Next Article in Journal
Matrix Metallopeptidase-Gene Signature Predicts Stage I Lung Adenocarcinoma Survival Outcomes
Next Article in Special Issue
Benefits of the Neurogenic Potential of Melatonin for Treating Neurological and Neuropsychiatric Disorders
Previous Article in Journal
Genome-Wide Characterization and Functional Analysis of ABCG Subfamily Reveal Its Role in Cutin Formation in Cotton
Previous Article in Special Issue
Multiplatform-Integrated Identification of Melatonin Targets for a Triad of Psychosocial-Sleep/Circadian-Cardiometabolic Disorders
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress

1
East Coast Life Sciences Institute, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
2
Department of Marine Bioscience, College of Life Science, Gangneung-Wonju National University, Gangneung 25457, Republic of Korea
*
Authors to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(3), 2381; https://doi.org/10.3390/ijms24032381
Submission received: 26 December 2022 / Revised: 17 January 2023 / Accepted: 21 January 2023 / Published: 25 January 2023
(This article belongs to the Special Issue Pleiotropic Benefits of Melatonin: From Basic Mechanisms to Disease)

Abstract

:
As people age, their risks of developing degenerative diseases such as cancer, diabetes, Parkinson’s Disease (PD), Alzheimer’s Disease (AD), rheumatoid arthritis, and osteoporosis are generally increasing. Millions of people worldwide suffer from these diseases as they age. In most countries, neurodegenerative diseases are generally recognized as the number one cause afflicting the elderly. Endoplasmic reticulum (ER) stress has been suggested to be associated with some human neurological diseases, such as PD and AD. Melatonin, a neuroendocrine hormone mainly synthesized in the pineal gland, is involved in pleiotropically biological functions, including the control of the circadian rhythm, immune enhancement, and antioxidant, anti-aging, and anti-tumor effects. Although there are many papers on the prevention or suppression of diseases by melatonin, there are very few papers about the effects of melatonin on ER stress in neurons and neurodegenerative diseases. This paper aims to summarize and present the effects of melatonin reported so far, focusing on its effects on neurons and neurodegenerative diseases related to ER stress. Studies have shown that the primary target molecule of ER stress for melatonin is CHOP, and PERK and GRP78/BiP are the secondary target molecules. Therefore, melatonin is crucial in protecting neurons and treating neurodegeneration against ER stress.

1. Introduction

The endoplasmic reticulum (ER) is critical for normal cellular function. ER stress due to dysfunction or loss of integrity caused by the accumulation of unfolded proteins or changes in calcium homeostasis within the ER can lead to apoptosis and cell signals associated with apoptosis [1,2,3]. Common causes of neurodegenerative diseases include the accumulation and deposition of misfolded proteins that affect neuronal connectivity, cell death, and faulty cell signaling [4,5]. Unfolded protein response (UPR) activity, typically dysfunctional due to erroneous protein aggregation or oxidative stress in cells, can cause more protein accumulation in the cells, leading to ER stress and disease exacerbation [6,7]. Melatonin, a neuroendocrine hormone mainly synthesized in the pineal gland, is involved in pleiotropically biological functions, including the control of the circadian rhythm, immune enhancement, antioxidant, anti-aging, and anti-tumor effects [8,9].
This paper aimed to collect and analyze data on the association between neurodegenerative diseases and ER stress in PubMed published up to 23 November 2022, focusing on molecular mechanisms involved in such association according to the year of publication and how melatonin might contribute to the relationship between ER stress and human neurological disorders.

2. Relationship between Endoplasmic Reticulum Stress and Neurodegenerative Diseases

As of 23 November 2022, the total number of published articles related to neurodegenerative diseases in PubMed was 400,813, of which 17,869 included oxidative stress among the cell mechanisms related to neurodegenerative diseases. There were 13,097 articles on apoptosis, 11,477 on mitochondria, 6481 on autophagy, and 1627 on ER stress. Among the neurodegenerative disease studies, ER stress-related research started in 1990 had fewer studies than the other studies. According to PubMed, there were 514 review articles. In 2005, there were only about 20 papers. However, the research has been quite explosive recently (Figure 1).
According to PubMed, Parkinson’s Disease (PD) was the most studied neurodegenerative disease, with a focus on ER stress, followed by Alzheimer’s Disease (AD), Amyotrophic Lateral Sclerosis (ALS), Transmissible Spongiform Encephalopathy (TSE), polyglutamine diseases, acute neurodegeneration, and neuronal storage diseases (Figure 2). This suggests that PD, AD, and ALS are the most relevant neurodegenerative diseases in the elderly. It also suggests that the research has been focused on ER stress for treating these diseases.
In the following, we will describe how ER stress in neurodegenerative diseases may contribute to the disease process of human neurological disorders.

