Abstract
Autophagy is a lysosome-based degradative process used to recycle obsolete cellular constituents and eliminate damaged organelles and aggregate-prone proteins. Their postmitotic nature and extremely polarized morphologies make neurons particularly vulnerable to disruptions caused by autophagy–lysosomal defects, especially as the brain ages. Consequently, mutations in genes regulating autophagy and lysosomal functions cause a wide range of neurodegenerative diseases. Here, we review the role of autophagy and lysosomes in neurodegenerative diseases such as Alzheimer disease, Parkinson disease and frontotemporal dementia. We also consider the strong impact of cellular ageing on lysosomes and autophagy as a tipping point for the late-age emergence of related neurodegenerative disorders. Many of these diseases have primary defects in autophagy, for example affecting autophagosome formation, and in lysosomal functions, especially pH regulation and calcium homeostasis. We have aimed to provide an integrative framework for understanding the central importance of autophagic–lysosomal function in neuronal health and disease.
Introduction
Macroautophagy, which is conserved from yeast to humans, is a process whereby cells capture typically cytoplasmic material in double-membraned autophagosomes (Box 1) and then traffic these organelles to perinuclear sites, where lysosomes are concentrated. This is followed by autophagosome–lysosome fusion, after which the autophagic contents are degraded by lysosomal hydrolases and the building blocks such as amino acids or sugar moieties are recycled (Fig. 1).
Box 1 |. Recent insights into autophagosome formation.
Autophagosomes evolve from open, finger-shaped, double-membrane structures called phagophores56. These originate from recycling endosome membranes where phosphatidylinositol 3-phosphate (PI3P) synthesis by a Beclin 1-dependent kinase complex enables PI3P-dependent and RAB11-dependent recruitment of WD repeat domain phosphoinositide-interacting protein 2 (WIPI2)299. WIPI2 recruits the ATG5–ATG12–ATG16L1 complex that conjugates ATG8 or LC3 family members to phagophore membranes — this ubiquitination-like conjugation event represents a defining step in autophagosome formation46. The phagophores have finger-like morphologies and do not appear to be single-opening cup-shaped structures, as they are conventionally portrayed. These ‘fingers’ close analogous to how one would make a fist in an endosomal sorting complexes required for transport (ESCRT)-dependent manner to enable autophagosome closure, a prerequisite for subsequent release from the recycling endosome56.
Most, if not all, autophagosomes are derived from RAB11-positive recycling endosome membranes, while receiving inputs from other membrane compartments299,300. For example, the endoplasmic reticulum (ER) is in close proximity to phagophores and regulates the early stages of autophagosome biogenesis301 (for example, the autophagy protein ATG2 transfers lipids between the ER and phagophores302), as do secretory pathway-derived machinery and regulators303,304. However, these organelles have distinct roles from the RAB11A membranes that are conjugated by LC3 and other ATG8 family members to evolve into autophagosomes.
Autophagosome release from the recycling endosome compartment is mediated by dynamin 2 (ref. 57). The autophagosomes are then trafficked on microtubules by the dynein machinery to the part of the cells where the lysosomes are concentrated, within the neuronal cell body58. Autophagosome–lysosome fusion appears to be mediated by kiss-and-run repeated interactions305 although such repeated interactions between these organelles have not been studied extensively in different cell types. The productive fusion leading to autolysosome formation depends on multiple machineries, including certain RAB proteins (for example, RAB7), phosphoinositides (for example, phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2)), tethering complex components and soluble N-ethylmaleimide-sensitive factor attachment protein receptors (SNAREs)306.
Fig. 1 |. Autophagy–lysosomal pathway in neurons.
Macroautophagy is initiated by the formation of a double-membrane enveloping structure, the phagophore, which occurs both in the synapse and throughout the neuron. However, most autophagosome formation appears to occur in the distal axon. Cytoplasmic constituents and organelles targeted for degradation are captured within an autophagosome, which is formed as the phagophore elongates and encircles the substrates. The autophagosome is degraded upon fusion with a lysosome to form an autolysosome. This process introduces acid hydrolases and proton pumps (vacuolar H+-ATPase (v-ATPase)), leading to luminal acidification, thereby activating an array of hydrolases that can fully digest most substrates into their building blocks (for example, amino acids), which are recycled for energy or new synthesis. An intermediate step particularly prevalent in the processes of neurons (in both dendrites and axons) is the fusion of an autophagosome with a RAB7-positive late endosome to form an amphisome, which shows high dynein-based retrograde motility17,278. In chaperone-mediated autophagy (CMA), proteins carrying a KFERQ-like sequence are recognized by the chaperone heat shock cognate 70-kDa protein (HSC70), which can then associate with the integral lysosomal-associated membrane protein 2A (LAMP2A). This interaction triggers its oligomerization, enabling translocation of the bound substrate into the lysosome. ‘Heterophagy’ via the endolysosomal pathway involves lysosomal degradation of plasma membrane components and exogenous substrates after they are internalized by receptor-mediated or bulk endocytosis. Cargo proteins are sorted selectively to different cellular destinations or recycled to the plasma membrane (not shown). Proteins targeted for degradation are trafficked to late endosomes or multivesicular bodies (MVBs), which mature to lysosomes, enabling complete degradation of cargo proteins. A build-up of lipofuscin reflecting normal presence of internal waste storage in this compacted form increases as neurons age (see details in main text). The figure illustrates unique features of the autophagy–lysosomal pathway in neurons, which are asymmetric postmitotic cells that cannot be replaced and therefore must maintain efficient proteostasis over many decades. Sites of highly active autophagy and endocytosis are separated by long distances from the most active degradative capability located in the soma where lysosomes are concentrated, implying a strong reliance on efficient retrograde transport — a process commonly affected in disease. The absence of mature fully activated lysosomes along axons limits the capability of a neuron for eliminating waste during the lengthy transit of autophagosomes and amphisomes until they reach lysosomes in the soma. Anterogradely moving Golgi carrier vesicles deliver lysosomal components to the amphisomes and late endosomes to facilitate their maturation towards a lysosome identity, but this does not suffice for efficient substrate degradation. The most vulnerable neuron populations in neurodegenerative disease are often ones with a very high axon to somal volume ratio requiring autophagic quality control over huge cytoplasmic volumes. ER, endoplasmic reticulum.
Autophagy is an important intracellular clearance route for intracytoplasmic aggregate-prone proteins, certain pathogens and dysfunctional organelles such as mitochondria. The accumulation of aggregate-prone proteins is a hallmark of most neurodegenerative diseases. Usually, these aggregates are intracellular but sometimes, as in the case of Alzheimer disease (AD), amyloid-β also appears extracellularly and the intraneuronal tau aggregates persist in the extracellular space after the neuron dies1. Most of these aggregate-prone proteins cause disease through toxic gain-of-function mechanisms and their pathogenicity is associated with their propensity to aggregate. Cells buffer themselves against such aggregate-prone proteins through the proteostasis network, which comprises components that regulate the synthesis, folding, aggregation and disposal of proteins, including through autophagy2.
In addition to macroautophagy, cytoplasmic proteins can be delivered to lysosomes via other ‘autophagy’ pathways. These include non-canonical autophagy, a poorly understood phenomenon where macroautophagy-like processes appear to occur in specific contexts in the absence of some of the core autophagy machinery3. In chaperone-mediated autophagy (CMA), substrates containing a KFERQ or similar pentapeptide motif are recognized by the heat shock cognate 70-kDa protein (HSC70)4. This allows targeting of such HSC70-bound proteins to the lysosomal-associated membrane protein 2A (LAMP2A) that acts as a translocation complex to enable import into the lysosome, where the substrates are recognized by lysosomal HSC70 (ref. 4). Although CMA is not the focus of this Review, it is important to stress that the efficiency of this pathway declines with neuronal ageing and contributes to the accumulation of aggregate-prone proteins in the brain5. Similarly, its activity is compromised by various disease-causing proteins, such as α-synuclein, tau and huntingtin (reviewed elsewhere4). In microautophagy and endosomal microautophagy, substrates are captured by lysosomes and endosomes through the formation of invaginations of their membranes4. The roles of these pathways in mammalian neurodegenerative diseases are still poorly understood and will not be discussed here. In the following, we refer to macroautophagy as autophagy for simplicity.
Although it is convenient to consider the different components of the autophagy–lysosome network as separate, they frequently engage in significant cross-talk. For example, mTORC1 positively regulates protein synthesis while restraining protein degradation via the autophagy–lysosome pathway6. Conversely, mTORC1 inhibition stimulates both autophagosome and lysosome biogenesis6. mTORC1 activity is enabled by the binding of its subunit Raptor to Rag proteins on the lysosome, as part of its nutrient-sensing functions7,8. Indeed, lysosomal mTORC1 tethering and mTORC1-containing lysosomal localization in the cell (perinuclear versus peripheral) are critical factors regulating mTORC1 activity and, consequently, autophagy9,10. Similarly, consideration of the key degradation pathways used by cells shows that proteasome inhibition activates macroautophagy11, whereas autophagy inhibition impairs flux in the ubiquitin–proteasome pathway12 (Fig. 1).
The autophagy–lysosome pathway is not simple or unidirectional. For example, the lysosome has emerged as a key signalling hub enabling activation of crucial autophagy regulators such as mTORC1 (ref. 13) and alterations of lysosomal distribution in cells can impact mTORC1 activity and autophagosome biogenesis, further underscoring the interconnections of this network10. Many cross-dependencies also exist between the autophagy–lysosome pathway and the endosomal–lysosomal system. For example, proper lysosome function depends on endocytosis, which is crucial for autophagosome biogenesis14. In addition, autophagy can impact aspects of endocytic trafficking15. Although this Review focuses squarely on the pathway from autophagosomes to lysosomes, it is reasonable to consider both pathways to lysosomes as an integrated endosomal–lysosomal–autophagy (ELA) network. Further details on the autophagy interface with the endocytic pathway to lysosomes are described in Box 2.
Box 2 |. Autophagy and endosomal pathways are tightly integrated and equally vulnerable.
Recent studies have amplified the number of known routes capable of sequestering and delivering damaged, toxic or obsolete cellular constituents to lysosomes for degradation and recycling. This includes the endosomal–lysosomal pathway. Although not formally considered part of the autophagy system, in fact the pathway tightly integrates with autophagy by contributing membrane components during autophagosome formation and by providing an alternative cytoplasmic substrate sequestration site. Indeed, as autophagosomes are derived from RAB11-positive recycling endosomes, some transmembrane proteins that traffic via endocytosis from the plasma membrane to recycling endosomes, such as C–C chemokine receptor type 5 (CCR5) and the transferrin receptor, are degraded via autophagy as part of the inner autophagosome membrane after endocytosis from the plasma membrane33,299. This contrasts with the canonical model of macroautophagic substrate captured by engulfment within autophagosomes. The interface with autophagy extends the role of endocytosis as a sorting mechanism for delivery of internalized cargoes to varied cellular destinations to include the routing of non-essential endocytosed materials to lysosomes for clearance.
Another interface between endolysosomal and autophagy pathways is when endosomes fuse with autophagosomes to form a hybrid organelle, the amphisome, which is especially important in neurons. Indeed, most of the autophagosomes generated in axonal terminal regions quickly acquire late endosomal markers (for example, RAB7), which engage the dynein complex and accelerate their retrograde transport to the soma17. Endosomes also serve as a default waste clearance site by providing exit routes from cells via exocytosis and the release of cargo-containing exosomes from multivesicular bodies (MVBs). This process is stimulated when autophagosome or endosome fusion with lysosomes is compromised307. Autophagy also shares machinery with endocytic-like pathways such as LC3-associated endocytosis and LC3-associated phagocytosis, where components of the LC3-conjugation system (but not the entire broader canonical autophagy machinery) are co-opted to these vesicles. This pathway probably has relevance to neurodegeneration, as LC3-associated endocytosis helps clearance of amyloid-β and protects against neurodegeneration in Alzheimer disease (AD) mouse models308. Conjugation of ATG8 (LC3) family membranes to single membranes (CASM) also occurs with other scenarios, including during membrane repair processes in damaged lysosomes (CASM) and in LC3-associated micropinocytosis309.
Given the tight integration of the endolysosomal pathway with autophagy, bidirectional interplay between these two pathways in addition to independent pathogenic contributions may substantially impact clearance. A few examples are briefly mentioned here but are reviewed in detail elsewhere310–313. One example is exocytic release of cargoes from late endosomes via either exosomes314,315, autophagosomes or amphisomes, which is activated as a default mechanism for waste clearance when delivery to lysosomes or subsequent degradation stalls in disease states316,317. Whether neurons can discard waste from lysosome-related compartments, as actively as occurs in non-neuronal cells from individuals with lysosomal storage disorders (LSDs), is less clear although likely. Another default pathway is inter-neuronal transfer of proteins released by exosomes or other exocytic paths. This occurs physiologically at low levels318, but is enhanced as another default clearance option that helps clear toxic proteins. However, this process may also propagate disease to other brain regions when the pathogenic protein (for example, synuclein, tau) is endocytosed and delivered to lysosomes of unaffected neurons319.
In this Review, we highlight how the autophagy–lysosome system is involved in various neurodegenerative diseases. We provide examples that illustrate key concepts but do not aim to be encyclopaedic, as this approach risks obscuring the key broad concepts we aim to convey. We have focused particularly on the roles of the autophagy–lysosome system in neurons. Although the roles of glia in neurodegeneration are becoming more apparent, the understanding of the autophagy–lysosome system in these cells is less developed in the context of neurodegenerative diseases. We will also discuss and speculate on the interplay between the autophagy–lysosome pathway and ageing in neurodegeneration, and the impact on specific neuronal and cell-type vulnerabilities in different diseases, and consider some challenges when aiming to exploit the therapeutic potential of this pathway in different diseases.
The autophagy–lysosome system in neurons
Much of what we know about autophagy has come from characterizations in non-neuronal cells. Newer studies have revealed a more complex autophagy–endolysosomal topography and more dynamic behaviour in neurons compared with other cells, reflecting adaptations to accommodate the extreme asymmetrical shape of neurons and their much larger cytoplasmic volumes.
The extreme lengths of projecting axonal and some dendritic processes, combined with total volumes as much as 10-fold to 50-fold larger than that of the neuronal cell body or the average size of glial cells16, make neurons especially dependent on efficient protein quality control mechanisms, including autophagy. This challenge is unique to neurons, as they must traffic autophagosomes and endosomes across long distances to reach the degradative lysosomes that are found in the cell body17. This challenge is partly the consequence of endosomes moving retrogradely along axons in vivo and only achieving mature lysosome identity and acidification as they reach their somal destination18–20. Once lysosomes are fully acidified and activated, they do not return to axons, explaining why lysosomes are rare in axons18,19 and relatively sparse in dendrites of the adult brain21. Instead, LAMP1-positive and cathepsin-positive Golgi carriers predominate in axons and, as in other cell types, these are the vesicles that deliver lysosomal components to axonal endosomes during lysosomal maturation18.
These adaptations make neurons especially vulnerable to autophagy–endolysosomal disruption and explain why gene mutations targeting dynein-based retrograde axonal transport mechanisms cause varied neurodegenerative disorders22 (Fig. 2). In addition, being predominantly postmitotic also prevents most populations of neurons from reducing cytoplasmic waste by cell division or by being replaced with a new neuron as is possible with the mitotic cells of the brain23. The need of neurons to survive for the length of an individual’s lifetime, yet with exceedingly limited capacity for neurogenesis, makes them exceptionally susceptible to the cumulative effects of cellular ageing, especially oxidative stress24, and to the emergence of late-life neurodegenerative diseases. The deterioration of autophagy and lysosomal functions, which is pivotal to the mechanism underlying cellular ageing, is central as a precipitant of overt disease in these late-life disorders. A better understanding of the molecular basis responsible for considerable deterioration of lysosomal function in the ageing brain requires the development of more cell-specific probes for this incipient pathobiology applied to in vivo model systems, rather than cultured cells, to appreciate the extent of its synergies with disease factors and what interventions may be effective to preserve lysosome health in normal ageing.
Fig. 2 |. Causal genes for adult-onset neurodegenerative disease commonly disrupt autophagic–lysosomal function.
Principally mutated genes considered causative for selected major adult-onset neurodegenerative diseases such as Alzheimer disease (AD), Parkinson disease (PD) and frontotemporal dementia (FTD) as well as several examples of proteins believed to contribute to disease are highlighted, where the evidence strongly pinpoints a critical pathogenic mechanism within the endosomal–lysosomal–autophagy (ELA) axis. There are additional unlisted genes featuring polymorphisms or other modifications that increase the disease risk but have smaller effect sizes or where the mechanism of pathway disruption is less well defined. The listed genes disrupt a broad range of lysosome-related pathways, including lysosome or autophagosome biogenesis, chaperone-mediated autophagy (CMA), substrate capture, mitophagy and macroautophagy, and trafficking routes, but mechanistic convergence at the lysosome is most common, causing failure to properly clear potentially toxic substrates and maintain important lysosomal-related cellular homeostatic signalling mechanisms. Although the diagram and this Review focus on this dysfunction in neurons, which is most closely linked to clinical progression of neurodegenerative disease, the less well characterized autophagy–lysosomal impairments of clearance mechanisms in microglia, astrocytes and vascular cells undoubtably contribute to disease progression. ALS, amyotrophic lateral sclerosis; APP, β-amyloid precursor protein; ER, endoplasmic reticulum; GBA, glucosylceramidase; GRN, granulin; HSC70, heat shock cognate 70-kDa protein; HD, Huntington disease; LAMP2A, lysosomal-associated membrane protein 2A; LRRK2, leucine-rich repeat serine/threonine-protein kinase 2; MVB, multivesicular body; NCL, neuronal ceroid lipofuscinosis; PSEN1/2, presenilin 1/2.
