Abstract
Autophagy is a constitutive process that degrades, recycles and clears damaged proteins or organelles, yet, despite activation of this pathway, abnormal proteins accumulate in neurons in neurodegenerative diseases and in oligodendrocytes in white matter disorders. Here, we discuss the role of autophagy in white matter disorders, including neurotropic infections, inflammatory diseases such as multiple sclerosis, and in hereditary metabolic disorders and acquired toxic‐metabolic disorders. Once triggered due to cell stress, autophagy can enhance cell survival or cell death that may contribute to oligodendrocyte damage and myelin loss in white matter diseases. For some disorders, the mechanisms leading to myelin loss are clear, whereas the aetiological agent and pathological mechanisms are unknown for other myelin disorders, although emerging studies indicate that a common mechanism underlying these disorders is dysregulation of autophagic pathways. In this review we discuss the alterations in the autophagic process in white matter disorders and the potential use of autophagy‐modulating agents as therapeutic approaches in these pathological conditions. © 2020 The Authors. The Journal of Pathology published by John Wiley & Sons, Ltd. on behalf of The Pathological Society of Great Britain and Ireland.
Keywords: white matter disorders, autophagy, demyelination, multiple sclerosis, therapy, demyelinating disease, leukodystrophy
Introduction
Neurological diseases pose a major public health challenge due to the increasing ageing community. A common pathological feature of many neurological diseases is myelin damage that occurs as a primary event or arises secondarily to neuronal, axonal damage and infections. Here, we focus on white matter disorders (WMDs) that primarily affect the myelin in the CNS [1] and are acquired as a result of infection or due to autoimmunity, or occur as a consequence of genetic mutations. Many of these diseases show increased cell death and have limited therapeutic options available (Table 1).
Table 1.
CNS WMDs, cell death mechanisms and therapies.
| Type | Disease example | Clinical symptoms | Pathology | Causative factor | Cell death mechanism | Therapy | Reference |
|---|---|---|---|---|---|---|---|
| Acquired disorders | |||||||
| Inflammatory | MS | Sensory loss, motor deficits, cognitive changes | Inflammation, demyelination, axonal loss and gliosis | Not known |
Viral infection Pathogenic T‐ and B‐cells |
Targeting immune response | [2] |
| NMO | Vision and spinal cord function loss |
Lesions in optic nerve and spinal cord. Loss of AQP4 expression |
Antibodies against AQP4 | Antibody mediated | Targeting immune response | [3] | |
| ADEM | Motor and neurocognitive deficits | Widespread CNS inflammation and demyelination | Infection | Autoimmune | Immune therapy | [4] | |
| AHL | Rapid onset fever, neck stiffness, fatigue, headache, nausea, vomiting, seizures, coma | Inflammatory haemorrhagic demyelination of the white matter | Follows viral and bacterial infections and vaccinations | Autoimmune reaction to viral antigens | Steroids and plasma exchange | [5] | |
| Infectious | PML | Progressive weakness, motor deficits, cognitive changes | Focal areas of demyelination | Polyomavirus JC virus replication | Viral cytotoxicity | No effective therapy | [6] |
| SSPE | Progressive neurological and psychological deterioration. Seizures, ataxia, photosensitivity, spasticity, coma | Viral inclusion bodies in neurons, neuronal damage and loss | Abnormal viral replication in neurons | Persistent infection with MeV |
No effective therapy. Anti‐convulsive therapy for palliative care |
[7] | |
| Congenital cytomegalovirus | Hearing loss, vision impairment, learning disability | Encephalitis, microglial activation | Virus inhibits NSPC proliferation and differentiation. Neuronal cell loss | Neuronal apoptosis, autophagy | Ganciclovir or valganciclovir | [8] | |
| Toxic‐metabolic | Paraneoplastic syndrome |
Depends on tumour, e.g. NMO |
Tumour expressing CNS antigens | Not reported | IVIG | [9] | |
| Hypoxic–ischaemic | Binswanger disease | Vascular cognitive impairment and dementia | Chronic microvascular leukoencephalopathy, white matter lesions, axonal damage, BBB damage | Endothelia cell dysfunction | Not reported | No effective therapy | [10] |
| Cerebral hypoxia and ischaemia in newborns | Cerebral palsy, visual, auditory, motor and behavioural problems. Epilepsy, developmental delay, autism | Diffuse white matter damage, gliosis, decreased oligodendrocytes | Damage to neural stem cells and oligodendrocyte progenitors in the SVZ | Increased glutamate, free radicals, apoptosis autophagy | Hypothermia | [11] | |
| Traumatic | Diffuse axonal injury, chronic traumatic encephalopathy | Dependent on location of injury ‐ motor, memory, neuropsychological changes | Axonal damage, tau accumulation, secondary white matter damage, astrogliosis | White matter loss associated with astrogliosis and microglia activation | Glutamate excitotoxicity, intracellular Ca2+ accumulation, ROS production | Anti‐CD11d, progesterone, valganciclovir tacrolimus, moderate hypothermia (32–33 °C 1 h) | [12] |
| Genetic disorders | |||||||
| Lysosomal storage | Metachromatic leukodystrophy | Gait abnormalities, spasticity, ataxia, polyneuropathy psychosis, cognitive decline | Demyelination, sparing of U fibres. Eosinophilic granules in macrophages, metachromasia | Decrease in arylsulfatase A1. Sulphated glycolipid accumulation in myelin | Sulphatide accumulation induces apoptosis in vitro | HSCT, enzyme replacement therapy, gene therapy | [13] |
| Peroxisomal | X‐ALD | Ataxia, dementia, behavioural changes, hyperactivity | Increased saturated VLCFA in serum. Progressive demyelination | Mutations in ABCD1 gene | VLCFA accumulation in CNS | Allogeneic HSCT | [14] |
| Mitochondrial dysfunction with leukoencephalopathy | Leber's hereditary optic neuropathy | Acute/subacute painless central visual loss | Loss of retinal ganglion cells. Optic nerve degeneration | Mitochondrial DNA mutations | Proposed to be apoptotic | Antioxidants, experimental gene therapy | [15] |
| Nuclear DNA repair defects | Cockayne syndrome | Growth and development failure, accelerated, aging | Patchy myelin loss, white matter atrophy, neuronal loss, astrocytic gliosis, microglia nodules | Mutations in CSA or CSB genes. Lack of repair of damaged nuclear and mitochondrial DNA | Apoptotic cell death | Diet restriction or high fat diet, vitamin D. Otherwise no cure | [16] |
| Defects in genes encoding myelin proteins | Pelizaeus–Merzbacher disease | Dystonia, ataxia, nystagmus, spasticity, mild cognitive decline | Splitting and decompaction of myelin sheaths, axonal spheroids | Mutations in PLP1 and accumulation of aberrant protein, or GJC2 mutation | UPR‐induced apoptotic pathway | Experimental neural stem cell and glial progenitor cell transplantation | [17] |
| Amino acid and organic acid metabolism disorders | Canavan disease | Macrocephaly, loss of head control, developmental delay, hypotonia and spasticity | Diffuse spongiform white matter degeneration, dysmyelination and intramyelinic oedema | Mutation in ASPA encoding aspartoacylase and accumulation of NAA | Not reported | No effective therapy | [18] |
| Miscellaneous | Alexander disease | Macrocephaly, dementia, spasticity, developmental delay | Myelin damage. Elevated GFAP in cerebrospinal fluid. Rosenthal fibres | GFAP mutation | Reduced GLT‐1, increased autophagy in astrocytes | No effective therapy | [19] |
| VWM | Spasticity, loss motor function, epilepsy, ataxia | Progressive demyelination | Mutations in EIF2B1–EIF2B5 | Increased cellular stress | No effective therapy | [20] | |
| CADASIL | Migraines, TIAs, dementia, apathy, depression | Diffuse white matter lesions, subcortical infarcts. Granular osmiophilic material in small vessels | NOTCH3 mutation | Protein misfolding and receptor aggregation | No effective therapy | [15] | |
BBB, blood–brain barrier; CADASIL, cerebral autosomal dominant arteriopathy with subcortical infarcts and leukoencephalopathy; GFAP, glial fibrillary acidic protein; HSCT, haematopoietic stem cell therapy; IVIG, intravenous immunoglobulins; NMO, neuromyelitis optica; NSPC, neural stem/progenitor cells; SSPE, subacute sclerosing panencephalitis; SVZ, subventricular zone; TIA, transient ischaemic attack.
A prominent CNS demyelinating disease in young adults is multiple sclerosis (MS), which affects one in 1000 people in the UK and the Netherlands, and an estimated 2.3 million cases worldwide. Myelin damage, inflammation and neurodegeneration are key pathological hallmarks in MS. Although MS has been described for many years, the pathogenic processes are still largely enigmatic, and the disease is still inadequately controlled. A similar disease to MS is acute disseminated encephalomyelitis (ADEM), an immune‐mediated demyelinating CNS disorder with a predilection to early childhood. Children with ADEM often present with acute neurological deficits following infections. Although the outcome after treatment is generally favourable, therapies are not absolute (Table 1).
More severe yet less common are genetic WMDs or leukoencephalopathies that cause devastating CNS damage in children but are also observed in adults. Leukodystrophies arise as a result of genetic mutations in myelin sheath formation and maintenance, some of which include abnormalities in autophagy pathways that lead to insufficient clearance of accumulated proteins. In these disorders, demyelination, axonal damage and neurodegeneration leads to a spectrum of severe clinical disabilities and early death [14, 20]. There is no effective therapy for these diseases, which include, for example, vanishing white matter (VWM), Alexander's disease and X‐linked adrenoleukodystrophy (X‐ALD).
WMDs also arise following neurotropic infections. These include progressive multifocal leukoencephalopathy (PML), measles, mumps, rubella and Lyme's disease. Although the aetiological agents in these diseases are known, the exact pathological mechanisms leading to myelin damage are unclear, except for PML, in which polyomavirus JC virus lyses the oligodendrocyte.
