Motor neuron diseases (MNDs) include a broad group of diseases in which neurodegeneration mainly affects upper and/or lower motor neurons (MNs). Although the involvement of specific MNs, symptoms, age of onset, and progression differ in MNDs, the main pathogenic mechanism common to most MNDs is represented by proteostasis alteration and proteotoxicity. This pathomechanism may be directly related to mutations in genes encoding proteins involved in the protein quality control system, particularly the autophagy-lysosomal pathway (ALP). Alternatively, proteostasis alteration can be caused by aberrant proteins that tend to misfold and to aggregate, two related processes that, over time, cannot be properly handled by the ALP. Here, we summarize the main ALP features, focusing on different routes utilized to deliver substrates to the lysosome and how the various ALP pathways intersect with the intracellular trafficking of membranes and vesicles. Next, we provide an overview of the mutated genes that have been found associated with MNDs, how these gene products are involved in different steps of ALP and related processes. Finally, we discuss how autophagy can be considered a valid therapeutic target for MNDs treatment focusing on traditional autophagy modulators and on emerging approaches to overcome their limitations.

Motor neuron diseases (MNDs) are a large class of neurodegenerative diseases (NDs) primarily affecting neurons that control the skeletal muscle fibers. These neurons comprise upper motor neurons (MNs) localized in the motor cortex and lower MNs localized in the bulbar regions or in the anterior horns of the spinal cord. Other cell types surrounding (e.g. glial and microglial cells) or connecting to (e.g. sensory neurons, interneurons, muscle cells, etc.) MNs may contribute to disease onset and progression, and ultimately to paralysis and death of the patients. These neural systems are governed by a complex architecture, thus various MN types are differentially and selectively involved in these disorders, leading to multifaceted clinical manifestations that considerably vary among affected individuals.

To simplify, MNDs can be roughly classified into three major categories that reflect both the clinical signs and the type of MNs affected: (i) with lower MNs involvement (e.g. spinal muscular atrophy (SMA), spinobulbar muscular atrophy (SBMA or Kennedy's disease), progressive muscular atrophy (PMA), pseudobulbar palsy (in the bulbar region), monomelic amyotrophy (MMA), lethal congenital contracture syndrome (LCCS), etc.), (ii) with upper MNs involvement (e.g. primary lateral sclerosis (PLS) and hereditary spastic paraplegias (HSP), etc.), and (iii) with combined upper and lower MNs involvement (e.g. amyotrophic lateral sclerosis (ALS), etc.) [1–3]. Overall, motor manifestations depend on which MNs are affected: lower MN loss results in muscle weakness, and upper MN loss in spasticity; mixed clinical signs associate initially to upper and lower MN loss, but when the disease progresses weakness and flaccid paralysis prevail over spasticity. Noteworthily, this basic classification does not fully reflect the existence of mixed forms, as, for example, ALS with frontotemporal dementia (FTD) [4].

No cure is available for most of these MNDs. Only for specific forms of SMA some highly innovative genetic approaches have recently proved to be therapeutically efficient [5–7].

The molecular bases of these MNDs may considerably differ and, while specific inherited (familial) forms are characterized by mutations in genes encoding proteins involved in a variety of intracellular and extracellular biological functions, some MNDs occur in a sporadic manner, making it very difficult to determine the causes of MN loss. Nevertheless, sporadic forms often display a dysregulation of the same proteins that have been identified mutated in familial forms of MNDs (see the case of sporadic and familial ALS) [8]. Intriguingly, many of these proteins and their mutated counterparts act in the neuronal response to proteotoxic stresses, which includes the protein quality control (PQC) system and particularly the autophagy-lysosomal pathway (ALP). ALP is essential to degrade damaged organelles or toxic protein aggregates and inclusions capable to alter cellular homeostasis causing MN death. This review will thus focus on the role of ALP in MNDs.

The term ‘autophagy' refers to various cellular processes involved in proteins or organelles clearance via the lysosomal system. Autophagy eliminates unused, excessive, and/or defective macromolecules (either endogenous or exogenous) and recycles their components for other biosynthetic pathways (Figure 1). Autophagy takes place in the cell cytoplasm, and it is an essential arm of the PQC and RNA quality control system, which works together with the proteasome in a finely tuned equilibrium. The fate of a given substrate is regulated by a plethora of chaperone proteins, that define how substrates will be cleared from cells. Autophagy dysregulations cause several acute and chronic human diseases and promote the aging process; these findings have stimulated the search for autophagy modulators as potential therapeutic tools to efficiently counteract them [9–11].

Autophagy is a complex system, in which the delivery of cargoes to lysosomes for degradation follows different routes named (i) macroautophagy (usually referred to as autophagy), (ii) microautophagy, and (iii) chaperone-mediated autophagy (CMA).

Macroautophagy implies that cargoes are sequestered by ‘omegasomes' released by the endoplasmic reticulum (ER) or other membranous compartments (e.g. late endosomes, Golgi apparatus, or plasma membrane). Omegasomes release double-membrane structures to enclose substrates generating the autophagosomes. This process is tightly regulated and controlled by the autophagy-related proteins (named ATGs) which orchestrate autophagosome assembly, trafficking, and fusion to lysosomes [10,12]. The type of cargo determines whether macroautophagy occurs ‘in bulk' (i.e. cytoplasm portions engulfed into autophagosomes), or ‘selectively', thanks to specific autophagy receptors (e.g. sequestosome 1 (SQSTM1/p62), optineurin (OPTN), etc.) that bridge cargoes to the autophagosome membranes. This is enabled by the presence of an LC3-interacting region (LIR) in most of the autophagy receptors. The LIR allows them to interact with members of the LC3/GABA Type A Receptor-Associated Protein (GABARAP) family (e.g. the microtubule-associated protein light chain 3 (MAP-LC3 or LC3)) anchored in their lipidated form (LC3-II) on the autophagosomes surface [13]. Through this mechanism, damaged organelles or other substrates are selectively removed from the cytoplasm [14]. For instance, terms such as ERphagy, mitophagy, lysophagy, ribophagy, pexophagy, RN/DNautophagy, xenophagy, aggrephagy describe the selective removal of damaged structures of ER, mitochondria, lysosomes, ribosomes, peroxisomes, nucleic acids, pathogens, and protein aggregates, respectively (Figure 1). Once substrates have been engulfed into autophagosomes, macroautophagy ends with the fusion of autophagosomes with lysosomes for cargo degradation.

In diseases characterized by the presence of misfolded proteins prone to aggregate (including MNDs), aggrephagy may utilize a specific form of cargo delivery to the microtubule organization center (MTOC), where aggresomes are assembled [15–19]. This route, known as chaperone-assisted selective autophagy (CASA) [20,21], is based on misfolded protein recognition by a heteromeric complex of the small heat shock protein B8 (HSPB8) and its obligatory partner Bcl-2-associated athanogene 3 (BAG3) [22,23]. Once formed, HSPB8/BAG3 associate to HSPAs/HSP70s bound to the carboxyl terminus of HSC70-interacting protein (CHIP or STUB1), a U-box-containing E3 ubiquitin-protein ligase that tags misfolded proteins with a polyubiquitin chain for autophagy receptor recognition (SQSTM1/p62 for CASA). This protein complex, named CASA complex, is then transported along microtubules to the MTOC by the dynein machinery bound to BAG3 [22,23]. Next, the autophagy receptors allow the LC3-II-mediated insertion into autophagosomes. The relevance of the CASA in cell homeostasis is underlined by the fact that, when mutated, most of its members cause neuronal or muscular disorders (see Figure 2 for details).

Regarding mammalian microautophagy, cargoes are specifically engulfed into lysosomes via the intervention of late endosomes. Protein recognition can be achieved either ‘in bulk' or (like CMA, see below) selectively thanks to a KFERQ-like motif in the target protein which is recognized by the constitutive form of HSPAs/HSP70s, the heat shock cognate 70 (HSPA8/HSC70) [24]. In microautophagy, HSPAs/HSP70s activities are mediated by an invagination of the late endosome membrane with the involvement of the endosomal sorting complex required for transport (ESCRT) machinery. Entrapped proteins and organelles can be then directly processed in late endosomes or after their fusion with lysosomes [9,25–27]. HSPA8/HSC70 plays a pivotal role also in CMA. In CMA, the HSPA8/HSC70-bound substrate is not subjected to endosomal engulfment. Instead, the HSPA8/HSC70 recognizes and unfolds the cargo that is directly routed to lysosomes where it is released to the CMA receptor LAMP2A, thus escaping the late endosomal engulfment [28]. In CMA, only the proteins possessing the KFERQ-like motif can be cleared [29], even if this motif is relatively frequent in mammalian proteins and there is the intriguing possibility to activate it via post-translational modifications (phosphorylation, acetylation) acting as an off/on switch to regulate protein degradation [24]. When the cargo is released from HSPA8/HSC70 it is associated to LAMP2A, which oligomerizes into the lysosomal membrane. Oligomerization promotes the translocation of the cargo to the lumen, where another HSPA8/HSC70 extracts the cargo allowing its degradation (Figure 1).

As mentioned above, in the past two decades, it emerged that many mutant genes causative of specific MND forms encode proteins differentially involved in the regulatory steps of autophagy.