Brain ageing in adult-onset neurodegenerative diseases
The direct neurotoxic actions of a ‘pathogenic protein’ are often blamed as the sole or primary cause of adult-onset neurodegenerative diseases. Less commonly considered is that the toxic form of the protein is usually produced in comparable amounts throughout life and is handled effectively without evident toxicity during early life. Disease emergence during adulthood coincides temporally with the progressive failure of proteostatic systems and, especially, autophagy at all levels of the pathway, which is facilitated by poorly understood ageing factors. For example, autophagosome biogenesis appears to decrease in mammalian brains and neurons with age25,26 and the delivery of waste-carrying vesicles to lysosomes slows26. A particularly critical driver of cell ageing in various lower species such as Caenorhabditis elegans27,28 and probably mammals29 is declining lysosomal function linked to failing intralumenal acidification26, which is believed to be responsible for the initial build-up of multiple pathogenic aggregation-prone proteins (β-amyloid precursor protein (APP) peptides (for example, amyloid-β and APP carboxy-terminal fragment (APP-βCTF), TAR DNA-binding protein 43 (TDP43), synuclein)) in the brain upon normal ageing. During normal brain ageing, the coalescence of ineffective autolysosomes containing hydrolysis-resistant substrates enables the compaction through extensive cross-linking modifications of the undigested cargoes within lipofuscin granules30. This process renders the substrates less bioactive and capable of release, which reduces, but does not eliminate, the damaging impact on cell function. The gradual accumulation of lipofuscin granules is a well-established correlate of neuronal age in many cell populations.
Many neurodegenerative diseases are probably driven by the accumulation of an autophagy substrate that itself compromises autophagy modestly (for example, mutant huntingtin accelerating the degradation of the autophagy regulator Beclin 1 (ref. 31)), providing the potential for positive feedback. This is seen with α-synuclein in Parkinson disease (PD)32, tau in various dementias33 and mutant huntingtin in Huntington disease (HD)31. It is likely that neurons maintain functional autophagy and/or buffer the accumulation of these proteins for many years by using other proteostasis mechanisms, including the ubiquitin–proteasome pathway and CMA. However, once these mechanisms can no longer manage the accumulating aggregate-prone proteins, more autophagy substrates will accumulate, leading to further autophagy impairment including failing mitophagy34. This creates a positive feedback loop as the toxic substrates accumulate more and boost oxidative stress35. This may explain why neurodegenerative diseases strike in late adulthood and that even individuals with autosomal dominant mutations in huntingtin, tau or α-synuclein are functionally normal until disease onset.
In the absence of a highly penetrant disease-causing gene mutation, declining function of autophagy and particularly lysosomes, known to be driven by ageing-related factors such as oxidative damage to substrates and ELA network components24,35–38, represents a basis for the emergence of manifest disease. In so-called sporadic forms of neurodegenerative diseases, a summation of ageing-related factors such as oxidative damage to substrates and components of the ELA network synergize with environmental and genetic risk factors to initiate disease. In this context, it is noteworthy that so many of the risk genes for the two most studied major late-life neurodegenerative disorders, AD and PD, have a direct gain-of-function or loss-of-function impact on the ELA network, the effects of which can summate or synergize. A pivotal role of ageing in triggering neurodegenerative disease critically involving lysosomal mechanisms is consistent with genes that cause a congenital devastating lysosomal storage disorder (LSD) when homozygously mutated, but are also major risk factors for a late age-onset neurodegenerative disorder, such as PD (for example, GB1) or frontotemporal dementia (FTD) (for example, GRN), when only one mutant copy is inherited39,40.
Defects in upstream autophagy in neurodegenerative disease
As strong genetic evidence implicates proteins of the autophagy–lysosome pathway and the broader ELA network in neurodegenerative disease pathogenesis, it is crucial to understand how this network maintains cell homeostasis and how its disruption may impact a wide range of neuronal and glial functions in disease and cause resulting phenotypes.
Many Mendelian neurological or neurodegenerative diseases, including HD, certain forms of parkinsonism and forms of amyotrophic lateral sclerosis (ALS) reviewed previously4, are caused by mutations that compromise autophagy either at stages of autophagosome biogenesis or at subsequent steps that orchestrate effective autophagic substrate degradation in autolysosomes. Mouse models with defects in these early steps suggest that they are sufficient to cause neurodegeneration41,42 and can partially explain the accumulation of aggregate-prone proteins in these conditions. However, the mutated proteins often have roles in other membrane trafficking processes, including alternative lysosomal degradation systems, and the combined deficits in multiple pathways result in multifactorial pathology.
In the following, we provide some examples of Mendelian diseases causing defects at different stages of the macroautophagy pathway (Fig. 1), using neurological or neurodegenerative diseases where possible. It is interesting to note that, in some cases, human mutations have helped illuminate new steps in autophagosome biogenesis (Box 1). Within this network of systems available for capturing substrates, lysosomes deserve special emphasis as the only degradative compartment shared by all autophagy and endocytic delivery routes. Investigators often incorrectly equate ‘autophagy’ with just the substrate sequestration steps of the process even though the process derives its name from its digestive ‘phagy’ (‘eating’) lysosomal step. Despite its obvious redundancy, a useful convention has often been to refer to an ‘autophagy–lysosomal pathway’ to underscore the crucial importance of the degradative step in autophagy. For the discussion below on neurodegenerative diseases, we find it useful to first consider the autophagy pathway defects affecting upstream steps of the pathway followed by the more extensive dysfunctions affecting the digestive lysosomal stage of autophagy, bearing in mind that both are integral stages of the same pathway.
Autophagosome initiation
Homozygous mutations in ATG5 (encoding autophagy protein 5)43, WIPI2 (encoding WD repeat domain phosphoinositide-interacting protein 2)44 and ATG7 (ref. 45), which are involved in the biogenesis of the phagophore, that is, the open autophagosome precursor, cause human diseases. ATG5 and ATG7 contribute to the formation of the ATG5–ATG12–ATG16L1 complex, which serves as the E3-like enzyme for the ubiquitin-like conjugation of LC3 family members to the lipid phosphatidylethanolamine in recycling endosome membranes, whereas the recruitment of the ATG5–ATG12–ATG16 complex to these membranes is mediated by WIPI2 (ref. 46). Mutation of ATG5 causes congenital ataxia, mental retardation and developmental delay, and loss of ATG7 is associated with complex neurodevelopmental disorders with brain, muscle and endocrine involvement45. The loss of WIPI2 function also causes a neurodevelopmental disorder that includes developmental delay, speech and language impairment, and cardiac, neurological, thyroid and skeletal abnormalities44. It is surprising that the loss of function of these core autophagy genes results in neurodevelopmental abnormalities in humans that are not obvious in autophagy-null mice47, where loss of autophagy causes neurodegeneration41,42. However, it is possible that neurodevelopmental signs were overlooked in whole body autophagy-null mice that die soon after birth, and that humans with ATG5, ATG7 and/or WIPI2 mutations may develop neurodegenerative features as they age, as they survive unexpectedly into quite advanced age (eighth decade of life in individuals where ATG7 is reduced and third decade where ATG7 is undetectable)45. As there are minimal longitudinal phenotypic data on such cases and no post-mortem analyses available, it is not possible to know if and when any progressive neurodegeneration may start to manifest. In some of these ultra-rare diseases, there may be insufficient neuropathological assessment so neurodegeneration may well have been missed.
Autophagosome closure and scission
The closure of the multiple openings between phagophore ‘fingers’ that enables sequestration of their substrates from the cytoplasm is mediated by the endosomal sorting complexes required for transport (ESCRT) complex48. When this step is blocked, autophagic flux is impaired and autophagic cargoes accumulate48. Mutations in the ESCRT complex cause diverse diseases, including forms of neurodegeneration, such as FTD (for example, CHMP2B), and it is possible that the autophagy defects have an important role in pathogenesis49. However, the ESCRT complex also has other roles, including in the formation of multivesicular bodies (MVBs) in endosomes, a route for degradation of ubiquitinated cargo receptors enabled by invagination and inward budding of vesicles into endosomes49. After fusion of endosomes with lysosomes, these vesicles and their cargoes are degraded. The ESCRT complex has also been shown to participate in the repair of lysosomal damage50,51, a process in which PD-related genes such as leucine-rich repeat serine/threonine-protein kinase 2 (LRRK2) have been implicated52–54. Further, disruption of the ESCRT pathway has been reported to promote endolysosomal escape in tau propagation, with tau aggregation being one of the hallmarks of AD55. Thus, non-autophagic functions of the ESCRT complex also probably contribute to these diseases — a principle that applies to some of the examples below.
Following autophagosome closure, they need to be released from the RAB11A-based recycling endosome platform from which autophagosomes evolve56 by dynamin 2-dependent scission57. A DNM2 mutation that targets this recently described step in the autophagy pathway causes centronuclear myopathy57 and has helped define this new rate-limiting step in autophagosome formation — the scission of closed autophagosomes from the recycling endosome compartment. So far, neurodegenerative diseases have not been associated with this step specifically. However, as autophagosome closure (which is affected in various neurodegenerative diseases described above) is a prerequisite for release from the recycling endosome, such closure-defective mutations result in the accumulation of open autophagosome precursors still attached to the recycling endosome56.
After closed autophagosomes are released from the recycling endosomes, they need to be trafficked by dyneins along microtubules in a retrograde direction to the perinuclear region of the cell where the lysosomes are clustered, to facilitate autophagosome–lysosome fusion58 (Fig. 2). This transport machinery is defective due to mutations in the dynein complex in Perry syndrome, which manifests with parkinsonism59,60, and results in impaired autophagosome–lysosome fusion.
Defects associated with autophagy receptors
Originally, most of the focus of autophagy research was on its role as a non-selective bulk degradation process. However, it is becoming increasingly evident that autophagosomes can selectively engulf substrates including aggregate-prone proteins (aggrephagy)61, dysfunctional mitochondria (mitophagy)62 and the endoplasmic reticulum (ER-phagy)63. The selectivity is mediated by so-called autophagy receptors that link the substrates (normally ubiquitinated proteins) to the autophagy machinery (such as LC3 family members). An ALS and FTD-associated mutation in p62 (also known as SQSTM1), the first of these receptors to be described, compromises selective autophagy of ubiquitinated proteins64. Optineurin is a key mitophagy receptor, whose phosphorylation by the serine/threonine-protein kinase TBK1 is crucial for its receptor role in mitophagy65. Mutations in the genes encoding optineurin and TBK1 can cause ALS or FTD65. Mutations of two proteins that act together to regulate ER-phagy, reticulophagy regulator 1 (also known as FAM134B) and ADP-ribosylation factor-like protein 6-interacting protein 1 (ARL6IP1), cause sensory neuropathy66,67. Such mutations have the potential to reveal important physiological roles of these forms of selective autophagy in neurological function.
Although the effect of some ER-phagy pathways on sensory neuropathies suggests the importance of this process in the peripheral nervous system, it is difficult to know how important loss of this selective autophagy subtype is for the genesis of neurodegenerative diseases of the central nervous system. Previously, much of the support for the idea that mitophagy was critical in neurons came from studies of two genes involved in early-onset parkinsonism, PINK1 (encoding the mitochondrial serine/threonine-protein kinase PINK1) and PRKN (encoding Parkin) which act in the same mitophagy pathway68. However, this particular mitophagy pathway may not be dominant in the brain, compared with other forms of mitophagy69, and PINK1 and PRKN may have mitophagy-independent roles as regulators of mitochondrial biogenesis70. Indeed, recent data suggested that nucleoid-enriched mitochondrial fragments are predominant cargoes in mouse brain autophagosomes in the absence of autophagy receptors. The authors speculate that this is due to mitochondrial fission occurring close to sites of non-selective autophagosome formation71. Moreover, distinct from previously described extracellular vesicle subtypes (for example, microvesicles, exosomes), these ‘mitovesicles’72 are upregulated in brains derived from a mouse model of Down syndrome, where mitochondrial damage is a prominent anomaly72. These studies suggest that bulk autophagic clearance of mitochondria is important for neuronal health.
Defects affecting multiple steps of the autophagy–lysosomal pathway
Some mutations associated with neurodegenerative disease impact multiple processes in autophagy and this may be more prevalent than one would first imagine. One reason is that key regulators of autophagy, such as mTORC1, act at multiple stages of the pathway73,74. Mutant huntingtin may regulate autophagy at multiple stages from autophagosome biogenesis by enhancing Beclin 1 degradation31, cargo selection possibly because of abnormal binding of mutant huntingtin to the cargo receptor p62 (ref. 75), to autophagosome trafficking to lysosomes76. In addition, the AD risk gene PICALM, encoding phosphatidylinositol-binding clathrin assembly protein, regulates both autophagosome biogenesis and autophagosome clearance by lysosomes by controlling the endocytosis of soluble N-ethylmaleimide-sensitive factor attachment protein receptor (SNARE) proteins77. Such SNARE proteins are required for the homotypic fusion between plasma membrane-derived ATG16L-containing vesicles and heterotypic fusions between ATG16L1-containing and ATG9A-containing vesicles that are required prior to the initiation of autophagosome formation78,79. In addition, SNAREs, whose trafficking is regulated by PICALM, impact autophagosome–lysosome fusion77. These upstream impairments compound the range of deficits corrupting the degradative steps in AD discussed below.
Most of the focus of the field has understandably been on cell-autonomous effects of disease-modifying or disease-causing factors on autophagy. However, neurons exist within an environment containing many different glial cells and recent data have suggested that surrounding cells may impact neuronal vulnerability in disease80. In this context, nitric oxide and chemokines such as C–C motif ligand 3 (CCL3), CCL4 and CCL5, which are secreted by reactive microglia, impair neuronal autophagy and countering these effects was found to be protective in disease models33,81. CCL3, CCL4 and CCL5 inhibit neuronal autophagy by binding and activating the GPCR C–C chemokine receptor type 5 (CCR5) on neurons33, which may represent the deleterious effect of chronic activation of a pathway that has been suggested to assist repair after acute neuronal injury82. These are mechanisms whereby microglia can accelerate neurodegeneration in models of HD and tauopathy by compromising neuronal proteostasis, driving the accumulation of the disease-causing protein. In the context of a mouse model of AD, autophagy buffers against microglial senescence and ameliorates extracellular amyloid-plaque pathology83.
Lysosomal defects in neurodegenerative disease
As the indispensable ‘downstream’ degradative (‘phagy’) step in autophagy (Fig. 1) and endosomal cargo clearance pathways, lysosomes are essential for neuronal survival. Lysosomal dysfunction in adult-onset neurodegenerative ‘proteinopathies’, such as AD, PD, HD and frontotemporal lobar degeneration (FTLD) (Fig. 2), was long presumed to be a later secondary response to eliminate aggregates and damaged organelles in severely compromised neurons. Newer evidence, however, showed that, for major disorders such as AD, PD and FTD, lysosomal components commonly are the primary target of causative and risk gene mutations84,85, as discussed below. The resulting lysosomal dysfunction drives disease onset by allowing pathogenic forms of the mutant protein and other substrates to resist breakdown, to become (further) covalently modified by reactive oxygen species (ROS), and to accumulate as even more toxic fragments or aggregates compared with the original pathogenic form. Combined effects from these instigating processes are essential to drive the downstream pathobiology leading to neuronal death.
The apt description of lysosomes by their discoverer, Christian de Duve, as potential ‘suicide bags’ anticipated the vulnerability of this fundamental degradative process in degenerative diseases86,87. This is now well validated and >40 lysosomal genes are known to cause neuron loss in inheritable neurodegenerative disorders across the age spectrum88. Further validating the link between lysosome-based mechanisms and adult neurodegenerative disease are lysosomal gene mutations that in homozygous form cause congenital LSD with severe neurodegenerative or neurodevelopmental phenotypes, but in heterozygous form cause an adult-onset neurodegenerative disease or substantially raise its risk39,40. Pathogenic mechanisms underlying varying major late age-onset neurodegenerative diseases are converging on common themes of pH and ion balance dysregulation that disrupt not only lysosomal hydrolytic enzyme function broadly but also the diverse signalling roles of the lysosome and related acidic vesicular compartments, as discussed below, which together instigate and further drive disease progression.
Lysosomal acidification and neuronal homeostasis failure in adult neurodegenerative diseases
Lysosomes maintain cellular homeostasis through the complex coordination of their essential roles in substrate hydrolysis to generate energy, the controlled release of nutrient transporters and signalling factors, which together enable metabolic coordination with other organelles to regulate vesicle dynamics and signalling events, and specific gene expression programmes. This range of physiological functions, beyond the scope of this Review of their implications for neurodegeneration, underscore the centrality of lysosome in metabolic regulation7. Among the lysosomal properties most crucial to this orchestration of neuronal functions and, arguably, the one most significantly implicated in disease pathogenesis is the strongly acidic intraluminal pH of the lysosome (pH 4.3–5.0)89, which is essential for hydrolytic enzyme activation, complete digestion of substrates, H+-coupled nutrient and drug transport, and the varied homeostatic signalling functions30,90,91. Acidification is achieved by the ATP-dependent proton pump, vacuolar H+-ATPase (v-ATPase)92,93, a 14-subunit complex regulated mainly by the association and dissociation of 2 subcomplexes. One of these subcomplexes (V1) is cytoplasmic and provides the ATPase activity needed to generate torque that opens a channel within the membrane-spanning V0 subcomplex (Fig. 3). The V0a1 subunit of the V0 subcomplex is especially critical because it tethers V1 to the membrane-anchored V0 subcomplex94. Not surprisingly, this subunit is broadly implicated in the pathogenesis of neurodegenerative diseases95. In addition to the v-ATPase complex, an array of ion channels in the lysosomal membrane are now known to influence lysosomal proton content and finely modulate lysosomal pH, including LAMPs, which were previously viewed only as structural proteins96. Remarkably, the varied modulators of ion balance within lysosomes have themselves been recently implicated as causative for major neurological diseases (Fig. 4).
