev. 159, 308–321 10.1016/j.addr.2020.03.002 [DOI] [PubMed] [Google Scholar]
- 69.Antoniou A., Auderset L., Kaurani L., Fischer A. and Schneider A. (2021) Neuronal extracellular vesicles mediate BDNF-dependent dendritogenesis and synapse maturation via microRNAs. bioRxiv 10.1101/2021.05.11.443606 [DOI] [Google Scholar]
- 70.Park M.H., Jin H.K. and Bae J.-S. (2020) Potential therapeutic target for aging and age-related neurodegenerative diseases: the role of acid sphingomyelinase. Exp. Mol. Med. 52, 380–389 10.1038/s12276-020-0399-8 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Bilderback T.R., Gazula V.R., Lisanti M.P. and Dobrowsky R.T. (1999) Caveolin interacts with Trk A and p75NTR and regulates neurotrophin signaling pathways. J. Biol Chem. 274, 257–263 10.1074/jbc.274.1.257 [DOI] [PubMed] [Google Scholar]
- 72.Park M.H., Jin H.K. and Bae J.-S. (2019) Acid sphingomyelinase-mediated blood-brain barrier disruption in aging. BMB Rep. 52, 111–112 10.5483/BMBRep.2019.52.2.033 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Alcalay R.N., Mallett V., Vanderperre B., Tavassoly O., Dauvilliers Y., Wu R.Y.J.et al. (2019) SMPD1 mutations, activity, and α-synuclein accumulation in Parkinson's disease. Mov. Disord. 34, 526–535 10.1002/mds.27642 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Cooper-Knock J., Zhang S., Kenna K.P., Moll T., Franklin J., Allen S.et al. (2020) Rare variant burden analysis within enhancers identifies CAV1 as a new ALS risk gene, Cell Rep. 33, 108456 10.1016/j.celrep.2020.108456 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Dennis E.A. (2015) Introduction to thematic review series: phospholipases: central role in lipid signaling and disease. J. Lipid Res. 56, 1245–1247 10.1194/jlr.E061101 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Dowhan W. (2017) Understanding phospholipid function: why are there so many lipids? J. Biol. Chem. 292, 10755–10766 10.1074/jbc.X117.794891 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Filkin S.Y., Lipkin A.V. and Fedorov A.N. (2020) Phospholipase superfamily: structure, functions, and biotechnological applications. Biochemistry (Moscow) 85, 177–195 10.1134/S0006297920140096 [DOI] [PubMed] [Google Scholar]
- 78.Canossa M. (2001) Regulated secretion of neurotrophins by metabotropic glutamate group I (mGluRI) and Trk receptor activation is mediated via phospholipase C signalling pathways. EMBO J. 20, 1640–1650 10.1093/emboj/20.7.1640 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Kashyap M.P., Roberts C., Waseem M. and Tyagi P. (2018) Drug targets in neurotrophin signaling in the central and peripheral nervous system. Mol. Neurobiol. 55, 6939–6955 10.1007/s12035-018-0885-3 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Joensuu M., Wallis T.P., Saber S.H. and Meunier F.A. (2020) Phospholipases in neuronal function: a role in learning and memory? J. Neurochem. 153, 300–333 10.1111/jnc.14918 [DOI] [PubMed] [Google Scholar]
- 81.Farese R.V. and Sajan M.P. (2010) Metabolic functions of atypical protein kinase C: “good” and “bad” as defined by nutritional status. Am. J. Physiol.-Endocrinol. Metab. 298, E385–E394 10.1152/ajpendo.00608.2009 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Tury A., Tolentino K. and Zou Y. (2014) Altered expression of atypical PKC and Ryk in the spinal cord of a mouse model of amyotrophic lateral sclerosis. Dev. Neurobiol. 74, 839–850 10.1002/dneu.22137 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Jin J.-K., Ahn B.-H., Na Y.-J., Kim J.-I., Kim Y.-S., Choi E.-K.et al. (2007) Phospholipase D1 is associated with amyloid precursor protein in Alzheimer's disease. Neurobiol. Aging 28, 1015–1027 10.1016/j.neurobiolaging.2006.05.022 [DOI] [PubMed] [Google Scholar]