Indeed, by merely looking at the many different HSP forms, several mutated genes, called Spastic ParapleGia (SPG) genes, encode proteins that are key regulators of the endolysosomal system and autophagy. An autosomal dominant form of HSP is caused by a mutation of the SPG42 gene encoding a protein involved in ER membrane transport, bone morphogenetic protein (BMP) signaling and autophagy [30–32]; other autosomal recessive forms are caused by mutant genes encoding proteins involved in autophagy as well as in lysosome shaping (e.g. SPG11, encoding spatacsin), in endosomal trafficking (e.g. SPG15, encoding spastizin), in vesicle formation and trafficking (e.g. SPG47, SPG50, SPG51, SPG52, SPG53, encoding AP4B1, AP4M1, AP4E1, AP4S1, VPS37A, respectively), in Golgi apparatus-multivesicular bodies (MVB) dynamics (e.g. SPG48, encoding AP5Z1), and lysosomal targeting to autophagosomes (e.g. SPG49, encoding TECPR2), or in membrane trafficking and mitochondrial function (e.g. SPG78, encoding ATP13A2) [30–34] (Figure 3 and Table 1). Mutations of these genes result through different mechanisms in MNs death and involve other neuronal populations, giving rise to the complex and mixed clinical manifestations which coexist with motor dysfunctions.

Even more complex is the autophagy involvement in ALS [4,11,84]. As for HSP, several familial ALS forms are caused by mutations in genes encoding proteins regulating the endolysosomal and autophagic pathways (as extensively reviewed in [85]). Examples are the C9ORF72 (encoding the C9orf72-SMCR8 complex subunit, guanine nucleotide exchange factor C9orf72, recently involved in autophagy), the DCTN1 and KIF5A (encoding, respectively, dynactin subunit 1 (a component of dynein motor complex) and kinesin family member 5A, involved in the autophago-lysosome fusion process and, for dynein/dynactin, in the routing of the CASA complex to the MTOC), VCP (encoding the valosin containing protein, a hexameric complex involved in misfolded protein extraction for their degradation via different pathways, including autophagy), SQSTM1/p62 and OPTN (encoding the Sequestosome 1 and the optineurin, two autophagy receptors), TBK1 (encoding the TANK binding kinase 1, involved in the innate immune response, inflammation, cell proliferation responses, and autophagy), FIG4 (encoding the factor-induced gene 4, a magnesium-dependent phosphatase converting phosphatidylinositol-3,5-bisphosphate (PI(3,5)P2) into phosphatidylinositol-3-phosphate (PI(3)P) on endosomal membranes, a process relevant for endosomal transport and autophagy), and others.

Moreover, several mutant genes encode proteins not directly involved in autophagy, but related to mechanisms that facilitate the process (e.g. genes encoding proteins involved in vesicle trafficking (and apoptosis, exocytosis, and cytokinesis), like ANXA11, encoding annexin A11; in endocytosis (and RNA metabolism), like ATXN2, encoding ataxin 2; in sorting of endosomal cargo proteins, like CHMP2B, encoding the charged MVB protein 2B, a component of the ESCRT complex III; in vesicle trafficking from the ER-membrane, like VAPB, encoding VAMP associated protein B/C) (Figure 3). Also, the molecular re-routing system from autophagy to proteasome and vice versa may indirectly cause autophagy alterations. In fact, this system maintains the fine-tuned equilibrium between these two degradative pathways. The best example is the routing system based on the activity of HSPAs/HSP70s-CHIP, which is differentially regulated by their selective association to HSPB8/BAG3 (CASA for autophagic degradation) or to BAG1 (for proteasome degradation) [22]. Autophagy could be required as an alternative mechanism, in case of mutations occurring in UBQLN2 or CCNF (encoding, respectively, Ubiquilin 2 and cyclin F, the first physically associated with both proteasome and ubiquitin ligases and involved in protein degradation via proteasome, the second catalyzing ubiquitin transfer to substrates for proteasomal degradation), even if it is still unclear whether the autophagic response may compensate the altered proteasome pathway.

The impact of some of the mutations/variants reported above on ALS onset and progression is debated. A recent database (ALSoD (https://alsod.ac.uk/)) has tried to correlate how the variants are linked to ALS, even when evidence is weak.

It should be underlined that mutations in some of the genes associated to HSP and ALS may also be responsible for the appearance of PLS (e.g. SPG7, TBK1, ALS2, ERLIN2, FIG4) [86]. Similarly, a FTD phenotype may be present at different degrees in ALS cases (even as pure FTD with some ALS-related mutant genes). Therefore, it is difficult to draw clear correlations between genotype and phenotype in these diseases.

Even in the case of SMA, the causative survival MN (SMN1) protein (a protein implicated in spliceosome assembly and ribonucleoproteins biogenesis, mRNA trafficking, local translation, and cytoskeletal dynamics) has been found involved in endocytosis and autophagy (see [87] for review), although the mechanism by which SMN controls or is regulated by autophagy is still poorly understood [88–90].

While some mutated genes causative of MNDs directly modulate ALP, many others indirectly impact on autophagy as a consequence of the aberrant biochemical properties acquired by the mutant protein. This occurs when a mutation modifies the native protein (i) altering its capability to properly fold, (ii) enhancing its hydrophobicity, (iii) affecting highly disordered domains, or (iv) altering its interaction with partners, etc. These mechanisms are not mutually exclusive and often increase the propensity of the mutant proteins to clamp together, forming aggregate structures of diverse nature: liquid–liquid droplets or fluid condensates, solid-like aggregates or densely packed insoluble protein inclusions, etc., based on their mechanism of formation and/or their stage of maturation. As a function of their biochemical state, aggregates can be dissolved or further processed to be cleared from cells via autophagy. However, aggregated proteins may impair the autophagic flux, causing severe consequences to the PQC system, with the degradative capability of affected neurons (or surrounding cells affected in MNDs) being overwhelmed. Indeed, this is the case of SBMA, caused by the transcription factor androgen receptor (AR) carrying an elongated polyglutamine tract (ARpolyQ) in its N-terminus. Soon after its translation, the polyQ is masked by specific chaperones (HSPAs/HSP70s, HSP90, etc.), that are released upon AR-binding with its natural androgenic ligands. These molecules activate the AR, allowing conformational changes that unmask the polyQ, causing ARpolyQ aggregation and impairment of the autophagic process [91–98]. Despite a deep investigation, it is still not totally clear whether aggregates cause autophagic flux blockage, or if a defective autophagy results in poor protein clearance and thus excessive protein aggregation. Likely, the two processes equally contribute to generate a toxic cycle in which defective autophagy allows protein accumulation, that in turn, perturbs the autophagic pathway, potentiating the deleterious effects of ARpolyQ in MNs (Figure 4). This toxic cycle could also involve the proteasome system, which has a low degradative capacity and only processes monomeric unfolded proteins. Thus, when the proteasome is saturated by an excessive amount of substrates to be degraded, these accumulate worsening the autophagic flux blockage previously mentioned [99–107]. Similar processes take place also in the case of various mutant proteins causative of familial ALS or HSP. In fact, like the ARpolyQ in SBMA, most of these mutated proteins tend to misfold and aggregate (e.g.: Superoxide Dismutase 1 (SOD1), TAR DNA-Binding Protein 43 (TDP-43, and its fragment of 35 and 25 kDa), fused in sarcoma (FUS), VCP, the dipeptide repeats (DPRs) products of the C9ORF72 mRNA, etc.) [100,108–129], possibly impairing the degradative systems including autophagy. Notably, the exogenous expression or a pharmacological transcriptional induction of HSPB8 [22,116,117,130], both robustly enhance mutant misfolded and aggregated protein clearance. Since HSPB8 is not an autophagy inducer, its enhancement facilitates the delivery of misfolded proteins along microtubules for their autophagic clearance [104,131–134], improving or restoring a normal autophagic flux.

Based on what reported above, autophagy activation may represent a valuable therapeutic approach for MNDs, but this approach has to be carefully evaluated considering the specific alteration present in each clinical form studied.

For example, in case of insufficient autophagy (e.g. lack of crucial factors linked to haploinsufficiency in MNDs associated with loss-of-function mutations; recessive forms involving proteins which can be replaced by the activity of other factors, etc.), it might be useful to stimulate the remaining functional wild type allele, or ‘redundant' factors to restore autophagy. Unfortunately, autophagy and its regulation may considerably differ in the various cell types affected in MNDs (neurons, MNs, glial cells, microglia, muscle cells) making it difficult to identify the proper therapeutic agent for each MND form [11].

In general, drugs stimulating autophagy are relatively poorly specific. This is the case of the mammalian target of rapamycin (mTOR) inhibitor rapamycin, tested in MND animal models [135] and under investigation in ND patients [136,137], that acts not only on autophagy, but on a plethora of other intracellular pathways. Lithium has also been tested in some forms of MNDs, even if its activity as an autophagy inducer is rather controversial [138–141].

Other autophagy inducers, like trehalose and the more stable derivatives melibiose and lactulose, appear to be more promising, since their molecular mechanism of action is more selective and impacts directly on the autophagy master regulator transcription factor EB (TFEB) [104,142–146]. Trehalose has already been successfully tested in several animal models of NDs [147–159]. No double-blind clinical trial has been started yet, even if a single-arm, open-label pilot study has been recently reported on Niemann–Pick disease types A and B patients to assess its potential efficacy after intravenous delivery for 3 months. Despite the size of the study was small, a positive (not statistically relevant) trend was reported [160], suggesting that larger studies are needed to verify trehalose efficacy.

In the case of some MND forms in which autophagy is already enhanced in response to the overload of aberrant aggregation-prone proteins, generic autophagy activators may not be effective treatments. Instead, the pharmacological targeting of factors specifically acting on misfolded protein recognition and delivery to autophagy will represent a valid therapeutic strategy. The drug colchicine has these properties since it has been identified as a potent activator of the human HSPB8 promoter in MNs. Its capability to enhance HSPB8 expression was correlated with an increased CASA-mediated removal of ALS-associated proteins (TDP-43 and its disease associated fragments) in ALS cell and fly models [161]. A double-blind clinical trial with colchicine is presently ongoing on a group of ALS patients [133].