Fig. 3 |. v-ATPase and lysosome acidification in adult neurodegenerative disease.
The vacuolar H+-ATPase (v-ATPase) complex is composed of an extralumenal V1 domain (consisting of subunits A–F) and an integral membrane-associated V0 domain (composed of subunits a, c, c″, d and e)279. ATP hydrolysis by the V1 subcomplex opens a channel within the V0 subcomplex through which protons pass. Proton pump activity is regulated by the reversible dissociation of the V1 and V0 domains. Reversible dissociation is rapid, does not require new protein synthesis and occurs in response to diverse cellular signals that toggle v-ATPase activity up or down. A few examples are nutritional states and assembly factors such as phosphatidylinositol 3-kinase, mTORC, cAMP, PKA, glucose and amino acid levels. Owing to its interactions with V1 domain subunits that mediate full complex assembly and proton pump activity, the V0a1 subunit is an especially vulnerable target in many diseases. Most of the proteins listed in the box interfere at different stages of V0a1 subunit maturation, complex assembly or interaction with the V1 subcomplex and their mutation or loss elevates pH in disease. The figure illustrates primary actions of two key proteins causing Alzheimer disease (AD), namely β-amyloid precursor protein (APP) and presenilin 1 (PSEN1). Disease variants of PSEN1 and APP directly disrupt v-ATPase activity and cause lysosomal pH elevation responsible for autophagy clearance failure, intracellular amyloid formation and, ultimately, neuronal cell death and extracellular senile (amyloid) plaque formation131 (more details in text and Fig. 6). PSEN1 deletion or mutations causing AD, which are loss-of-function defects, impair the activity of the presenilin 1 holoprotein in the endoplasmic reticulum (ER), which acts as a chaperone for V0a1 subunit glycosylation (by the oligosaccharyltransferase (OST) complex) and folding. The instability of the subunit structure triggers premature degradation by endoplasmic reticulum-associated degradation (ERAD) and results in insufficient delivery of V0a1 subunits for assembling adequate complexes on lysosomes and ensuring proper proton pumping activity. In addition, PSEN1 has a separate function in the cleavage of APP: the initial cleavage of APP by β-secretase (BACE) generates a carboxy-terminal fragment (APP-βCTF, along with the cleavage product sAPP-β) that, in its constitutively phosphorylated form (pTYR682), specifically binds to V0a1 and impedes V1 subcomplex association with V0, leading to similar pathological consequences as in the mutant PSEN1 loss-of-function (PSEN1-LoF) condition. Levels of APP-βCTF and pTYR682-βCTF are substantially elevated in early-onset AD due to its mutation-mediated overproduction and impaired turnover. In the common late-onset forms of AD, neuronal APP-βCTF is also elevated due to multiple factors that include overproduction based on elevated β-cleavage of APP and its accumulation in the failing lysosomes of vulnerable neuron populations121,132,133,136,139,280–292, setting up a vicious cycle similar to that triggered in early-onset AD. ALS, amyotrophic lateral sclerosis; FTD, frontotemporal dementia; GBA, glucosylceramidase; GRN, granulin; KO, knock out; LRRK2, leucine-rich repeat serine/threonine-protein kinase 2; LSD, lysosomal storage disorder; PD, Parkinson disease; TMEM106B, transmembrane protein 106B.
Fig. 4 |. Lysosomal ion imbalances in neurodegenerative disease.
Effective lysosomal acidification requires not only the vacuolar H+-ATPase (v-ATPase) described in Fig. 3 but also counterion flows involving chloride, potassium, sodium and, possibly, other ions. This figure depicts components of the lysosome implicated in neurodegenerative disease that affect lysosomal ion flux, including v-ATPase, the non-selective cation channels transient receptor potential channel mucolipin 1 (TRPML1) (which transports mainly Ca2+ but also other cations, for details see text) and two-pore channel (TPC2). TPC2 acts as a Ca2+-permeable channel when activated by NAADP190,221,293,294 and can act as a Na+ channel when activated by phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2). In addition, the ion transporters H+/Cl− exchange transporter 7 (ClC7), ClC5 that transports Cl− and the K+ channel TMEM175 that also acts as a proton leak channel are found in the lysosomal membrane. Arrows indicate the direction of ion fluxes. Further details on their roles in neurodegenerative diseases are provided in the text. Lysosomal pH is also influenced by Donnan particles, which are negatively charged proteins and molecules that affect ion homeostasis through changes in the lysosomal membrane potential295. The buffering capacity of the luminal contents of the lysosome can also substantially affect the rate of pH changes after fusion with substrate-carrying organelles296. Various molecules on the lysosome surface that regulate ionic balance fine-tune the extent of v-ATPase-mediated pH changes and these can be primary targets of disease-causing genes (see text). v-ATPase-mediated acidification of lysosomes, which hyperpolarizes the lysosomal membrane, is countered by ClC7 that exchanges two cytosolic Cl− ions for one luminal H+ ion201 and TPCs that transport Na+ from the lysosomes. Furthermore, when v-ATPase activity results in excessive number of protons, TMEM175 shuttles protons back out to prevent excessive acidification. Once the pH rises to optimal levels, TMEM175 stops releasing protons. The barrier to proton leak by TMEM175 is aided by proper levels of lysosomal-associated membrane protein 1 (LAMP1) and LAMP2, which interact directly with TMEM175, blocking its proton conduction and facilitating acidification96. Counter-balancing the potentially toxic effects of excessive calcium release are independent, possibly adaptive, protective actions of calcium release from TRPML1 and TPC2 channels. AD, Alzheimer disease; ER, endoplasmic reticulum; FTD, frontotemporal dementia; LSD, lysosomal storage disorder; PD, Parkinson disease; RyR, ryanodine receptor.
The need for tight regulation of lysosomal pH can be appreciated from the diverse regulatory roles that vesicle acidity has in neuronal function. The widely varied pH optima of lysosomal hydrolases (pH 3.5–6.0) imply that a small rise in pH, as might chronically be imposed in some diseases affecting v-ATPase or other ion channels, can alter patterns of substrate cleavage and promote aggregation, proteolytic resistance of the substrate, enzyme denaturation and turnover97 (Fig. 5). Hydrolytic activity, in turn, regulates neuronal homeostasis during nutrient and lysosomal stress98 by generating amino acids that activate signalling platforms on lysosomes, thereby controlling homeostatic balance13,99–102. Declining proteolysis and amino acid generation, for example, releases lysosome-docked mTORC1, resulting in lower mTORC1 kinase activity13. This decreases phosphorylation of transcription factors, such as TFEB, TFE3 and others103, by this kinase, enabling their nuclear translocation to drive expression of genes encoding many components of lysosomes and additional autophagosome machinery13,103–105. Feedback onto mTOR also influences the crucial balance between autophagy and protein synthesis. Apart from clearance functions, high levels of acidification in synapses are also required for synaptic vesicle fusion and recycling106 during neurotransmitter exocytic release107, which may explain why lysosomal enhancement via pH correction ameliorates synaptic dysfunction and cognition, as well as clearance of aggregate-prone proteins, in mouse models of AD and PD94,108,109.
Fig. 5 |. Pathological consequences of chronic lysosomal dysfunction.
In ageing neurons, gradual impairment of lysosomal acidification caused by oxidative damage to vacuolar H+-ATPase (v-ATPase) initiates a vicious cycle involving suppression of substrate hydrolysis, accelerated aggregation of modified proteins that are increasingly protease-resistant and accumulation of poorly degraded oxidizable lipids, which are a major source of free radicals248,297. Limited proteolysis required to activate proenzymes (for example, pro-cathepsin D (Cat D)) and generate new bioactive polypeptides (for example, conversion of progranulin (PRGN) to granulins (GRNs)) begins to diminish. The subsequent superimposition of disease-promoting genetic and environmental factors exacerbates this pathogenic cycle. Persistent residence of these accumulated oxidized substrates inactivates autolysosomes, promoting their accumulation and enlargement and accelerating production of free radicals such as reactive oxygen species (ROS). In turn, ROS cause bidirectional toxicity to mitochondria that then release additional free radicals. Lipofuscin granules, within which undegraded substrates are compacted through a series of cross-linking events, renders this waste less toxic albeit still potentially damaging to membranes297. The collective injury to the lysosomal limiting membrane from these sources induces lysosomal membrane permeability (LMP) — a state of injured membranes that allows relatively small proteins such as cathepsins to pass through into the cytoplasm. Calcium release via transient receptor potential channel mucolipin 1 (TRPML1) and two-pore channel 2 (TPC2) channels activates calpains and calcium-dependent protein kinases such as cyclin-dependent kinase 5 (CDK5), promoting hyper-phosphorylation of pathogenic proteins such as tau and activation of RIPK1, which initiates necrosis-associated neurodegeneration. TRPML1-mediated and TPC2-mediated calcium release is linked to mitochondrial dysfunction, and mTORC1 activation. Figure adapted from ref. 297, CC BY 4.0 (https://creativecommons.org/licenses/by/4.0/). AD, Alzheimer disease; ALS, amyotrophic lateral sclerosis; FTD, frontotemporal dementia; PD, Parkinson disease.
Lysosomal acidification failure in Alzheimer disease.
v-ATPase complex vulnerability underlying neurodegenerative disease gained initial attention from studies of presenilin 1 (PSEN1), mutations of which cause the most common inherited form of AD110. This multi-span transmembrane protein111 is the catalytic subunit of the γ-secretase complex responsible for intramembrane proteolysis of many substrates110–112, the best known being cleavage of APP-βCTF to amyloid-β (Fig. 3). The γ-secretase, including the PSEN1 homologue PSEN2 (ref. 113), mutations of which are also associated with AD, is enriched in lysosomes114,115, has broad substrate specificity and becomes highly abundant in autophagic vacuoles when autophagy is upregulated116, all of which points strongly to γ-secretase having a broad role in lysosomal catabolism rather than a selective one. Independently of its function in the γ-secretase complex, PSEN1 itself is required for complete lysosomal acidification108,117,118. The PSEN1 holoprotein located in the ER acts as a chaperone for the v-ATPase subunit V0a1, the crucial anchoring subunit (Fig. 3), and enables normal v-ATPase complex assembly108,117–121. PSEN1 deletion or loss-of-function mutations causing AD reduce V0a1 delivery to lysosomes, lysosomal V0a1 content, v-ATPase activity and lysosomal acidification94,108,117–120,122–125. These effects have been documented in primary skin fibroblasts from patients with early-onset AD caused by a familial autosomal dominant mutation of presenilin 1 (PSEN1-FAD)94,108,117,118, human PSEN1-FAD neurons125 and mouse models of PSEN1-FAD (ref. 118), and are reversed by re-establishing normal lysosomal pH directly with lysosome-targeted acidic nanoparticles94,108 or by pharmacologically restoring lysosomal pH94,119,122–126. An earlier unconfirmed claim that PSEN1 deletion has no effect on pH127 was subsequently challenged on multiple technical grounds108,117,128 and by the wealth of subsequent contrary evidence cited above. As discussed below, PSEN1 loss of function also releases calcium from lysosomes19,108,117,127 via transient receptor potential channel mucolipin 1 (TRPML1; also known as mucolipin 1)19,108 or two-pore channel protein 2 (TPC2) channels129. Normalizing the rise in lysosomal pH in PSEN1 knockout cells mentioned above also corrected lysosomal Ca2+ release, whereas blocking lysosomal Ca2+ release did not correct the abnormal pH rise19,108. PSEN1-mediated release of calcium from the ER123,126 may also promote lysosomal pH elevation and reduce lysosomal function, which is partially rescued by blocking ryanodine123,126 or inositol triphosphate receptors126, which are two of the multiple calcium channels mainly in the ER that maintain cellular calcium homeostasis by releasing calcium and refilling depleted calcium stores in lysosomes or mitochondria130. Whereas several mechanisms potentially contribute to PSEN1-mediated lysosomal deacidification in still unclear ways, there is strong consensus that pH rise inhibits waste elimination by lysosomes including the two proteotoxic metabolites of APP, namely APP-βCTF and amyloid-β, which drive disease progression121,131–133 (Figs. 5,6).
Fig. 6 |. Evolution of autophagy–lysosome dysfunction in Alzheimer disease leading to neurodegeneration.
a, The primary disruptive action of elevated β-amyloid precursor protein (APP) carboxy-terminal fragment (APP-βCTF) on lysosomal acidification in Alzheimer disease (AD) (see Fig. 3), coupled with multiple genetic, environmental and cell ageing factors121,132,133,135,136,139,280–292, begins to corrupt lysosomal function at the earliest stage of the disease and before signs of neuropathology appear. Increased autophagy induction, a neuroprotective cellular stress response, becomes counterproductive later as poorly acidified autolysosomes and lysosomes progressively fail to clear the growing waste burden. The result is a massive build-up of autophagic vacuoles, mainly poorly acidified autolysosomes, causing a unique pattern of extreme perikaryal membrane blebbing and trafficking deficits producing autophagic vacuole-filled swellings along axons (dystrophic neurites). b, These processes also cause accelerated intra-vesicle amyloid-β accumulation, and the formation of fibrillar aggregates of amyloid-β within an expanding network of endoplasmic reticulum (ER) tubules (see inset) which reflects an apparent stalling of ER-phagy — a normally highly active constitutive process of ER turnover by autophagy. c, This intracellular pathobiology evolves within still intact neurons, preceding the advanced neuronal degeneration associated with lysosomal membrane permeability (LMP) and lysosomal-associated neuronal cell death, which initiates the transformation of each dying neuron into a senile (‘amyloid’) plaque. An ensuing inflammatory response involving the recruitment of reactive astrocytes and phagocytic microglia to the disintegrating neuron induces release of damaging cytokines and hydrolases from surrounding microglia in the effort to clear the extracellular debris. This microglial phagocytic process creates bystander toxicity and accelerates autophagy pathology in the nearby less affected neurons. The recruitment of these neighbouring neurons to the lesion expands the senile plaque (not shown) and gradually condenses protease-resistant debris, especially amyloid-β, within the central ‘core’ of the plaque. v-ATPase, vacuolar H+-ATPase. The neuron diagrams in this figure are reprinted from ref. 131, Springer Nature Limited. Senile plaque adapted with permission from ref. 298, Oxford University Press, which highlights the projection of membrane blebs outward from the central region of the degenerating soma, as detailed in panel b.
APP, arguably the most important AD gene, also directly inhibits v-ATPase, although through a different mechanism to PSEN1. The initial APP cleavage product, APP-βCTF, binds selectively to the V0a1 subunit within the v-ATPase complex and reduces association of the V1 subcomplex with the V0 subcomplex, thereby lowering v-ATPase activity94. In healthy cells, small fluctuations in APP-βCTF level can tonically modulate v-ATPase activity and pH within a physiological range94,121 (Fig. 3). Early in AD, APP-βCTF accumulation to abnormally high levels in neuronal lysosomes more strongly inhibits the association of the V1 subcomplex with the V0 subcomplex, causing a pathological lysosomal pH rise121 and impaired substrate hydrolysis. In response to pathological lysosomal pH elevation in neurons of AD brain, a uniquely extreme ‘autophagic stress’131 develops in the neuronal soma131, characterized by massive accumulation of inadequately acidified autolysosomes unable to degrade substrates, including APP-βCTF and amyloid-β, leading to intracellular formation of amyloid-β131 (Figs. 5,6). Ensuing free radical-mediated and proteotoxic damage to lysosomal membranes promotes the early autophagic–lysosomal death of affected neurons containing amyloid-β aggregates and their transformation into extracellular senile (‘amyloid-β’) plaques131,132,134–136 (Fig. 6). A similar neuropathological sequence also evolves early in sporadic AD131.
Mounting evidence for the pathogenicity of APP-βCTF in AD includes its prominent role in triggering endosome dysfunction beginning at the earliest stages of AD132,137–140 and regulating interactions at mitochondria-associated ER membranes, which, in AD, disrupts calcium and lipid homeostasis and alters mitochondrial behaviours that amplify oxidative phosphorylation and ROS141. Moreover, APP-βCTF is a suspected factor mediating the increased late-onset AD risk due to polymorphisms of the neuronal lysosomal protein phospholipase D3 (PLD3): PLD3, a transmembrane protein delivered from the Golgi and proteolytically cleaved in lysosomes, yields a stable soluble polypeptide with 5′–3′ exonuclease activity but unclear roles in lysosomes142. APP-βCTF build-up in lysosomes of PLD3-overexpressing mice is associated with an AD-like phenotype characterized by robust accumulation of autophagic vacuoles in the soma and within swellings along axons resembling ‘dystrophic neurites’ of AD, suggesting selectively impaired retrograde transport of these autophagic vacuoles143 (Fig. 6). A similar autophagic phenotype is accelerated in brains from individuals carrying the risk PLD3 gene polymorphism142,143.
Primary lysosomal pH dysregulation is common in neurodegenerative disease.