2.1. ER Stress and Parkinson’s Disease (PD)

PD is an age-related, chronic, progressive, degenerative disease caused by the decline of dopaminergic neurons in a specific brain area, i.e., the substantia nigra pars compacta. Typically, PD is characterized by intraneuronal cytoplasmic inclusion bodies, known as Lewy bodies, caused by the abnormal deposition of a protein called alpha-synuclein in the brain [10]. However, the exact cellular mechanisms that cause selective neuronal death in PD are still not fully understood yet [11].
According to PubMed, research on ER stress in PD began in 2000 and gradually increased. Since 2016, more than 46 papers have been intensively published every year (Figure 3).
The following papers are organized by year of publication in PubMed: 6-Hydroxydopamine (6-OHDA) and 1-methyl-4-phenyl-pyridinium (MPP+) are widely used to induce dopaminergic neuron death in in vitro and in vivo PD models. An association between ER stress and the UPR has been reported in neuronal PC12 cells [12], sympathetic neurons from PERK null mice [13], and the MN9D cells’ dopaminergic cell line [13]. ER-localized molecular chaperones (GRP78/BiP and GRP94) have been revealed in SH-SY5Y cells [14]. The relationship between Parkin protein and ER stress in PD has also been proven [15]. IRE1alpha, an ER stress molecule, participates in unfolded protein responses through its interaction with the Jun activation domain-binding protein-1 (JAB1) [16]. Alpha-synuclein cytotoxicity is related to ER stress in PD [17,18,19,20,21]. The ER stress molecules CHOP/Gadd153 and GRP78/BiP are increased in human neuroblastoma SH-SY5Y cells treated with 6-OHDA, indicating that ER dysfunction is involved in the mechanisms induced by 6-OHDA in SH-SY5Y cells [22]. The relationship between alpha-synuclein and PD concerning ER stress has been reviewed [23,24,25]. As ER stress markers, the phosphorylated pancreatic ER kinase (p-PERK) and phosphorylated eukaryotic initiation factor 2alpha (p-eIF2alpha) have been detected in the substantia nigra of PD patients, suggesting that the UPR’s activation is closely related to the accumulation of alpha-synuclein aggregates [26]. The emerging mechanisms of ER stress in PD have been reviewed [27,28] and demonstrated [29].
Parkin has a role in the crosstalk between ER and mitochondrial stress, suggesting that Parkin has a role in the crosstalk between ER and mitochondrial stress, suggesting that both ER and mitochondrial stress can contribute to the pathogenesis of PD [30]. GRP78/BiP can bind to α-synuclein and increase the aggregated α-synuclein’s accumulation in in vitro and in vivo models, indicating that the activation of the UPR pathway in the PD brain is associated with α-synuclein’s accumulation occurring, in part, within the ER [31]. Calì et al. [32] have reviewed the interplay between the mitochondria, ER, and proteasome systems in PD-associated neurodegeneration. When molecular events occurring during ER stress and the unfolded protein response are evaluated, it has been found that the ER stress response plays a role in neurodegenerative disorders, including AD, PD, ALS, and prion diseases [33,34,35,36,37]. Alpha-synuclein can inhibit ATF6, a protective branch of the UPR, suggesting a link between ER stress and the role of the UPR in PD [38]. Tsujii et al. have reviewed the involvement of ER stress with in vitro and in vivo PD models [39]. GRP78/BiP and p-PERK are increased in the Lewy bodies of patients with dementia [40]. An increase in activating transcription factor 4 (ATF4) as a member of the PERK signaling pathway has been identified in experimental animal models and the postmortem melanin-containing neurons of PD patients [41,42]. The IRE1 pathway can drive PD’s progression by coupling the ER stress [43]. The significant role of ER stress in PD pathogenesis has been highlighted [44]. eIF2α phosphorylation mediated by PERK can protect DA neurons against chronic heat stress in Drosophila [45]. The neurodegenerative disease can be activated or inhibited through the PERK pathway [46].
Therefore, the target molecules of ER stress for PD are GRP78/BiP, CHOP, PERK, IRE1, ATF6, and ATF4, suggesting that these ER stress molecules can act mainly as pathological factors in PD.