- 84.Shibata N., Kakita A., Takahashi H., Ihara Y., Nobukuni K., Fujimura H.et al. (2010) Increased expression and activation of cytosolic phospholipase A2 in the spinal cord of patients with sporadic amyotrophic lateral sclerosis. Acta Neuropathol. (Berl) 119, 345–354 10.1007/s00401-009-0636-7 [DOI] [PubMed] [Google Scholar]
- 85.Oliveira T.G., Chan R.B., Tian H., Laredo M., Shui G., Staniszewski A.et al. (2010) Phospholipase D2 Ablation Ameliorates Alzheimer's Disease-Linked Synaptic Dysfunction and Cognitive Deficits. J. Neurosci. 30, 16419–16428 10.1523/JNEUROSCI.3317-10.2010 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 86.Bennett S.A.L., Valenzuela N., Xu H., Franko B., Fai S. and Figeys D. (2013) Using neurolipidomics to identify phospholipid mediators of synaptic (dys)function in Alzheimer's disease. Front. Physiol. 4, 168 10.3389/fphys.2013.00168 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 87.Cruchaga C., Karch C.M., Jin S.C., Benitez B.A., Cai Y., Guerreiro R.et al. (2014) Rare coding variants in the phospholipase D3 gene confer risk for Alzheimer's disease. Nature 505, 550–554 10.1038/nature12825 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88.Jang H.-J., Yang Y.R., Kim J.K., Choi J.H., Seo Y.-K., Lee Y.H.et al. (2013) Phospholipase C-γ1 involved in brain disorders. Adv. Biol. Regul. 53, 51–62 10.1016/j.jbior.2012.09.008 [DOI] [PubMed] [Google Scholar]
- 89.Rankin S.L., Guy C.S., Rahimtula M. and Mearow K.M. (2008) Neurotrophin-induced upregulation of p75NTR via a protein kinase C-delta-dependent mechanism. Brain Res. 1217, 10–24 10.1016/j.brainres.2008.03.076 [DOI] [PubMed] [Google Scholar]
- 90.Li Y., Maher P. and Schubert D. (1998) Phosphatidylcholine-specific phospholipase C regulates glutamate-induced nerve cell death. Proc. Natl. Acad. Sci. 95, 7748–7753 10.1073/pnas.95.13.7748 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Van Den Bosch L., Van Damme P., Bogaert E. and Robberecht W. (2006) The role of excitotoxicity in the pathogenesis of amyotrophic lateral sclerosis. Biochim. Biophys. Acta 1762, 1068–1082 10.1016/j.bbadis.2006.05.002 [DOI] [PubMed] [Google Scholar]
- 92.Mateos M.V., Giusto N.M. and Salvador G.A. (2012) Distinctive roles of PLD signaling elicited by oxidative stress in synaptic endings from adult and aged rats. Biochim. Biophys. Acta (BBA) - Mol. Cell Res. 1823, 2136–2148 10.1016/j.bbamcr.2012.09.005 [DOI] [PubMed] [Google Scholar]
- 93.Mateos M.V., Uranga R.M., Salvador G.A. and Giusto N.M. (2008) Activation of phosphatidylcholine signalling during oxidative stress in synaptic endings. Neurochem. Int. 53, 199–206 10.1016/j.neuint.2008.07.005 [DOI] [PubMed] [Google Scholar]
- 94.Kriem B., Sponne I., Fifre A., Malaplate-Armand C., Lozac'h-Pillot K., Koziel V.et al. (2005) Cytosolic phospholipase A2 mediates neuronal apoptosis induced by soluble oligomers of the amyloid-beta peptide. FASEB J. 19, 85–87 10.1096/fj.04-1807fje [DOI] [PubMed] [Google Scholar]