In conclusion, autophagy is an essential cellular process that maintains proteostasis in neurons and other cell types typically affected in MNDs. Its role in MNDs is extremely complex as both its dysregulation and its insufficient or excessive activity play multiple roles in the appearance and/or progression of different forms of MNDs and other NDs. Moreover, it is still unclear why this relatively conserved mechanism among the various cell types of the human body associates to so many different pathological conditions. Since these diseases are characterized by the selective vulnerability of different neuronal populations, other modifier factors might differentially contribute to exacerbate or attenuate, respectively, the deleterious or protective activities of autophagy in each neuronal type. Therefore, the identification of a proper target to be tackled by drugs capable of modulating the autophagic pathways is still challenging. However, it is emerging that approaches aimed to facilitate an already activated autophagy (e.g. those acting on the selective delivery of misfolded proteins to the autophago-lysosomes for their clearance) will provide more tailored treatments to counteract the neurotoxicity of aggregation-prone proteins in a wide variety of MNDs linked to defective response to proteotoxic stimuli.

  • MNDs comprise several untreatable neurodegenerative diseases characterized by the loss of neurons controlling voluntary movements. Several MND forms are associated to alteration of the main degradative systems, particularly the ALP.

  • Several genes found causative of MNDs are involved in the regulation of the various forms of autophagy. Other genes code proteins that perturb proteostasis in neurons indirectly altering autophagy.

  • Different compounds have been developed to stimulate and/or facilitate the ALP, some have already been successfully tested in animal models of MNDs and may be utilized soon for advanced clinical trials in MND patients.

The authors declare that there are no competing interests associated with the manuscript.

This research was funded by: Fondazione Telethon, Italy (no. GGP19128 to A.P.); Kennedy's disease association (2018 grant to R.C.; 2020 grant to M.G.); Fondazione Cariplo, Italy (no. 2021–1544 to R.C.); Fondazione AriSLA, Italy (MLOpathy to A.P.; Target-RAN to A.P.); Association Française contre les Myopathies, France (AFM Telethon no. 23236 to A.P.); Bando Straordinario per Progetti Interdipartimentali (Gender-ALS to M.G.); Italian Ministry of University and Research (MIUR), PRIN - Progetti di ricerca di interesse nazionale (no. 2017F2A2C5 to A.P.; no. 2020PBS5MJ to V.C.); Agenzia Italiana del Farmaco (AIFA) (Co_ALS to A.P.); Italian Ministry of University and Research (Progetto Dipartimenti di Eccellenza) Italian Ministry of University and Research - PNRR – Ecosistema dell’innovazione (CN3 RNA, NAME OF THE NC: National Center for Gene Therapy and Drugs based on RNA Technology) (to A.P.).

ALP

autophagy-lysosomal pathway

ALS

amyotrophic lateral sclerosis

ANXA11

annexin A11

AR

androgen receptor

ATGs

autophagy-related proteins

ATXN2

ataxin 2

BAG3

Bcl-2-associated athanogene 3

BMP

bone morphogenetic protein

CASA

chaperone-assisted selective autophagy

CCNF

cyclin F

CHIP/STUB1

carboxyl terminus of Hsc70-interacting protein

CHMP2B

charged multivesicular body protein 2B

CMA

chaperone-mediated autophagy

DCTN1

dynactin subunit 1

DPRs

dipeptide repeats

ER

endoplasmic reticulum

ESCRT

endosomal sorting complex required for transport

FIG4

factor-induced gene 4

FTD

frontotemporal dementia

FUS

fused in sarcoma

GABARAP

GABA Type A Receptor-Associated Protein

HSP

hereditary spastic paraplegia

HSPA8/HSC70

heat shock cognate 70

HSPAs/HSP70s

heat shock protein 70

HSPB8

heat shock protein B8

KIF5A

kinesin family member 5A

LAMP2A

lysosome-associated membrane protein type 2A

LCCS

lethal congenital contracture syndrome

LIR

LC3-interacting region

MAP-LC3

microtubule-associated protein light chain 3

MMA

monomelic amyotrophy

MNDs

motor neuron diseases

MNs

motor neurons

MTOC

microtubule organization center

mTOR

mammalian target of rapamycin

MVBs

multivesicular bodies

NDs

neurodegenerative diseases

OPTN

optineurin

PI(3)P

phosphatidylinositol-3-phosphate

PI(3,5)P2

phosphatidylinositol-3,5-bisphosphate

PLS

primary lateral sclerosis

PMA

progressive muscular atrophy

polyQ

polyglutamine

PQC

protein quality control

SBMA

spinobulbar muscular atrophy

SMA

spinal muscular atrophy

SMN

survival MN

SOD1

superoxide dismutase 1

SPG

spastic paraplegia [gene]