The pathogenic targeting of lysosome acidification, and particularly the V0a1 v-ATPase subunit, extends beyond AD-related conditions to mechanisms underlying the pathogenic actions of causal genes in a host of other neurodegenerative diseases95 (Figs. 2,3). Analogous to the chaperone role of PSEN1, palmitoylation of V0a1 in the Golgi assists its maturation144. Loss-of-function mutations of the palmitoyl-protein thioesterase 1 gene (PPT) that cause a severe neurodegenerative LSD, called CLN1 neuronal ceroid lipofuscinosis (NCL), impair maturation of V0a1 and other Ppt-modified proteins, leading to deficient assembly and activity of lysosomal v-ATPase145. Also, in PD, the causative Arg1441Cys mutation in LRRK2 disrupts a LRRK2–v-ATPase interaction leading to the observed v-ATPase deficiency and acidification defects144. Given the mechanism in CLN1, a possible role for v-ATPase palmitoylation in the disruption is possible but has not been demonstrated144. Furthermore, PD-causing LRRK2 mutations hyperactivate LRRK2 function as a kinase for numerous different Ras-related (RAB) proteins, thus probably impacting diverse components of the endolysosomal system146. The relationship between PD and LSDs is supported by Kufor–Rakeb syndrome, a rare neurodegenerative dementia associated with early-onset parkinsonism147 and a cause of NCL in a strain of dogs and at least one human family148–150. The PD risk gene ATP13A2, encoding a lysosomal P5-type ATPase involved in polyamine (for example, spermine, spermidine) transport from the lysosome151, thereby maintains lysosome health by preventing toxic polyamine accumulation151. ATP13A2 is considered a pH modulator given that its mutation and/or loss leads to accumulation of these polyamines152–154, pH elevation, autophagosome and lysosome build-up155–157 and earlier cell death by apoptosis than treated wild-type cells151. As polyamines act as scavengers of heavy metals, polyamine accumulation in lysosomes in PD might explain previously reported changes in heavy metal content in PD158.
Yet another likely modulator of lysosomal pH, and a risk factor for the synucleinopathies PD and Lewy body dementia (LBD), is the endosomal/lysosomal proton channel TMEM175 (refs. 159–161) (Fig. 4). The working mechanism and function of TMEM175 in different pathophysiological contexts are not yet fully resolved. Although originally reported to be a non-canonical potassium channel162,163, recent reports emphasize its role as a selective channel to leak H+ out of lysosomes when the lysosomal pH drops below normal164,165. Conversely, TMEM175 is proposed to expel potassium to retain H+ if the pH rises excessively164,165. TMEM175-deficient cells have hypoactive hydrolases including lysosomal acid glucosylceramidase (GBA)144,166 and an accumulation of pathological α-synuclein164,167 potentially accounting for the loss of dopaminergic neurons seen in mice with TMEM175 mutations163,164,168. Whether this phenotype reflects hyper-acidification or elevated lysosomal pH may depend on the experimental context164,167. A recently discovered facet of TMEM175 biology is its association with LAMP1 and LAMP2. LAMPs were originally believed just to be structural proteins that protect lysosomal membrane from resident hydrolytic enzymes, although evidence now reveals that they have a key physiological role in suppressing proton leak through TMEM175 channels to facilitate re-acidification when lysosomal pH rises inappropriately96.
Another example of lysosomal dysfunction and pH disruption as central to the mechanism of adult neurodegenerative disease is FTLD, the most common, inherited form of which is caused by mutations of granulin (GRN), the gene encoding progranulin (PGRN) and causing the disorder known as FTLD-GRN169 (Fig. 5). Growing evidence indicates that PGRN regulates multiple aspects of lysosomal function, including cathepsin and GCase activities and lysosomal pH40,170. Recent unbiased multimodal proteomics and functional analyses of lysosomes in PGRN-deficient neurons or human PGRN-mutant induced pluripotent stem cell neurons uncovered a marked impairment of lysosomal acidification resulting in impaired hydrolytic activity despite an upregulated expression of acidification machinery proteins171. Loss of PGRN is proposed to impede attachment of the V1 subcomplex to the membrane-anchored V0 domain172 (Fig. 3). Furthermore, PGRN directly interacts with the lysosomal pH regulator, transmembrane protein 106B (TMEM106B), a ubiquitous type 2 (endo)lysosomal integral membrane protein expressed at especially high levels in neurons and oligodendroglial cells173–175. Single-nucleotide TMEM106B polymorphisms modify the risk and lower the age of onset of FTLD-GRN176–178. TMEM106 gene deletion downregulates V0a1, V0c and V0d1, and dramatically exacerbates central nervous system neurodegeneration and autophagic stress when combined with loss of PGRN in mice, presumably compounding the acidification deficit common to both genetic manipulations179–181. Tmem106b deletion alone induces a cascade of effects stemming from insufficient lysosomal acidification172,179 and autophagic stress resembling effects seen in AD182,183, as well as defective lysosomal transport183. Lysosomal processing of TMEM106B yields a C-terminal fragment that binds to v-ATPase172 and its accessory protein 1 (AP1) component172 as well as to cathepsin D (CTSD)179, one of approximately a dozen proteases in lysosomes that are activated at low pH and collectively are capable of degrading endogenous and internalized exogenous proteins completely to amino acids. Their range of functions in maintaining cellular homeostasis is very broad184. CTSD, although ubiquitously expressed in cells, exhibits high expression in the brain and decreases in its function are frequently suspected in mechanisms of neurodegeneration179,185. Notably, a second accessory protein of the V0 domain, ATP6AP2, is a risk gene for PD154. Finally, the v-ATPase is also targeted in individuals with X-linked dominantly inherited ALS with FTD by mutation in the UBQLN2 gene (encoding ubiquilin 2) that functions in both the proteasome and autophagy pathways by clearing misfolded proteins186. Mutation of UBQLN2 reduces expression of ATP6v1G1, a critical subunit of the ATPase pump that also regulates vacuolar acidification and is required for autophagosome maturation187. Modelling the disease in flies and mammalian cells also revealed that ubiquilins are required to maintain proper levels of the V0a/V100 subunit of v-ATPase. Mutations or deletion of UBQLN2 elevated the lysosomal pH and lowered cathepsin B activity despite greater autophagy induction188.
That so many disease-causing lysosomal genes converge directly on pH regulation within this vital organelle is remarkable, and novel evidence continues to emerge. It is important also to recognize that the impact of these genes on other ELA network components and, most importantly, on downstream neuronal functions is informative about brain disease pathogenesis and particular vulnerabilities exhibited by different neurodegenerative diseases. Several major interconnecting mechanisms are considered below.
Lysosomal calcim and neurodegeneration
Lysosomes represent a rich store of calcium in neurons, which is tightly regulated in close coordination with ER and mitochondrial calcium regulation and lysosomal pH108,189–191 (Figs. 4,5). Calcium efflux from lysosomes influences waste clearance through regulatory actions on lysosomal motility, fusion with other organelles and cell signalling controlling autophagy flux192–194. As an example for the latter role, calcium released locally around lysosomes in response to cell stress activates calcineurin, which dephosphorylates the lysosome-docked transcription factors TFEB and TFE3 and enables their translocation to the nucleus195. There, these factors promote gene transcription supporting lysosome biogenesis, including expression of catabolic enzymes and v-ATPase complex subunits. The calcium release also promotes lysosomal exocytosis (that is, secretion of lysosomal content upon lysosome fusion with plasma membrane), an alternative route for waste clearance194, which is upregulated when conventional autophagy routes are impaired194. TRPML1, which transports mainly Ca2+ but also Fe2+ and Zn2+, initiates this process194 and its expression itself is regulated by TFEB195. Loss-of-function mutations in TRPML1 cause mucolipidosis type IV, a severe neurodegenerative LSD196 with impaired lysosomal trafficking and hydrolysis197.
TRPML1 (ref. 198) can be activated by phosphatidylinositol 3,5-bisphosphate (PI(3,5)P2)199–205. Importantly, however, TRPML1 channel opening also increases independently of agonists in response to metabolic stressors, including lysosomal deacidification206 and oxidative stress — another major contributor to lysosomal pH elevation207–210. TRPML1 signalling induces lysosome biogenesis and mTORC1-mediated autophagy211 to maintain cell homeostasis under conditions of mild stress. Sustained TRPML1 channel opening is triggered by extreme ROS generation from oxidants such as hydrogen peroxide207 or following damage to mitochondria212,213, which can cause neurodegeneration by over-activating calcium-dependent calpain proteases that cleave structural proteins, dysregulate essential enzymes and, thereby, induce cell necrosis108,214,215. Its inhibition is neuroprotective in AD, PD and HD models216–218. Calcium (and calpains) also over-activate the protein kinase CDK5, which injures mitochondria by hyper-phosphorylating key components and also can cause postmitotic neurons to re-enter the cell cycle108,216–219. A buffer against rises in cytoplasmic calcium or altered organellar calcium homeostasis is the bidirectional exchange of calcium through multiple ER channels (IP3Rs, ryanodine receptors (RyRs)). Repleting levels of ER calcium attenuates disease-related ER stress220, whereas refilling lysosomal calcium stores may facilitate acidification. TPCs are a class of NAADP-stimulated Ca2+ release channels that modulate varied physiological processes, including endolysosomal trafficking and plasma membrane excitability203, and, similar to TRPML1, promote autophagy and, possibly, lysosomal exocytosis mainly under cellular stress conditions221.
The intertwined nature of lysosomal calcium and pH or autophagy dysregulation seen in varied neurodegenerative diseases is exemplified by inherited (familial) mutations of PSEN1 that cause AD (that is, PSEN1-FAD) (Fig. 4). A knock-in mouse model of PSEN1-FAD was found to develop a late age-onset AD-like axonal dystrophy stemming from selectively impaired retrograde transport of autophagic and/or amphisomal vesicles and their accumulation within focal axon swellings19. The defect involves pH-dependent TRPML1-mediated efflux of calcium that activates c-Jun amino-terminal kinase ( JNK)-mediated phosphorylation of dynein intermediate chains, and thereby impedes retrograde motility19. Restoring proper lysosome acidity rescues the subsequent pathological events19. Recent findings that lysosome and autophagy defects in PSEN1-FAD models could be substantially rescued by blocking calcium release from the ER through overactivated RyRs123 raised the question of whether the release of calcium from the ER is a cause or an effect of lysosomal pH elevation and calcium release. Supporting the latter possibility, an initial Ca2+ release from activated TRPML1 channels can trigger subsequent Ca2+ release through IP3Rs or RyRs in the ER222–225. Moreover, activating TRPML1 channels experimentally increases TFEB-mediated transcription of v-ATPase components225, a known neuroprotective response that is thought to rescue acidification and enhance autophagy. A primary disruption of v-ATPase and lysosomal pH in PSEN1-FAD is also supported by disruptions of v-ATPase via direct binding with the FAD-mutant pathogenic protein PSEN1 and the β-cleaved CTF of APP. Interestingly, as in the FAD mouse model, lysosomal TRPML1 activation, by inducing autophagy, also promotes autophagy stress and the focal axonal swellings filled with autophagic vacuoles, which are associated with coding variants of PLD3 that increase AD risk142,143. Reduced lysosomal Ca2+ levels, probably reflecting excessive calcium efflux, are also seen in forms of PD caused by mutations in GBA1 (refs. 226,227). The accumulating mitochondrial damage and ROS generated by other causal Parkinson genes that disrupt mitophagy226 are a further basis for abnormal pH elevation and TRPML1 over-activation (Figs. 4,5). In several other cellular states (for example, low-density lipoprotein (LDL) overload in non-neural cells228, APOE4 expression), by contrast, the TRPML1 agonist ML-SA1 prevents and TRPML1 silencing potentiates lysosomal dysfunction228,229. These seemingly contradictory data obscure an answer to whether stimulating or preventing Ca+2 efflux from lysosomes would be a useful intervention in any particular neurological disease context, especially in view of the potential neurotoxicity of excessive calcium release from lysosomes108. Given the important cross-regulation among major calcium stores and the potential for lysosomal dysfunction by varied alterations in degenerative disease states, this interplay warrants more intensive research focus to clarify the long-term impact of modifying calcium fluxes as a disease intervention.
Lysosomal hydrolysis failure
In addition to the sizable impact of pH changes on general hydrolytic activity in some disorders, disease genes or pathogenic substrates, or a combination of both, may directly disrupt lysosomal hydrolytic enzymes. Either of these two sources of disruption may induce varied responses from different enzymes, such as compensatory upregulated lysosomal gene transcription, altered hydrolase maturation and proenzyme activation, or inactivation and build-up of the hydrolase and its specific substrates. The disease literature is replete with reports of altered cathepsin levels; however, the use of mainly artificial assay systems makes interpretation difficult and few reports document direct pathogenic protein–enzyme interactions within a specific intact cell type. As in the diseases described above, a chronically low-level rise in baseline pH can substantially inactivate the hydrolases with the most acidic optima, such as CTSD and CTSL. Also, by inhibiting the turnover of the protease in the lysosome, elevated pH raises levels of the partially inactive enzyme, which is mistaken for an elevation in expression. That said, mutations in 3 of 15 cathepsins (CTSD, CTSA and CTSF) are known to cause degenerative LSDs, including most notably neuronal ceroid lipofuscinosis 10 (CLN10). CTSD polymorphisms are additionally associated with increased risk of AD, PD and FTD185,230. Additional gene mutations and polymorphisms disrupting a specific hydrolase or calcium channel involved in autophagy–lysosome clearance expand the important theme linking pathogenesis of neurodegenerative LSDs with disorders in adults. Mutations affecting enzymes involved in lipid clearance also disrupt overall hydrolysis231–237 and clearing these lipids can help ameliorate proteolysis deficits238. The physiological regulation and cross-talk among individual hydrolases remains largely uncharted investigative territory and is likely to reveal important new roles in signalling and neuronal dysfunctions more subtle than degeneration239,240.
Later consequences of autophagy–lysosomal failure
In the foregoing disease scenarios, the accumulation of autophagic substrates in autophagic vacuoles, mainly autolysosomes, is a common pathological outcome in the most affected neurons in adult neurodegenerative diseases and a harbinger of eventual neuron death. This phenomenon, termed autophagic stress, reflects an imbalance favouring autophagosome production over the rate of clearance, and its effects on the severity of autophagic vacuole build-up varies in different diseases241. Given the established function of autophagy as a neuroprotective cellular response through specific anti-apoptosis molecular mechanisms242, autophagy upregulation is an early response against cell ageing and the added compromise due to disease. In the case of AD, however, the persistence of autophagy induction in the face of growing lysosome failure becomes counterproductive, leading to a uniquely massive autolysosome accumulation in the most compromised neurons (Fig. 6). In PD and HD, autophagy induction does not change and may actually be lowered, possibly accounting for why autophagic stress is much less prominent than in AD despite lysosomal dysfunction243,244. Lysosomal and autolysosomal damage causing lysosomal membrane permeability (LMP) leads to gradual leakage of toxic hydrolytic enzymes. This is the likely final common pathway triggering a slowly progressive neuronal degeneration and cell death in the above-described diseases. LMP leading to a gradual lysosomal-associated cell death can be mediated by lysosomal hydrolase release directly245,246. Greater release via more severe LMP can trigger cascades (for example, apoptosis, necroptosis, ferroptosis) that cause rapid demise of the neuron, which has morphological features or markers of multiple death cascades131,247. LMP is often attributed to damage from the accumulated pathogenic protein (for example, amyloid-β, α-synuclein); however, hydrolytic failure generates numerous damaging derivatives, most notably oxidized lipids and proteins that further inhibit hydrolases by oxidizing or otherwise damaging resident proteins24,36–38,248,249. Oxidized lipid accumulations in lysosomes elevate Fe2+ entry into cells250,251 through the endolysosomal system252,253, which helps further drive oxidative stress via the Fenton reaction254,255 and further promotes lysosomal deacidification256 and promotes a more acute death involving ferroptosis257. The oxidative attack on v-ATPase and lysosomal enzymes creates a vicious cycle of progressive lysosomal corruption that initiates other pathogenic events as described in Fig. 5. The sequence of autophagy–lysosomal-triggered pathogenic events evolving in AD highlights the protracted intraneuronal deterioration of this system. This failure precedes very early death of a select population of vulnerable neurons and the transformation of each of these dying neurons into an extracellular senile plaque (Fig. 6). This pathological sequence of intraneuronal events aligns AD pathobiology with other ageing-related neurodegenerative proteinopathies, many of which are being recognized as disrupting the autophagic–lysosomal system and inter-related endosomal–lysosomal system functions131,134,241,243,258–260.
Future directions for clinical translation
As accumulated toxic misfolded proteins are a hallmark of most neurodegenerative diseases, translational efforts have focused on enhancing degradation of these proteins. As lesions that cause neurodegenerative diseases may impact autophagy at different stages of the pathway, the sites of such defects need to be understood to maximize therapeutic opportunities. For example, in the case of disease where there is defective formation of autophagosomes at an early stage of the pathway but no compromise in the pathway after autophagosome formation or not enough of a defect to substantially impair subsequent stages of autophagic flux, then therapeutically boosting autophagosome formation may be suitable (for example, in models of HD or in PD caused by α-synuclein mutations)261. By contrast, in diseases where there are defects after the stage of autophagosome formation, for instance in AD and LSDs, then autophagosomes may accumulate unproductively and cause toxicity especially if autophagosome formation is boosted as part of positive feedback loops aimed to promote clearance of accumulating pathogenic substrates. For example, autophagosome membranes may serve as a platform for p62-dependent caspase 8 recruitment to form an intracellular death-inducing signalling complex262 and apoptosis may thus occur under conditions leading to excessive autophagic membranes. However, this theoretical possibility needs to be rigorously tested in different disease-relevant scenarios, as there is evidence that inhibition of autophagic flux caused by some lysosomal defects may be amenable to amelioration with autophagosome formation inducers263 if the compound can also unblock the lysosome264. Nevertheless, it is desirable to stratify diseases by genotype, where possible, in order to prioritize the cases that are most likely to benefit from induction of autophagosome formation. For example, in the case of PD, patients with α-synuclein mutation may be suitable for this strategy265, whereas those with ATP13A2 defects affecting lysosomal activity266 may be less suitable.