- 95.Conde M.A., Alza N.P., Iglesias González P.A., Scodelaro Bilbao P.G., Campos S.S., Uranga R.M.et al. (2018) Phospholipase D1 downregulation by α-synuclein: implications for neurodegeneration in Parkinson's disease. Biochim. Biophys. Acta (BBA) - Mol. Cell Biol. Lipids 1863, 639–650 10.1016/j.bbalip.2018.03.006 [DOI] [PubMed] [Google Scholar]
- 96.Gómez-Muñoz A., Kong J., Salh B. and Steinbrecher U.P. (2003) Sphingosine-1-phosphate inhibits acid sphingomyelinase and blocks apoptosis in macrophages. FEBS Lett. 539, 56–60 10.1016/S0014-5793(03)00197-2 [DOI] [PubMed] [Google Scholar]
- 97.Malaplate-Armand C., Florent-Béchard S., Youssef I., Koziel V., Sponne I., Kriem B.et al. (2006) Soluble oligomers of amyloid-β peptide induce neuronal apoptosis by activating a cPLA2-dependent sphingomyelinase-ceramide pathway. Neurobiol. Dis. 23, 178–189 10.1016/j.nbd.2006.02.010 [DOI] [PubMed] [Google Scholar]
- 98.Grimm M.O.W., Grimm H.S. and Hartmann T. (2007) Amyloid beta as a regulator of lipid homeostasis. Trends Mol. Med. 13, 337–344 10.1016/j.molmed.2007.06.004 [DOI] [PubMed] [Google Scholar]
- 99.Kornhuber J., Muehlbacher M., Trapp S., Pechmann S., Friedl A., Reichel M.et al. (2011) Identification of novel functional inhibitors of acid sphingomyelinase. PLoS ONE 6, e23852 10.1371/journal.pone.0023852 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Staats K.A., Van Helleputte L., Jones A.R., Bento-Abreu A., Van Hoecke A., Shatunov A.et al. (2013) Genetic ablation of phospholipase C delta 1 increases survival in SOD1(G93A) mice. Neurobiol. Dis. 60, 11–17 10.1016/j.nbd.2013.08.006 [DOI] [PubMed] [Google Scholar]
- 101.Takadera T. and Ohyashiki T. (2007) Caspase-dependent apoptosis induced by calcineurin inhibitors was prevented by glycogen synthase kinase-3 inhibitors in cultured rat cortical cells. Brain Res. 1133, 20–26 10.1016/j.brainres.2006.11.037 [DOI] [PubMed] [Google Scholar]
- 102.He B., Lu N. and Zhou Z. (2009) Cellular and nuclear degradation during apoptosis. Curr. Opin. Cell Biol. 21, 900–912 10.1016/j.ceb.2009.08.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Drachman D.B., Frank K., Dykes-Hoberg M., Teismann P., Almer G., Przedborski S.et al. (2002) Cyclooxygenase 2 inhibition protects motor neurons and prolongs survival in a transgenic mouse model of ALS. Ann. Neurol. 52, 771–778 10.1002/ana.10374 [DOI] [PubMed] [Google Scholar]
- 104.Solomonov Y., Hadad N. and Levy R. (2016) Reduction of cytosolic phospholipase A2α upregulation delays the onset of symptoms in SOD1G93A mouse model of amyotrophic lateral sclerosis. J. Neuroinflammation 13, 134 10.1186/s12974-016-0602-y [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Zhang S., Cooper-Knock J., Weimer A.K., Shi M., Moll T., Harvey C.et al. (2021) Genome-wide identification of the genetic basis of amyotrophic lateral sclerosis. bioRxiv 10.1101/2020.11.14.382606 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Achi E.Y. and Rudnicki S.A. (2012) ALS and frontotemporal dysfunction: a review. Neurol. Res. Int. 2012, 806306 10.1155/2012/806306 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107.Kiaei M., Kipiani K., Petri S., Choi D.-K., Chen J., Calingasan N.Y.et al. (2005) Integrative role of cPLA with COX-2 and the effect of non-steriodal anti-inflammatory drugs in a transgenic mouse model of amyotrophic lateral sclerosis. J. Neurochem. 93, 403–411 10.1111/j.1471-4159.2005.03024.x [DOI] [PubMed] [Google Scholar]