For most WMDs there is an urgent unmet clinical need for therapy, as these diseases are currently inadequately controlled (Table 1). A common pathological feature observed in many WMDs is increased cellular stress [21, 22, 23, 24], a process that activates the unfolded protein response (UPR). When this response is impaired, apoptotic pathways are activated to remove damaged cells [23] and in many cases increased stress also activates autophagic pathways. Autophagy is a fundamental and conserved lysosomal degradation pathway activated in response to stress. It is important for cell health and growth by maintaining a balance between synthesis and degradation of proteins and organelles [25]. The process acts as a garbage collector to recycle damaged proteins and organelles, a critical event during development, homeostasis and differentiation of cells in mammals. Autophagy can be distinguished into three different categories based on the route of action: chaperone‐mediated autophagy, microautophagy and macroautophagy. As macroautophagy has been the most extensively studied and understood, this review will focus on macroautophagy (hereafter termed autophagy) in WMDs. When autophagy is impaired, there is no capacity to degrade damaged organelles, misfolded proteins or invading microorganisms, leading to long‐term negative consequences and cell death. The impact of autophagy depends on the disease. Autophagy has been proposed to play a pathogenic role in neurodegenerative diseases including Parkinson's disease [26], Alzheimer's disease [27] and amyotrophic lateral sclerosis [28]. For other diseases, such as MS, X‐ALD, VWM or post‐infectious WMDs, little is known about the role of autophagy.
Autophagy can have a dual function based on the circumstances. On one hand the autophagic process can be protective; on the other it can be destructive for cells. Autophagy can act as a survival mechanism to prevent oxidative stress and genomic instability by constitutively eliminating defective proteins and organelles, generating nutrients and providing energy for the cell. In this way, autophagy plays a neuroprotective role, promotes cell survival and protects from neurodegeneration. In contrast, under certain conditions, for example excessive upregulation of autophagy or long‐term exposure to autophagy, cell death can be triggered. The mechanism by which autophagy induces cell death is not clearly understood. Autophagy‐induced cell death might be apoptosis related as autophagy and apoptosis share regulators such as Bcl‐2, Beclin‐1, ATG5, etc. Based on the consequences of autophagy in WMDs, it might be possible to either inhibit or stimulate autophagy for therapeutic purposes. Autophagy modulation is currently used for cancer therapy and several clinical trials are underway to examine autophagy‐modulating compounds.
Here, we review the role of autophagic processes in WMDs of the CNS taking into account the Janus face of autophagy that on one hand it may lead to oligodendrocyte and neuronal death, and on the other hand the pro‐survival function of autophagy may aid survival of pathogenic immune cells. We also review how autophagy pathways are modulated experimentally and how these therapeutic approaches can be applied in WMDs.
Autophagy pathways and regulators
Autophagy is regulated by autophagy‐related genes (ATGs) that are the core of the autophagic processes. More than 30 ATGs have been identified [29], some of which are involved in several pathophysiological conditions such as neurodegeneration [26, 27, 28] and neural infection [30]. Although autophagy is constitutively active at a basal level, several factors influence the rate of the process, including nutrient deprivation, growth factor withdrawal [31], ER stress [32, 33], ROS production, hypoxia and mitochondrial dysfunction. During inflammation, which is a key feature of many WMDs, several immune pathways are known to influence autophagy, including the quality and extent of inflammatory signals (e.g. IFN‐γ, IL‐1β) and activation of TLR and other pathogen recognition receptors [34].
Autophagy is characterised by the formation of an autophagosome and occurs in six stages: initiation, nucleation, elongation, closure, maturation and degradation (Figure 1). During initiation and nucleation, a double membrane phagophore is formed, and this process continues to form a spherical autophagosome. Subsequently, the autophagosome fuses with the lysosome to generate a single membrane autolysosome in which the autophagic cargo will be degraded by lytic hydrolases [35]. Initiation of autophagy is dependent on the Unc‐51‐like kinase 1 and 2 (ULK1/2) complex and the class III PI3K complex (Figure 1). The ULK1/2 complex, which consists of ULK1/2, ATG13, FIP200 and ATG101 [35], is responsible for controlling levels of autophagy and is highly upregulated during stress. Under nutrient‐rich conditions, the mammalian target of rapamycin (mTOR) interacts, phosphorylates and inactivates the ULK1/2 complex. When mTOR is inhibited, ULK1 and ULK2 activate and phosphorylate ATG13 and FIP200. The ULK1/2 complex consequently localises to the phagophore. ATG101 serves a stabilising role for ATG13 in the ULK1/2 complex and is essential for mammalian autophagy [36]. Additionally, AMP kinase (AMPK), a cellular metabolism regulator and energy sensor, phosphorylates ULK1 to induce autophagy. AMPK‐dependent and ‐independent processes activate ULK1 kinase in situations of glucose starvation and amino acid starvation, respectively [37]. Another common pathway that negatively impacts on autophagy is the RAS/cAMP protein kinase A (PKA) pathway that plays a role in sensing the availability of glucose, and is thus activated during nutrient deprivation [38]. When nutrients are readily available, Ras1 and Ras2 induce cAMP generation by adenylyl cyclase. This induces an inhibitory effect on PKA, which then downregulates the autophagy machinery (Figure 1).
Figure 1.

Macroautophagy machinery. Autophagy is regulated in six distinctive stages. Peripheral signals (e.g. starvation, growth factors) stimulate mTOR to activate the ULK1/2 complex, which in turn induces the class III PI3K complex. Nucleation is then initiated by a class III PI3K complex that induces a cascade of conjugations of AGSs, which lead to the formation of the dimeric ATG5–ATG12 complex and the production of LC3‐II, which aids in elongation of the now forming phagophore. Once the expanding phagophore is large enough it closes and an autophagosome is formed. The outer ATG5–ATG12 and LC3‐II are then cleaved from the membrane, after which it is ready to fuse with a lysosome, which is filled with lysosomal hydrolases. Fusion between the autophagosome and the lysosome creates an autolysosome, termed maturation. After fusion, the cargo (e.g. mitochondria or ribosomes) is degraded by lysosomal hydrolases and the end products are ready for reuse after export to the cytoplasm.
Phagophore assembly is regulated by the class III PI3K complex consisting of Beclin‐1, vacuolar protein sorting 34 (Vps34), p150 and ATG14 [35, 38]. Beclin‐1 is inhibited by Bcl‐2, which dissociates from Beclin‐1 during nutrient starvation. The class III PI3K complex then scouts for two ubiquitination‐like conjugation systems, ATG5–ATG12 and microtubule‐associated protein 1 light chain 3 (LC3). The ATG5–ATG12 complex is formed with the help of ATG7 and ATG10 and subsequently interacts with ATG16 followed by association with the phagophores. Furthermore, LC3‐I interacts with ATG7 and ATG3, after which it conjugates to phosphatidyl‐ethanolamine to form LC3‐II. LC3‐II localises on the forming autophagosome, where it serves as a recognition site for autophagosome synthesis to ensure elongation and expansion of the membrane [39]. LC3‐II levels are strongly correlated with the extent of autophagosome formation and LC3‐II is therefore often used to measure autophagic activity [40]. Lastly, ATG9 functions as a carrier by trafficking membranes from the trans‐Golgi network to the late endosomes, which are labelled with LC3‐II [41]. Once the phagophore transitions into the autophagosome it docks and fuses with a lysosome in the presence of LAMP‐2 and Rab7 proteins. After fusion of the autophagosome and lysosome into the autolysosome is complete, the content is degraded by lysosomal proteases (cathepsins B, D and L) and the lipids are exported to the cytosol for reuse.
Emerging evidence suggests a dual role of autophagy in cancer [42] but also in neurodegenerative diseases [43]. Autophagy can act as a survival mechanism by generating nutrients and energy during nutritional scarcity and increasing cell longevity and viability, and conversely contributing to cell damage when autophagy is dysregulated.
Autophagy in WMDs
Autophagy is a multistep process that has been suggested to dysfunction in neurodegenerative diseases [26, 27, 28]. In health, autophagy has been demonstrated to downregulate neuroinflammatory processes to minimise damage from pro‐inflammatory factors. However, the exact contribution of autophagy in acquired and genetic WMDs of the CNS has not been well characterised. Uncovering the role of autophagy in WMDs may help to understand the pathological processes, as well as allowing approaches to modulate autophagy pathways to treat these diseases.
Multiple sclerosis
MS is widely considered to be an autoimmune disorder of the CNS characterised by neurological damage, chronic inflammation and demyelination. Although the aetiology is unclear, the clinical episodes of disease are associated with the entry of pathogenic myelin‐reactive auto‐aggressive T‐ and B‐cells into the CNS. Although autophagy has been associated with neuroinflammation in MS, whether autophagy actively contributes to cell damage in MS (e.g. by prolonging the survival of autoreactive T‐cells) or acts as a rescue mechanism to counteract glial cell activation (e.g. dampening the neuroinflammatory response), is not yet known [44]. For example, peripheral T‐cells in people with active relapsing–remitting MS express increased ATG5, suggesting that autophagy may prolong the survival of autoreactive pathogenic T‐cells. However, although an association in gene variants for ATG5 was found in the optic tract in neuromyelitis optica, an autoimmune disease closely related to MS, this was not observed in MS [45]. Decreased levels of ATG16L2 in the serum of people with MS are related to the abnormal activation of T‐cells [46]. Furthermore, the pathogen receptor and inducer of autophagy CD46 was reported to impair regulatory functioning in T‐cells of people with MS upon pathogen recognition [34, 47]. To investigate the role of autophagy in MS, several studies have utilised experimental autoimmune encephalomyelitis (EAE), the autoimmune animal model of MS that is induced by immunising susceptible animals with myelin proteins or peptides administered in strong adjuvants. Such immunisation triggers myelin‐reactive pathogenic T‐cells that enter the CNS, inducing neuroinflammation and myelin damage [48]. Similar to MS, a role for autophagy has been proposed in EAE as circulating T‐cells have increased expression of Atg5 [49], whereas a reduction in LC3‐II/LC3‐I ratio [50] and reduced Beclin‐1 [51] resulting in reduced autophagy correlate with disease severity. Thus, the balance between reduced and increased markers of autophagy probably represent differential expression in activated immune cells versus damage to CNS cells. Autophagy is also implicated in CNS repair, reflected by the finding that rapamycin, an mTOR inhibitor, enhances remyelination in experimental peripheral neuropathy and tuberous sclerosis in mice [52, 53]. Additionally, increased levels of LC3‐II and p62, a protein that interacts with autophagosomes and ubiquitinated proteins that are ready for degradation, were observed in oligodendrocytes in myelin mutant Long‐Evans rats. After intermittent fasting, p62 levels in the rats were reduced, indicating an upregulation of autophagy that was associated with increased thickness in myelin sheaths [54], supporting the idea that autophagy pathways are able to aid in repair mechanisms in the CNS. Furthermore, inducing mTOR signalling with oestrogen receptor β ligand diarylpropionitrile in oligodendrocytes improved remyelination during EAE in mice [55]. These studies underscore a key role for the mTOR signalling pathway in oligodendrocyte survival and axon myelination. The chronic inflammation in MS is associated with increased levels of cytokines, including IL‐17 in T‐cells [56] that is reported to suppress autophagy by activating TAK‐binding proteins 2 and 3 and inducing MAPK [57]. Thus, increased IL‐17 exacerbated the effects of increased ATG5, and reduced LC3‐II in pathogenic T‐cells (Table 2).