Much activity has focused on identifying drugs to induce autophagosome formation and improve lysosome function4,267,268. Frequently, such drugs act at signalling nodes that affect other pathways, hence the need to consider safety and specificity. However, one of the attractions of this strategy is that one can induce the autophagy pathway in a pulsatile fashion, which may mitigate some side effects. This has been demonstrated with mTOR inhibition in a tauopathy mouse model269. The timing of autophagosome induction treatment may be critical in some diseases. Upregulated autophagosome biogenesis might be protective in the early stages of diseases such as AD (as suggested by mouse studies270), but may be ineffective or compound the lysosome failure that becomes overt at later stages. One possible way that may enable simultaneous induction of autophagosome biogenesis along with improved lysosomal function would be by increasing TFEB and/or TFE3 activities271. It is also worth considering the strategy of increasing specific removal of substrates by using molecular glues to link substrates to autophagy components such as LC3 (ref. 4).
Pharmacological intervention programmes have historically been wary of considering lysosomal remediation, due to the risks of overshooting normalization (for example, decreasing pH too far in diseases where lysosomes are too alkaline) by therapeutically enhancing lysosomal proteolytic activity. However, scant evidence has been produced to justify the concern and, in fact, effective targeting of lysosomal dysfunction has been demonstrated in animal models of neurodegenerative disease260,268,272–277. A growing number of strategies to abrogate pH elevation and calcium efflux abnormalities and the toxic substrate storage are suggested by the discussion above or detailed in recent reviews. Some preclinical interventions have attenuated lysosomal dysfunction and rescued additional deficits in synaptic function and cognition, attesting to the promise of this approach.
Despite enormous progress in understanding autophagy, the movement of more agents into the clinic is still impeded by a relative lack of tools to measure autophagy activity in vivo and to establish target engagement by drugs in specific cell types in intact brain85. Different or even reciprocal responses to a drug may well occur in different neural cell types. Biochemical analyses of tissues from preclinical models may obscure these cell-type differences. For future clinical trials, surrogate biomarkers of ELA dysfunction in accessible fluids or revealed by neuroimaging are urgently needed to validate the effective modification of an ELA-related disease target non-invasively. Nevertheless, for advances towards targeting mechanisms underlying phenotypes in neurodegenerative diseases, the diversity of possible ways that autophagy might be targeted therapeutically provides optimism that autophagy modulation may, ultimately, prove to be an effective therapeutic approach for neurodegenerative diseases.
Acknowledgements
Research in the Nixon laboratory has been supported by grants (P01 AG017617; R01 AG062376) from the National Institute on Aging, New Vision Research Foundation (Leonard Litwin Scholar award), Cure Huntington’s Disease Initiative (CHDI) Foundation, Takeda Corp. and Johnson & Johnson. D.C.R. received funding from the UK Dementia Research Institute (funded by the MRC, Alzheimer’s Research UK and the Alzheimer’s Society), Parkinson’s UK, the National Institute for Health and Care Research (NIHR) Cambridge Biomedical Research Centre (NIHR203312), Rosetrees Trust and the Tau Consortium/Rainwater Charitable Foundation. The views expressed are those of the authors and not necessarily those of the NIHR or the Department of Health and Social Care. The authors acknowledge the expert assistance of S. Jain in preparing this manuscript for publication. They thank G. Di Paolo and A. Henry for critical reading of the manuscript and suggestions. The authors appreciate additional contributions made to the subjects reviewed that regretfully could not be cited due to space and reference limitations.
Glossary
- α-Synuclein
A highly soluble neuronal protein that regulates synaptic vesicle trafficking and subsequent neurotransmitter release, and accumulates in Lewy bodies and Lewy neurites in Parkinson disease (PD) and other synucleinopathies. Mutations of α-synuclein are linked to familial PD
- β-Amyloid precursor protein (APP)
A single-pass transmembrane protein highly expressed in the brain but of mainly unknown function. APP undergoes rapid cleavage into multiple bioactive products by sequential proteases, including the intramembranous γ-secretase complex that generates β-carboxy-terminal fragment (βCTF) and amyloid-β, two polypeptides implicated in Alzheimer disease (AD) pathogenesis and overproduced by APP mutations that cause autosomal dominant AD
- Alzheimer disease (AD)
The most common form of dementia involving neurodegeneration of brain regions controlling thought, memory and language. AD progresses from memory loss to impaired language expression, comprehension and inability to perform activities of daily living. Autosomal dominant gene mutations (β-amyloid precursor protein (APP), presenilin 1 (PSEN1), PSEN2) induce adult early-onset (age <65 years) AD, whereas most cases are late-onset (age >65 years) AD involving influences of varied genetic and environmental risk factors
- Conjugation of ATG8 (LC3) family membranes to single membranes (CASM)
A non-canonical autophagy pathway that shares some of the common ATG machinery but bears key mechanistic and functional distinctions, and is characterized by conjugation of ATG8 (LC3) to single membranes such as lysosomes and phagosomes
- Endolysosomal escape
Escape of substrates from vesicles in the endocytic and lysosomal systems
- Endoplasmic reticulum-associated degradation (ERAD)
The recognition of substrates in the lumen and membrane of the endoplasmic reticulum (ER), and their translocation into the cytosol, ubiquitination and delivery to the proteasome for degradation
- Exosomes
Membrane-bound extracellular vesicles that are produced in the endosomal compartment of eukaryotic cells
- Frontotemporal lobar degeneration (FTLD)
Also known as Pick disease. A group of brain disorders caused by degeneration of the frontal and anterior temporal lobes of the brain and characterized by progressive decline in behaviour (for example, personality changes, apathy) or movement, speaking or language comprehension
- Huntingtin
A ubiquitously expressed protein with varied roles in synaptic transmission, transport and cell survival. Abnormal expansion of a glutamine stretch (polyQ) in mutant huntingtin causes Huntington disease (HD)
- Huntington disease (HD)
A monogenic neurodegenerative disorder caused by the huntingtin gene, HTT, characterized by loss of striatal neurons, and resulting in motor, psychiatric and cognitive symptoms
- Induced pluripotent stem cells
A type of stem cells derived from adult somatic cells which have been reprogrammed through inducing genes and factors to be pluripotent
- LC3 family members
Members of the ATG8 gene family that are classical markers for autophagosomes
- Lewy body dementia (LBD)
A progressive dementia involving a decline in thinking, movement, behaviour and mood and associated with abnormal deposits of α-synuclein in the brain, called Lewy bodies
- Lysosomal storage disorder (LSD)
A group of more than 50 mainly childhood disorders that are inborn errors of metabolism characterized by abnormal accumulation of substrates due to defective lysosomes and usually involving deficiency of a single lipid metabolizing enzyme
- Micropinocytosis
A process in which macromolecules are engulfed by small vesicles from the plasma membrane
- Microvesicles
Diverse membrane-enclosed vesicles that are released from cells into the extracellular space
- Neuronal ceroid lipofuscinosis (NCL).
Also called Batten disease. A group of 14 inherited lysosomal storage disorders (LSDs) characterized by intracellular accumulation of autofluorescent lipopigment (ceroid and lipofuscin) and progressive neurodegeneration
- Nucleoid
A structure comprising mitochondrial DNA (mtDNA) and numerous nucleoid-associated proteins that enables submitochondrial organization of mtDNA
- Parkinson disease (PD)
A chronic degenerative disorder targeting dopaminergic neural circuits and initially causing tremors, rigidity and slowed movement, and later additional intellectual functions, including dementia in a minor population of affected individuals
- Presenilin 1 (PSEN1)
A multifunctional transmembrane protein that, in loss-of-function mutant form, is one of three autosomal dominant causes of Alzheimer disease (AD). PSEN1 is one of four core proteins in the γ-secretase endoprotease complex that performs intramembrane cleavage of dozens of integral membrane proteins, including β-amyloid precursor protein (APP) that sequentially generates the β-carboxy-terminal fragment (βCTF) and amyloid-β, two polypeptides implicated in AD pathogenesis
- Ryanodine receptors (RyRs)
Ion channels residing in the sarcoplasmic/endoplasmic reticulum membrane and responsible for Ca2+ release from intracellular stores in excitable tissues, such as muscles and neurons
- Tau (MAPT)
A group of six highly soluble protein isoforms produced by alternative splicing from MAPT that help stabilize the microtubule cytoskeleton of neurons and compose neurofibrillary tangles, a hallmark of Alzheimer disease (AD)
- TFEB, TFE3
Members of the MiT-TFE family of helix–loop–helix leucine zipper transcription factors that regulate expression of genes involved in the biogenesis and function of lysosomes and autophagosomes
Footnotes
Competing interests
D.C.R. is a consultant for Aladdin Healthcare Technologies Ltd, Mindrank AI, Nido Biosciences, Drishti Discoveries, Retro Biosciences, PAQ Therapeutics and Alexion Pharma Intl Ops Ltd. R.A.N. declares no competing interests.
Additional information
Peer review information Nature Reviews Molecular Cell Biology thanks Zhenyu Yue and the other, anonymous, reviewers for their contribution to the peer review of this work.
References
- 1.Ross CA & Poirier MA Protein aggregation and neurodegenerative disease. Nat. Med 10 Suppl, S10–S17 (2004). [DOI] [PubMed] [Google Scholar]
- 2.Jayaraj GG, Hipp MS & Hartl FU Functional modules of the proteostasis network. Cold Spring Harb. Perspect. Biol 12, 30833457 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 3.Nishida Y. et al. Discovery of Atg5/Atg7-independent alternative macroautophagy. Nature 461, 654–658 (2009). [DOI] [PubMed] [Google Scholar]
- 4.Fleming A. et al. The different autophagy degradation pathways and neurodegeneration. Neuron 110, 935–966 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 5.Bourdenx M. et al. Chaperone-mediated autophagy prevents collapse of the neuronal metastable proteome. Cell 184, 2696–2714.e25 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 6.Liu GY & Sabatini DM mTOR at the nexus of nutrition, growth, ageing and disease. Nat. Rev. Mol. Cell Biol 21, 183–203 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 7.Settembre C. & Perera RM Lysosomes as coordinators of cellular catabolism, metabolic signalling and organ physiology. Nat. Rev. Mol. Cell Biol 25, 223–245 (2024). [DOI] [PubMed] [Google Scholar]
- 8.Son SM et al. Leucine signals to mTORC1 via its metabolite acetyl-coenzyme A. Cell Metab. 29, 192–201.e7 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Son SM et al. Leucine regulates autophagy via acetylation of the mTORC1 component raptor. Nat. Commun 11, 3148 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 10.Korolchuk VI et al. Lysosomal positioning coordinates cellular nutrient responses. Nat. Cell Biol 13, 453–460 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11.Ding WX et al. Oncogenic transformation confers a selective susceptibility to the combined suppression of the proteasome and autophagy. Mol. Cancer Ther 8, 2036–2045 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Korolchuk VI, Mansilla A, Menzies FM & Rubinsztein DC Autophagy inhibition compromises degradation of ubiquitin–proteasome pathway substrates. Mol. Cell 33, 517–527 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 13.Ballabio A. & Bonifacino JS Lysosomes as dynamic regulators of cell and organismal homeostasis. Nat. Rev. Mol. Cell Biol 21, 101–118 (2020). [DOI] [PubMed] [Google Scholar]
- 14.Ravikumar B, Moreau K, Jahreiss L, Puri C. & Rubinsztein DC Plasma membrane contributes to the formation of pre-autophagosomal structures. Nat. Cell Biol 12, 747–757 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15.Fraser J. et al. Targeting of early endosomes by autophagy facilitates EGFR recycling and signalling. EMBO Rep. 20, e47734 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16.Lees MB Basic Neurochemistry 5th edn, Vol. 39 (Raven Press, 1994). [Google Scholar]
- 17.Lee S, Sato Y. & Nixon RA Lysosomal proteolysis inhibition selectively disrupts axonal transport of degradative organelles and causes an Alzheimer’s-like axonal dystrophy. J. Neurosci 31, 7817–7830 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Lie PPY et al. Post-Golgi carriers, not lysosomes, confer lysosomal properties to pre-degradative organelles in normal and dystrophic axons. Cell Rep. 35, 109034 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 19.Lie PPY et al. Axonal transport of late endosomes and amphisomes is selectively modulated by local Ca2+ efflux and disrupted by PSEN1 loss of function. Sci. Adv 8, eabj5716 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 20.Overly CC & Hollenbeck PJ Dynamic organization of endocytic pathways in axons of cultured sympathetic neurons. J. Neurosci 16, 6056–6064 (1996). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Yap CC, Mason AJ & Winckler B. Dynamics and distribution of endosomes and lysosomes in dendrites. Curr. Opin. Neurobiol 74, 102537 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 22.Lipka J, Kuijpers M, Jaworski J. & Hoogenraad CC Mutations in cytoplasmic dynein and its regulators cause malformations of cortical development and neurodegenerative diseases. Biochem. Soc. Trans 41, 1605–1612 (2013). [DOI] [PubMed] [Google Scholar]
- 23.Aranda-Anzaldo A. The post-mitotic state in neurons correlates with a stable nuclear higher-order structure. Commun. Integr. Biol 5, 134–139 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24.Plascencia-Villa G. & Perry G. Roles of oxidative stress in synaptic dysfunction and neuronal cell death in Alzheimer’s disease. Antioxid 12, 1628 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25.Park SJ et al. Vinexin contributes to autophagic decline in brain ageing across species. Cell Death Differ. 29, 1055–1070 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Tsong H, Holzbaur EL & Stavoe AK Aging differentially affects axonal autophagosome formation and maturation. Autophagy 19, 3079–3095 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27.Carmona-Gutierrez D, Hughes AL, Madeo F. & Ruckenstuhl C. The crucial impact of lysosomes in aging and longevity. Ageing Res. Rev 32, 2–12 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Hansen M, Rubinsztein DC & Walker DW Autophagy as a promoter of longevity insights from model organisms. Nat. Rev. Mol. Cell Biol 19, 579–593 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Burrinha T. et al. Deacidification of endolysosomes by neuronal aging drives synapse loss. Traffic 24, 334–354 (2023). [DOI] [PubMed] [Google Scholar]
- 30.Nixon RA The aging lysosome: an essential catalyst for late-onset neurodegenerative diseases. Biochim. Biophys. Acta Proteins Proteom 1868, 140443 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.Ashkenazi A. et al. Polyglutamine tracts regulate beclin 1-dependent autophagy. Nature 545, 108–111 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 32.Winslow AR et al. α-Synuclein impairs macroautophagy: implications for Parkinson’s disease. J. Cell Biol 190, 1023–1037 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33.Festa BP et al. Microglial-to-neuronal CCR5 signaling regulates autophagy in neurodegeneration. Neuron 111, 2021–2037.e12 (2023). [DOI] [PubMed] [Google Scholar]
- 34.Plascencia-Villa G. & Perry G. Exploring molecular targets for mitochondrial therapies in neurodegenerative diseases. Int. J. Mol. Sci 24, 12486 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 35.Reed TT et al. Proteomic identification of nitrated brain proteins in early Alzheimer’s disease inferior parietal lobule. J. Cell Mol. Med 13, 2019–2029 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36.Di Domenico F. et al. Impairment of proteostasis network in Down syndrome prior to the development of Alzheimer’s disease neuropathology: redox proteomics analysis of human brain. Biochim. Biophys. Acta 1832, 1249–1259 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 37.Perluigi M. et al. Redox proteomics in aging rat brain: involvement of mitochondrial reduced glutathione status and mitochondrial protein oxidation in the aging process. J. Neurosci. Res 88, 3498–3507 (2010). [DOI] [PubMed] [Google Scholar]
- 38.Butterfield DA Oxidative stress in brain in amnestic mild cognitive impairment. Antioxidants 12, 462 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39.Klein M. & Hermey G. Converging links between adult-onset neurodegenerative Alzheimer’s disease and early life neurodegenerative neuronal ceroid lipofuscinosis? Neural Regen. Res 18, 1463–1471 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 40.Simon MJ, Logan T, DeVos SL & Di Paolo G. Lysosomal functions of progranulin and implications for treatment of frontotemporal dementia. Trends Cell Biol. 33, 324–339 (2023). [DOI] [PubMed] [Google Scholar]
- 41.Hara T. et al. Suppression of basal autophagy in neural cells causes neurodegenerative disease in mice. Nature 441, 885–889 (2006). [DOI] [PubMed] [Google Scholar]
- 42.Komatsu M. et al. Loss of autophagy in the central nervous system causes neurodegeneration in mice. Nature 441, 880–884 (2006). [DOI] [PubMed] [Google Scholar]