SQSTM1/p62

sequestosome 1

TBK1

TANK binding kinase 1

TDP-43

TAR DNA-binding protein 43

TFEB

transcription factor EB

UBQLN2

ubiquilin 2

VAPB

VAMP associated protein B/C

VCP

valosin containing protein

1
Figlewicz
,
D.A.
and
Orrell
,
R.W.
(
2003
)
The genetics of motor neuron diseases
.
Amyotroph. Lateral Scler. Other Motor Neuron Disord.
4
,
225
231
2
James
,
P.A.
and
Talbot
,
K.
(
2006
)
The molecular genetics of non-ALS motor neuron diseases
.
Biochim. Biophys. Acta
1762
,
986
1000
3
Dion
,
P.A.
,
Daoud
,
H.
and
Rouleau
,
G.A.
(
2009
)
Genetics of motor neuron disorders: new insights into pathogenic mechanisms
.
Nat. Rev. Genet.
10
,
769
782
4
Casterton
,
R.L.
,
Hunt
,
R.J.
and
Fanto
,
M.
(
2020
)
Pathomechanism heterogeneity in the amyotrophic lateral sclerosis and frontotemporal dementia disease spectrum: providing focus through the lens of autophagy
.
J. Mol. Biol.
432
,
2692
2713
5
Aslesh
,
T.
and
Yokota
,
T.
(
2022
)
Restoring SMN expression: an overview of the therapeutic developments for the treatment of spinal muscular atrophy
.
Cells
11
,
417
434
6
Chilcott
,
E.M.
,
Muiruri
,
E.W.
,
Hirst
,
T.C.
and
Yanez-Munoz
,
R.J.
(
2021
)
Systematic review and meta-analysis determining the benefits of in vivo genetic therapy in spinal muscular atrophy rodent models
.
Gene Ther.
Advance online publication.
7
Poletti
,
A.
and
Fischbeck
,
K.H.
(
2020
)
Combinatorial treatment for spinal muscular atrophy: an editorial for ‘Combined treatment with the histone deacetylase inhibitor LBH589 and a splice-switch antisense oligonucleotide enhances SMN2 splicing and SMN expression in Spinal Muscular Atrophy cells’ on page 264
.
J. Neurochem.
153
,
146
149
8
Robberecht
,
W.
and
Philips
,
T.
(
2013
)
The changing scene of amyotrophic lateral sclerosis
.
Nat. Rev. Neurosci.
14
,
248
264
9
Fleming
,
A.
,
Bourdenx
,
M.
,
Fujimaki
,
M.
,
Karabiyik
,
C.
,
Krause
,
G.J.
,
Lopez
,
A.
et al (
2022
)
The different autophagy degradation pathways and neurodegeneration
.
Neuron
110
,
935
966
10
Kaushik
,
S.
,
Tasset
,
I.
,
Arias
,
E.
,
Pampliega
,
O.
,
Wong
,
E.
,
Martinez-Vicente
,
M.
et al (
2021
)
Autophagy and the hallmarks of aging
.
Ageing Res. Rev.
72
,
101468
11
Chua
,
J.P.
,
De Calbiac
,
H.
,
Kabashi
,
E.
and
Barmada
,
S.J.
(
2022
)
Autophagy and ALS: mechanistic insights and therapeutic implications
.
Autophagy
18
,
254
282
12
Mizushima
,
N.
(
2020
)
The ATG conjugation systems in autophagy
.
Curr. Opin. Cell Biol.
63
,
1
10
13
Stolz
,
A.
,
Ernst
,
A.
and
Dikic
,
I.
(
2014
)
Cargo recognition and trafficking in selective autophagy
.
Nat. Cell Biol.
16
,
495
501
14
Lamark
,
T.
and
Johansen
,
T.
(
2021
)
Mechanisms of selective autophagy
.
Annu. Rev. Cell Dev. Biol.
37
,
143
169
15
Bence
,
N.F.
,
Sampat
,
R.M.
and
Kopito
,
R.R.
(
2001
)
Impairment of the ubiquitin-proteasome system by protein aggregation
.
Science
292
,
1552
1555
16
Bennett
,
E.J.
,
Bence
,
N.F.
,
Jayakumar
,
R.
and
Kopito
,
R.R.
(
2005
)
Global impairment of the ubiquitin-proteasome system by nuclear or cytoplasmic protein aggregates precedes inclusion body formation
.
Mol. Cell
17
,
351
365
17
Johnston
,
J.A.
,
Illing
,
M.E.
and
Kopito
,
R.R.
(
2002
)
Cytoplasmic dynein/dynactin mediates the assembly of aggresomes
.
Cell Motil. Cytoskelet.
53
,
26
38
18
Johnston
,
J.A.
,
Ward
,
C.L.
and
Kopito
,
R.R.
(
1998
)
Aggresomes: a cellular response to misfolded proteins
.
J. Cell Biol.
143
,
1883
1898
19
Kopito
,
R.R.
(
2000
)
Aggresomes, inclusion bodies and protein aggregation
.
Trends Cell Biol.
10
,
524
530
20
Arndt
,
V.
,
Dick
,
N.
,
Tawo
,
R.
,
Dreiseidler
,
M.
,
Wenzel
,
D.
,
Hesse
,
M.
et al (
2010
)
Chaperone-assisted selective autophagy is essential for muscle maintenance
.
Curr. Biol.
20
,
143
148
21
Carra
,
S.
,
Seguin
,
S.J.
and
Landry
,
J.
(
2008
)
Hspb8 and Bag3: a new chaperone complex targeting misfolded proteins to macroautophagy
.
Autophagy
4
,
237
239
22
Cristofani
,
R.
,
Crippa
,
V.
,
Rusmini
,
P.
,
Cicardi
,
M.E.
,
Meroni
,
M.
,
Licata
,
N.V.
et al (
2017
)
Inhibition of retrograde transport modulates misfolded protein accumulation and clearance in motoneuron diseases
.
Autophagy
13
,
1280
1303
23
Cristofani
,
R.
,
Piccolella
,
M.
,
Crippa
,
V.
,
Tedesco
,
B.
,
Montagnani Marelli
,
M.
,
Poletti
,
A.
et al (
2021
)
The role of HSPB8, a component of the chaperone-assisted selective autophagy machinery, in cancer
.
Cells
10
,
335
358
24
Kaushik
,
S.
and
Cuervo
,
A.M.
(
2018
)
The coming of age of chaperone-mediated autophagy
.
Nat. Rev. Mol. Cell Biol.
19
,
365
381
25
Kunz
,
J.B.
,
Schwarz
,
H.
and
Mayer
,
A.
(
2004
)
Determination of four sequential stages during microautophagy in vitro
.
J. Biol. Chem.
279
,
9987
9996
26
Sahu
,
R.
,
Kaushik
,
S.
,
Clement
,
C.C.
,
Cannizzo
,
E.S.
,
Scharf
,
B.
,
Follenzi
,
A.
et al (
2011
)
Microautophagy of cytosolic proteins by late endosomes
.
Dev. Cell
20
,
131
139
27
Park
,
J.S.
,
Kim
,
D.H.
and
Yoon
,
S.Y.
(
2016
)
Regulation of amyloid precursor protein processing by its KFERQ motif
.
BMB Rep.
49
,
337
342
28
Chiang
,
H.L.
and
Dice
,
J.F.
(
1988
)
Peptide sequences that target proteins for enhanced degradation during serum withdrawal
.
J. Biol. Chem.
263
,
6797
6805
29
Cuervo
,
A.M.
and
Dice
,
J.F.
(
1996
)
A receptor for the selective uptake and degradation of proteins by lysosomes
.
Science.
273
,
501
503
30
Marchesi L
,
T.
,
Leblanc
,
M.
and
Stevanin
,
G.
(
2021
)
Current knowledge of endolysosomal and autophagy defects in hereditary spastic paraplegia
.
Cells
10
,
1678
1702
31
Boutry
,
M.
,
Morais
,
S.
and
Stevanin
,
G.
(
2019
)
Update on the genetics of spastic paraplegias
.
Curr. Neurol. Neurosci. Rep.
19
,
18
32
Teinert
,
J.
,
Behne
,
R.
,
Wimmer
,
M.
and
Ebrahimi-Fakhari
,
D.
(
2020
)
Novel insights into the clinical and molecular spectrum of congenital disorders of autophagy
.
J. Inherit. Metab. Dis.
43
,
51
62
33
Fraiberg
,
M.
,
Tamim-Yecheskel
,
B.C.
,
Kokabi
,
K.
,
Subic
,
N.
,
Heimer
,
G.
,
Eck
,
F.
et al (
2021
)
Lysosomal targeting of autophagosomes by the TECPR domain of TECPR2
.
Autophagy
17
,
3096
3108
34
Tamim-Yecheskel
,
B.C.
,
Fraiberg
,
M.
,
Kokabi
,
K.
,
Freud
,
S.
,
Shatz
,
O.
,
Marvaldi
,
L.
et al (
2021
)
A tecpr2 knockout mouse exhibits age-dependent neuroaxonal dystrophy associated with autophagosome accumulation
.
Autophagy
17
,
3082
3095
35
Hadano
,
S.
,
Hand
,
C.K.
,
Osuga
,
H.
,
Yanagisawa
,
Y.
,
Otomo
,
A.
,
Devon
,
R.S.
et al (
2001
)
A gene encoding a putative GTPase regulator is mutated in familial amyotrophic lateral sclerosis 2
.
Nat. Genet.
29
,
166
173
36
Yang
,
Y.
,
Hentati
,
A.
,
Deng
,
H.X.
,
Dabbagh
,
O.
,
Sasaki
,
T.
,
Hirano
,
M.
et al (
2001
)
The gene encoding alsin, a protein with three guanine-nucleotide exchange factor domains, is mutated in a form of recessive amyotrophic lateral sclerosis
.
Nat. Genet.
29
,
160
165
37
Smith
,
B.N.
,
Topp
,
S.D.
,
Fallini
,
C.
,
Shibata
,
H.
,
Chen
,
H.J.
,
Troakes
,
C.
et al (
2017
)
Mutations in the vesicular trafficking protein annexin A11 are associated with amyotrophic lateral sclerosis
.
Sci. Transl. Med.
9
,
eaad9157
38
La Spada
,
A.R.
,
Wilson
,
E.M.
,
Lubahn
,
D.B.
,
Harding
,
A.E.