Table 2.
Autophagy pathways in CNS WMDs.
| Type | Disease | Autophagy pathways associated with disease | Impact on autophagy | Reference |
|---|---|---|---|---|
| Inflammatory | MS | Increased ATG5 in T‐cells in relapsing–remitting MS | Supports (autoimmune) T‐cell survival | [49] |
| Reduced LC3‐II/LC3‐I ratio | ↓ | [50] | ||
| Increased IL‐17 | ↓ | [56, 57] | ||
| Decreased ATG16L2 | ↓ | [46] | ||
| NMO | ATG5 variants | [45] | ||
| ADEM | Unknown | |||
| AHL | Unknown | |||
| Infectious | PML | Reduced Bag3 | ↓ | [58] |
| MeV | Increased LC3‐II | ↑ | [47] | |
| Rubella virus | Reduced LC3‐II | ↓ | [59] | |
| Reduced Atg5 | ↓ | [59] | ||
| Reduced Atg12 | ↓ | [59] | ||
| Peroxisomal | X‐ALD | Increased p62 | ↓ | [60] |
| Reduced LC3‐II | ↓ | [61] | ||
| Increased mTOR signalling | ↓ | [62] | ||
| Miscellaneous | VWM | Decreased ATG3 and ATG7 in EIF2B3 oligodendrocytes and reduced LC3‐II | Depressed autophagy flux | [63, 64] |
NMO, neuromyelitis optica.
A possible mechanism of autophagy‐induced oligodendrocyte damage in the CNS in MS is shown in Figure 2. Increased levels of ATG5 may influence apoptosis as ATG5 is also involved in apoptotic pathways [65]. Atg5 can be cleaved by calpains and the resulting truncated version of ATG5 then interacts with Bcl‐xL and translocates from the cytosol to mitochondria where it may trigger apoptosis [66]. Another mechanism in which increased Atg5 can result in apoptosis is by interacting with Fas‐associated protein with death domain (FADD), which induces apoptosis through a cascade of interactions (for a review see [67]). Thus, the overexpression of ATG5 in MS, together with a decrease in ATG16L2 and LC3‐II/LC3‐I ratio, could trigger excessive activation of apoptotic pathways by increased binding to FADD or Bcl‐xL. Consequently, apoptotic signalling may induce cell death and white matter damage characteristic for MS.
Figure 2.

A mechanism of autophagy‐induced oligodendrocyte death in MS. ATG5 is involved in both autophagic and apoptotic pathways. (A) Atg5 regulates autophagy by interacting with ATG12 and ATG16 to initiate the nucleation process of autophagy and to form an autophagophore. (B) ATG5 is cleaved by calpains, leading to binding of truncated ATG5 with Bcl‐xL, which will dissociate from Bcl‐2 resulting in the initiation of apoptotic pathways. (C) ATG5 interacts with FADD, which results in the initiation of apoptotic pathways. Both apoptotic pathways can result in the death of oligodendrocytes in MS.
In summary, pathogenic survival mechanisms facilitated by increased autophagy are present in T‐cells contributing to CNS damage. On the other hand, there are pro‐survival mechanisms that need an increase in autophagy in the CNS that contribute to remyelination. Targeting remyelination through autophagy in the CNS will be beneficial for slowing down the disease progression of MS.
VWM disease
VWM disease is an autosomal recessive leukoencephalopathy leading to progressive ataxia, spasticity and seizures. Mutations in eukaryotic factor 2B (eIF2B) are present in VWM [20] and, although eIF2B is ubiquitously expressed, the pathology is almost exclusively in astrocytes and oligodendrocytes. eIF2B is essential for protein synthesis and the cellular response to stress, indicating that mutations in eIF2B may impair protein synthesis and the adaptability to cellular stress. Furthermore, ER stress [68] and upregulation of the UPR [63, 69], reported to trigger autophagy, may play a role in oligodendrocyte damage in VWM. In vitro, human oligodendrocytes expressing mutant eIF2B have reduced tolerance to ER stress [63, 70] and decreased levels of LC3‐II, indicating a decreased autophagy response [63]. In support of this, mutant eIF2B oligodendrocytes pre‐treated with autophagy inducers rapamycin and ridaforolimus showed normal levels of autophagy in vitro, resulting in higher tolerance to ER stress and cell survival [70].
Adrenoleukodystrophy
The pathology of X‐ALD arises due to the dysfunction of the peroxisomal ABCD1 transporter [62] that is critical for transporting very long chain fatty acids (VLCFAs). Such dysfunctions lead to an accumulation of VLCFAs as they cannot be transported into peroxisomes for degradation. VLCFA accumulation triggers oxidative stress and mitochondrial toxicity that may alter autophagy, although this has not been extensively investigated in human tissues. In vitro, VLCFAs activate autophagy pathways and induce lysosomal membrane destabilisation in rat oligodendrocytes [71], indicating that this may also be the cause of cell damage in X‐ALD. Indeed, impaired autophagy has been reported in human brain samples from X‐ALD cases, as well as in Abcd1 − and Abcd1−/Abcd2 −/− mouse models of X‐ALD [60]. During progressive neurological disease, people with X‐ALD, as well as Abcd1 − mice, have decreased LC3‐II and increased p62 levels in the CNS, indicative of impaired autophagy [60]. Thus, the pathology of X‐ALD might arise due to aberrant activation of the mTOR pathway as VLCFAs accumulate in the cell, or a gradual increase in mTOR levels that inhibit autophagic processes leading to axonal degeneration.
Systemic lupus erythematosus
Although systemic lupus erythematosus (SLE) is not a classical WMD, it is pertinent to discuss the CNS manifestations, as transverse myelitis is a rare myelopathy complication that may arise early in SLE and is associated with a poor recovery rate and high mortality [72, 73]. SLE has been associated with autophagy genes in numerous genome‐wide association studies, many of them implicating a role for ATG5 [74, 75, 76, 77, 78, 79]. Another autophagy‐related gene is leucine‐rich repeat kinase 2 (LRRK2). Several studies have shown the critical role of LRRK2 in regulating autophagy [80, 81]. In SLE, LRRK2 is upregulated in primary B‐cells and correlates with disease severity [82]. Furthermore, sera from people with SLE induced autophagy in neuroblastoma cells in vitro [83], suggesting a role in the neurological symptoms in people with SLE. Recently, it has also been suggested that mTOR signalling might be affected in SLE [84]. Activation of mTOR signalling leads to aberrant activation of lymphocytes and reduces regulatory T‐cell expansion, which is characteristic for SLE [85, 86]. Increased type 1 IFNs, for example IFN‐α, play a crucial role in SLE pathogenesis [87, 88]. Inhibition of the mTOR pathway has also been reported to supress stimulator of IFN genes (STING), an ATG5‐dependent autophagy pathway, effectively reducing IFN‐α expression in SLE monocytes [89, 90]. In summary, mutations in the ATG5 gene in SLE impact on autophagy and disease, and a genetic association between the autophagy‐related LRRK2 gene and SLE susceptibility indicates a strong association with a dysfunctional autophagy pathway in this disease.
Infectious WMDs
PML is a rare and generally fatal viral disease observed in people with severe immunodeficiency. PML arises due to human polyomavirus JC virus replication in the CNS leading to severe demyelination. In healthy individuals, the virus establishes a latent infection; however, viral reactivation and replication in glial cells leads to PML [91]. Overexpression of Bag3, a modulator of the cellular response to stress, induced autophagic degradation of viral proteins by activation of LC3‐II [92]. However, Bag3 is downregulated in cells infected with JC virus [58], possibly contributing to increased viral load.
Other pathogens have also developed strategies to utilise the autophagy machinery to their own advantage. Measles virus (MeV) is highly contagious and infectious and leads to the severe and often fatal disease subacute sclerosing panencephalitis, in which neurons and oligodendrocytes in the CNS are destroyed [93]. An important hallmark of the route of action of MeV is autophagic induction by the MeV receptor CD46 that contributes and sustains viral particle formation in infected cells [34, 47, 94]. After CD46‐dependent induction of autophagy, a second wave of autophagy induction may follow, resulting in a significant increase of viral transcripts [94]. This later wave of autophagy is reported to be dependent on interaction between the MeV C‐protein and the human immunity‐related GTPase family M [95]. Significantly increased LC3‐II was found in MeV‐infected host cells [47]. In contrast, rubella virus infection of oligodendrocytes utilises the autophagy machinery by reducing LC3‐II, ATG12 and ATG5 levels in vitro [59]. Rubella virus infection also inhibits autophagic flux and reduces the number and size of autophagosomes in the SIRC cell line, which may be beneficial for viral replication [59]. These viruses exemplify the intricate strategies that they have evolved to exploit autophagic pathways for self‐replication.
ADEM is a rare WMD that arises following a viral or bacterial infection and in rare cases by vaccination [96, 97]. ADEM causes an acute widespread inflammation of the CNS with severe demyelination and white matter destruction. Although symptoms resemble those of MS, ADEM follows a monophasic course, with full recovery rates from 50 to 70% [96]. Studies of the impact of autophagy in the pathogenesis of ADEM are lacking. However, as ADEM is closely related to other post‐infectious demyelinating diseases, the involvement of autophagy requires further exploration. Likewise, acute haemorrhagic leukoencephalitis (AHL), a fulminant and aggressive form of ADEM with a high mortality rate, is characterised by perivenular demyelination and inflammation. Case reports reveal that aggressive immunosuppressant therapy fails to halt the course of the disease [98]. The similarities with MS and infectious WMDs suggest that autophagy modulation in combination with immunosuppressive therapy might be more effective in controlling the disease and offer a more favourable outcome for ADEM and AHL.
Autophagy as a therapeutic target
Strong evidence indicates that autophagy plays an important role in the pathogenesis of WMDs and may thus be a target for therapeutic strategies. Emerging reports reveal that targeting the autophagy pathways in neurodegenerative diseases may reduce toxic accumulation of proteins (e.g. in Parkinson's disease, Alzheimer's disease, Huntington's disease) (for a recent review see [99]). Such approaches, as discussed below, may therefore also be beneficial in WMDs in which neuronal and axonal damage occur.