- 43.Kim M. et al. Mutation in ATG5 reduces autophagy and leads to ataxia with developmental delay. eLife 5, e12245 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44.Jelani M. et al. A mutation in the major autophagy gene, WIPI2, associated with global developmental abnormalities. Brain 142, 1242–1254 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45.Collier JJ et al. Developmental consequences of defective ATG7-mediated autophagy in humans. N. Engl. J. Med 384, 2406–2417 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 46.Dooley HC et al. WIPI2 links LC3 conjugation with PI3P, autophagosome formation, and pathogen clearance by recruiting Atg12–5–16L1. Mol. Cell 55, 238–252 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 47.Kuma A. et al. The role of autophagy during the early neonatal starvation period. Nature 432, 1032–1036 (2004). [DOI] [PubMed] [Google Scholar]
- 48.Takahashi Y. et al. An autophagy assay reveals the ESCRT-III component CHMP2A as a regulator of phagophore closure. Nat. Commun 9, 2855 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49.Migliano SM, Wenzel EM & Stenmark H. Biophysical and molecular mechanisms of ESCRT functions, and their implications for disease. Curr. Opin. Cell Biol 75, 102062 (2022). [DOI] [PubMed] [Google Scholar]
- 50.Radulovic M. et al. ESCRT-mediated lysosome repair precedes lysophagy and promotes cell survival. Embo J. 37, e99753 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51.Eriksson I, Wäster P. & Öllinger K. Restoration of lysosomal function after damage is accompanied by recycling of lysosomal membrane proteins. Cell Death Dis. 11, 370 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 52.Bonet-Ponce L. et al. LRRK2 mediates tubulation and vesicle sorting from lysosomes. Sci. Adv 6, eabb2454 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53.Herbst S. et al. LRRK2 activation controls the repair of damaged endomembranes in macrophages. EMBO J. 39, e104494 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54.Morrison R. et al. LRRK2 kinase dependent and independent function on endolysosomal repair promotes macrophage cell death. Preprint at bioRxiv 10.1101/2023.09.27.559807 (2023). [DOI] [Google Scholar]
- 55.Chen JJ et al. Compromised function of the ESCRT pathway promotes endolysosomal escape of tau seeds and propagation of tau aggregation. J. Biol. Chem 294, 18952–18966 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56.Puri C, Gratian MJ & Rubinsztein DC Mammalian autophagosomes form from finger-like phagophores. Dev. Cell 58 2746–2760.e5 (2023). [DOI] [PubMed] [Google Scholar]
- 57.Puri C. et al. A DNM2 centronuclear myopathy mutation reveals a link between recycling endosome scission and autophagy. Dev. Cell 53, 154–168.e6 (2020). [DOI] [PubMed] [Google Scholar]
- 58.Ravikumar B. et al. Dynein mutations impair autophagic clearance of aggregate-prone proteins. Nat. Genet 37, 771–776 (2005). [DOI] [PubMed] [Google Scholar]
- 59.Ishikawa KI et al. p150glued deficiency impairs effective fusion between autophagosomes and lysosomes due to their redistribution to the cell periphery. Neurosci. Lett 690, 181–187 (2019). [DOI] [PubMed] [Google Scholar]
- 60.Cierny M. et al. Novel destabilizing dynactin variant (DCTN1 p.Tyr78His) in patient with Perry syndrome. Parkinsonism Relat. Disord 77, 110–113 (2020). [DOI] [PubMed] [Google Scholar]
- 61.Cóppola-Segovia V. & Reggiori F. Molecular insights into aggrephagy: their cellular functions in the context of neurodegenerative diseases. J. Mol. Biol 436, 168493 (2024). [DOI] [PubMed] [Google Scholar]
- 62.D’Arcy MS Mitophagy in health and disease. Molecular mechanisms, regulatory pathways, and therapeutic implications. Apoptosis 10.1007/s10495-024-01977-y (2024). [DOI] [PubMed] [Google Scholar]
- 63.Iavarone F, Di Lorenzo G. & Settembre C. Regulatory events controlling ER-phagy. Curr. Opin. Cell Biol 76, 102084 (2022). [DOI] [PubMed] [Google Scholar]
- 64.Deng Z. et al. ALS-FTLD-linked mutations of SQSTM1/p62 disrupt selective autophagy and NFE2L2/NRF2 anti-oxidative stress pathway. Autophagy 16, 917–931 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 65.Lazarou M. et al. The ubiquitin kinase PINK1 recruits autophagy receptors to induce mitophagy. Nature 524, 309–314 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 66.Khaminets A. et al. Regulation of endoplasmic reticulum turnover by selective autophagy. Nature 522, 354–358 (2015). [DOI] [PubMed] [Google Scholar]
- 67.Foronda H. et al. Heteromeric clusters of ubiquitinated ER-shaping proteins drive ER-phagy. Nature 618, 402–410 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68.Pickrell AM & Youle RJ The roles of PINK1, parkin, and mitochondrial fidelity in Parkinson’s disease. Neuron 85, 257–273 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 69.McWilliams TG et al. Basal mitophagy occurs independently of PINK1 in mouse tissues of high metabolic demand. Cell Metab. 27, 439–449.e5 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 70.Pirooznia SK et al. PARIS induced defects in mitochondrial biogenesis drive dopamine neuron loss under conditions of parkin or PINK1 deficiency. Mol. Neurodegener 15, 17 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 71.Goldsmith J, Ordureau A, Harper JW & Holzbaur ELF Brain-derived autophagosome profiling reveals the engulfment of nucleoid-enriched mitochondrial fragments by basal autophagy in neurons. Neuron 110, 967–976.e8 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 72.D’Acunzo P. et al. Mitovesicles are a novel population of extracellular vesicles of mitochondrial origin altered in Down syndrome. Sci. Adv 7, eabe5085 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 73.Hosokawa N. et al. Nutrient-dependent mTORC1 association with the ULK1–Atg13–FIP200 complex required for autophagy. Mol. Biol. Cell 20, 1981–1991 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74.Pena-Llopis S. et al. Regulation of TFEB and v-ATPases by mTORC1. EMBO J. 30, 3242–3258 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Martinez-Vicente M. et al. Cargo recognition failure is responsible for inefficient autophagy in Huntington’s disease. Nat. Neurosci 13, 567–576 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Wong YC & Holzbaur EL The regulation of autophagosome dynamics by huntingtin and HAP1 is disrupted by expression of mutant huntingtin, leading to defective cargo degradation. J. Neurosci 34, 1293–1305 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77.Moreau K. et al. PICALM modulates autophagy activity and tau accumulation. Nat. Commun 5, 4998 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Moreau K, Ravikumar B, Renna M, Puri C. & Rubinsztein DC Autophagosome precursor maturation requires homotypic fusion. Cell 146, 303–317 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79.Puri C, Renna M, Bento CF, Moreau K. & Rubinsztein DC Diverse autophagosome membrane sources coalesce in recycling endosomes. Cell 154, 1285–1299 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80.Simons M, Levin J. & Dichgans M. Tipping points in neurodegeneration. Neuron 111, 2954–2968 (2023). [DOI] [PubMed] [Google Scholar]
- 81.Sarkar S. et al. Complex inhibitory effects of nitric oxide on autophagy. Mol. Cell 43, 19–32 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Ping S, Qiu X, Kyle M. & Zhao LR Brain-derived CCR5 contributes to neuroprotection and brain repair after experimental stroke. Aging Dis. 12, 72–92 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83.Choi I. et al. Autophagy enables microglia to engage amyloid plaques and prevents microglial senescence. Nat. Cell Biol 25, 963–974 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 84.Wang C, Telpoukhovskaia MA, Bahr BA, Chen X. & Gan L. Endo-lysosomal dysfunction: a converging mechanism in neurodegenerative diseases. Curr. Opin. Neurobiol 48, 52–58 (2018). [DOI] [PubMed] [Google Scholar]
- 85.Nixon RA The role of autophagy in neurodegenerative disease. Nat. Med 19, 983–997 (2013). [DOI] [PubMed] [Google Scholar]
- 86.de Duve C. The lysosome turns fifty. Nat. Cell Biol 7, 847–849 (2005). [DOI] [PubMed] [Google Scholar]
- 87.de Duve C. Lysosomes, a New Group of Cytoplasmic Particle (Ronald Press, 1959). [Google Scholar]
- 88.Nixon RA, Yang DS & Lee JH Neurodegenerative lysosomal disorders: a continuum from development to late age. Autophagy 4, 590–599 (2008). [DOI] [PubMed] [Google Scholar]
- 89.Ponsford AH et al. Live imaging of intra-lysosome pH in cell lines and primary neuronal culture using a novel genetically encoded biosensor. Autophagy 17, 1500–1518 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Yoshimori T, Yamamoto A, Moriyama Y, Futai M. & Tashiro Y. Bafilomycin A1, a specific inhibitor of vacuolar-type H+-ATPase, inhibits acidification and protein degradation in lysosomes of cultured cells. J. Biol. Chem 266, 17707–17712 (1991). [PubMed] [Google Scholar]
- 91.Stoka V, Turk V. & Turk B. Lysosomal cathepsins and their regulation in aging and neurodegeneration. Ageing Res. Rev 32, 22–37 (2016). [DOI] [PubMed] [Google Scholar]
- 92.Nishi T. & Forgac M. The vacuolar (H+)-ATPases-nature’s most versatile proton pumps. Nat. Rev. Mol. Cell Biol 3, 94–103 (2002). [DOI] [PubMed] [Google Scholar]
- 93.Collins MP & Forgac M. Regulation and function of v-ATPases in physiology and disease. Biochim. Biophys. Acta Biomembr 1862, 183341 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Coffey EE, Beckel JM, Laties AM & Mitchell CH Lysosomal alkalization and dysfunction in human fibroblasts with the Alzheimer’s disease-linked presenilin 1 A246E mutation can be reversed with cAMP. Neuroscience 263, 111–124 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Colacurcio DJ & Nixon RA Disorders of lysosomal acidification-the emerging role of v-ATPase in aging and neurodegenerative disease. Ageing Res. Rev 32, 75–88 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96.Zhang J. et al. Lysosomal LAMP proteins regulate lysosomal pH by direct inhibition of the TMEM175 channel. Mol. Cell 83, 2524–2539.e7 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Turk B. et al. Acidic pH as a physiological regulator of human cathepsin L activity. Eur. J. Biochem 259, 926–932 (1999). [DOI] [PubMed] [Google Scholar]
- 98.Lie PPY & Nixon RA Lysosome trafficking and signaling in health and neurodegenerative diseases. Neurobiol. Dis 122, 94–105 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 99.Zoncu R. et al. mTORC1 senses lysosomal amino acids through an inside-out mechanism that requires the vacuolar H+-ATPase. Science 334, 678–683 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 100.Jung J, Genau HM & Behrends C. Amino acid-dependent mTORC1 regulation by the lysosomal membrane protein SLC38A9. Mol. Cell Biol 35, 2479–2494 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 101.Rebsamen M. et al. SLC38A9 is a component of the lysosomal amino acid sensing machinery that controls mTORC1. Nature 519, 477–481 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 102.Wang S. et al. Metabolism. Lysosomal amino acid transporter SLC38A9 signals arginine sufficiency to mTORC1. Science 347, 188–194 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Bendl J. et al. The three-dimensional landscape of cortical chromatin accessibility in Alzheimer’s disease. Nat. Neurosci 25, 1366–1378 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 104.Lapierre LR et al. The TFEB orthologue HLH-30 regulates autophagy and modulates longevity in Caenorhabditis elegans. Nat. Commun 4, 2267 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105.Vilchez D, Simic MS & Dillin A. Proteostasis and aging of stem cells. Trends Cell Biol. 24, 161–170 (2014). [DOI] [PubMed] [Google Scholar]
- 106.El Far O. & Seagar M. A role for v-ATPase subunits in synaptic vesicle fusion? J. Neurochem 117, 603–612 (2011). [DOI] [PubMed] [Google Scholar]
- 107.Higashida H, Yokoyama S, Tsuji C. & Muramatsu SI Neurotransmitter release: vacuolar ATPase V0 sector c-subunits in possible gene or cell therapies for Parkinson’s, Alzheimer’s, and psychiatric diseases. J. Physiol. Sci 67, 11–17 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 108.Lee JH et al. Presenilin 1 maintains lysosomal Ca2+ homeostasis via TRPML1 by regulating vATPase-mediated lysosome acidification. Cell Rep. 12, 1430–1444 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Bourdenx M. et al. Nanoparticles restore lysosomal acidification defects: Implication for Parkinson and other lysosomal-related diseases. Autophagy 12, 472–483 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110.Schroder B. & Saftig P. Intramembrane proteolysis within lysosomes. Ageing Res. Rev 32, 51–64 (2016). [DOI] [PubMed] [Google Scholar]
- 111.De Strooper B, Iwatsubo T. & Wolfe MS Presenilins and γ-secretase: structure, function, and role in Alzheimer disease. Cold Spring Harb. Perspect. Med 2, a006304 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112.Haapasalo A. & Kovacs DM The many substrates of presenilin/γ-secretase. J. Alzheimers Dis 25, 3–28 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 113.Sannerud R. et al. Restricted location of PSEN2/γ-secretase determines substrate specificity and generates an intracellular Aβ pool. Cell 166, 193–208 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114.Pasternak SH et al. Presenilin-1, nicastrin, amyloid precursor protein, and γ-secretase activity are co-localized in the lysosomal membrane. J. Biol. Chem 278, 26687–26694 (2003). [DOI] [PubMed] [Google Scholar]
- 115.Bagshaw RD, Pasternak SH, Mahuran DJ & Callahan JW Nicastrin is a resident lysosomal membrane protein. Biochem. Biophys. Res. Commun 300, 615–618 (2003). [DOI] [PubMed] [Google Scholar]
- 116.Yu WH et al. Autophagic vacuoles are enriched in amyloid precursor protein-secretase activities: implications for β-amyloid peptide over-production and localization in Alzheimer’s disease. Int. J. Biochem. Cell Biol 36, 2531–2540 (2004). [DOI] [PubMed] [Google Scholar]
- 117.Wolfe DM et al. Autophagy failure in Alzheimer’s disease and the role of defective lysosomal acidification. Eur. J. Neurosci 37, 1949–1961 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 118.Lee JH et al. Lysosomal proteolysis and autophagy require presenilin 1 and are disrupted by Alzheimer-related PS1 mutations. Cell 141, 1146–1158 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 119.Avrahami L. et al. Inhibition of glycogen synthase kinase-3 ameliorates β-amyloid pathology and restores lysosomal acidification and mammalian target of rapamycin activity in the Alzheimer disease mouse model: in vivo and in vitro studies. J. Biol. Chem 288, 1295–1306 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 120.Sharma D. et al. γ-Secretase orthologs are required for lysosomal activity and autophagic degradation in Dictyostelium discoideum, independent of PSEN (presenilin) proteolytic function. Autophagy 15, 1407–1418 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 121.Im E. et al. Lysosomal dysfunction in Down syndrome and Alzheimer mouse models is caused by selective v-ATPase inhibition by Tyr682 phosphorylated APP βCTF. Sci. Adv 9, eadg1925 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122.Lee JH et al. Transgenic expression of a ratiometric autophagy probe specifically in neurons enables the interrogation of brain autophagy in vivo. Autophagy 15, 543–557 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123.Mustaly-Kalimi S. et al. Protein mishandling and impaired lysosomal proteolysis generated through calcium dysregulation in Alzheimer’s disease. Proc. Natl Acad. Sci. USA 119, e2211999119 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 124.Malampati S. et al. Pharmacological reacidification of lysosomes attenuates intraneuronal amyloidosis, early neuron death, and amyloid plaque formation in 5xFAD mice. In Alzheimer’s Association International Conference (2024). [Google Scholar]
- 125.Chou CC et al. Proteostasis and lysosomal quality control deficits in Alzheimer’s disease neurons. Preprint at bioRxiv 10.1101/2023.03.27.534444 (2023). [DOI] [Google Scholar]
- 126.Yang M. et al. Alzheimer’s disease presenilin-1 mutation sensitizes neurons to impaired autophagy flux and propofol neurotoxicity: role of calcium dysregulation. J. Alzheimers Dis 67, 137–147 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 127.Coen K. et al. Lysosomal calcium homeostasis defects, not proton pump defects, cause endo-lysosomal dysfunction in PSEN-deficient cells. J. Cell Biol 198, 23–35 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128.Nixon RA, Lee JH & Wolfe D. Comments on presenilins and lysosome pH revisited again. J. Neurosci 10.1523/JNEUROSCI.0556-12.2012 (2012). [DOI] [Google Scholar]
- 129.Tong BC et al. Lysosomal TPCN (two pore segment channel) inhibition ameliorates β-amyloid pathology and mitigates memory impairment in Alzheimer disease. Autophagy 18, 624–642 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 130.Karagas NE & Venkatachalam K. Roles for the endoplasmic reticulum in regulation of neuronal calcium homeostasis. Cells 8, 1232 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 131.Lee JH et al. Faulty autolysosome acidification in Alzheimer’s disease mouse models induces autophagic build-up of Aβ in neurons, yielding senile plaques. Nat. Neurosci 25, 688–701 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 132.Jiang Y. et al. Lysosomal dysfunction in down syndrome is APP-dependent and mediated by APP-βCTF (C99). J. Neurosci 39, 5255–5268 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 133.Pulina MV, Hopkins M, Haroutunian V, Greengard P. & Bustos V. C99 selectively accumulates in vulnerable neurons in Alzheimer’s disease. Alzheimers Dement. 16, 273–282 (2020). [DOI] [PubMed] [Google Scholar]