2.2. ER Stress and Alzheimer’s Disease (AD)

AD is a neurodegenerative disease that can lead to progressive impairments in memory, behavioral, and thinking functions. AD causes the loss of neurons in several brain regions within the prefrontal cortex, hippocampus, and basal forebrain area [47]. Pathological features of AD patients’ brain tissues include extracellular senile plaques with amyloid β-peptide (Aβ) deposits and intracellular accumulation of the tau protein with neurofibrillary tangles (NFTs). AD includes early-onset familial AD (FAD) and later-onset sporadic AD. Mutations in presenilin PS1 and PS2 are associated with FAD [47].
According to PubMed, research on ER stress in AD was started in 1997. The number of such research studies then gradually increased. Since 2016, more than 50 papers on ER stress in AD have been published intensively annually (Figure 4).
The following papers are organized according to the year presented in PubMed: For the pathogenesis of neurodegeneration in AD, presenilin has been suggested to be related to ER [48,49]. PS1 mutations or loss of the PS1 function can affect the UPR, such as the expression of GRP78/BiP or CHOP, suggesting that PS1 can increase vulnerability to ER stress in association with the UPR [50,51]. Activation of the UPR in AD and increased GRP78/BiP and p-PERK in normal neurons suggest that the UPR is involved in AD neurodegeneration [52]. ER stress can be a critical trigger in the neuronal response to inflammatory activity. This has been reviewed in terms of the pathology of AD [53].
Aβs can activate the ER stress response factor X-box binding protein 1 (XBP-1) in transgenic flies and mammalian-cultured neurons [54]. AD pathogenesis involves ER stress and hyperphosphorylation of the tau protein [55]. Although the oligomeric form of Aβs has been thought to play a critical role in AD, the mechanisms involved remain unclear in the pathogenesis of AD. Aβs can induce ER stress in AD, suggesting that targeting this toxic component can enable the treatment of various neurodegenerative diseases [56]. Differential manifestations of ER stress and docosahexaenoic acid’s (DHA’s) reactiveness may help explain the variable clinical results obtained with DHA treatment, suggesting that DHA might be adequate for a subset of AD patients [57]. The IgG Fcγ receptor II-b (FcγRIIb) can mediate Aβ neurotoxicity and neurodegeneration. Soluble Aβ oligomers can interact with FcγRIIb in vitro and in AD brains to activate ER stress and caspase-12 [58]. The pharmacological inhibition of brain inflammation and ER stress in mice can prevent glucose intolerance, demonstrating that AD-associated Aβ oligomers can affect the central nervous system [59,60]. This result suggests a novel molecular mechanism between metabolic disease and hypothalamic dysfunction in AD. The complex roles of ER stress, including XBP1 and PERK, during Aβ pathogenesis, indicate the status of disease progression [61]. Inflammasome-mediated peripheral inflammation can contribute to AD pathology [62]. ER stress and the UPR are critical to AD development and onset pathogenesis [63,64]. ER stress and the UPR in cellular aging and neuroinflammatory processes can lead to memory impairment and synapse dysfunction in AD [65].
Therefore, the target molecules of ER stress for AD are GRP78/BiP, CHOP, PERK, and XBP-1, suggesting that these ER stress molecules can act mainly as pathological factors in AD.