Autophagy inducers
A well‐known class of autophagy inducers are drugs that target mTOR signalling, indicating that these might prove useful to support oligodendrocytes and myelin repair in MS. Rapamycin functions as an immunosuppressive agent as well as increasing autophagy by inhibiting mTOR during nutrient‐rich conditions [100]. mTOR serves a central role in the regulation of various house‐keeping functions, including autophagy. Although rapamycin has been studied in MS, those studies focussed on its impact as an immunomodulatory agent and not on the effects on autophagy [101]. Rapamycin reduced EAE severity and decreases of LC3‐II, p62 and Beclin‐1 (Table 3) [102]. Similarly, rapamycin also showed significant anti‐inflammatory actions in EAE and MS, in which disease progression was slowed [148, 149, 150, 151]. Thus, rapamycin may control aberrant autophagy in addition to its immunomodulatory functions. The rapamycin ester temsirolimus has been shown to be effective in an X‐ALD mouse model, restoring autophagy and reducing the axonal degenerative processes [60], indicating that these drugs may also be useful in other neurodegenerative disease and WMDs. Other drugs that act through a similar working mechanism and that may have similar effects are PP242 and Torin 1 [104, 105]. Resveratrol acts by directly binding to the ATP binding pocket of mTOR, thereby competing with ATP and inducing autophagy [106].
Table 3.
Therapies targeting autophagy pathways in CNS WMDs.
| Compound | Mode of action | Effects in WMDs | Reference | |
|---|---|---|---|---|
| Autophagy inducers | ||||
| Targets of mTOR | Rapamycin | Inhibits mTOR |
Ameliorates EAE by blocking immune cell activation. May be effective in PND. Improves viability of M03.13/EIF2B3 cells |
[51, 102, 103] |
| Ridaforolimus | Inhibitor of mTOR complexes | Improves viability of M03.13/EIF2B3 cells | [70] | |
| Temsirolimus | Rapamycin analogue | Prevents protein accumulation, energetic failure and proteasome malfunctioning in Abcd1 − mice | [60] | |
| Pp242 | Inhibits the active site of mTOR | Blocks Akt phosphorylation | [104] | |
| Torin 1 | mTOR inhibitor | Unknown | [105] | |
| mTOR ATP competition | Resveratrol | Inhibits mTOR–ULK1 pathway | Unknown | [106] |
| AMPK activation | Pinosylvin | Induces conversion of LC3‐I to LC3‐II, and activates AMPK | Unknown | [107] |
| Chebulagic acid | Increases phosphorylated AMPK | Protects against cytotoxicity in SH‐SY5Y cells | [108] | |
| Metformin | Activates AMPK by increasing cytosolic AMP |
Reduces lesions in cuprizone model Rejuvenates OPC ageing Reduced lesion load in MS |
[109, 110, 111, 112] | |
| Akt–mTOR modulation | Gefitinib | Inhibits PI3K/Akt/mTOR pathways | Unknown | [113] |
| RY10‐4 | Inhibits phosphorylation of Akt and mTOR | Unknown | [114] | |
| Berberine | Inhibits upstream mTOR signalling and MAPK phosphorylation | Unknown | [115] | |
| Concanavalin A | Inhibits PI3K/Akt/mTOR pathways | Unknown | [116] | |
| Curcumin | Inhibits PI3K/Akt/mTOR pathways | Ameliorates EAE progression and delays onset | [51] | |
| Tunicamycin | Induces ER stress | Unknown | [117] | |
| Erlotinib | Inhibits PI3K/Akt/mTOR pathways | Unknown | [113] | |
| Piperlongumine | Inhibits PI3K/Akt/mTOR pathways | Unknown | [118] | |
| Baicalein | Decreases expression of Akt/ULK1 and 4EBP1 | Unknown | [119] | |
| Plumbagin | Inhibits PI3K/AKT/mTOR pathways | Unknown | [120] | |
| Emodin | Increases LC3‐II | Unknown | [121] | |
| Ivermectin | Promotes degradation of PAK1 | Unknown | [122] | |
| Corynoxine | Inhibits PI3K/AKT/mTOR pathways | Unknown | [64] | |
| NOS inhibitor | L‐NAME | Inhibits NOS activity | Unknown | [123] |
| Reducing cAMP | Rilmenidine | Binds to imidazoline‐1 receptor to reduce cAMP | Unknown | [124, 125] |
| Clonidine | Binds to imidazoline‐1 receptor to reduce cAMP | Unknown | [124] | |
| H89 | Inhibits cAMP‐dependent protein kinase | Unknown | [126] | |
| HU‐308 | Activates CB2R | Ameliorates EAE progression | [127] | |
| Targeting ULK1/2 or class III PI3K complexes | BH3 mimetics | Inhibits Beclin‐1 interaction with Bcl‐2 | Unknown | [128] |
| Glycyrrhizic acid | Induces Beclin‐1 production | Unknown | [129] | |
| MicroRNAs | miRNA‐18 | Inhibits mTOR signalling pathway | Unknown | [130] |
| miRNA‐21 | Upregulates Bcl‐2 expression | Unknown | [131] | |
| Enhanced ROS production | Cucurbitacin B | Increases expression of Beclin‐1, ULK1, reduces expression of mTOR | Unknown | [132] |
| Lowering inositol synthesis | Lithium | Inhibits inositol monophosphate | Reduces aggregates of GFAP in a mouse model of Alexander disease | [133, 134] |
| Carbamazepine | Inhibits inositol monophosphate | Unknown | [135, 136] | |
| Sodium valproate | Inhibits inositol monophosphate | Unknown | [135, 136] | |
| Autophagy inhibitors | ||||
| Akt–mTOR modulation | Pollen typhae | Increases LC‐3 and Beclin‐1 | Unknown | [137] |
| Gastrodin | Decreases LC3‐II, p62 and Beclin‐1 | Protects astrocytes in vitro from LPS‐induced cell death | [138] | |
| Bafilomycin A1 | Blocks lysosome fusion with autophagosomes | Unknown | [139] | |
| Spautin‐1 | Promotes degradation of Vps34 by inhibiting ubiquitin‐specific peptidases | Unknown | [140] | |
| Chloroquine | Blocks lysosome fusion with autophagosomes | Ameliorates EAE disease severity | [141] | |
| ERK kinase inhibitor | U0126 | Inhibits ERK | Unknown | [142] |
| PI3K inhibitor | 3‐methyladenine | Blocks class III PI3K | Increased severity of EAE and inflammation | [102] |
| Wortmannin | Blocks autophagosome formation | Unknown | [138] | |
| Matrine | Blocks autophagosome formation | Reduces stress in oligodendrocytes in EAE | [143] | |
| Other | Angiotensin (1–7) | Reduces oxidative stress | Unknown | [144] |
| MicroRNAs | miRNA‐30a | Suppresses expression of TP53INP1 | Unknown | [145] |
| miRNA‐205 | Suppresses expression of TP53INP1 | Unknown | [145] | |
| miRNA‐101 | Suppresses RAB5A | Unknown | [146] | |
| miRNA‐223 | Targets ATG16L1 | Unknown | [147] | |
GFAP, glial fibrillary acidic protein; LPS, lipopolysaccharide; OPC, oligodendrocyte precursor cell; PND, paraneoplastic neurological disorder.
Although lithium was the first drug found to induce autophagy in an mTOR‐independent manner by lowering inositol synthesis [133], other FDA‐approved drugs may also regulate autophagy in mTOR‐independent manners [124]. Two of these, carbamazepine and sodium valproate, act similarly to lithium, whereas xestospongin B decreased intracellular IP3 levels [152]. The compounds rilmenidine and clonidine reduce cAMP levels by inhibiting G‐protein signalling pathways that in turn inhibit adenylyl cyclase, after which autophagy is initiated [124]. Rilmenidine is known for its positive characteristics and safe long‐term use in chronic diseases and has been tested in neuronal cell cultures [125]. The cannabinoid receptor 2 (CB2R) inhibits adenylyl cyclase directly, which reduces cAMP levels similarly to the working mechanism of rilmenidine, clonidine and H89 [124, 126]. Activation of CB2R with HU‐308 alleviated clinical signs of EAE through activation of autophagy and restoring the reduced LC3‐II/LC3‐I ratio [127]. Cannabis, which targets CB2R, is used to treat MS and has been reported to reduce disease progression via autophagic‐related pathways [153]. Additionally, cAMP‐dependent PKA inhibitor H89 was found to induce autophagy through downstream modulation of Akt [126].
Another class of autophagy inducers targets the initiation process by increasing the activity of ULK1 or Beclin‐1. Corynoxine and BH3 mimetics were found to induce autophagy in a Beclin‐1‐dependent manner [64, 128]. Glycyrrhizic acid was found to not only induce Beclin‐1 production but also has antiviral properties that could prove useful in post‐infectious demyelinating diseases [129]. Similarly, cucurbitacin B increased Beclin‐1 and ULK1 levels, although its working mechanism is probably mediated by increased ROS production [132].
Recently, gefitinib and erlotinib [113], RY10‐4 [114], berberine [115], concanavalin A [116], curcumin [154], tunicamycin [117], piperlongumine [118], baicalein [119], plumbagin [120], emodin [121] and ivermectin [122] have all been found to induce autophagy through modulation of the Akt–mTOR pathway. Pinosylvin and chebulagic acid were found to induce autophagy through AMPK activation reducing necrotic progress and pathological symptoms of Parkinson's disease, respectively [107, 108]. Another AMPK activator is the anti‐diabetic drug metformin, which also induces autophagy [155]. Additionally, autophagy might be regulated by indirect targeting through microRNAs (miRNA‐18, miRNA‐21) [130, 131], nitric oxide synthase inhibitors such as L‐NAME that reduce cell stress [124] or disaccharides that may act as chaperones to induce autophagy [156].