- 134.Nixon RA Amyloid precursor protein and endosomal–lysosomal dysfunction in Alzheimer’s disease: inseparable partners in a multifactorial disease. FASEB J. 31, 2729–2743 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135.Lauritzen I. et al. The β-secretase-derived C-terminal fragment of βAPP, C99, but not Aβ, is a key contributor to early intraneuronal lesions in triple-transgenic mouse hippocampus. J. Neurosci 32, 16243–16255a (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 136.Lauritzen I. et al. Intraneuronal aggregation of the β-CTF fragment of APP (C99) induces Aβ-independent lysosomal-autophagic pathology. Acta Neuropathol. 132, 257–276 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 137.Tamayev R, Matsuda S, Arancio O. & D’Adamio L. β- but not γ-secretase proteolysis of APP causes synaptic and memory deficits in a mouse model of dementia. EMBO Mol. Med 4, 171–179 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 138.Lauritzen I. et al. Targeting γ-secretase triggers the selective enrichment of oligomeric APP-CTFs in brain extracellular vesicles from Alzheimer cell and mouse models. Transl. Neurodegener 8, 35 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 139.Jiang Y. et al. Partial BACE1 reduction in a Down syndrome mouse model blocks Alzheimer-related endosomal anomalies and cholinergic neurodegeneration: role of APP-CTF. Neurobiol. Aging 39, 90–98 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140.Jiang Y. et al. Alzheimer’s-related endosome dysfunction in down syndrome is Aβ-independent but requires APP and is reversed by BACE-1 inhibition. Proc. Natl Acad. Sci. USA 107, 1630–1635 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141.Area-Gomez E. et al. A key role for MAM in mediating mitochondrial dysfunction in Alzheimer disease. Cell Death Dis. 9, 335 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 142.Van Acker ZP et al. Phospholipase D3 degrades mitochondrial DNA to regulate nucleotide signaling and APP metabolism. Nat. Commun 14, 2847 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 143.Yuan P. et al. PLD3 affects axonal spheroids and network defects in Alzheimer’s disease. Nature 612, 328–337 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 144.Wallings R, Connor-Robson N. & Wade-Martins R. LRRK2 interacts with the vacuolar-type H+-ATPase pump a1 subunit to regulate lysosomal function. Hum. Mol. Genet 28, 2696–2710 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 145.Bagh MB et al. Misrouting of v-ATPase subunit V0a1 dysregulates lysosomal acidification in a neurodegenerative lysosomal storage disease model. Nat. Commun 8, 14612 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 146.Steger M. et al. Phosphoproteomics reveals that Parkinson’s disease kinase LRRK2 regulates a subset of Rab GTPases. eLife 5, e12813 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 147.Ramirez A. et al. Hereditary parkinsonism with dementia is caused by mutations in ATP13A2, encoding a lysosomal type 5 P-type ATPase. Nat. Genet 38, 1184–1191 (2006). [DOI] [PubMed] [Google Scholar]
- 148.Farias FH et al. A truncating mutation in ATP13A2 is responsible for adult-onset neuronal ceroid lipofuscinosis in Tibetan terriers. Neurobiol. Dis 42, 468–474 (2011). [DOI] [PubMed] [Google Scholar]
- 149.Schultheis PJ et al. Atp13a2-deficient mice exhibit neuronal ceroid lipofuscinosis, limited α-synuclein accumulation and age-dependent sensorimotor deficits. Hum. Mol. Genet 22, 2067–2082 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 150.Bras J, Verloes A, Schneider SA, Mole SE & Guerreiro RJ Mutation of the parkinsonism gene ATP13A2 causes neuronal ceroid-lipofuscinosis. Hum. Mol. Genet 21, 2646–2650 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 151.van Veen S. et al. ATP13A2 deficiency disrupts lysosomal polyamine export. Nature 578, 419–424 (2020). [DOI] [PubMed] [Google Scholar]
- 152.van Veen S. et al. Cellular function and pathological role of ATP13A2 and related P-type transport ATPases in Parkinson’s disease and other neurological disorders. Front. Mol. Neurosci 7, 48 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 153.Dubos A. et al. Conditional depletion of intellectual disability and parkinsonism candidate gene ATP6AP2 in fly and mouse induces cognitive impairment and neurodegeneration. Hum. Mol. Genet 24, 6736–6755 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 154.Korvatska O. et al. Altered splicing of ATP6AP2 causes X-linked parkinsonism with spasticity (XPDS). Hum. Mol. Genet 22, 3259–3268 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 155.Park JS et al. Pathogenic effects of novel mutations in the P-type ATPase ATP13A2 (PARK9) causing Kufor–Rakeb syndrome, a form of early-onset parkinsonism. Hum. Mutat 32, 956–964 (2011). [DOI] [PubMed] [Google Scholar]
- 156.Dehay B. et al. Loss of P-type ATPase ATP13A2/PARK9 function induces general lysosomal deficiency and leads to Parkinson disease neurodegeneration. Proc. Natl Acad. Sci. USA 109, 9611–9616 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 157.Usenovic M, Tresse E, Mazzulli JR, Taylor JP & Krainc D. Deficiency of ATP13A2 leads to lysosomal dysfunction, α-synuclein accumulation, and neurotoxicity. J. Neurosci 32, 4240–4246 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158.Vrijsen S, Houdou M, Cascalho A, Eggermont J. & Vangheluwe P. Polyamines in Parkinson’s disease: balancing between neurotoxicity and neuroprotection. Annu. Rev. Biochem 92, 435–464 (2023). [DOI] [PubMed] [Google Scholar]
- 159.Jinn S. et al. Functionalization of the TMEM175 p.M393T variant as a risk factor for Parkinson disease. Hum. Mol. Genet 28, 3244–3254 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 160.Krohn L. et al. Genetic, structural, and functional evidence link TMEM175 to synucleinopathies. Ann. Neurol 87, 139–153 (2020). [DOI] [PubMed] [Google Scholar]
- 161.Palomba NP et al. Common and rare variants in TMEM175 gene concur to the pathogenesis of Parkinson’s disease in Italian patients. Mol. Neurobiol 60, 2150–2173 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 162.Cang C, Aranda K, Seo YJ, Gasnier B. & Ren D. TMEM175 Is an organelle K+ channel regulating lysosomal function. Cell 162, 1101–1112 (2015). [DOI] [PubMed] [Google Scholar]
- 163.Jinn S. et al. TMEM175 deficiency impairs lysosomal and mitochondrial function and increases α-synuclein aggregation. Proc. Natl Acad. Sci. USA 114, 2389–2394 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 164.Hu M. et al. Parkinson’s disease-risk protein TMEM175 is a proton-activated proton channel in lysosomes. Cell 185, 2292–2308.e20 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 165.Zheng W. et al. pH regulates potassium conductance and drives a constitutive proton current in human TMEM175. Sci. Adv 8, eabm1568 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 166.Tang T, Jian B. & Liu Z. Transmembrane protein 175, a lysosomal ion channel related to Parkinson’s disease. Biomolecules 13, 802 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 167.Wie J. et al. A growth-factor-activated lysosomal K+ channel regulates Parkinson’s pathology. Nature 591, 431–437 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 168.Henderson MX et al. Spread of α-synuclein pathology through the brain connectome is modulated by selective vulnerability and predicted by network analysis. Nat. Neurosci 22, 1248–1257 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 169.Rohrer JD et al. The heritability and genetics of frontotemporal lobar degeneration. Neurology 73, 1451–1456 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 170.Logan T. et al. Rescue of a lysosomal storage disorder caused by Grn loss of function with a brain penetrant progranulin biologic. Cell 184, 4651–4668.e25 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 171.Hasan S. et al. Multi-modal proteomic characterization of lysosomal function and proteostasis in progranulin-deficient neurons. Mol. Neurodegener 18, 87 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 172.Klein ZA et al. Loss of TMEM106B ameliorates lysosomal and frontotemporal dementia-related phenotypes in progranulin-deficient mice. Neuron 95, 281–296.e6 (2017). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 173.Brady OA, Zheng Y, Murphy K, Huang M. & Hu F. The frontotemporal lobar degeneration risk factor, TMEM106B, regulates lysosomal morphology and function. Hum. Mol. Genet 22, 685–695 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 174.Schwenk BM et al. The FTLD risk factor TMEM106B and MAP6 control dendritic trafficking of lysosomes. EMBO J. 33, 450–467 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 175.Lang CM et al. Membrane orientation and subcellular localization of transmembrane protein 106B (TMEM106B), a major risk factor for frontotemporal lobar degeneration. J. Biol. Chem 287, 19355–19365 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 176.Van Deerlin VM et al. Common variants at 7p21 are associated with frontotemporal lobar degeneration with TDP-43 inclusions. Nat. Genet 42, 234–239 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 177.Cruchaga C. et al. Association of TMEM106B gene polymorphism with age at onset in granulin mutation carriers and plasma granulin protein levels. Arch. Neurol 68, 581–586 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 178.Finch N. et al. TMEM106B regulates progranulin levels and the penetrance of FTLD in GRN mutation carriers. Neurology 76, 467–474 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 179.Feng T. et al. Loss of TMEM106B and PGRN leads to severe lysosomal abnormalities and neurodegeneration in mice. EMBO Rep. 21, e50219 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 180.Werner G. et al. Loss of TMEM106B potentiates lysosomal and FTLD-like pathology in progranulin-deficient mice. EMBO Rep. 21, e50241 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 181.Zhou X. et al. Loss of TMEM106B leads to myelination deficits: implications for frontotemporal dementia treatment strategies. Brain 143, 1905–1919 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 182.Stroobants S, D’Hooge R. & Damme M. Aged Tmem106b knockout mice display gait deficits in coincidence with Purkinje cell loss and only limited signs of non-motor dysfunction. Brain Pathol. 31, 223–238 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 183.Luningschror P. et al. The FTLD risk factor TMEM106B regulates the transport of lysosomes at the axon initial segment of motoneurons. Cell Rep. 30, 3506–3519.e6 (2020). [DOI] [PubMed] [Google Scholar]
- 184.Yadati T, Houben T, Bitorina A. & Shiri-Sverdlov R. The ins and outs of cathepsins: physiological function and role in disease management. Cells 9, 1679 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 185.Vidoni C, Follo C, Savino M, Melone MA & Isidoro C. The role of cathepsin D in the pathogenesis of human neurodegenerative disorders. Med. Res. Rev 36, 845–870 (2016). [DOI] [PubMed] [Google Scholar]
- 186.Zhang KY, Yang S, Warraich ST & Blair IP Ubiquilin 2: a component of the ubiquitin–proteasome system with an emerging role in neurodegeneration. Int. J. Biochem. Cell Biol 50, 123–126 (2014). [DOI] [PubMed] [Google Scholar]
- 187.Wu JJ et al. ALS/FTD mutations in UBQLN2 impede autophagy by reducing autophagosome acidification through loss of function. Proc. Natl Acad. Sci. USA 117, 15230–15241 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 188.Şentürk M. et al. Ubiquilins regulate autophagic flux through mTOR signalling and lysosomal acidification. Nat. Cell Biol 21, 384–396 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 189.Christensen KA, Myers JT & Swanson JA pH-dependent regulation of lysosomal calcium in macrophages. J. Cell Sci 115, 599–607 (2002). [DOI] [PubMed] [Google Scholar]
- 190.Churchill GC et al. NAADP mobilizes Ca2+ from reserve granules, lysosome-related organelles, in sea urchin eggs. Cell 111, 703–708 (2002). [DOI] [PubMed] [Google Scholar]
- 191.Guse AH & Lee HC NAADP: a universal Ca2+ trigger. Sci. Signal 1, re10 (2008). [DOI] [PubMed] [Google Scholar]
- 192.Feng X. & Yang J. Lysosomal calcium in neurodegeneration. Messenger 5, 56–66 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 193.Tedeschi V. & Secondo A. Emerging role of lysosomal calcium store as a hub of neuroprotection. Neural Regen. Res 17, 1259–1260 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 194.Lloyd-Evans E. & Waller-Evans H. Lysosomal Ca2+ homeostasis and signaling in health and disease. Cold Spring Harb. Perspect. Biol 12, a035311 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 195.Medina DL et al. Lysosomal calcium signalling regulates autophagy through calcineurin and TFEB. Nat. Cell Biol 17, 288–299 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 196.Berman ER, Livni N, Shapira E, Merin S. & Levij IS Congenital corneal clouding with abnormal systemic storage bodies: a new variant of mucolipidosis. J. Pediatr 84, 519–526 (1974). [DOI] [PubMed] [Google Scholar]
- 197.Shen D. et al. Lipid storage disorders block lysosomal trafficking by inhibiting a TRP channel and lysosomal calcium release. Nat. Commun 3, 731 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 198.Samie MA et al. The tissue-specific expression of TRPML2 (MCOLN-2) gene is influenced by the presence of TRPML1. Pflug. Arch 459, 79–91 (2009). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 199.Grimm C, Hassan S, Wahl-Schott C. & Biel M. Role of TRPML and two-pore channels in endolysosomal cation homeostasis. J. Pharmacol. Exp. Ther 342, 236–244 (2012). [DOI] [PubMed] [Google Scholar]
- 200.Guse AH Second messenger signaling: multiple receptors for NAADP. Curr. Biol 19, R521–R523 (2009). [DOI] [PubMed] [Google Scholar]
- 201.Xiong J. & Zhu MX Regulation of lysosomal ion homeostasis by channels and transporters. Sci. China Life Sci 59, 777–791 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 202.Cheng X, Zhang X, Yu L. & Xu H. Calcium signaling in membrane repair. Semin. Cell Dev. Biol 45, 24–31 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 203.Morgan AJ, Platt FM, Lloyd-Evans E. & Galione A. Molecular mechanisms of endolysosomal Ca2+ signalling in health and disease. Biochem. J 439, 349–374 (2011). [DOI] [PubMed] [Google Scholar]
- 204.Patel S. & Cai X. Evolution of acidic Ca2+ stores and their resident Ca2+-permeable channels. Cell Calcium 57, 222–230 (2015). [DOI] [PubMed] [Google Scholar]
- 205.Waller-Evans H. & Lloyd-Evans E. Regulation of TRPML1 function. Biochem. Soc. Trans 43, 442–446 (2015). [DOI] [PubMed] [Google Scholar]
- 206.Soyombo AA et al. TRP-ML1 regulates lysosomal pH and acidic lysosomal lipid hydrolytic activity. J. Biol. Chem 281, 7294–7301 (2006). [DOI] [PubMed] [Google Scholar]
- 207.Zhang X. et al. MCOLN1 is a ROS sensor in lysosomes that regulates autophagy. Nat. Commun 7, 12109 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 208.Butterfield DA, Di Domenico F, Swomley AM, Head E. & Perluigi M. Redox proteomics analysis to decipher the neurobiology of Alzheimer-like neurodegeneration: overlaps in Down’s syndrome and Alzheimer’s disease brain. Biochem. J 463, 177–189 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 209.Mundy DI, Li WP, Luby-Phelps K. & Anderson RG Caveolin targeting to late endosome/lysosomal membranes is induced by perturbations of lysosomal pH and cholesterol content. Mol. Biol. Cell 23, 864–880 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 210.Ollinger K. & Brunk UT Cellular injury induced by oxidative stress is mediated through lysosomal damage. Free. Radic. Biol. Med 19, 565–574 (1995). [DOI] [PubMed] [Google Scholar]
- 211.Samie M. et al. A TRP channel in the lysosome regulates large particle phagocytosis via focal exocytosis. Dev. Cell 26, 511–524 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 212.Elbaz-Alon Y. et al. A dynamic interface between vacuoles and mitochondria in yeast. Dev. Cell 30, 95–102 (2014). [DOI] [PubMed] [Google Scholar]
- 213.Li L, Tan J, Miao Y, Lei P. & Zhang Q. ROS and autophagy: interactions and molecular regulatory mechanisms. Cell Mol. Neurobiol 35, 615–621 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 214.Potz BA, Abid MR & Sellke FW Role of calpain in pathogenesis of human disease processes. J. Nat. Sci 2, e218 (2016). [PMC free article] [PubMed] [Google Scholar]
- 215.Nixon RA The calpains in aging and aging-related diseases. Ageing Res. Rev 2, 407–418 (2003). [DOI] [PubMed] [Google Scholar]
- 216.Diepenbroek M. et al. Overexpression of the calpain-specific inhibitor calpastatin reduces human α-synuclein processing, aggregation and synaptic impairment in [A30P] αSyn transgenic mice. Hum. Mol. Genet 23, 3975–3989 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 217.Menzies FM et al. Calpain inhibition mediates autophagy-dependent protection against polyglutamine toxicity. Cell Death Differ. 22, 433–444 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 218.Rao MV et al. Specific calpain inhibition by calpastatin prevents tauopathy and neurodegeneration and restores normal lifespan in tau P301L mice. J. Neurosci 34, 9222–9234 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 219.Requejo-Aguilar R. Cdk5 and aberrant cell cycle activation at the core of neurodegeneration. Neural Regen. Res 18, 1186–1190 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 220.Yang J, Zhao Z, Gu M, Feng X. & Xu H. Release and uptake mechanisms of vesicular Ca2+ stores. Protein Cell 10, 8–19 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 221.Ogunbayo OA et al. mTORC1 controls lysosomal Ca2+ release through the two-pore channel TPC2. Sci. Signal 11, eaao5775 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 222.Thakore P. et al. TRPML1 channels initiate Ca2+ sparks in vascular smooth muscle cells. Sci. Signal 10.1126/scisignal.aba1015 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 223.Griffin CS et al. The intracellular Ca2+ release channel TRPML1 regulates lower urinary tract smooth muscle contractility. Proc. Natl Acad. Sci. USA 117, 30775–30786 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 224.Li G. & Li PL Lysosomal TRPML1 channel: implications in cardiovascular and kidney diseases. Adv. Exp. Med. Biol 1349, 275–301 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 225.Scotto Rosato A. et al. TRPML1 links lysosomal calcium to autophagosome biogenesis through the activation of the CaMKKβ/VPS34 pathway. Nat. Commun 10, 5630 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 226.Kilpatrick BS Connecting Ca2+ and lysosomes to Parkinson disease. Messenger 5, 76–86 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 227.Kilpatrick BS et al. Endoplasmic reticulum and lysosomal Ca2+ stores are remodelled in GBA1-linked Parkinson disease patient fibroblasts. Cell Calcium 59, 12–20 (2016). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 228.Hui L. et al. Acidifying endolysosomes prevented low-density lipoprotein-induced amyloidogenesis. J. Alzheimers Dis 67, 393–410 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 229.Somogyi A. et al. The synthetic TRPML1 agonist ML-SA1 rescues Alzheimer-related alterations of the endosomal–autophagic–lysosomal system. J. Cell Sci 136, jcs259875 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 230.Drobny A. et al. The role of lysosomal cathepsins in neurodegeneration: mechanistic insights, diagnostic potential and therapeutic approaches. Biochim. Biophys. Acta Mol. Cell Res 1869, 119243 (2022). [DOI] [PubMed] [Google Scholar]