2.3. ER Stress and Amyotrophic Lateral Sclerosis (ALS)

ALS is a progressive neurodegenerative disease of neurons in the spinal cord and the brain, resulting in a loss of muscle control. Motor neuron loss continues until the person loses the ability to eat, speak, move, and breathe. ALS eventually causes paralysis and premature death due to respiratory failure [66,67,68]. ALS is most common in people aged 40 to 70 years, regardless of racial or ethnic group, although it can occur at younger ages. ALS has two types: sporadic ALS and familial ALS. Sporadic ALS is the most common form in the United States, accounting for approximately 90 to 95% of all cases. It occurs randomly without a known cause or family history. Familial ALS affects 5 to 10% of people and is inherited [69,70]. Although the disease usually progresses rapidly, the underlying causes of neuronal death are not fully known.
According to PubMed, research on ER stress in ALS started in 2003 and gradually increased. Since 2013, more than 20 papers have been published yearly (Figure 5).
The following papers are organized according to the year presented in PubMed: Motor neurons in the spinal cords of transgenic mice with the mutant SOD1 linked to familial amyotrophic local sclerosis (FALS) show a significant increase in GRP78/BiP just before motor symptoms begin, suggesting that ER stress is significantly related to the pathogenesis of FALS with SOD1 mutation [71]. When caspase-12 activation occurs in the spinal cord of a transformed ALS mouse, oxidation and ER-induced stress are induced, resulting in neuronal cell death and disease progression [72]. This suggests that caspase-12 and the ER stress pathways for neuronal apoptosis might be potential new targets for ALS treatment. The balance between the anti- and pro-apoptotic proteins associated with ER stress can induce damage by affecting the presymptomatic phase in an ALS mouse model, suggesting that this imbalance might be involved in the pathological cause of motor neuron degeneration in ALS [73]. Alterations in the fatty acid composition, mitochondrial function, and proteasome activity can be induced by excitotoxicity in the neurons, thereby inducing oxidative stress and, finally, endoplasmic reticulum stress in sporadic ALS [74]. The BH3-only protein, Puma, associated with ER stress, is critically involved in neurodegenerative disease progression in SOD1G93A mice, indicating that Puma is an essential factor in controlling chronic neurodegeneration in ALS and other neurodegenerative disorders, including defects in the protein quality control [75]. SOD1-induced protein misfolding associated with ALS mutations can induce ER stress, suggesting that it might contribute to motor neuron degeneration in ALS pathogenesis [76].
The interaction between Derlin-1 and SOD1 mutation can induce ER stress. Subsequent ASK1 activation is critical for disease progression in familial ALS [77]. Vesicle-associated membrane protein-associated protein B (VAPB), a novel ALS causative gene, plays a pivotal role in the UPR to ER stress. The ALS-linked P56S mutation in VAPB can abolish the function of VAPB, resulting in motor neuron vulnerability to ER stress [78]. A review article has presented ER stress and UPR dysfunction concerning ALS pathological mechanisms [79]. Although all motoneurons in ALS can be preferentially affected, the ER stress response in the specific motoneuron subtypes of ALS can affect the gradual onset of weakness and paralysis [80]. A new function of XBP-1 in autophagy indicates the critical correlation between the increased autophagy in the motoneurons and a reduced accumulation of mutant SOD1 aggregates, which can protect the neurons against neurodegeneration in ALS [81,82]. The upregulation of CHOP in the motor neurons and glial cells might play a pivotal role in the pathogenesis of ALS [83]. ER stress may act as a possible risk factor for the development of ALS by increasing the susceptibility of the wild-type SOD1 to aggregation during aging [84].
Therefore, the target molecules of ER stress for ALS are GRP78/BiP, CHOP, and XBP-1, suggesting that these ER stress molecules can act mainly as pathological factors in ALS.