Autophagy inhibitors
On the other end of the spectrum are autophagy modulators that reduce autophagy‐associated pathways and may be useful to salvage neuronal and oligodendrocyte death in WMDs. For example, Pollen typhae and bafilomycin A1 were shown to reduce autophagy by acting on the Akt/mTOR pathway [137, 139]. That this pathway is essential for myelination suggests that this approach may aid remyelination in WMDs [157]. U0126, an ERK kinase inhibitor, was found to reduce Beclin‐1 and LC3 levels, which indicates a suppressive effect on autophagy [142]. Similarly, gastrodin, normally used as an anti‐convulsant, prevents lipopolysaccharide‐induced autophagy by reducing LC3‐II, p62 and Beclin‐1 levels [138]. Spautin‐1 indirectly targets Beclin‐1 through degradation of ubiquitin‐specific peptidases USP10 and USP13 [158]. 3‐MA blocks class III PI3K to prevent autophagosome formation, which shows neuroprotective effects [159] similar to Wortmannin [138]. Angiotensin (1–7) targets autophagy by reducing oxidative stress in the cell [144]. Matrine was found to have a similar mechanism of action by reducing stress in oligodendrocytes and ameliorating EAE [143]. One drug commonly used in clinical oncology trials to block autophagy is chloroquine, an anti‐malaria drug. Recently, chloroquine was used to block autophagy in a model for myelin mutants [54]. Similarly, chloroquine was found to reduce EAE symptoms [160]. However, the precise working mechanism is not known. Several miRNAs (miRNA‐30a, miRNA‐205, miRNA‐101 and miRNA‐223) also inhibit autophagy [145, 146, 147]. Although research in this field is scarce, recent evidence in other diseases shows that these strategies might also be beneficial for WMDs given the role of autophagy in these diseases.
Conclusion
Emerging evidence supports the role of autophagy in WMDs. An increased understanding of the mechanisms driving autophagy has resulted in increased insight into the pathogenic processes that are affected in disease. A pressing problem is the heterogeneity between WMDs, which is also apparent in neurodegenerative disorders [161]. Indeed, in a disease such as MS there is evidence that some aspects of autophagy are upregulated whereas others are downregulated. This heterogeneity emphasises the need for thorough investigative research focusing on autophagy in WMDs to better understand the progression of autophagy malfunctioning during the disease and whether this is a causal aspect or a consequence of other facets of WMDs.
The development of pharmacological agents that modulate autophagy has been very successful. However, there are important obstacles to highlight. Most WMD cases are sporadic, but treatment early in the disease relies on the identification of the disease prior to symptomatic onset. Therefore, drugs that target autophagy will probably not rescue cells that have already undergone permanent damage. Furthermore, some autophagy modulators might have non‐specific targets that thus carry risks associated with unwanted side‐effects. Although there are obstacles to overcome, the recent surge in modulatory compounds for autophagy shows that there is no lack of treatment possibilities. If we can better understand the underlying pathological processes in autophagy for WMDs there is a great potential in finding new treatment strategies.
Author contributions statement
EN performed the literature search, generated figures, wrote and edited the review. EN, MCM, SA and SC critically reviewed and edited the final submitted version.
Abbreviations
ADEM, acute disseminated encephalomyelitis; AHL, acute haemorrhagic leukoencephalitis; AMPK, AMP kinase; CB2R, cannabinoid receptor 2; EAE, experimental autoimmune encephalomyelitis; eIF2B, eukaryotic factor 2B; FADD, Fas‐associated protein with death domain; LC3, light chain 3; LRRK2, leucine‐rich repeat kinase 2; MeV, measles virus; MS, multiple sclerosis; mTOR, mammalian target of rapamycin; PKA, protein kinase A; PML, progressive multifocal leukoencephalopathy; SLE, systemic lupus erythematosus; ULK, Unc‐51‐like kinase; UPR, unfolded protein response; VLCFA, very long chain fatty acid; Vps34, vacuolar protein sorting 34; VWM, vanishing white matter; WMDs, white matter disorders; X‐ALD, X‐linked adrenoleukodystrophy.
No conflicts of interest were declared.
References
- 1. Knaap MS, van der Valk J. Classification of myelin disorders In Magnetic Resonance of Myelination and Myelin Disorders (3rd edn). Springer: Berlin Heidelberg, 2005; 20–24. [Google Scholar]
- 2. Goldenberg MM. Multiple sclerosis review. P T 2012; 37 : 175–184. [PMC free article] [PubMed] [Google Scholar]
- 3. Kimbrough DJ, Fujihara K, Jacob A, et al Treatment of neuromyelitis optica: review and recommendations. Mult Scler Relat Disord 2012; 1 : 180–187. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 4. Pohl D, Alper G, Van Haren K, et al Acute disseminated encephalomyelitis: updates on an inflammatory CNS syndrome. Neurology 2016; 87 : S38–S45. [DOI] [PubMed] [Google Scholar]
- 5. Hofer M, Weber A, Haffner K, et al Acute hemorrhagic leukoencephalitis (Hurst's disease) linked to Epstein‐Barr virus infection. Acta Neuropathol 2005; 109 : 226–230. [DOI] [PubMed] [Google Scholar]
- 6. Major EO, Yousry TA, Clifford DB. Pathogenesis of progressive multifocal leukoencephalopathy and risks associated with treatments for multiple sclerosis: a decade of lessons learned. Lancet Neurol 2018; 17 : 467–480. [DOI] [PubMed] [Google Scholar]
- 7. Garg RK, Mahadevan A, Malhotra HS, et al Subacute sclerosing panencephalitis. Rev Med Virol 2019; 29 : e2058. [DOI] [PubMed] [Google Scholar]
- 8. Rawlinson WD, Boppana SB, Fowler KB, et al Congenital cytomegalovirus infection in pregnancy and the neonate: consensus recommendations for prevention, diagnosis, and therapy. Lancet Infect Dis 2017; 17 : e177–e188. [DOI] [PubMed] [Google Scholar]
- 9. Cai G, He D, Chu L, et al Paraneoplastic neuromyelitis optica spectrum disorders: three new cases and a review of the literature. Int J Neurosci 2016; 126 : 660–668. [DOI] [PubMed] [Google Scholar]
- 10. Rosenberg GA. Binswanger's disease: biomarkers in the inflammatory form of vascular cognitive impairment and dementia. J Neurochem 2018; 144 : 634–643. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 11. Negro S, Benders M, Tataranno ML, et al Early prediction of hypoxic‐ischemic brain injury by a new panel of biomarkers in a population of term newborns. Oxid Med Cell Longev 2018; 2018 : 7608108. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12. Armstrong RC, Mierzwa AJ, Sullivan GM, et al Myelin and oligodendrocyte lineage cells in white matter pathology and plasticity after traumatic brain injury. Neuropharmacology 2016; 110 : 654–659. [DOI] [PubMed] [Google Scholar]
- 13. Penati R, Fumagalli F, Calbi V, et al Gene therapy for lysosomal storage disorders: recent advances for metachromatic leukodystrophy and mucopolysaccaridosis I. J Inherit Metab Dis 2017; 40 : 543–554. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 14. Engelen M, Kemp S, de Visser M, et al X‐linked adrenoleukodystrophy (X‐ALD): clinical presentation and guidelines for diagnosis, follow‐up and management. Orphanet J Rare Dis 2012; 7 : 51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 15. Wan X, Pei H, Zhao M‐j, et al Efficacy and safety of rAAV2‐ND4 treatment for Leber's hereditary optic neuropathy. Sci Rep 2016; 6 : 1–10. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 16. Karikkineth AC, Scheibye‐Knudsen M, Fivenson E, et al Cockayne syndrome: clinical features, model systems and pathways. Ageing Res Rev 2017; 33 : 3–17. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17. Laukka JJ, Kamholz J, Bessert D, et al Novel pathologic findings in patients with Pelizaeus–Merzbacher disease. Neurosci Lett 2016; 627 : 222–232. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18. Matalon D, Matalon KM, Matalon R. Canavan disease In Rosenberg's Molecular and Genetic Basis of Neurological and Psychiatric Disease, Rosenberg RN, Pascual JM. (eds). Elsevier: London, 2020; 909–916. [Google Scholar]
- 19. Messing A. Alexander disease In Handbook of Clinical Neurology, Geschwind DH, Paulson HL, Klein C. (eds). Elsevier: London, 2018; 693–700. [DOI] [PubMed] [Google Scholar]
- 20. van der Knaap MS, Pronk JC, Scheper GC. Vanishing white matter disease. Lancet Neurol 2006; 5 : 413–423. [DOI] [PubMed] [Google Scholar]
- 21. Gonsette RE. Neurodegeneration in multiple sclerosis: the role of oxidative stress and excitotoxicity. J Neurol Sci 2008; 274 : 48–53. [DOI] [PubMed] [Google Scholar]
- 22. Kantor L, Harding HP, Ron D, et al Heightened stress response in primary fibroblasts expressing mutant eIF2B genes from CACH/VWM leukodystrophy patients. Hum Genet 2005; 118 : 99–106. [DOI] [PubMed] [Google Scholar]
- 23. Lin W, Popko B. Endoplasmic reticulum stress in disorders of myelinating cells. Nat Neurosci 2009; 12 : 379–385. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 24. Patel J, Balabanov R. Molecular mechanisms of oligodendrocyte injury in multiple sclerosis and experimental autoimmune encephalomyelitis. Int J Mol Sci 2012; 13 : 10647–10659. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 25. Parzych KR, Klionsky DJ. An overview of autophagy: morphology, mechanism, and regulation. Antioxid Redox Signal 2014; 20 : 460–473. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26. Lynch‐Day MA, Mao K, Wang K, et al The role of autophagy in Parkinson's disease. Cold Spring Harb Perspect Med 2012; 2 : a009357. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 27. Nixon RA, Wegiel J, Kumar A, et al Extensive involvement of autophagy in Alzheimer disease: an immuno‐electron microscopy study. J Neuropathol Exp Neurol 2005; 64 : 113–122. [DOI] [PubMed] [Google Scholar]