and
Fischbeck
,
K.H.
(
1991
)
Androgen receptor gene mutations in X-linked spinal and bulbar muscular atrophy
.
Nature
352
,
77
79
39
Elden
,
A.C.
,
Kim
,
H.J.
,
Hart
,
M.P.
,
Chen-Plotkin
,
A.S.
,
Johnson
,
B.S.
,
Fang
,
X.
et al (
2010
)
Ataxin-2 intermediate-length polyglutamine expansions are associated with increased risk for ALS
.
Nature
466
,
1069
1075
40
Tazen
,
S.
,
Figueroa
,
K.
,
Kwan
,
J.Y.
,
Goldman
,
J.
,
Hunt
,
A.
,
Sampson
,
J.
et al (
2013
)
Amyotrophic lateral sclerosis and spinocerebellar ataxia type 2 in a family with full CAG repeat expansions of ATXN2
.
JAMA Neurol.
70
,
1302
1304
41
Renton
,
A.E.
,
Majounie
,
E.
,
Waite
,
A.
,
Simon-Sanchez
,
J.
,
Rollinson
,
S.
,
Gibbs
,
J.R.
et al (
2011
)
A hexanucleotide repeat expansion in C9ORF72 is the cause of chromosome 9p21-linked ALS-FTD
.
Neuron
72
,
257
268
42
DeJesus-Hernandez
,
M.
,
Mackenzie
,
I.R.
,
Boeve
,
B.F.
,
Boxer
,
A.L.
,
Baker
,
M.
,
Rutherford
,
N.J.
et al (
2011
)
Expanded GGGGCC hexanucleotide repeat in noncoding region of C9ORF72 causes chromosome 9p-linked FTD and ALS
.
Neuron
72
,
245
256
43
Williams
,
K.L.
,
Topp
,
S.
,
Yang
,
S.
,
Smith
,
B.
,
Fifita
,
J.A.
,
Warraich
,
S.T.
et al (
2016
)
CCNF mutations in amyotrophic lateral sclerosis and frontotemporal dementia
.
Nat. Commun.
7
,
11253
44
Parkinson
,
N.
,
Ince
,
P.G.
,
Smith
,
M.O.
,
Highley
,
R.
,
Skibinski
,
G.
,
Andersen
,
P.M.
et al (
2006
)
ALS phenotypes with mutations in CHMP2B (charged multivesicular body protein 2B)
.
Neurology
67
,
1074
1077
45
Munch
,
C.
,
Sedlmeier
,
R.
,
Meyer
,
T.
,
Homberg
,
V.
,
Sperfeld
,
A.D.
,
Kurt
,
A.
et al (
2004
)
Point mutations of the p150 subunit of dynactin (DCTN1) gene in ALS
.
Neurology
63
,
724
726
46
Puls
,
I.
,
Jonnakuty
,
C.
,
LaMonte
,
B.H.
,
Holzbaur
,
E.L.
,
Tokito
,
M.
,
Mann
,
E.
et al (
2003
)
Mutant dynactin in motor neuron disease
.
Nat. Genet.
33
,
455
456
47
Alazami
,
A.M.
,
Adly
,
N.
,
Al Dhalaan
,
H.
and
Alkuraya
,
F.S.
(
2011
)
A nullimorphic ERLIN2 mutation defines a complicated hereditary spastic paraplegia locus (SPG18)
.
Neurogenetics
12
,
333
336
48
Al-Saif
,
A.
,
Bohlega
,
S.
and
Al-Mohanna
,
F.
(
2012
)
Loss of ERLIN2 function leads to juvenile primary lateral sclerosis
.
Ann. Neurol.
72
,
510
516
49
Chow
,
C.Y.
,
Landers
,
J.E.
,
Bergren
,
S.K.
,
Sapp
,
P.C.
,
Grant
,
A.E.
,
Jones
,
J.M.
et al (
2009
)
Deleterious variants of FIG4, a phosphoinositide phosphatase, in patients with ALS
.
Am. J. Hum. Genet.
84
,
85
88
50
Vance
,
C.
,
Rogelj
,
B.
,
Hortobagyi
,
T.
,
De Vos
,
K.J.
,
Nishimura
,
A.L.
,
Sreedharan
,
J.
et al (
2009
)
Mutations in FUS, an RNA processing protein, cause familial amyotrophic lateral sclerosis type 6
.
Science
323
,
1208
1211
51
Kwiatkowski TJ
,
J.
,
Bosco
,
D.A.
,
Leclerc
,
A.L.
,
Tamrazian
,
E.
,
Vanderburg
,
C.R.
,
Russ
,
C.
et al (
2009
)
Mutations in the FUS/TLS gene on chromosome 16 cause familial amyotrophic lateral sclerosis
.
Science
323
,
1205
1208
52
Nicolas
,
A.
,
Kenna
,
K.P.
,
Renton
,
A.E.
,
Ticozzi
,
N.
,
Faghri
,
F.
,
Chia
,
R.
et al (
2018
)
Genome-wide analyses identify KIF5A as a novel ALS gene
.
Neuron
97
,
1268
1283.e6
53
Brenner
,
D.
,
Yilmaz
,
R.
,
Muller
,
K.
,
Grehl
,
T.
,
Petri
,
S.
,
Meyer
,
T.
et al (
2018
)
Hot-spot KIF5A mutations cause familial ALS
.
Brain
141
,
688
697
54
Maruyama
,
H.
,
Morino
,
H.
,
Ito
,
H.
,
Izumi
,
Y.
,
Kato
,
H.
,
Watanabe
,
Y.
et al (
2010
)
Mutations of optineurin in amyotrophic lateral sclerosis
.
Nature
465
,
223
226
55
Rodrigues
,
N.R.
,
Owen
,
N.
,
Talbot
,
K.
,
Ignatius
,
J.
,
Dubowitz
,
V.
and
Davies
,
K.E.
(
1995
)
Deletions in the survival motor neuron gene on 5q13 in autosomal recessive spinal muscular atrophy
.
Hum. Mol. Genet.
4
,
631
634
56
Lefebvre
,
S.
,
Burglen
,
L.
,
Reboullet
,
S.
,
Clermont
,
O.
,
Burlet
,
P.
,
Viollet
,
L.
et al (
1995
)
Identification and characterization of a spinal muscular atrophy-determining gene
.
Cell
80
,
155
165
57
Rosen
,
D.R.
,
Siddique
,
T.
,
Patterson
,
D.
,
Figlewicz
,
D.A.
,
Sapp
,
P.
,
Hentati
,
A.
et al (
1993
)
Mutations in Cu/Zn superoxide dismutase gene are associated with familial amyotrophic lateral sclerosis
.
Nature
362
,
59
62
58
Stevanin
,
G.
,
Santorelli
,
F.M.
,
Azzedine
,
H.
,
Coutinho
,
P.
,
Chomilier
,
J.
,
Denora
,
P.S.
et al (
2007
)
Mutations in SPG11, encoding spatacsin, are a major cause of spastic paraplegia with thin corpus callosum
.
Nat. Genet.
39
,
366
372
59
Del Bo
,
R.
,
Di Fonzo
,
A.
,
Ghezzi
,
S.
,
Locatelli
,
F.
,
Stevanin
,
G.
,
Costa
,
A.
et al (
2007
)
SPG11: a consistent clinical phenotype in a family with homozygous spatacsin truncating mutation
.
Neurogenetics
8
,
301
305
60
Orlacchio
,
A.
,
Babalini
,
C.
,
Borreca
,
A.
,
Patrono
,
C.
,
Massa
,
R.
,
Basaran
,
S.
et al (
2010
)
SPATACSIN mutations cause autosomal recessive juvenile amyotrophic lateral sclerosis
.
Brain
133
,
591
598
61
Hanein
,
S.
,
Martin
,
E.
,
Boukhris
,
A.
,
Byrne
,
P.
,
Goizet
,
C.
,
Hamri
,
A.
et al (
2008
)
Identification of the SPG15 gene, encoding spastizin, as a frequent cause of complicated autosomal-recessive spastic paraplegia, including Kjellin syndrome
.
Am. J. Hum. Genet.
82
,
992
1002
62
Hughes
,
C.A.
,
Byrne
,
P.C.
,
Webb
,
S.
,
McMonagle
,
P.
,
Patterson
,
V.
,
Hutchinson
,
M.
et al (
2001
)
SPG15, a new locus for autosomal recessive complicated HSP on chromosome 14q
.
Neurology
56
,
1230
1233
63
Elleuch
,
N.
,
Bouslam
,
N.
,
Hanein
,
S.
,
Lossos
,
A.
,
Hamri
,
A.
,
Klebe
,
S.
et al (
2007
)
Refinement of the SPG15 candidate interval and phenotypic heterogeneity in three large Arab families
.
Neurogenetics
8
,
307
315
64
Lin
,
P.
,
Li
,
J.
,
Liu
,
Q.
,
Mao
,
F.
,
Li
,
J.
,
Qiu
,
R.
et al (
2008
)
A missense mutation in SLC33A1, which encodes the acetyl-CoA transporter, causes autosomal-dominant spastic paraplegia (SPG42)
.
Am. J. Hum. Genet.
83
,
752
759
65
Jamra R
,
A.
,
Philippe
,
O.
,
Raas-Rothschild
,
A.
,
Eck
,
S.H.
,
Graf
,
E.
,
Buchert
,
R.
et al (
2011
)
Adaptor protein complex 4 deficiency causes severe autosomal-recessive intellectual disability, progressive spastic paraplegia, shy character, and short stature
.
Am. J. Hum. Genet.
88
,
788
795
66
Slabicki
,
M.
,
Theis
,
M.
,
Krastev
,
D.B.
,
Samsonov
,
S.
,
Mundwiller
,
E.
,
Junqueira
,
M.
et al (
2010
)
A genome-scale DNA repair RNAi screen identifies SPG48 as a novel gene associated with hereditary spastic paraplegia
.
PLoS Biol.
8
,
e1000408
67
Oz-Levi
,
D.
,
Ben-Zeev
,
B.
,
Ruzzo
,
E.K.
,
Hitomi
,
Y.
,
Gelman
,
A.
,
Pelak
,
K.
et al (
2012
)
Mutation in TECPR2 reveals a role for autophagy in hereditary spastic paraparesis
.
Am. J. Hum. Genet.
91
,
1065
1072
68
Verkerk
,
A.J.
,
Schot
,
R.
,
Dumee
,
B.
,
Schellekens
,
K.
,
Swagemakers
,
S.
,
Bertoli-Avella
,
A.M.
et al (
2009
)
Mutation in the AP4M1 gene provides a model for neuroaxonal injury in cerebral palsy
.
Am. J. Hum. Genet.
85
,
40
52
69
Moreno-De-Luca
,
A.
,
Helmers
,
S.L.
,
Mao
,
H.
,
Burns
,
T.G.
,
Melton
,
A.M.
,
Schmidt
,
K.R.
et al (
2011
)
Adaptor protein complex-4 (AP-4) deficiency causes a novel autosomal recessive cerebral palsy syndrome with microcephaly and intellectual disability
.
J. Med. Genet.