2.4. ER Stress and Transmissible Spongiform Encephalopathies (TSEs)

TSEs or prion diseases are rare progressive neurodegenerative brain disorders affecting humans and animals with a spongy form in the brain. TSEs include Creutzfeldt–Jakob disease, bovine spongiform encephalopathy (BSE), and scrapie. The pathological hallmark of TSEs is the accumulation of a misfolded protease-resistant form of prion protein (PrP) in the cerebrum, leading to extensive neuronal cell death as the disease progresses [85,86,87].
According to PubMed, ER stress in TSE was not studied much until recently. However, in 2000, papers began to be published gradually. In 2015, 12 papers were published. The number of published papers gradually decreased until recently (Figure 6).
The following articles are organized by year of publication in PubMed: Caspase-12 is increased in cells showing misfolded prion protein (PrPSc). It is also increased in the brain tissues of PrPSc-infected mice and Creutzfeldt–Jakob disease patients [88]. This is the first study to present that ER stress and the activation of caspase-12 can cause neuronal cell death by accumulating mutant prion protein in the neurons. The overexpression of GRP58 can protect the cells from PrPSc toxicity and reduce caspase-12, suggesting that the expression of GRP58 can act as a neuroprotective factor against prion neurotoxicity [89]. Studies on GRP58 have suggested that it can be used to develop new targets for treating prion diseases [89]. The induction of ER stress in transgenic mice expressing PrP variants is accompanied by reduced translocation, a finding that could link age-dependent clinical and histological signs to PrP-mediated neurodegeneration [90]. One mechanism of prion-mediated neurodegeneration involves indirect ER stress-dependent effects on early PrP biosynthesis and metabolism [90]. The role of GRP78/BiP has been demonstrated to be important in prion diseases through in vivo and in vitro approaches [91]. The upregulation of PERK, GRP78/BiP, the ER protein, disulfide isomerase, and ubiquitin in prion disease indicates that ER stress and proteasome damage can initiate the early stages of spontaneous prion disease [92].
Therefore, the target molecules of ER stress for TSEs are GRP58, GRP78/BiP, and PERK, suggesting that these ER stress molecules can act mainly as pathological factors in TSEs.

2.5. ER Stress and Polyglutamine Diseases

Polyglutamine diseases are manifested by progressive neurodegeneration that can lead to behavioral and physical impairments. Polyglutamine diseases include Huntington’s disease (HD), dentatorubral-pallidoluysian atrophy, spinal and bulbar muscular atrophy, and spinocerebellar ataxias 1, 2, 3, 6, 7, and 17 [93]. The disease features selective neuronal cell death and the accumulation of intracellular protein aggregates in cultured cells, transgenic animals, and human postmortem brain tissues. The pathophysiology of polyglutamine disorders with polyglutamine expansion and protein deposits remains unclear [94]. ASK1 is essential for neuropathological changes in polyglutamine diseases. It is crucial for killing neurons by inducing ER stress [95]. In HD, the N-terminal huntingtin protein can increase ER stress-related GRP78/BiP, CHOP, c-Jun-N-terminal kinase (JNK) phosphorylation, and caspase-12 activation [96]. Polyglutamine-expanded huntingtin fragments expressing yeast cells and neuron-like PC12 cells can impair the ER-associated degradation (ERAD) proteins Npl4, Ufd1, and p97 [97]. Soluble oligomeric polyglutamine-expanded huntingtin can induce ER stress before its aggregation through p97/VCP [98].
Therefore, the target molecules of ER stress for polyglutamine diseases are ASK1, GRP78/BiP, CHOP, and ERAD, suggesting that these ER stress molecules can act mainly as pathological factors in polyglutamine diseases.

2.6. ER Stress and Neuronal Storage Diseases or Lysosomal Storage Disease (LSD)

Lysosomal storage disease (LSD) is a hereditary metabolic disorder with a defective lysosomal function. LSD includes more than 70 diseases with an abnormal lysosomal function, most of which are inherited as autosomal recessive traits. Although these disorders are rare individually, collectively, they occur in one in five thousand births [99]. Most LSDs show clinically progressive neurodegeneration, although symptoms of other organ systems are also frequently present. LSD has been classified and described in considerable detail previously [99].
Pelizaeus–Merzbacher disease (PMD) is an X-linked recessive pediatric and neurodegenerative disorder characterized by diffuse hypomyelination of the central nervous system. Mutations, duplications, or deletions of the phospholipid protein (PLP) gene can cause PMD by accumulating the PLP and/or DM20 proteins in the ER. CHOP in oligodendrocytes is directly involved in mice with the gene deletion causing PMD [100]. The accumulation of GM2-gangliosidosis in a mouse model of Sandhoff disease is caused by reduced calcium uptake into the ER due to decreased sarcoplasmic/ER calcium ATPase (SERCA), resulting in neuronal death [101]. GM1-ganglioside (GM1) is a significant sialoglycolipid in nerve cell membranes that can regulate calcium homeostasis. In a mouse model with GM1-gangliosidosis, the upregulation of GRP78/BiP and CHOP and the activation of JNK2 and caspase-12 can induce neuronal cell death, suggesting that the UPR can induce the accumulation of the sialoglylipid GM1 in the neuronal cells and then cause neuronal apoptosis [102]. CLN6 is a non-glycosylated ER-resident membrane protein with an unknown function. Mutant Cln6 can prevent the accumulation of the misfolded Cln6 protein by proteasomal degradation, impair constitutive autophagy by lysosomal dysfunction, and promote neurodegeneration [103]. A deficiency in the hexosaminidase activity can disrupt the normal metabolism of GM2-ganglioside, leading to progressive neurodegenerative diseases [104]. In cultured neurons, GM2-ganglioside accumulation in the ER can induce luminal Ca2+ depletion, activating PERK. Therefore, it can induce neurite atrophy and apoptosis [104]. Pelizaeus–Merzbacher-like disease type 1 (PMLD1) is a hypomyelination disorder in patients with mutations in the GJC2 coding for Connexin47 (Cx47). PMLD1 can cause nystagmus, cerebellar ataxia, convulsions, and changes in the CNS’s white matter. In three PMLD1-related mutants (p.P87S, p.Y269D, and p.M283T), Cx47 can induce ER stress, including the UPR, and activate the apoptotic pathway [105].
Therefore, the target molecules of ER stress for LSD are GRP78/BiP, CHOP, PERK, IRE1, ATF6, and ATF4, suggesting that these ER stress molecules can act mainly as pathological factors in LSD.