- 28. Song CY, Guo JF, Liu Y, et al Autophagy and its comprehensive impact on ALS. Int J Neurosci 2012; 122 : 695–703. [DOI] [PubMed] [Google Scholar]
- 29. Klionsky DJ, Cregg JM, Dunn WA Jr, et al A unified nomenclature for yeast autophagy‐related genes. Dev Cell 2003; 5 : 539–545. [DOI] [PubMed] [Google Scholar]
- 30. Orvedahl A, Alexander D, Talloczy Z, et al HSV‐1 ICP34.5 confers neurovirulence by targeting the Beclin 1 autophagy protein. Cell Host Microbe 2007; 1 : 23–35. [DOI] [PubMed] [Google Scholar]
- 31. Lum JJ, Bauer DE, Kong M, et al Growth factor regulation of autophagy and cell survival in the absence of apoptosis. Cell 2005; 120 : 237–248. [DOI] [PubMed] [Google Scholar]
- 32. Ogata M, Hino S, Saito A, et al Autophagy is activated for cell survival after endoplasmic reticulum stress. Mol Cell Biol 2006; 26 : 9220–9231. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 33. Yorimitsu T, Nair U, Yang Z, et al Endoplasmic reticulum stress triggers autophagy. J Biol Chem 2006; 281 : 30299–30304. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 34. Meiffren G, Joubert PE, Gregoire IP, et al Pathogen recognition by the cell surface receptor CD46 induces autophagy. Autophagy 2010; 6 : 299–300. [DOI] [PubMed] [Google Scholar]
- 35. Feng Y, He D, Yao Z, et al The machinery of macroautophagy. Cell Res 2014; 24 : 24–41. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 36. Mercer CA, Kaliappan A, Dennis PB. A novel, human Atg13 binding protein, Atg101, interacts with ULK1 and is essential for macroautophagy. Autophagy 2009; 5 : 649–662. [DOI] [PubMed] [Google Scholar]
- 37. Kim J, Kundu M, Viollet B, et al AMPK and mTOR regulate autophagy through direct phosphorylation of Ulk1. Nat Cell Biol 2011; 13 : 132–141. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 38. He C, Klionsky DJ. Regulation mechanisms and signaling pathways of autophagy. Annu Rev Genet 2009; 43 : 67–93. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 39. Rubinsztein DC, Shpilka T, Elazar Z. Mechanisms of autophagosome biogenesis. Curr Biol 2012; 22 : R29–R34. [DOI] [PubMed] [Google Scholar]
- 40. Kabeya Y, Mizushima N, Ueno T, et al LC3, a mammalian homologue of yeast Apg8p, is localized in autophagosome membranes after processing. EMBO J 2000; 19 : 5720–5728. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 41. Schneider JL, Cuervo AM. Autophagy and human disease: emerging themes. Curr Opin Genet Dev 2014; 26 : 16–23. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 42. Singh SS, Vats S, Chia AY, et al Dual role of autophagy in hallmarks of cancer. Oncogene 2018; 37 : 1142–1158. [DOI] [PubMed] [Google Scholar]
- 43. Bar‐Yosef T, Damri O, Agam G. Dual role of autophagy in diseases of the central nervous system. Front Cell Neurosci 2019; 13 : 196. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 44. Liang P, Le W. Role of autophagy in the pathogenesis of multiple sclerosis. Neurosci Bull 2015; 31 : 435–444. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 45. Cai PP, Wang HX, Zhuang JC, et al Variants of autophagy‐related gene 5 are associated with neuromyelitis optica in the Southern Han Chinese population. Autoimmunity 2014; 47 : 563–566. [DOI] [PubMed] [Google Scholar]
- 46. Yin L, Liu J, Dong H, et al Autophagy‐related gene16L2, a potential serum biomarker of multiple sclerosis evaluated by bead‐based proteomic technology. Neurosci Lett 2014; 562 : 34–38. [DOI] [PubMed] [Google Scholar]
- 47. Joubert PE, Meiffren G, Gregoire IP, et al Autophagy induction by the pathogen receptor CD46. Cell Host Microbe 2009; 6 : 354–366. [DOI] [PubMed] [Google Scholar]
- 48. Constantinescu CS, Farooqi N, O'Brien K, et al Experimental autoimmune encephalomyelitis (EAE) as a model for multiple sclerosis (MS). Br J Pharmacol 2011; 164 : 1079–1106. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 49. Alirezaei M, Fox HS, Flynn CT, et al Elevated ATG5 expression in autoimmune demyelination and multiple sclerosis. Autophagy 2009; 5 : 152–158. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 50. Dasgupta A, Zheng J, Perrone‐Bizzozero NI, et al Increased carbonylation, protein aggregation and apoptosis in the spinal cord of mice with experimental autoimmune encephalomyelitis. ASN Neuro 2013; 5 : e00111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 51. Boyao Y, Mengjiao S, Caicai B, et al Dynamic expression of autophagy‐related factors in autoimmune encephalomyelitis and exploration of curcumin therapy. J Neuroimmunol 2019; 337 : 577067. [DOI] [PubMed] [Google Scholar]
- 52. Meikle L, Pollizzi K, Egnor A, et al Response of a neuronal model of tuberous sclerosis to mammalian target of rapamycin (mTOR) inhibitors: effects on mTORC1 and Akt signaling lead to improved survival and function. J Neurosci 2008; 28 : 5422–5432. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 53. Rangaraju S, Verrier JD, Madorsky I, et al Rapamycin activates autophagy and improves myelination in explant cultures from neuropathic mice. J Neurosci 2010; 30 : 11388–11397. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 54. Smith CM, Mayer JA, Duncan ID. Autophagy promotes oligodendrocyte survival and function following dysmyelination in a long‐lived myelin mutant. J Neurosci 2013; 33 : 8088–8100. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 55. Kumar S, Patel R, Moore S, et al Estrogen receptor beta ligand therapy activates PI3K/Akt/mTOR signaling in oligodendrocytes and promotes remyelination in a mouse model of multiple sclerosis. Neurobiol Dis 2013; 56 : 131–144. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 56. Lock C, Hermans G, Pedotti R, et al Gene‐microarray analysis of multiple sclerosis lesions yields new targets validated in autoimmune encephalomyelitis. Nat Med 2002; 8 : 500–508. [DOI] [PubMed] [Google Scholar]
- 57. Zhou Y, Wu PW, Yuan XW, et al Interleukin‐17A inhibits cell autophagy under starvation and promotes cell migration via TAB2/TAB3‐p38 mitogen‐activated protein kinase pathways in hepatocellular carcinoma. Eur Rev Med Pharmacol Sci 2016; 20 : 250–263. [PubMed] [Google Scholar]
- 58. Basile A, Darbinian N, Kaminski R, et al Evidence for modulation of BAG3 by polyomavirus JC early protein. J Gen Virol 2009; 90 : 1629–1640. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 59. Pásztor K, Orosz L, Seprényi G, et al Rubella virus perturbs autophagy. Med Microbiol Immunol 2014; 203 : 323–331. [DOI] [PubMed] [Google Scholar]
- 60. Launay N, Aguado C, Fourcade S, et al Autophagy induction halts axonal degeneration in a mouse model of X‐adrenoleukodystrophy. Acta Neuropathol 2015; 129 : 399–415. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61. Kroemer G. Autophagy: a druggable process that is deregulated in aging and human disease. J Clin Invest 2015; 125 : 1–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 62. Fourcade S, Ferrer I, Pujol A. Oxidative stress, mitochondrial and proteostasis malfunction in adrenoleukodystrophy: a paradigm for axonal degeneration. Free Radic Biol Med 2015; 88 : 18–29. [DOI] [PubMed] [Google Scholar]
- 63. Chen N, Jiang YW, Hao HJ, et al Different eukaryotic initiation factor 2Bepsilon mutations lead to various degrees of intolerance to the stress of endoplasmic reticulum in oligodendrocytes. Chin Med J (Engl) 2015; 128 : 1772–1777. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 64. Chen LL, Song JX, Lu JH, et al Corynoxine, a natural autophagy enhancer, promotes the clearance of alpha‐synuclein via Akt/mTOR pathway. J Neuroimmune Pharmacol 2014; 9 : 380–387. [DOI] [PubMed] [Google Scholar]
- 65. Pyo JO, Jang MH, Kwon YK, et al Essential roles of Atg5 and FADD in autophagic cell death: dissection of autophagic cell death into vacuole formation and cell death. J Biol Chem 2005; 280 : 20722–20729. [DOI] [PubMed] [Google Scholar]
- 66. Yousefi S, Perozzo R, Schmid I, et al Calpain‐mediated cleavage of Atg5 switches autophagy to apoptosis. Nat Cell Biol 2006; 8 : 1124–1132. [DOI] [PubMed] [Google Scholar]
- 67. Gump JM, Thorburn A. Autophagy and apoptosis: what is the connection? Trends Cell Biol 2011; 21 : 387–392. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 68. van der Voorn JP, van Kollenburg B, Bertrand G, et al The unfolded protein response in vanishing white matter disease. J Neuropathol Exp Neurol 2005; 64 : 770–775. [DOI] [PubMed] [Google Scholar]
- 69. van Kollenburg B, van Dijk J, Garbern J, et al Glia‐specific activation of all pathways of the unfolded protein response in vanishing white matter disease. J Neuropathol Exp Neurol 2006; 65 : 707–715. [DOI] [PubMed] [Google Scholar]
- 70. Chen N, Dai L, Jiang Y, et al Endoplasmic reticulum stress intolerance in EIF2B3 mutant oligodendrocytes is modulated by depressed autophagy. Brain Dev 2016; 38 : 507–515. [DOI] [PubMed] [Google Scholar]
- 71. Baarine M, Ragot K, Athias A, et al Incidence of Abcd1 level on the induction of cell death and organelle dysfunctions triggered by very long chain fatty acids and TNF‐alpha on oligodendrocytes and astrocytes. Neurotoxicology 2012; 33 : 212–228. [DOI] [PubMed] [Google Scholar]
- 72. D'Cruz DP, Mellor‐Pita S, Joven B, et al Transverse myelitis as the first manifestation of systemic lupus erythematosus or lupus‐like disease: good functional outcome and relevance of antiphospholipid antibodies. J Rheumatol 2004; 31 : 280–285. [PubMed] [Google Scholar]