- 231.Samie MA & Xu H. Lysosomal exocytosis and lipid storage disorders. J. Lipid Res 55, 995–1009 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 232.Hoglinger D. et al. NPC1 regulates ER contacts with endocytic organelles to mediate cholesterol egress. Nat. Commun 10, 4276 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 233.Tancini B. et al. Insight into the role of extracellular vesicles in lysosomal storage disorders. Genes 10, 510 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 234.Lloyd-Evans E. & Platt FM Lipids on trial: the search for the offending metabolite in Niemann–Pick type C disease. Traffic 11, 419–428 (2010). [DOI] [PubMed] [Google Scholar]
- 235.Wheeler S, Schmid R. & Sillence DJ Lipid–protein interactions in Niemann–Pick type C disease: insights from molecular modeling. Int. J. Mol. Sci 20, 717 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 236.Butterfield DA, Gu L, Di Domenico F. & Robinson RA Mass spectrometry and redox proteomics: applications in disease. Mass. Spectrom. Rev 33, 277–301 (2014). [DOI] [PubMed] [Google Scholar]
- 237.Di Domenico F. et al. Redox proteomics analyses of the influence of co-expression of wild-type or mutated LRRK2 and Tau on C. elegans protein expression and oxidative modification: relevance to Parkinson disease. Antioxid. Redox Signal 17, 1490–1506 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 238.Yang DS et al. Defective macroautophagic turnover of brain lipids in the TgCRND8 Alzheimer mouse model: prevention by correcting lysosomal proteolytic deficits. Brain 137, 3300–3318 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 239.Cox TM Lysosomal diseases and neuropsychiatry: opportunities to rebalance the mind. Front. Mol. Biosci 7, 177 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 240.Matutino Santos P, Pereira Campos G. & Nascimento C. Endo-lysosomal and autophagy pathway and ubiquitin-proteasome system in mood disorders: a review article. Neuropsychiatr. Dis. Treat 19, 133–151 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 241.Chu CT Autophagic stress in neuronal injury and disease. J. Neuropathol. Exp. Neurol 65, 423–432 (2006). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 242.Kang R, Zeh HJ, Lotze MT & Tang D. The Beclin 1 network regulates autophagy and apoptosis. Cell Death Differ. 18, 571–580 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 243.Park H, Kang JH & Lee S. Autophagy in neurodegenerative diseases: a hunter for aggregates. Int. J. Mol. Sci 21, 3369 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 244.Dehay B. et al. Pathogenic lysosomal depletion in Parkinson’s disease. J. Neurosci 30, 12535–12544 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 245.Papadopoulos C. & Meyer H. Detection and clearance of damaged lysosomes by the endo-lysosomal damage response and lysophagy. Curr. Biol 27, R1330–R1341 (2017). [DOI] [PubMed] [Google Scholar]
- 246.Alu A. et al. The role of lysosome in regulated necrosis. Acta Pharm. Sin. B 10, 1880–1903 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 247.Ketterer S. et al. Inherited diseases caused by mutations in cathepsin protease genes. FEBS J. 284, 1437–1454 (2017). [DOI] [PubMed] [Google Scholar]
- 248.Perluigi M, Di Domenico F. & Butterfield DA Oxidative damage in neurodegeneration: roles in the pathogenesis and progression of Alzheimer disease. Physiol. Rev 104, 103–197 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 249.Perry G, Cash AD & Smith MA Alzheimer disease and oxidative stress. J. Biomed. Biotechnol 2, 120–123 (2002). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 250.J. S. Does high iron push a person with pathology into dementia? ALZFORUM https://www.alzforum.org/news/research-news/does-high-iron-push-person-pathology-dementia (2019). [Google Scholar]
- 251.Zhang G. et al. The potential role of ferroptosis in Alzheimer’s disease. J. Alzheimers Dis 80, 907–925 (2021). [DOI] [PubMed] [Google Scholar]
- 252.Sheftel AD, Zhang AS, Brown C, Shirihai OS & Ponka P. Direct interorganellar transfer of iron from endosome to mitochondrion. Blood 110, 125–132 (2007). [DOI] [PubMed] [Google Scholar]
- 253.Ashraf A, Clark M. & So PW The aging of iron man. Front. Aging Neurosci 10, 65 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 254.Schubert D. & Chevion M. The role of iron in β-amyloid toxicity. Biochem. Biophys. Res. Commun 216, 702–707 (1995). [DOI] [PubMed] [Google Scholar]
- 255.Smith MA & Perry G. Free radical damage, iron, and Alzheimer’s disease. J. Neurol. Sci 134, 92–94 (1995). [DOI] [PubMed] [Google Scholar]
- 256.Yambire KF et al. Impaired lysosomal acidification triggers iron deficiency and inflammation in vivo. eLife 8, e51031 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 257.Ou M. et al. Role and mechanism of ferroptosis in neurological diseases. Mol. Metab 61, 101502 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 258.Pensalfini A. et al. Intracellular amyloid and the neuronal origin of Alzheimer neuritic plaques. Neurobiol. Dis 71, 53–61 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 259.Eckman EA et al. Nascent Aβ42 fibrillization in synaptic endosomes precedes plaque formation in a mouse model of Alzheimer’s-like β-amyloidosis. J. Neurosci 43, 8812–8824 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 260.Decressac M. et al. TFEB-mediated autophagy rescues midbrain dopamine neurons from α-synuclein toxicity. Proc. Natl Acad. Sci. USA 110, E1817–E1826 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 261.Siddiqi FH et al. Felodipine induces autophagy in mouse brains with pharmacokinetics amenable to repurposing. Nat. Commun 10, 1817 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 262.Young MM et al. Autophagosomal membrane serves as platform for intracellular death-inducing signaling complex (iDISC)-mediated caspase-8 activation and apoptosis. J. Biol. Chem 287, 12455–12468 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 263.Maetzel D. et al. Genetic and chemical correction of cholesterol accumulation and impaired autophagy in hepatic and neural cells derived from Niemann–Pick type C patient-specific iPS cells. Stem Cell Rep. 2, 866–880 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 264.Elrick MJ, Yu T, Chung C. & Lieberman AP Impaired proteolysis underlies autophagic dysfunction in Niemann–Pick type C disease. Hum. Mol. Genet 21, 4876–4887 (2012). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 265.Song JX et al. HMGB1 is involved in autophagy inhibition caused by SNCA/α-synuclein overexpression: a process modulated by the natural autophagy inducer corynoxine B. Autophagy 10, 144–154 (2014). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 266.Fujii T. et al. Parkinson’s disease-associated ATP13A2/PARK9 functions as a lysosomal H+,K+-ATPase. Nat. Commun 14, 2174 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 267.Menzies FM et al. Autophagy and neurodegeneration: pathogenic mechanisms and therapeutic opportunities. Neuron 93, 1015–1034 (2017). [DOI] [PubMed] [Google Scholar]
- 268.Boland B. et al. Promoting the clearance of neurotoxic proteins in neurodegenerative disorders of ageing. Nat. Rev. Drug Discov 17, 660–688 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 269.Morawe MP et al. Pharmacological mTOR-inhibition facilitates clearance of AD-related tau aggregates in the mouse brain. Eur. J. Pharmacol 934, 175301 (2022). [DOI] [PubMed] [Google Scholar]
- 270.Majumder S, Richardson A, Strong R. & Oddo S. Inducing autophagy by rapamycin before, but not after, the formation of plaques and tangles ameliorates cognitive deficits. PLoS ONE 6, e25416 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 271.Settembre C. et al. TFEB links autophagy to lysosomal biogenesis. Science 332, 1429–1433 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 272.Wang H, Zhu Y, Liu H, Liang T. & Wei Y. Advances in drug discovery targeting lysosomal membrane proteins. Pharmaceuticals 16, 601 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 273.Xue W, Zhang J. & Li Y. Enhancement of lysosome biogenesis as a potential therapeutic approach for neurodegenerative diseases. Neural Regen. Res 18, 2370–2376 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 274.Peng W, Minakaki G, Nguyen M. & Krainc D. Preserving lysosomal function in the aging brain: insights from neurodegeneration. Neurotherapeutics 16, 611–634 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 275.Bonam SR, Wang F. & Muller S. Lysosomes as a therapeutic target. Nat. Rev. Drug Discov 18, 923–948 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 276.Lee JH et al. β2-Adrenergic agonists rescue lysosome acidification and function in PSEN1 deficiency by reversing defective ER-to-lysosome delivery of ClC-7. J. Mol. Biol 432, 2633–2650 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 277.Yang DS et al. Reversal of autophagy dysfunction in the TgCRND8 mouse model of Alzheimer’s disease ameliorates amyloid pathologies and memory deficits. Brain 134, 258–277 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 278.Cason SE et al. Sequential dynein effectors regulate axonal autophagosome motility in a maturation-dependent pathway. J. Cell Biol 220, e202010179 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 279.Forgac M. Vacuolar ATPases: rotary proton pumps in physiology and pathophysiology. Nat. Rev. Mol. Cell Biol 8, 917–929 (2007). [DOI] [PubMed] [Google Scholar]
- 280.Checler F, Afram E, Pardossi-Piquard R. & Lauritzen I. Is γ-secretase a beneficial inactivating enzyme of the toxic APP C-terminal fragment C99? J. Biol. Chem 296, 100489 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 281.Bourgeois A. et al. Intraneuronal accumulation of C99 contributes to synaptic alterations, apathy-like behavior, and spatial learning deficits in 3xTgAD and 2xTgAD mice. Neurobiol. Aging 71, 21–31 (2018). [DOI] [PubMed] [Google Scholar]
- 282.Jin LW, Shie FS, Maezawa I, Vincent I. & Bird T. Intracellular accumulation of amyloidogenic fragments of amyloid-β precursor protein in neurons with Niemann–Pick type C defects is associated with endosomal abnormalities. Am. J. Pathol 164, 975–985 (2004). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 283.Holsinger RM, McLean CA, Beyreuther K, Masters CL & Evin G. Increased expression of the amyloid precursor β-secretase in Alzheimer’s disease. Ann. Neurol 51, 783–786 (2002). [DOI] [PubMed] [Google Scholar]
- 284.Tian Y, Bustos V, Flajolet M. & Greengard P. A small-molecule enhancer of autophagy decreases levels of Aβ and APP-CTF via Atg5-dependent autophagy pathway. FASEB J. 25, 1934–1942 (2011). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 285.Lauritzen I, Pardossi-Piquard R, Bourgeois A, Bécot A. & Checler F. Does intraneuronal accumulation of carboxyl-terminal fragments of the amyloid precursor protein trigger early neurotoxicity in Alzheimer’s disease? Curr. Alzheimer Res 16, 453–457 (2019). [DOI] [PubMed] [Google Scholar]
- 286.Lardelli M. An alternative view of familial Alzheimer’s disease genetics. J. Alzheimers Dis 96, 13–39 (2023). [DOI] [PubMed] [Google Scholar]
- 287.Vaillant-Beuchot L. et al. Accumulation of amyloid precursor protein C-terminal fragments triggers mitochondrial structure, function, and mitophagy defects in Alzheimer’s disease models and human brains. Acta Neuropathol. 141, 39–65 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 288.Fukumoto H, Cheung BS, Hyman BT & Irizarry MC β-Secretase protein and activity are increased in the neocortex in Alzheimer disease. Arch. Neurol 59, 1381–1389 (2002). [DOI] [PubMed] [Google Scholar]
- 289.Ahmed RR et al. BACE1 and BACE2 enzymatic activities in Alzheimer’s disease. J. Neurochem 112, 1045–1053 (2010). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 290.Kim HS et al. Carboxyl-terminal fragment of Alzheimer’s APP destabilizes calcium homeostasis and renders neuronal cells vulnerable to excitotoxicity. FASEB J. 14, 1508–1517 (2000). [DOI] [PubMed] [Google Scholar]
- 291.Takasugi N. et al. The pursuit of the “inside” of the amyloid hypothesis — is C99 a promising therapeutic target for Alzheimer’s disease? Cells 12, 454 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 292.Pera M. et al. MAM and C99, key players in the pathogenesis of Alzheimer’s disease. Int. Rev. Neurobiol 154, 235–278 (2020). [DOI] [PubMed] [Google Scholar]
- 293.Patel S, Yuan Y, Gunaratne GS, Rahman T. & Marchant JS Activation of endo-lysosomal two-pore channels by NAADP and PI(3,5)P2. Five things to know. Cell Calcium 103, 102543 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 294.Lu Y, Hao B, Graeff R. & Yue J. NAADP/TPC2/Ca2+ signaling inhibits autophagy. Commun. Integr. Biol 6, e27595 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 295.Moriyama Y, Maeda M. & Futai M. Involvement of a non-proton pump factor (possibly Donnan-type equilibrium) in maintenance of an acidic pH in lysosomes. FEBS Lett. 302, 18–20 (1992). [DOI] [PubMed] [Google Scholar]
- 296.Grabe M. & Oster G. Regulation of organelle acidity. J. Gen. Physiol 117, 329–344 (2001). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 297.Guerrero-Navarro L, Jansen-Dürr P. & Cavinato M. Age-related lysosomal dysfunctions. Cells 11, 1977 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 298.Fischer P-DD Oskar Miliare Nekrosen mit drusigen Wucherungen der Neurofibrillen, eine regelmässige Veränderung der Hirnrinde bei seniler Demenz [German]. Monatsschrift f.ür. Psychiatr. und Neurologie 22, 361–372 (1907). [Google Scholar]
- 299.Puri C. et al. The RAB11A-positive compartment is a primary platform for autophagosome assembly mediated by WIPI2 recognition of PI3P-RAB11A. Dev. Cell 45, 114–131.e8 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 300.Gomez-Sanchez R, Tooze SA & Reggiori F. Membrane supply and remodeling during autophagosome biogenesis. Curr. Opin. Cell Biol 71, 112–119 (2021). [DOI] [PubMed] [Google Scholar]
- 301.Zheng Q. et al. Calcium transients on the ER surface trigger liquid–liquid phase separation of FIP200 to specify autophagosome initiation sites. Cell 185, 4082–4098.e22 (2022). [DOI] [PubMed] [Google Scholar]
- 302.Osawa T, Matoba K. & Noda NN Lipid transport from endoplasmic reticulum to autophagic membranes. Cold Spring Harb. Perspect. Biol 14, a041254 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 303.Li S. et al. A new type of ERGIC–ERES membrane contact mediated by TMED9 and SEC12 is required for autophagosome biogenesis. Cell Res. 32, 119–138 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 304.Kumar S. et al. Mammalian hybrid pre-autophagosomal structure HyPAS generates autophagosomes. Cell 184, 5950–5969.e22 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 305.Jahreiss L, Menzies FM & Rubinsztein DC The itinerary of autophagosomes: from peripheral formation to kiss-and-run fusion with lysosomes. Traffic 9, 574–587 (2008). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 306.Zhao YG, Codogno P. & Zhang H. Machinery, regulation and pathophysiological implications of autophagosome maturation. Nat. Rev. Mol. Cell Biol 22, 733–750 (2021). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 307.Liang W. et al. Mitochondria are secreted in extracellular vesicles when lysosomal function is impaired. Nat. Commun 14, 5031 (2023). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 308.Heckmann BL et al. LC3-associated endocytosis facilitates β-amyloid clearance and mitigates neurodegeneration in murine Alzheimer’s disease. Cell 183, 1733–1734 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 309.Deretic V. et al. Membrane atg8ylation in canonical and noncanonical autophagy. J. Mol. Biol 10.1016/j.jmb.2024.168532 (2024). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 310.Almeida MF, Bahr BA & Kinsey ST Endosomal–lysosomal dysfunction in metabolic diseases and Alzheimer’s disease. Int. Rev. Neurobiol 154, 303–324 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 311.Qureshi YH, Baez P. & Reitz C. Endosomal trafficking in Alzheimer’s disease, Parkinson’s disease, and neuronal ceroid lipofuscinosis. Mol. Cell Biol 40 e00262–20 (2020). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 312.Winckler B. et al. The endolysosomal system and proteostasis: from development to degeneration. J. Neurosci 38, 9364–9374 (2018). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 313.Small SA & Petsko GA Retromer in Alzheimer disease, Parkinson disease and other neurological disorders. Nat. Rev. Neurosci 16, 126–132 (2015). [DOI] [PubMed] [Google Scholar]
- 314.Mathews PM & Levy E. Exosome production is key to neuronal endosomal pathway integrity in neurodegenerative diseases. Front. Neurosci 13, 1347 (2019). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 315.Izco M, Carlos E. & Alvarez-Erviti L. Impact of endolysosomal dysfunction upon exosomes in neurodegenerative diseases. Neurobiol. Dis 166, 105651 (2022). [DOI] [PubMed] [Google Scholar]
- 316.Annunziata I. et al. Lysosomal NEU1 deficiency affects amyloid precursor protein levels and amyloid-β secretion via deregulated lysosomal exocytosis. Nat. Commun 4, 2734 (2013). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 317.Bodzeta A, Kahms M. & Klingauf J. The presynaptic v-ATPase reversibly disassembles and thereby modulates exocytosis but is not part of the fusion machinery. Cell Rep. 20, 1348–1359 (2017). [DOI] [PubMed] [Google Scholar]
- 318.Schiapparelli LM et al. Proteomic screen reveals diverse protein transport between connected neurons in the visual system. Cell Rep. 38, 110287 (2022). [DOI] [PMC free article] [PubMed] [Google Scholar]
- 319.Bae EJ et al. Haploinsufficiency of cathepsin D leads to lysosomal dysfunction and promotes cell-to-cell transmission of α-synuclein aggregates. Cell Death Dis. 6, e1901 (2015). [DOI] [PMC free article] [PubMed] [Google Scholar]