2.7. ER Stress and Acute Neurodegeneration

ER stress can induce acute brain disorders such as cerebral ischemia and traumatic brain injury, apart from chronic neurodegenerative diseases. Oxygen-regulated protein 150 kD (ORP150) and 94 kDa glucose-regulated protein (GRP94) as ER chaperones can be directly involved in the ER response in brain injury. ORP150 is elevated in the neurons of a human brain with ischemic stress [106]. In human neuroblastoma cells exposed to hypoxia/reoxygenation, GRP94 is increased [107]. Caspase-12-mediated ER apoptosis might be involved in rat traumatic brain injury pathology [108]. Primary hippocampal neurons in CHOP-deficient mice are resistant to hypoxia-reoxygenation-induced apoptosis, indicating that ischemia-induced hippocampal neuronal death is caused by the ER stress pathway associated with CHOP induction [109]. Global cerebral ischemia in transgenic rats can increase the expression of ATF4 and CHOP, indicating that superoxide is involved in ER stress-induced cell death [110].
Therefore, the target molecules of ER stress for acute neurodegeneration are GRP94, CHOP, and ATF4, suggesting that these ER stress molecules can act mainly as pathological factors in acute neurodegeneration.

3. Melatonin

Melatonin is a potent antioxidant and free radical scavenger [111,112] and protects against inflammation, apoptosis, or autophagy in physiological and pathophysiological conditions [113,114,115,116]. In addition, melatonin modulates ER stress and UPR dysfunction in cancers, liver diseases, and other pathologies [115,116,117]. In particular, review articles introduce that melatonin can suppress various diseases related to ER stress: chronic intestinal inflammation and colon cancer [118], breast cancer [119], osteoarthritis [120], hepatocellular carcinoma [121], delaying ovarian aging [122], acute myocardial infarction [123], and diabetic cardiomyopathy [124].
Although there are many papers on the prevention or suppression of diseases by melatonin, there are very few papers about the effects of melatonin on ER stress in neurons and neurodegenerative diseases. The following describes the effects of melatonin reported so far, focusing on its effects on neurons and neurodegenerative diseases related to ER stress.