- 73. Provenzale J, Bouldin TW. Lupus‐related myelopathy: report of three cases and review of the literature. J Neurol Neurosurg Psychiatry 1992; 55 : 830–835. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 74. Gateva V, Sandling JK, Hom G, et al A large‐scale replication study identifies TNIP1, PRDM1, JAZF1, UHRF1BP1 and IL10 as risk loci for systemic lupus erythematosus. Nat Genet 2009; 41 : 1228–1233. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75. Han JW, Zheng HF, Cui Y, et al Genome‐wide association study in a Chinese Han population identifies nine new susceptibility loci for systemic lupus erythematosus. Nat Genet 2009; 41 : 1234–1237. [DOI] [PubMed] [Google Scholar]
- 76. Zhang YM, Cheng FJ, Zhou XJ, et al Rare variants of ATG5 are likely to be associated with Chinese patients with systemic lupus erythematosus. Medicine (Baltimore) 2015; 94 : e939. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 77. Zhou XJ, Lu XL, Lv JC, et al Genetic association of PRDM1‐ATG5 intergenic region and autophagy with systemic lupus erythematosus in a Chinese population. Ann Rheum Dis 2011; 70 : 1330–1337. [DOI] [PubMed] [Google Scholar]
- 78. International Consortium for Systemic Lupus Erythematosus Genetics , Harley JB, Alarcón‐Riquelme ME, et al Genome‐wide association scan in women with systemic lupus erythematosus identifies susceptibility variants in ITGAM, PXK, KIAA1542 and other loci. Nat Genet 2008; 40 : 204–210. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 79. Yang W, Tang H, Zhang Y, et al Meta‐analysis followed by replication identifies loci in or near CDKN1B, TET3, CD80, DRAM1, and ARID5B as associated with systemic lupus erythematosus in Asians. Am J Hum Genet 2013; 92 : 41–51. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 80. Gan‐Or Z, Dion PA, Rouleau GA. Genetic perspective on the role of the autophagy‐lysosome pathway in Parkinson disease. Autophagy 2015; 11 : 1443–1457. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 81. Manzoni C. LRRK2 and autophagy: a common pathway for disease. Biochem Soc Trans 2012; 40 : 1147–1151. [DOI] [PubMed] [Google Scholar]
- 82. Zhang M, Yao C, Cai J, et al LRRK2 is involved in the pathogenesis of system lupus erythematosus through promoting pathogenic antibody production. J Transl Med 2019; 17 : 37. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 83. Towns R, Kabeya Y, Yoshimori T, et al Sera from patients with type 2 diabetes and neuropathy induce autophagy and colocalization with mitochondria in SY5Y cells. Autophagy 2005; 1 : 163–170. [DOI] [PubMed] [Google Scholar]
- 84. Pierdominici M, Vomero M, Barbati C, et al Role of autophagy in immunity and autoimmunity, with a special focus on systemic lupus erythematosus. FASEB J 2012; 26 : 1400–1412. [DOI] [PubMed] [Google Scholar]
- 85. Fernandez D, Perl A. mTOR signaling: a central pathway to pathogenesis in systemic lupus erythematosus? Discov Med 2010; 9 : 173–178. [PMC free article] [PubMed] [Google Scholar]
- 86. Pierdominici M, Vacirca D, Delunardo F, et al mTOR signaling and metabolic regulation of T cells: new potential therapeutic targets in autoimmune diseases. Curr Pharm Des 2011; 17 : 3888–3897. [DOI] [PubMed] [Google Scholar]
- 87. Crow MK. Type I interferon in the pathogenesis of lupus. J Immunol 2014; 192 : 5459–5468. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 88. Feng X, Wu H, Grossman JM, et al Association of increased interferon‐inducible gene expression with disease activity and lupus nephritis in patients with systemic lupus erythematosus. Arthritis Rheum 2006; 54 : 2951–2962. [DOI] [PubMed] [Google Scholar]
- 89. Liu D, Wu H, Wang C, et al STING directly activates autophagy to tune the innate immune response. Cell Death Differ 2019; 26 : 1735–1749. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90. Murayama G, Chiba A, Kuga T, et al Inhibition of mTOR suppresses IFNalpha production and the STING pathway in monocytes from systemic lupus erythematosus patients. Rheumatology (Oxford) 2020; 59 : 2992–3002. [DOI] [PubMed] [Google Scholar]
- 91. Beltrami S, Gordon J. Immune surveillance and response to JC virus infection and PML. J Neurovirol 2014; 20 : 137–149. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 92. Sariyer IK, Merabova N, Patel PK, et al Bag3‐induced autophagy is associated with degradation of JCV oncoprotein, T‐Ag. PLoS One 2012; 7 : e45000. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 93. Allen IV, McQuaid S, McMahon J, et al The significance of measles virus antigen and genome distribution in the CNS in SSPE for mechanisms of viral spread and demyelination. J Neuropathol Exp Neurol 1996; 55 : 471–480. [DOI] [PubMed] [Google Scholar]
- 94. Richetta C, Grégoire IP, Verlhac P, et al Sustained autophagy contributes to measles virus infectivity. PLoS Pathog 2013; 9 : e1003599. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95. Grégoire IP, Richetta C, Meyniel‐Schicklin L, et al IRGM is a common target of RNA viruses that subvert the autophagy network. PLoS Pathog 2011; 7 : e1002422. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 96. Menge T, Kieseier BC, Nessler S, et al Acute disseminated encephalomyelitis: an acute hit against the brain. Curr Opin Neurol 2007; 20 : 247–254. [DOI] [PubMed] [Google Scholar]
- 97. Tenembaum S, Chamoles N, Fejerman N. Acute disseminated encephalomyelitis: a long‐term follow‐up study of 84 pediatric patients. Neurology 2002; 59 : 1224–1231. [DOI] [PubMed] [Google Scholar]
- 98. Sureshbabu S, Babu R, Garg A, et al Acute hemorrhagic leukoencephalitis unresponsive to aggressive immunosuppression. Clin Exp Neuroimmunol 2017; 8 : 63–66. [Google Scholar]
- 99. Menzies FM, Fleming A, Rubinsztein DC. Compromised autophagy and neurodegenerative diseases. Nat Rev Neurosci 2015; 16 : 345–357. [DOI] [PubMed] [Google Scholar]
- 100. Mizushima N. The role of the Atg1/ULK1 complex in autophagy regulation. Curr Opin Cell Biol 2010; 22 : 132–139. [DOI] [PubMed] [Google Scholar]
- 101. Dello Russo C, Lisi L, Feinstein DL, et al mTOR kinase, a key player in the regulation of glial functions: relevance for the therapy of multiple sclerosis. Glia 2013; 61 : 301–311. [DOI] [PubMed] [Google Scholar]
- 102. Feng X, Hou H, Zou Y, et al Defective autophagy is associated with neuronal injury in a mouse model of multiple sclerosis. Bosn J Basic Med Sci 2017; 17 : 95–103. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103. Noack M, Richter‐Landsberg C. Activation of autophagy by rapamycin does not protect oligodendrocytes against protein aggregate formation and cell death induced by proteasomal inhibition. J Mol Neurosci 2015; 55 : 99–108. [DOI] [PubMed] [Google Scholar]
- 104. Feldman ME, Apsel B, Uotila A, et al Active‐site inhibitors of mTOR target rapamycin‐resistant outputs of mTORC1 and mTORC2. PLoS Biol 2009; 7 : e1000038. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 105. Thoreen CC, Kang SA, Chang JW, et al An ATP‐competitive mammalian target of rapamycin inhibitor reveals rapamycin‐resistant functions of mTORC1. J Biol Chem 2009; 284 : 8023–8032. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106. Park D, Jeong H, Lee MN, et al Resveratrol induces autophagy by directly inhibiting mTOR through ATP competition. Sci Rep 2016; 6 : 21772. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 107. Park J, Pyee J, Park H. Pinosylvin at a high concentration induces AMPK‐mediated autophagy for preventing necrosis in bovine aortic endothelial cells. Can J Physiol Pharmacol 2014; 92 : 993–999. [DOI] [PubMed] [Google Scholar]
- 108. Kim HJ, Kim J, Kang KS, et al Neuroprotective effect of chebulagic acid via autophagy induction in SH‐SY5Y cells. Biomol Ther (Seoul) 2014; 22 : 275–281. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109. Houshmand F, Barati M, Golab F, et al Metformin‐induced AMPK activation stimulates remyelination through induction of neurotrophic factors, downregulation of NogoA and recruitment of Olig2+ precursor cells in the cuprizone murine model of multiple sclerosis. Daru 2019; 27 : 583–592. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 110. Largani SHH, Borhani‐Haghighi M, Pasbakhsh P, et al Oligoprotective effect of metformin through the AMPK‐dependent on restoration of mitochondrial hemostasis in the cuprizone‐induced multiple sclerosis model. J Mol Histol 2019; 50 : 263–271. [DOI] [PubMed] [Google Scholar]
- 111. Neumann B, Baror R, Zhao C, et al Metformin restores CNS remyelination capacity by rejuvenating aged stem cells. Cell Stem Cell 2019; 25 : 473–485.e8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 112. Sanadgol N, Barati M, Houshmand F, et al Metformin accelerates myelin recovery and ameliorates behavioral deficits in the animal model of multiple sclerosis via adjustment of AMPK/Nrf2/mTOR signaling and maintenance of endogenous oligodendrogenesis during brain self‐repairing period. Pharmacol Rep 2020; 72 : 641–658. [DOI] [PubMed] [Google Scholar]
- 113. Han W, Pan H, Chen Y, et al EGFR tyrosine kinase inhibitors activate autophagy as a cytoprotective response in human lung cancer cells. PLoS One 2011; 6 : e18691. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 114. Zhang X, Wei H, Liu Z, et al A novel protoapigenone analog RY10‐4 induces breast cancer MCF‐7 cell death through autophagy via the Akt/mTOR pathway. Toxicol Appl Pharmacol 2013; 270 : 122–128. [DOI] [PubMed] [Google Scholar]
- 115. Li MH, Zhang YJ, Yu YH, et al Berberine improves pressure overload‐induced cardiac hypertrophy and dysfunction through enhanced autophagy. Eur J Pharmacol 2014; 728 : 67–76. [DOI] [PubMed] [Google Scholar]
- 116. Roy B, Pattanaik AK, Das J, et al Role of PI3K/Akt/mTOR and MEK/ERK pathway in Concanavalin A induced autophagy in HeLa cells. Chem Biol Interact 2014; 210 : 96–102. [DOI] [PubMed] [Google Scholar]
- 117. Ma M, Song L, Yan H, et al Low dose tunicamycin enhances atherosclerotic plaque stability by inducing autophagy. Biochem Pharmacol 2016; 100 : 51–60. [DOI] [PubMed] [Google Scholar]