- 108.Desbène C., Malaplate-Armand C., Youssef I., Garcia P., Stenger C., Sauvée M.et al. (2012) Critical role of cPLA2 in Aβ oligomer-induced neurodegeneration and memory deficit. Neurobiol. Aging 33, 1123.e17–1129.e17 10.1016/j.neurobiolaging.2011.11.008 [DOI] [PubMed] [Google Scholar]
- 109.Chuang D.Y., Simonyi A., Kotzbauer P.T., Gu Z. and Sun G.Y. (2015) Cytosolic phospholipase A2 plays a crucial role in ROS/NO signaling during microglial activation through the lipoxygenase pathway. J. Neuroinflammation 12, 199 10.1186/s12974-015-0419-0 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Klivenyi P., Beal M.F., Ferrante R.J., Andreassen O.A., Wermer M., Chin M.R.et al. (1998) Mice deficient in group IV cytosolic phospholipase A2 are resistant to MPTP neurotoxicity. J. Neurochem. 71, 2634–2637 10.1046/j.1471-4159.1998.71062634.x [DOI] [PubMed] [Google Scholar]
- 111.Bonventre J.V. (1999) The 85-kD cytosolic phospholipase A2 knockout mouse: a new tool for physiology and cell biology. J. Am. Soc. Nephrol. 10, 404–412 10.1681/ASN.V102404 [DOI] [PubMed] [Google Scholar]
- 112.Sanchez-Mejia R.O., Newman J.W., Toh S., Yu G.-Q., Zhou Y., Halabisky B.et al. (2008) Phospholipase A2 reduction ameliorates cognitive deficits in a mouse model of Alzheimer's disease. Nat. Neurosci. 11, 1311–1318 10.1038/nn.2213 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Clemens J.A., Stephenson D.T., Barry Smalstig E., Roberts E.F., Johnstone E.M., Sharp J.D.et al. (1996) Reactive glia express cytosolic phospholipase A2 after transient global forebrain ischemia in the rat. Stroke 27, 527–535 10.1161/01.STR.27.3.527 [DOI] [PubMed] [Google Scholar]
- 114.Stephenson D., Rash K., Smalstig B., Roberts E., Johnstone E., Sharp J.et al. (1999) Cytosolic phospholipase A2 is induced in reactive glia following different forms of neurodegeneration. Glia 27, 110–128 [DOI] [PubMed] [Google Scholar]
- 115.Sagy-Bross C., Hadad N. and Levy R. (2013) Cytosolic phospholipase A2α upregulation mediates apoptotic neuronal death induced by aggregated amyloid-β peptide1-42. Neurochem. Int. 63, 541–550 10.1016/j.neuint.2013.09.007 [DOI] [PubMed] [Google Scholar]
- 116.Filippov V., Song M.A., Zhang K., Vinters H.V., Tung S., Kirsch W.M.et al. (2012) Increased ceramide in brains with Alzheimer's and other neurodegenerative diseases. J. Alzheimers Dis. 29, 537–547 10.3233/JAD-2011-111202 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 117.Landry Y.D., Denis M., Nandi S., Bell S., Vaughan A.M. and Zha X. (2006) ATP-binding cassette transporter A1 expression disrupts raft membrane microdomains through its ATPase-related functions. J. Biol. Chem. 281, 36091–36101 10.1074/jbc.M602247200 [DOI] [PubMed] [Google Scholar]
- 118.Wu A., Wojtowicz K., Savary S., Hamon Y. and Trombik T. (2020) Do ABC transporters regulate plasma membrane organization? Cell. Mol. Biol. Lett. 25, 37 10.1186/s11658-020-00224-x [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Mao C. and Obeid L.M. (2008) Ceramidases: regulators of cellular responses mediated by ceramide, sphingosine, and sphingosine-1-phosphate. Biochim. Biophys. Acta 1781, 424–434 10.1016/j.bbalip.2008.06.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Liu N.-K., Zhang Y.P., Han S., Pei J., Xu L.Y., Lu P.-H.et al. (2007) Annexin A1 reduces inflammatory reaction and tissue damage through inhibition of phospholipase A2 activation in adult rats following spinal cord injury. J. Neuropathol. Exp. Neurol. 66, 932–943 10.1097/nen.0b013e3181567d59 [DOI] [PubMed] [Google Scholar]