3.1. Melatonin and ER Stress in Neurons

Melatonin can significantly reduce the neuron splicing of the XBP-1 mRNA, increase eIF2α phosphorylation, and elevate the expression of the chaperone proteins GRP78/BiP and Hsp70 in hypoxia-ischemia of a rat brain [125]. In addition, melatonin can significantly reduce CHOP expression. Melatonin can reduce ER stress induced by neonatal hypoxia-ischemia in newborn rats through SIRT-1 as the modulation and neuroprotection [125]. Melatonin can protect neuroblastoma cells against methamphetamine-induced ER stress and apoptosis by modulating CHOP, spliced XBP1, caspase-12, and caspase-3 [126]. Methamphetamine can induce ER stress in glial cells by stimulating the UPR to increase PERK phosphorylation, ATF6 expression, and the phosphorylated inositol-requiring enzyme 1 (p-IRE1). Moreover, the expression levels of GRP78/BiP and CHOP, caspase-12 activation, eIF2α phosphorylation, and XBP-1 mRNA expression are increased. However, melatonin can reduce ER stress through methamphetamine toxicity by reducing the expression of the ER stress response genes and proteins [127]. Ischemia/reperfusion in acute neuronal injury after an ischemic stroke can induce neuronal apoptosis by increasing ER stress, including the phosphorylation of PERK and IRE1 and the expression of ATF6 and CHOP. Melatonin pretreatment can attenuate ischemia/reperfusion-induced ER stress [128]. Melatonin treatment can attenuate the insulin-induced mRNA or protein expression levels of ER stress markers such as p-eIF2α, ATF4, CHOP, sXBP1, p-IRE1, and p-ASK1. Therefore, melatonin can regulate neuronal cell death induced by ER stress under insulin resistance by reducing the ER stress in SH-SY5Y neuroblastoma cells [129].
Melatonin can effectively downregulate the levels of ER stress molecules GRP78/BiP, CHOP, and caspase-12 proteins in kainic acid-induced N2a cells [130]. Melatonin can also inhibit tau hyperphosphorylation, the phosphorylation of PERK and eIF2α, and the expression levels of ATF4 and GRP78/BiP in the kainic acid-treated mouse hippocampus [131]. Melatonin can attenuate ER stress, such as the phosphorylation of PERK and eIF2α and the expression levels of ATF4 and CHOP in neurons exposed to oxygen and glucose deprivation and in rats subjected to transient focal cerebral ischemia [132]. These results indicate that melatonin suppresses post-ischemic ER stress after an ischemic stroke. The administration of melatonin can significantly decrease the mRNA and protein levels of ATF6 and CHOP in intracerebral hemorrhage rats, indicating that melatonin can protect neurons against apoptosis by suppressing ATF6 and CHOP [133]. Melatonin can decrease ER stress, such as the p-IRE1, p-PERK, GRP78/BiP, and CHOP, in the chronic cerebral hypoperfusion of rats, suggesting that melatonin can improve cognitive impairment following the induction of bilateral typical carotid artery occlusion by attenuating AD markers and reducing ER stress [134].
Therefore, these studies show that the first target molecule of ER stress for melatonin is CHOP, and the second target molecules are PERK and GRP78/BiP. Therefore, melatonin is a crucial strategy to protect and treat neurons against ER stress.

3.2. Melatonin as ER Stress Modulator against Neurodegenerative Diseases

Direct oxidative damage and organelle lesions in rats can increase the expression of ER stress-related proteins, including GRP78/BiP and CHOP. The addition of melatonin can partially suppress ER stress-related behavioral and molecular disturbances. These results suggest that melatonin is directly related to AD-like behavioral and molecular pathologies with ER stress-related mechanisms [135]. One review article has presented the effects of melatonin on the fundamental ER stress mechanisms, focusing on its ability to modulate autophagy and apoptotic processes in the development of cancer cells, neurodegeneration, liver disease, and other pathologies [117].
Melatonin can protect mesenchymal stem cells from ischemic damage by inhibiting ER stress (such as decreasing the phosphorylation of PERK/eIF2α/IRE1α and the expression levels of ATF4 and CHOP proteins) and autophagy both in vivo and in vitro. In addition, melatonin can increase the cellular prion protein (PrPC) expression to prevent apoptotic cell death induced by ER stress and autophagy [136]. Melatonin is an endogenously produced molecule that can act as a copper chelator, a potent antioxidant, and an inhibitor of ER stress and the UPR in the liver and brain. Therefore, melatonin can potentially lower copper levels and limit the progression of Wilson’s disease [137]. Melatonin can reduce Aβ peptide accumulation and ER stress markers such as the protein levels of GRP78/BiP, CHOP, and caspase-12 in an AlCl3-treated AD model. Therefore, adding melatonin might be an alternative way to alleviate the onset of Alzheimer’s disease [138].
Therefore, an in vivo study of melatonin has also revealed that the molecules of ER stress are CHOP, GRP78/BiP, and PERK. Therefore, melatonin has a significant strategic value in inhibiting and treating neurodegeneration caused by ER stress.