- 118. Wang F, Mao Y, You Q, et al Piperlongumine induces apoptosis and autophagy in human lung cancer cells through inhibition of PI3K/Akt/mTOR pathway. Int J Immunopathol Pharmacol 2015; 28 : 362–373. [DOI] [PubMed] [Google Scholar]
- 119. Wang YF, Li T, Tang ZH, et al Baicalein triggers autophagy and inhibits the protein kinase B/mammalian target of rapamycin pathway in hepatocellular carcinoma HepG2 cells. Phytother Res 2015; 29 : 674–679. [DOI] [PubMed] [Google Scholar]
- 120. Zhou ZW, Li XX, He ZX, et al Induction of apoptosis and autophagy via sirtuin1‐ and PI3K/Akt/mTOR‐mediated pathways by plumbagin in human prostate cancer cells. Drug Des Devel Ther 2015; 9 : 1511–1554. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
- 121. Liu H, Gu LB, Tu Y, et al Emodin ameliorates cisplatin‐induced apoptosis of rat renal tubular cells in vitro by activating autophagy. Acta Pharmacol Sin 2016; 37 : 235–245. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 122. Wang K, Gao W, Dou Q, et al Ivermectin induces PAK1‐mediated cytostatic autophagy in breast cancer. Autophagy 2016; 12 : 2498–2499. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 123. Shariatpanahi M, Khodagholi F, Ashabi G, et al Ameliorating of memory impairment and apoptosis in amyloid β‐injected rats via inhibition of nitric oxide synthase: possible participation of autophagy. Iran J Pharm Res 2015; 14 : 811–824. [PMC free article] [PubMed] [Google Scholar]
- 124. Williams A, Sarkar S, Cuddon P, et al Novel targets for Huntington's disease in an mTOR‐independent autophagy pathway. Nat Chem Biol 2008; 4 : 295–305. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 125. Rose C, Menzies FM, Renna M, et al Rilmenidine attenuates toxicity of polyglutamine expansions in a mouse model of Huntington's disease. Hum Mol Genet 2010; 19 : 2144–2153. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 126. Inoue H, Hase K, Segawa A, et al H89 (N‐[2‐p‐bromocinnamylamino‐ethyl]‐5‐isoquinolinesulphonamide) induces autophagy independently of protein kinase A inhibition. Eur J Pharmacol 2013; 714 : 170–177. [DOI] [PubMed] [Google Scholar]
- 127. Shao BZ, Wei W, Ke P, et al Activating cannabinoid receptor 2 alleviates pathogenesis of experimental autoimmune encephalomyelitis via activation of autophagy and inhibiting NLRP3 inflammasome. CNS Neurosci Ther 2014; 20 : 1021–1028. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 128. Malik SA, Orhon I, Morselli E, et al BH3 mimetics activate multiple pro‐autophagic pathways. Oncogene 2011; 30 : 3918–3929. [DOI] [PubMed] [Google Scholar]
- 129. Laconi S, Madeddu MA, Pompei R. Autophagy activation and antiviral activity by a licorice triterpene. Phytother Res 2014; 28 : 1890–1892. [DOI] [PubMed] [Google Scholar]
- 130. Fan YX, Dai YZ, Wang XL, et al MiR‐18a upregulation enhances autophagy in triple negative cancer cells via inhibiting mTOR signaling pathway. Eur Rev Med Pharmacol Sci 2016; 20 : 2194–2200. [PubMed] [Google Scholar]
- 131. Seca H, Lima RT, Lopes‐Rodrigues V, et al Targeting miR‐21 induces autophagy and chemosensitivity of leukemia cells. Curr Drug Targets 2013; 14 : 1135–1143. [DOI] [PubMed] [Google Scholar]
- 132. Wu CH, Bao JL, He CW, et al Cucurbitacin B inhibits proliferation, induces G2/M cycle arrest and autophagy without affecting apoptosis but enhances MTT reduction in PC12 cells. Bangladesh J Pharmacol 2016; 11 : 110–118. [Google Scholar]
- 133. Sarkar S, Floto RA, Berger Z, et al Lithium induces autophagy by inhibiting inositol monophosphatase. J Cell Biol 2005; 170 : 1101–1111. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 134. LaPash Daniels CM, Paffenroth E, Austin EV, et al Lithium decreases glial fibrillary acidic protein in a mouse model of Alexander disease. PLoS One 2015; 10 : e0138132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 135. Sarkar S, Ravikumar B, Floto RA, et al Rapamycin and mTOR‐independent autophagy inducers ameliorate toxicity of polyglutamine‐expanded huntingtin and related proteinopathies. Cell Death Differ 2009; 16 : 46–56. [DOI] [PubMed] [Google Scholar]
- 136. Zhu XC, Yu JT, Jiang T, et al Autophagy modulation for Alzheimer's disease therapy. Mol Neurobiol 2013; 48 : 702–714. [DOI] [PubMed] [Google Scholar]
- 137. Wang W, Guo Z, Xu Z, et al Effect of pollen typhae on inhibiting autophagy in spinal cord injury of rats and its mechanisms. Int J Clin Exp Pathol 2015; 8 : 2375–2383. [PMC free article] [PubMed] [Google Scholar]
- 138. Wang XS, Tian Z, Zhang N, et al Protective effects of gastrodin against autophagy‐mediated astrocyte death. Phytother Res 2016; 30 : 386–396. [DOI] [PubMed] [Google Scholar]
- 139. Degtyarev M, De Maziere A, Orr C, et al Akt inhibition promotes autophagy and sensitizes PTEN‐null tumors to lysosomotropic agents. J Cell Biol 2008; 183 : 101–116. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 140. López‐Erauskin J, Fourcade S, Galino J, et al Antioxidants halt axonal degeneration in a mouse model of X‐adrenoleukodystrophy. Ann Neurol 2011; 70 : 84–92. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 141. Abdel‐Aziz AK, Shouman S, El‐Demerdash E, et al Chloroquine synergizes sunitinib cytotoxicity via modulating autophagic, apoptotic and angiogenic machineries. Chem Biol Interact 2014; 217 : 28–40. [DOI] [PubMed] [Google Scholar]
- 142. Wang A, Zhang H, Liang Z, et al U0126 attenuates ischemia/reperfusion‐induced apoptosis and autophagy in myocardium through MEK/ERK/EGR‐1 pathway. Eur J Pharmacol 2016; 788 : 280–285. [DOI] [PubMed] [Google Scholar]
- 143. Wang MR, Zhang XJ, Liu HC, et al Matrine protects oligodendrocytes by inhibiting their apoptosis and enhancing mitochondrial autophagy. Brain Res Bull 2019; 153 : 30–38. [DOI] [PubMed] [Google Scholar]
- 144. Jiang T, Gao L, Zhu XC, et al Angiotensin‐(1‐7) inhibits autophagy in the brain of spontaneously hypertensive rats. Pharmacol Res 2013; 71 : 61–68. [DOI] [PubMed] [Google Scholar]
- 145. Xu CG, Yang MF, Fan JX, et al MiR‐30a and miR‐205 are downregulated in hypoxia and modulate radiosensitivity of prostate cancer cells by inhibiting autophagy via TP53INP1. Eur Rev Med Pharmacol Sci 2016; 20 : 1501–1508. [PubMed] [Google Scholar]
- 146. Jing Z, Han W, Sui X, et al Interaction of autophagy with microRNAs and their potential therapeutic implications in human cancers. Cancer Lett 2015; 356 : 332–338. [DOI] [PubMed] [Google Scholar]
- 147. Li Y, Zhou D, Ren Y, et al Mir223 restrains autophagy and promotes CNS inflammation by targeting ATG16L1. Autophagy 2019; 15 : 478–492. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 148. Donia M, Mangano K, Amoroso A, et al Treatment with rapamycin ameliorates clinical and histological signs of protracted relapsing experimental allergic encephalomyelitis in Dark Agouti rats and induces expansion of peripheral CD4+CD25+Foxp3+ regulatory T cells. J Autoimmun 2009; 33 : 135–140. [DOI] [PubMed] [Google Scholar]
- 149. Esposito M, Ruffini F, Bellone M, et al Rapamycin inhibits relapsing experimental autoimmune encephalomyelitis by both effector and regulatory T cells modulation. J Neuroimmunol 2010; 220 : 52–63. [DOI] [PubMed] [Google Scholar]
- 150. Lisi L, Navarra P, Cirocchi R, et al Rapamycin reduces clinical signs and neuropathic pain in a chronic model of experimental autoimmune encephalomyelitis. J Neuroimmunol 2012; 243 : 43–51. [DOI] [PubMed] [Google Scholar]
- 151. Salehi M, Bagherpour B, Shayghannejad V, et al Th1, Th2 and Th17 cytokine profile in patients with multiple sclerosis following treatment with rapamycin. Iran J Immunol 2016; 13 : 141–147. [PubMed] [Google Scholar]
- 152. Criollo A, Maiuri MC, Tasdemir E, et al Regulation of autophagy by the inositol trisphosphate receptor. Cell Death Differ 2007; 14 : 1029–1039. [DOI] [PubMed] [Google Scholar]
- 153. Pryce G, Riddall DR, Selwood DL, et al Neuroprotection in experimental autoimmune encephalomyelitis and progressive multiple sclerosis by cannabis‐based cannabinoids. J Neuroimmune Pharmacol 2015; 10 : 281–292. [DOI] [PubMed] [Google Scholar]
- 154. Wang C, Zhang X, Teng Z, et al Downregulation of PI3K/Akt/mTOR signaling pathway in curcumin‐induced autophagy in APP/PS1 double transgenic mice. Eur J Pharmacol 2014; 740 : 312–320. [DOI] [PubMed] [Google Scholar]
- 155. Tomic T, Botton T, Cerezo M, et al Metformin inhibits melanoma development through autophagy and apoptosis mechanisms. Cell Death Dis 2011; 2 : e199. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 156. Rodríguez‐Navarro JA, Rodríguez L, Casarejos MJ, et al Trehalose ameliorates dopaminergic and tau pathology in parkin deleted/tau overexpressing mice through autophagy activation. Neurobiol Dis 2010; 39 : 423–438. [DOI] [PubMed] [Google Scholar]
- 157. Sachs HH, Bercury KK, Popescu DC, et al A new model of cuprizone‐mediated demyelination/remyelination. ASN Neuro 2014; 6 : 1759091414551955. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 158. Liu J, Xia H, Kim M, et al Beclin1 controls the levels of p53 by regulating the deubiquitination activity of USP10 and USP13. Cell 2011; 147 : 223–234. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 159. Wei X, Zhou Z, Li L, et al Intrathecal injection of 3‐methyladenine reduces neuronal damage and promotes functional recovery via autophagy attenuation after spinal cord ischemia/reperfusion injury in rats. Biol Pharm Bull 2016; 39 : 665–673. [DOI] [PubMed] [Google Scholar]
- 160. Thome R, Moraes AS, Bombeiro AL, et al Chloroquine treatment enhances regulatory T cells and reduces the severity of experimental autoimmune encephalomyelitis. PLoS One 2013; 8 : e65913. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 161. Frake RA, Ricketts T, Menzies FM, et al Autophagy and neurodegeneration. J Clin Invest 2015; 125 : 65–74. [DOI] [PMC free article] [PubMed] [Google Scholar]