48
,
141
144
70
Zivony-Elboum
,
Y.
,
Westbroek
,
W.
,
Kfir
,
N.
,
Savitzki
,
D.
,
Shoval
,
Y.
,
Bloom
,
A.
et al (
2012
)
A founder mutation in Vps37A causes autosomal recessive complex hereditary spastic paraparesis
.
J. Med. Genet.
49
,
462
472
71
Yang
,
Y.
,
Zhang
,
L.
,
Lynch
,
D.R.
,
Lukas
,
T.
,
Ahmeti
,
K.
,
Sleiman
,
P.M.
et al (
2016
)
Compound heterozygote mutations in SPG7 in a family with adult-onset primary lateral sclerosis
.
Neurol. Genet.
2
,
e60
72
Kara
,
E.
,
Tucci
,
A.
,
Manzoni
,
C.
,
Lynch
,
D.S.
,
Elpidorou
,
M.
,
Bettencourt
,
C.
et al (
2016
)
Genetic and phenotypic characterization of complex hereditary spastic paraplegia
.
Brain
139
,
1904
1918
73
Estrada-Cuzcano
,
A.
,
Martin
,
S.
,
Chamova
,
T.
,
Synofzik
,
M.
,
Timmann
,
D.
,
Holemans
,
T.
et al (
2017
)
Loss-of-function mutations in the ATP13A2/PARK9 gene cause complicated hereditary spastic paraplegia (SPG78)
.
Brain
140
,
287
305
74
Fecto
,
F.
,
Yan
,
J.
,
Vemula
,
S.P.
,
Liu
,
E.
,
Yang
,
Y.
,
Chen
,
W.
et al (
2011
)
SQSTM1 mutations in familial and sporadic amyotrophic lateral sclerosis
.
Arch. Neurol.
68
,
1440
1446
75
Sreedharan
,
J.
,
Blair
,
I.P.
,
Tripathi
,
V.B.
,
Hu
,
X.
,
Vance
,
C.
,
Rogelj
,
B.
et al (
2008
)
TDP-43 mutations in familial and sporadic amyotrophic lateral sclerosis
.
Science
319
,
1668
1672
76
Van Deerlin
,
V.M.
,
Leverenz
,
J.B.
,
Bekris
,
L.M.
,
Bird
,
T.D.
,
Yuan
,
W.
,
Elman
,
L.B.
et al (
2008
)
TARDBP mutations in amyotrophic lateral sclerosis with TDP-43 neuropathology: a genetic and histopathological analysis
.
Lancet Neurol.
7
,
409
416
77
Kabashi
,
E.
,
Valdmanis
,
P.N.
,
Dion
,
P.
,
Spiegelman
,
D.
,
McConkey
,
B.J.
,
Vande Velde
,
C.
et al (
2008
)
TARDBP mutations in individuals with sporadic and familial amyotrophic lateral sclerosis
.
Nat. Genet.
40
,
572
574
78
Gitcho
,
M.A.
,
Baloh
,
R.H.
,
Chakraverty
,
S.
,
Mayo
,
K.
,
Norton
,
J.B.
,
Levitch
,
D.
et al (
2008
)
TDP-43 A315T mutation in familial motor neuron disease
.
Ann. Neurol.
63
,
535
538
79
Cirulli
,
E.T.
,
Lasseigne
,
B.N.
,
Petrovski
,
S.
,
Sapp
,
P.C.
,
Dion
,
P.A.
,
Leblond
,
C.S.
et al (
2015
)
Exome sequencing in amyotrophic lateral sclerosis identifies risk genes and pathways
.
Science
347
,
1436
1441
80
Freischmidt
,
A.
,
Wieland
,
T.
,
Richter
,
B.
,
Ruf
,
W.
,
Schaeffer
,
V.
,
Muller
,
K.
et al (
2015
)
Haploinsufficiency of TBK1 causes familial ALS and fronto-temporal dementia
.
Nat. Neurosci.
18
,
631
636
81
Deng
,
H.X.
,
Chen
,
W.
,
Hong
,
S.T.
,
Boycott
,
K.M.
,
Gorrie
,
G.H.
,
Siddique
,
N.
et al (
2011
)
Mutations in UBQLN2 cause dominant X-linked juvenile and adult-onset ALS and ALS/dementia
.
Nature
477
,
211
215
82
Nishimura
,
A.L.
,
Mitne-Neto
,
M.
,
Silva
,
H.C.
,
Richieri-Costa
,
A.
,
Middleton
,
S.
,
Cascio
,
D.
et al (
2004
)
A mutation in the vesicle-trafficking protein VAPB causes late-onset spinal muscular atrophy and amyotrophic lateral sclerosis
.
Am. J. Hum. Genet.
75
,
822
831
83
Johnson
,
J.O.
,
Mandrioli
,
J.
,
Benatar
,
M.
,
Abramzon
,
Y.
,
Van Deerlin
,
V.M.
,
Trojanowski
,
J.Q.
et al (
2010
)
Exome sequencing reveals VCP mutations as a cause of familial ALS
.
Neuron
68
,
857
864
84
Vegeto
,
E.
,
Villa
,
A.
,
Della Torre
,
S.
,
Crippa
,
V.
,
Rusmini
,
P.
,
Cristofani
,
R.
et al (
2020
)
The role of sex and sex hormones in neurodegenerative diseases
.
Endocr. Rev.
41
,
273
319
85
Cozzi
,
M.
and
Ferrari
,
V.
(
2022 Jul
)
Autophagy dysfunction in ALS: from transport to protein degradation
.
J. Mol. Neurosci.
72
,
1456
1481
86
Liu
,
Y.
,
Xu
,
J.
,
Tao
,
W.
,
Fu
,
C.
,
Liu
,
J.
,
Yu
,
R.
et al (
2019
)
Exome sequencing identifies a mutation (Y740C) in spastic paraplegia 7 gene associated with adult-onset primary lateral sclerosis in a Chinese family
.
Eur. Neurol.
81
,
87
93
87
Chaytow
,
H.
,
Huang
,
Y.T.
,
Gillingwater
,
T.H.
and
Faller
,
K.M.E.
(
2018
)
The role of survival motor neuron protein (SMN) in protein homeostasis
.
Cell. Mol. Life Sci.
75
,
3877
3894
88
Periyakaruppiah
,
A.
,
de la Fuente
,
S.
,
Arumugam
,
S.
,
Bahi
,
N.
,
Garcera
,
A.
and
Soler
,
R.M.
(
2016
)
Autophagy modulators regulate survival motor neuron protein stability in motoneurons
.
Exp. Neurol.
283
,
287
297
89
Sansa
,
A.
,
Hidalgo
,
I.
,
Miralles
,
M.P.
,
de la Fuente
,
S.
,
Perez-Garcia
,
M.J.
,
Munell
,
F.
et al (
2021
)
Spinal muscular atrophy autophagy profile is tissue-dependent: differential regulation between muscle and motoneurons
.
Acta Neuropathol. Commun.
9
,
122
90
Rodriguez-Muela
,
N.
,
Parkhitko
,
A.
,
Grass
,
T.
,
Gibbs
,
R.M.
,
Norabuena
,
E.M.
,
Perrimon
,
N.
et al (
2018
)
Blocking p62-dependent SMN degradation ameliorates spinal muscular atrophy disease phenotypes
.
J. Clin. Investig.
128
,
3008
3023
91
Li
,
M.
,
Miwa
,
S.
,
Kobayashi
,
Y.
,
Merry
,
D.
,
Yamamoto
,
M.
,
Tanaka
,
F.
et al (
1998
)
Nuclear inclusions of the androgen receptor protein in spinal and bulbar muscular atrophy
.
Ann. Neurol.
44
,
249
254
92
Li
,
M.
,
Nakagomi
,
Y.
,
Kobayashi
,
Y.
,
Merry
,
D.E.
,
Tanaka
,
F.
,
Doyu
,
M.
et al (
1998
)
Nonneural nuclear inclusions of androgen receptor protein in spinal and bulbar muscular atrophy
.
Am. J. Pathol.
153
,
695
701
93
Simeoni
,
S.
,
Mancini
,
M.A.
,
Stenoien
,
D.L.
,
Marcelli
,
M.
,
Weigel
,
N.L.
,
Zanisi
,
M.
et al (
2000
)
Motoneuronal cell death is not correlated with aggregate formation of androgen receptors containing an elongated polyglutamine tract
.
Hum. Mol. Genet.
9
,
133
144
94
Stenoien
,
D.L.
,
Cummings
,
C.J.
,
Adams
,
H.P.
,
Mancini
,
M.G.
,
Patel
,
K.
,
DeMartino
,
G.N.
et al (
1999
)
Polyglutamine-expanded androgen receptors form aggregates that sequester heat shock proteins, proteasome components and SRC-1, and are suppressed by the HDJ-2 chaperone
.
Hum. Mol. Genet.
8
,
731
741
95
Piccioni
,
F.
,
Pinton
,
P.
,
Simeoni
,
S.
,
Pozzi
,
P.
,
Fascio
,
U.
,
Vismara
,
G.
et al (
2002
)
Androgen receptor with elongated polyglutamine tract forms aggregates that alter axonal trafficking and mitochondrial distribution in motor neuronal processes
.
FASEB J.
16
,
1418
1420
96
Poletti
,
A.
(
2004
)
The polyglutamine tract of androgen receptor: from functions to dysfunctions in motor neurons
.
Front. Neuroendocrinol.
25
,
1
26
97
Adachi
,
H.
,
Katsuno
,
M.
,
Minamiyama
,
M.
,
Waza
,
M.
,
Sang
,
C.
,
Nakagomi
,
Y.
et al (
2005
)
Widespread nuclear and cytoplasmic accumulation of mutant androgen receptor in SBMA patients
.
Brain
128
,
659
670
98
Adachi
,
H.
,
Waza
,
M.
,
Katsuno
,
M.
,
Tanaka
,
F.
,
Doyu
,
M.
and
Sobue
,
G.
(
2007
)
Pathogenesis and molecular targeted therapy of spinal and bulbar muscular atrophy
.
Neuropathol. Appl. Neurobiol.
33
,
135
151
99
Chua
,
J.P.
,
Reddy
,
S.L.
,
Merry
,
D.E.
,
Adachi
,
H.
,
Katsuno
,
M.
,
Sobue
,
G.
et al (
2014
)
Transcriptional activation of TFEB/ZKSCAN3 target genes underlies enhanced autophagy in spinobulbar muscular atrophy
.
Hum. Mol. Genet.
23
,
1376
1386
100
Cicardi
,
M.E.
,
Cristofani
,
R.
,
Crippa
,
V.
,
Ferrari
,
V.
,
Tedesco
,
B.
,
Casarotto
,
E.
et al (
2019
)
Autophagic and proteasomal mediated removal of mutant androgen receptor in muscle models of spinal and bulbar muscular atrophy
.
Front. Endocrinol.