- 121.Levy M. and Futerman A.H. (2010) Mammalian ceramide synthases. IUBMB Life 62, 347–356 10.1002/iub.319 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Martin E., Schüle R., Smets K., Rastetter A., Boukhris A., Loureiro J.L.et al. (2013) Loss of function of glucocerebrosidase GBA2 is responsible for motor neuron defects in hereditary spastic paraplegia. Am. J. Hum. Genet. 92, 238–244 10.1016/j.ajhg.2012.11.021 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Höglinger D., Burgoyne T., Sanchez-Heras E., Hartwig P., Colaco A., Newton J.et al. (2019) NPC1 regulates ER contacts with endocytic organelles to mediate cholesterol egress. Nat Commun. 10, 1–14 10.1038/s41467-019-12152-2 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Yang X.-Y., Zhao E.Y., Zhuang W.-X., Sun F.-X., Han H.-L., Han H.-R.et al. (2015) LPA signaling is required for dopaminergic neuron development and is reduced through low expression of the LPA1 receptor in a 6-OHDA lesion model of Parkinson's disease. Neurol. Sci. 36, 2027–2033 10.1007/s10072-015-2295-x [DOI] [PubMed] [Google Scholar]
- 125.Xiang H., Lu Y., Shao M. and Wu T. (2020) Lysophosphatidic acid receptors: biochemical and clinical implications in different diseases. J. Cancer 11, 3519–3535 10.7150/jca.41841 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126.Li X.-M., Ramakrishnan M., Brockman H.L., Brown R.E. and Swamy M.J. (2002) N-myristoylated phosphatidylethanolamine: interfacial behavior and interaction with cholesterol. Langmuir 18, 231–238 10.1021/la010937t [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Palese F., Pontis S., Realini N. and Piomelli D. (2019) A protective role for N-acylphosphatidylethanolamine phospholipase D in 6-OHDA-induced neurodegeneration. Sci. Rep. 9, 15927 10.1038/s41598-019-51799-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Chen Z., Simmons M.S., Perry R.T., Wiener H.W., Harrell L.E. and Go R.C.P. (2008) Genetic association of neurotrophic tyrosine kinase receptor type 2 (NTRK2) With Alzheimer's disease. Am. J. Med. Genet. B Neuropsychiatr. Genet. 147, 363–369 10.1002/ajmg.b.30607 [DOI] [PubMed] [Google Scholar]
- 129.Hall E.A., Nahorski M.S., Murray L.M., Shaheen R., Perkins E., Dissanayake K.N.et al. (2017) PLAA mutations cause a lethal infantile epileptic encephalopathy by disrupting ubiquitin-mediated endolysosomal degradation of synaptic proteins. Am. J. Hum. Genet. 100, 706–724 10.1016/j.ajhg.2017.03.008 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.He Y., Cui X., Lin Y., Wang Y., Wu D. and Fang Y. (2021) Using elevated cholesterol synthesis as a prognostic marker in Wilms' tumor: a bioinformatic analysis. Biomed. Res. Int. 2021, 8826286 10.1155/2021/8826286 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Zhao J., Singleton P.A., Brown M.E., Dudek S.M. and Garcia J.G.N. (2009) Phosphotyrosine protein dynamics in cell membrane rafts of sphingosine-1-phosphate-stimulated human endothelium: role in barrier enhancement. Cell. Signal. 21, 1945–1960 10.1016/j.cellsig.2009.09.002 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Thanopoulou K., Fragkouli A., Stylianopoulou F. and Georgopoulos S. (2010) Scavenger receptor class B type I (SR-BI) regulates perivascular macrophages and modifies amyloid pathology in an Alzheimer mouse model. Proc. Natl. Acad. Sci. U.S.A. 107, 20816–20821 10.1073/pnas.1005888107 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Al-Saif A., Al-Mohanna F. and Bohlega S. (2011) A mutation in sigma-1 receptor causes juvenile amyotrophic lateral sclerosis. Ann. Neurol. 70, 913–919 10.1002/ana.22534 [DOI] [PubMed] [Google Scholar]