10
,
569
101
Cortes
,
C.J.
,
Miranda
,
H.C.
,
Frankowski
,
H.
,
Batlevi
,
Y.
,
Young
,
J.E.
,
Le
,
A.
et al (
2014
)
Polyglutamine-expanded androgen receptor interferes with TFEB to elicit autophagy defects in SBMA
.
Nat. Neurosci.
17
,
1180
1189
102
Giorgetti
,
E.
,
Rusmini
,
P.
,
Crippa
,
V.
,
Cristofani
,
R.
,
Boncoraglio
,
A.
,
Cicardi
,
M.E.
et al (
2015
)
Synergic prodegradative activity of bicalutamide and trehalose on the mutant androgen receptor responsible for spinal and bulbar muscular atrophy
.
Hum. Mol. Genet.
24
,
64
75
103
Montie
,
H.L.
,
Cho
,
M.S.
,
Holder
,
L.
,
Liu
,
Y.
,
Tsvetkov
,
A.S.
,
Finkbeiner
,
S.
et al (
2009
)
Cytoplasmic retention of polyglutamine-expanded androgen receptor ameliorates disease via autophagy in a mouse model of spinal and bulbar muscular atrophy
.
Hum. Mol. Genet.
18
,
1937
1950
104
Rusmini
,
P.
,
Cortese
,
K.
,
Crippa
,
V.
,
Cristofani
,
R.
,
Cicardi
,
M.E.
,
Ferrari
,
V.
et al (
2019
)
Trehalose induces autophagy via lysosomal-mediated TFEB activation in models of motoneuron degeneration
.
Autophagy
15
,
631
651
105
Rusmini
,
P.
,
Crippa
,
V.
,
Giorgetti
,
E.
,
Boncoraglio
,
A.
,
Cristofani
,
R.
,
Carra
,
S.
et al (
2013
)
Clearance of the mutant androgen receptor in motoneuronal models of spinal and bulbar muscular atrophy
.
Neurobiol. Aging
34
,
2585
2603
106
Rusmini
,
P.
,
Polanco
,
M.J.
,
Cristofani
,
R.
,
Cicardi
,
M.E.
,
Meroni
,
M.
,
Galbiati
,
M.
et al (
2015
)
Aberrant autophagic response in the muscle of A knock-in mouse model of spinal and bulbar muscular atrophy
.
Sci. Rep.
5
,
15174
107
Yu
,
Z.
,
Wang
,
A.M.
,
Adachi
,
H.
,
Katsuno
,
M.
,
Sobue
,
G.
,
Yue
,
Z.
et al (
2011
)
Macroautophagy is regulated by the UPR-mediator CHOP and accentuates the phenotype of SBMA mice
.
PLoS Genet.
7
,
e1002321
108
Crippa
,
V.
,
Carra
,
S.
,
Rusmini
,
P.
,
Sau
,
D.
,
Bolzoni
,
E.
,
Bendotti
,
C.
et al (
2010
)
A role of small heat shock protein B8 (HspB8) in the autophagic removal of misfolded proteins responsible for neurodegenerative diseases
.
Autophagy
6
,
958
960
109
Crippa
,
V.
,
Galbiati
,
M.
,
Boncoraglio
,
A.
,
Rusmini
,
P.
,
Onesto
,
E.
,
Giorgetti
,
E.
et al (
2013
)
Motoneuronal and muscle-selective removal of ALS-related misfolded proteins
.
Biochem. Soc. Trans.
41
,
1598
1604
110
Kabuta
,
T.
,
Suzuki
,
Y.
and
Wada
,
K.
(
2006
)
Degradation of amyotrophic lateral sclerosis-linked mutant Cu,Zn-superoxide dismutase proteins by macroautophagy and the proteasome
.
J. Biol. Chem.
281
,
30524
30533
111
Kim
,
J.
,
Kim
,
T.Y.
,
Cho
,
K.S.
,
Kim
,
H.N.
and
Koh
,
J.Y.
(
2013
)
Autophagy activation and neuroprotection by progesterone in the G93A-SOD1 transgenic mouse model of amyotrophic lateral sclerosis
.
Neurobiol. Dis.
59
,
80
85
112
Li
,
L.
,
Zhang
,
X.
and
Le
,
W.
(
2008
)
Altered macroautophagy in the spinal cord of SOD1 mutant mice
.
Autophagy
4
,
290
293
113
Morimoto
,
N.
,
Nagai
,
M.
,
Ohta
,
Y.
,
Miyazaki
,
K.
,
Kurata
,
T.
,
Morimoto
,
M.
et al (
2007
)
Increased autophagy in transgenic mice with a G93A mutant SOD1 gene
.
Brain Res.
1167
,
112
117
114
Brady
,
O.A.
,
Meng
,
P.
,
Zheng
,
Y.
,
Mao
,
Y.
and
Hu
,
F.
(
2011
)
Regulation of TDP-43 aggregation by phosphorylation and p62/SQSTM1
.
J. Neurochem.
116
,
248
259
115
Cicardi
,
M.E.
,
Cristofani
,
R.
,
Rusmini
,
P.
,
Meroni
,
M.
,
Ferrari
,
V.
,
Vezzoli
,
G.
et al (
2018
)
Tdp-25 routing to autophagy and proteasome ameliorates its aggregation in amyotrophic lateral sclerosis target cells
.
Sci. Rep.
8
,
12390
116
Crippa
,
V.
,
Sau
,
D.
,
Rusmini
,
P.
,
Boncoraglio
,
A.
,
Onesto
,
E.
,
Bolzoni
,
E.
et al (
2010
)
The small heat shock protein B8 (HspB8) promotes autophagic removal of misfolded proteins involved in amyotrophic lateral sclerosis (ALS)
.
Hum. Mol. Genet.
19
,
3440
3456
117
Cristofani
,
R.
,
Crippa
,
V.
,
Vezzoli
,
G.
,
Rusmini
,
P.
,
Galbiati
,
M.
,
Cicardi
,
M.E.
et al (
2018
)
The small heat shock protein B8 (HSPB8) efficiently removes aggregating species of dipeptides produced in C9ORF72-related neurodegenerative diseases
.
Cell Stress Chaperones
23
,
1
12
118
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
119
Ju
,
J.S.
,
Fuentealba
,
R.A.
,
Miller
,
S.E.
,
Jackson
,
E.
,
Piwnica-Worms
,
D.
,
Baloh
,
R.H.
et al (
2009
)
Valosin-containing protein (VCP) is required for autophagy and is disrupted in VCP disease
.
J. Cell Biol.
187
,
875
888
120
Ormeno
,
F.
,
Hormazabal
,
J.
,
Moreno
,
J.
,
Riquelme
,
F.
,
Rios
,
J.
,
Criollo
,
A.
et al (
2020
)
Chaperone mediated autophagy degrades TDP-43 protein and is affected by TDP-43 aggregation
.
Front. Mol. Neurosci.
13
,
19
121
Urushitani
,
M.
,
Sato
,
T.
,
Bamba
,
H.
,
Hisa
,
Y.
and
Tooyama
,
I.
(
2010
)
Synergistic effect between proteasome and autophagosome in the clearance of polyubiquitinated TDP-43
.
J. Neurosci. Res.
88
,
784
797
122
Wang
,
I.F.
,
Guo
,
B.S.
,
Liu
,
Y.C.
,
Wu
,
C.C.
,
Yang
,
C.H.
,
Tsai
,
K.J.
et al (
2012
)
Autophagy activators rescue and alleviate pathogenesis of a mouse model with proteinopathies of the TAR DNA-binding protein 43
.
Proc. Natl Acad. Sci. U.S.A.
109
,
15024
15029
123
Wang
,
X.
,
Fan
,
H.
,
Ying
,
Z.
,
Li
,
B.
,
Wang
,
H.
and
Wang
,
G.
(
2010
)
Degradation of TDP-43 and its pathogenic form by autophagy and the ubiquitin-proteasome system
.
Neurosci. Lett.
469
,
112
116
124
Wang
,
Y.
,
Liu
,
F.T.
,
Wang
,
Y.X.
,
Guan
,
R.Y.
,
Chen
,
C.
,
Li
,
D.K.
et al (
2018
)
Autophagic modulation by trehalose reduces accumulation of TDP-43 in a cell model of amyotrophic lateral sclerosis via TFEB activation
.
Neurotox Res.
34
,
109
120
125
Alberti
,
S.
,
Mateju
,
D.
,
Mediani
,
L.
and
Carra
,
S.
(
2017
)
Granulostasis: protein quality control of RNP granules
.
Front. Mol. Neurosci.
10
,
84
126
Seguin
,
S.J.
,
Morelli
,
F.F.
,
Vinet
,
J.
,
Amore
,
D.
,
De Biasi
,
S.
,
Poletti
,
A.
et al (
2014
)
Inhibition of autophagy, lysosome and VCP function impairs stress granule assembly
.
Cell Death Diff.
21
,
1838
1851
127
Tresse
,
E.
,
Salomons
,
F.A.
,
Vesa
,
J.
,
Bott
,
L.C.
,
Kimonis
,
V.
,
Yao
,
T.P.
et al (
2010
)
VCP/p97 is essential for maturation of ubiquitin-containing autophagosomes and this function is impaired by mutations that cause IBMPFD
.
Autophagy
6
,
217
227
128
Kirola
,
L.
,
Mukherjee
,
A.
and
Mutsuddi
,
M.
(
2022
)
Recent updates on the genetics of amyotrophic lateral sclerosis and frontotemporal dementia
.
Mol. Neurobiol.
59
,
5673
5694
129
Liu
,
M.
,
Pi
,
H.
,
Xi
,
Y.
,
Wang
,
L.
,
Tian
,
L.
,
Chen
,
M.
et al (
2021
)
KIF5A-dependent axonal transport deficiency disrupts autophagic flux in trimethyltin chloride-induced neurotoxicity
.
Autophagy
17
,
903
924
130
Carra
,
S.
(
2009
)
The stress-inducible HspB8-Bag3 complex induces the eIF2alpha kinase pathway: implications for protein quality control and viral factory degradation?
Autophagy
5
,
428
429
131
Crippa
,
V.
,
D'Agostino
,
V.G.
,
Cristofani
,
R.
,
Rusmini
,
P.
,
Cicardi
,
M.E.
,
Messi
,
E.
et al (
2016
)
Transcriptional induction of the heat shock protein B8 mediates the clearance of misfolded proteins responsible for motor neuron diseases
.
Sci. Rep.
6
,
22827
132
Cristofani
,
R.
,
Rusmini
,
P.
,
Galbiati
,
M.
,
Cicardi
,