- 134.Vollrath J.T., Sechi A., Dreser A., Katona I., Wiemuth D., Vervoorts J.et al. (2014) Loss of function of the ALS protein SigR1 leads to ER pathology associated with defective autophagy and lipid raft disturbances. Cell Death Dis. 5, e1290 10.1038/cddis.2014.243 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Huang Y., Zheng L., Halliday G., Dobson-Stone C., Wang Y., Tang H.-D.et al. (2011) Genetic polymorphisms in sigma-1 receptor and apolipoprotein E interact to influence the severity of Alzheimer's disease. Curr. Alzheimer Res. 8, 765–770 10.2174/156720511797633232 [DOI] [PubMed] [Google Scholar]
- 136.Penke B., Fulop L., Szucs M. and Frecska E. (2018) The role of Sigma-1 receptor, an intracellular chaperone in neurodegenerative diseases. Curr. Neuropharmacol. 16, 97–116 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Schenkel L.C., Singh R.K., Michel V., Zeisel S.H., da Costa K.-A., Johnson A.R.et al. (2015) Mechanism of choline deficiency and membrane alteration in postural orthostatic tachycardia syndrome primary skin fibroblasts. FASEB J. 29, 1663–1675 10.1096/fj.14-258566 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Grzybek M., Chorzalska A., Bok E., Hryniewicz-Jankowska A., Czogalla A., Diakowski W.et al. (2006) Spectrin-phospholipid interactions: existence of multiple kinds of binding sites? Chem. Phys. Lipids. 141, 133–141 10.1016/j.chemphyslip.2006.02.008 [DOI] [PubMed] [Google Scholar]
- 139.Leveille E., Estiar M.A., Krohn L., Spiegelman D., Dionne-Laporte A., Dupré N.et al. (2019) SPTAN1 variants as a potential cause for autosomal recessive hereditary spastic paraplegia. J. Hum. Genet. 64, 1145–1151 10.1038/s10038-019-0669-2 [DOI] [PubMed] [Google Scholar]
- 140.Miazek A., Zalas M., Skrzymowska J., Bogin B.A., Grzymajło K., Goszczynski T.M.et al. (2021) Age-dependent ataxia and neurodegeneration caused by an αII spectrin mutation with impaired regulation of its calpain sensitivity. Sci. Rep. 11, 7312 10.1038/s41598-021-86470-1 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Nonami A., Taketomi T., Kimura A., Saeki K., Takaki H., Sanada T.et al. (2005) The Sprouty-related protein, Spred-1, localizes in a lipid raft/caveola and inhibits ERK activation in collaboration with caveolin-1. Genes Cells 10, 887–895 10.1111/j.1365-2443.2005.00886.x [DOI] [PubMed] [Google Scholar]
- 142.Lee G.-H., Fujita M., Nakanishi H., Miyata H., Ikawa M., Maeda Y.et al. (2020) PGAP6, a GPI-specific phospholipase A2, has narrow substrate specificity against GPI-anchored proteins. J. Biol. Chem. 295, 14501–14509 10.1074/jbc.RA120.014643 [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Grillet M., Dominguez Gonzalez B., Sicart A., Pöttler M., Cascalho A., Billion K.et al. (2016) Torsins are essential regulators of cellular lipid metabolism. Dev. Cell. 38, 235–247 10.1016/j.devcel.2016.06.017 [DOI] [PubMed] [Google Scholar]
- 144.Wilson S.H., Bailey A.M., Nourse C.R., Mattei M.G. and Byrne J.A. (2001) Identification of MAL2, a novel member of the mal proteolipid family, though interactions with TPD52-like proteins in the yeast two-hybrid system. Genomics 76, 81–88 10.1006/geno.2001.6610 [DOI] [PubMed] [Google Scholar]