M.E.
,
Ferrari
,
V.
,
Tedesco
,
B.
et al (
2019
)
The regulation of the small heat shock protein B8 in misfolding protein diseases causing motoneuronal and muscle cell death
.
Front. Neurosci.
13
,
796
133
Mandrioli
,
J.
,
Crippa
,
V.
,
Cereda
,
C.
,
Bonetto
,
V.
,
Zucchi
,
E.
,
Gessani
,
A.
et al (
2019
)
Proteostasis and ALS: protocol for a phase II, randomised, double-blind, placebo-controlled, multicentre clinical trial for colchicine in ALS (Co-ALS)
.
BMJ Open
9
,
e028486
134
Adriaenssens
,
E.
,
Tedesco
,
B.
,
Mediani
,
L.
,
Asselbergh
,
B.
,
Crippa
,
V.
,
Antoniani
,
F.
et al (
2020
)
BAG3 pro209 mutants associated with myopathy and neuropathy relocate chaperones of the CASA-complex to aggresomes
.
Sci. Rep.
10
,
8755
135
Zhang
,
X.
,
Li
,
L.
,
Chen
,
S.
,
Yang
,
D.
,
Wang
,
Y.
,
Zhang
,
X.
et al (
2011
)
Rapamycin treatment augments motor neuron degeneration in SOD1(G93A) mouse model of amyotrophic lateral sclerosis
.
Autophagy
7
,
412
425
136
Mandrioli
,
J.
,
D'Amico
,
R.
,
Zucchi
,
E.
,
Gessani
,
A.
,
Fini
,
N.
,
Fasano
,
A.
et al (
2018
)
Rapamycin treatment for amyotrophic lateral sclerosis: protocol for a phase II randomized, double-blind, placebo-controlled, multicenter, clinical trial (RAP-ALS trial)
.
Medicine
97
,
e11119
137
Palma
,
J.A.
,
Martinez
,
J.
,
Millar Vernetti
,
P.
,
Ma
,
T.
,
Perez
,
M.A.
,
Zhong
,
J.
et al (
2022
)
mTOR inhibition with sirolimus in multiple system atrophy: a randomized, double-blind, placebo-controlled futility trial and 1-year biomarker longitudinal analysis
.
Mov. Disord.
37
,
778
789
138
Damri
,
O.
,
Shemesh
,
N.
and
Agam
,
G.
(
2020
)
Is there justification to treat neurodegenerative disorders by repurposing drugs? The case of Alzheimer's disease, lithium, and autophagy
.
Int. J. Mol. Sci.
22
,
189
206
139
Costa
,
A.J.
,
Erustes
,
A.G.
,
Sinigaglia
,
R.
,
Girardi
,
C.E.N.
,
Pereira
,
G.
,
Ureshino
,
R.P.
et al (
2021
)
Lack of autophagy induction by lithium decreases neuroprotective effects in the striatum of aged rats
.
Pharmaceutics
13
,
135
148
140
Pasquali
,
L.
,
Longone
,
P.
,
Isidoro
,
C.
,
Ruggieri
,
S.
,
Paparelli
,
A.
and
Fornai
,
F.
(
2009
)
Autophagy, lithium, and amyotrophic lateral sclerosis
.
Muscle Nerve
40
,
173
194
141
Motoi
,
Y.
,
Shimada
,
K.
,
Ishiguro
,
K.
and
Hattori
,
N.
(
2014
)
Lithium and autophagy
.
ACS Chem. Neurosci.
5
,
434
442
142
Medina
,
D.L.
,
Fraldi
,
A.
,
Bouche
,
V.
,
Annunziata
,
F.
,
Mansueto
,
G.
,
Spampanato
,
C.
et al (
2011
)
Transcriptional activation of lysosomal exocytosis promotes cellular clearance
.
Dev. Cell
21
,
421
430
143
Palmieri
,
M.
,
Impey
,
S.
,
Kang
,
H.
,
di Ronza
,
A.
,
Pelz
,
C.
,
Sardiello
,
M.
et al (
2011
)
Characterization of the CLEAR network reveals an integrated control of cellular clearance pathways
.
Hum. Mol. Genet.
20
,
3852
3866
144
Settembre
,
C.
and
Ballabio
,
A.
(
2011
)
TFEB regulates autophagy: an integrated coordination of cellular degradation and recycling processes
.
Autophagy
7
,
1379
1381
145
Medina
,
D.L.
and
Ballabio
,
A.
(
2015
)
Lysosomal calcium regulates autophagy
.
Autophagy
11
,
970
971
146
Medina
,
D.L.
,
Di Paola
,
S.
,
Peluso
,
I.
,
Armani
,
A.
,
De Stefani
,
D.
,
Venditti
,
R.
et al (
2015
)
Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB
.
Nat. Cell Biol.
17
,
288
299
147
Tanaka
,
M.
,
Machida
,
Y.
,
Niu
,
S.
,
Ikeda
,
T.
,
Jana
,
N.R.
,
Doi
,
H.
et al (
2004
)
Trehalose alleviates polyglutamine-mediated pathology in a mouse model of Huntington disease
.
Nat. Med.
10
,
148
154
148
Liu
,
R.
,
Barkhordarian
,
H.
,
Emadi
,
S.
,
Park
,
C.B.
and
Sierks
,
M.R.
(
2005
)
Trehalose differentially inhibits aggregation and neurotoxicity of beta-amyloid 40 and 42
.
Neurobiol. Dis.
20
,
74
81
149
Davies
,
J.E.
,
Sarkar
,
S.
and
Rubinsztein
,
D.C.
(
2006
)
Trehalose reduces aggregate formation and delays pathology in a transgenic mouse model of oculopharyngeal muscular dystrophy
.
Hum. Mol. Genet.
15
,
23
31
150
Sarkar
,
S.
,
Davies
,
J.E.
,
Huang
,
Z.
,
Tunnacliffe
,
A.
and
Rubinsztein
,
D.C.
(
2007
)
Trehalose, a novel mTOR-independent autophagy enhancer, accelerates the clearance of mutant huntingtin and alpha-synuclein
.
J. Biol. Chem.
282
,
5641
5652
151
Rodriguez-Navarro
,
J.A.
,
Rodriguez
,
L.
,
Casarejos
,
M.J.
,
Solano
,
R.M.
,
Gomez
,
A.
,
Perucho
,
J.
et al (
2010
)
Trehalose ameliorates dopaminergic and tau pathology in parkin deleted/tau overexpressing mice through autophagy activation
.
Neurobiol. Dis.
39
,
423
438
152
Perucho
,
J.
,
Casarejos
,
M.J.
,
Gomez
,
A.
,
Solano
,
R.M.
,
de Yebenes
,
J.G.
and
Mena
,
M.A.
(
2012
)
Trehalose protects from aggravation of amyloid pathology induced by isoflurane anesthesia in APP(swe) mutant mice
.
Curr. Alzheimer Res.
9
,
334
343
153
Schaeffer
,
V.
and
Goedert
,
M.
(
2012
)
Stimulation of autophagy is neuroprotective in a mouse model of human tauopathy
.
Autophagy
8
,
1686
1687
154
Schaeffer
,
V.
,
Lavenir
,
I.
,
Ozcelik
,
S.
,
Tolnay
,
M.
,
Winkler
,
D.T.
and
Goedert
,
M.
(
2012
)
Stimulation of autophagy reduces neurodegeneration in a mouse model of human tauopathy
.
Brain
135
,
2169
2177
155
Castillo
,
K.
,
Nassif
,
M.
,
Valenzuela
,
V.
,
Rojas
,
F.
,
Matus
,
S.
,
Mercado
,
G.
et al (
2013
)
Trehalose delays the progression of amyotrophic lateral sclerosis by enhancing autophagy in motoneurons
.
Autophagy
9
,
1308
1320
156
Sarkar
,
S.
,
Chigurupati
,
S.
,
Raymick
,
J.
,
Mann
,
D.
,
Bowyer
,
J.F.
,
Schmitt
,
T.
et al (
2014
)
Neuroprotective effect of the chemical chaperone, trehalose in a chronic MPTP-induced Parkinson's disease mouse model
.
Neurotoxicology
44
,
250
262
157
Li
,
Y.
,
Guo
,
Y.
,
Wang
,
X.
,
Yu
,
X.
,
Duan
,
W.
,
Hong
,
K.
et al (
2015
)
Trehalose decreases mutant SOD1 expression and alleviates motor deficiency in early but not end-stage amyotrophic lateral sclerosis in a SOD1-G93A mouse model
.
Neuroscience
298
,
12
25
158
He
,
Q.
,
Koprich
,
J.B.
,
Wang
,
Y.
,
Yu
,
W.B.
,
Xiao
,
B.G.
,
Brotchie
,
J.M.
et al (
2016
)
Treatment with trehalose prevents behavioral and neurochemical deficits produced in an AAV alpha-synuclein rat model of Parkinson's disease
.
Mol. Neurobiol.
53
,
2258
2268
159
Debnath
,
K.
,
Pradhan
,
N.
,
Singh
,
B.K.
,
Jana
,
N.R.
and
Jana
,
N.R.
(
2017
)
Poly(trehalose) nanoparticles prevent amyloid aggregation and suppress polyglutamine aggregation in a Huntington's disease model mouse
.
ACS Appl. Mater. Interfaces
9
,
24126
24139
160
Mobini
,
M.
,
Radbakhsh
,
S.
,
Kubaski
,
F.
,
Eshraghi
,
P.
,
Vakili
,
S.
,
Vakili
,
R.
et al (
2022
)
Impact of intravenous trehalose administration in patients with Niemann-Pick disease types A and B
.
J. Clin. Med.
11
,
247
258
161
Crippa
,
V.
,
Cicardi
,
M.E.
,
Ramesh
,
N.
,
Seguin
,
S.J.
,
Ganassi
,
M.
,
Bigi
,
I.
et al (
2016
)
The chaperone HSPB8 reduces the accumulation of truncated TDP-43 species in cells and protects against TDP-43-mediated toxicity
.
Hum. Mol. Genet.
25
,
3908
3924

Author notes

*

These authors contributed equally to this work.

This is an open access article published by Portland Press Limited on behalf of the Biochemical Society and distributed under the Creative Commons Attribution License 4.0 (CC BY).