4. Conclusions

ER participates in various cellular processes, from which proteins are synthesized and transported to their final destinations after correct processing and modification. In neurodegenerative diseases, misprocessing occurs in many cellular processes related to ER, which can act as ER stress and finally cause neuronal cell death. However, two questions remain in this process: (1) how much cellular degeneration will occur due to ER stress? and (2) how much would another pathway induce cellular degeneration? The proteins that regulate or participate in the ER–mitochondrial interaction that causes cell death are not fully understood yet. ER stress molecules are involved in apoptosis, leading to a loss of ER function and the activation of the apoptosis cascade. Likewise, neuronal death occurs when proteins associated with neurodegenerative diseases accumulate to form cellular aggregates. This probably depends on the nature and function of the protein in question. Therefore, we think it is necessary to clearly understand and solve the mutual relationship between the molecules involved in ER stress and neuronal cell death.
Taken together, the reviewed findings suggest that we can target specific stages of the disease by focusing on the ER stress target molecules of melatonin to prevent or treat neurodegenerative diseases. This review suggests that CHOP, GRP78/BiP, and PERK are the ER stress target molecules of melatonin. Of course, more research studies are needed to test this possibility.

Author Contributions

Y.-M.Y. and S.S.J. conceived the manuscript’s concept, wrote the paper, performed scientific and language editing, and revised the paper. All authors have read and agreed to the published version of the manuscript.

Funding

This research was supported by the Basic Science Research Program through the National Research Foundation of Korea (NRF), funded by the Ministry of Education (2020R1I1A1A01060627).

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

Not applicable.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Paschen, W.; Frandsen, A. Endoplasmic reticulum dysfunction—A common denominator for cell injury in acute and degenerative diseases of the brain? J. Neurochem. 2001, 79, 719–725. [Google Scholar] [CrossRef]
  2. Breckenridge, D.G.; Germain, M.; Mathai, J.P.; Nguyen, M.; Shore, G.C. Regulation of apoptosis by endoplasmic reticulum pathways. Oncogene 2003, 22, 8608–8618. [Google Scholar] [CrossRef] [Green Version]
  3. Rao, R.V.; Ellerby, H.M.; Bredesen, D.E. Coupling endoplasmic reticulum stress to the cell death program. Cell Death Differ. 2004, 11, 372–380. [Google Scholar] [CrossRef] [Green Version]
  4. Soto, C. Unfolding the role of protein misfolding in neurodegenerative diseases. Nat. Rev. Neurosci. 2003, 4, 49–60. [Google Scholar] [CrossRef]
  5. Bence, N.F.; Sampat, R.M.; Kopito, R.R. Impairment of the ubiquitin-proteasome system by protein aggregation. Science 2001, 292, 1552–1555. [Google Scholar] [CrossRef]
  6. Ciechanover, A.; Brundin, P. The ubiquitin proteasome system in neurodegenerative diseases: Sometimes the chicken, sometimes the egg. Neuron 2003, 40, 427–446. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  7. Imai, Y.; Soda, M.; Inoue, H.; Hattori, N.; Mizuno, Y.; Takahashi, R. An unfolded putative transmembrane polypeptide, which can lead to endoplasmic reticulum stress, is a substrate of Parkin. Cell 2001, 105, 891–902. [Google Scholar] [CrossRef] [Green Version]
  8. Di Bella, G.; Mascia, F.; Gualano, L.; Di Bella, L. Melatonin anticancer effects: Review. Int. J. Mol. Sci. 2013, 14, 2410–2440. [Google Scholar] [CrossRef] [Green Version]
  9. Lee, Y. Roles of circadian clocks in cancer pathogenesis and treatment. Exp. Mol. Med. 2021, 53, 1529–1538. [Google Scholar] [CrossRef]
  10. Healy, D.G.; Abou-Sleiman, P.M.; Wood, N.W. PINK, PANK, or PARK? A clinicians’ guide to familial parkinsonism. Lancet Neurol. 2004, 3, 652–662. [Google Scholar] [CrossRef]
  11. Greenamyre, J.T.; Hastings, T.G. Biomedicine. Parkinson’s--divergent causes, convergent mechanisms. Science 2004, 304, 1120–1122. [Google Scholar] [CrossRef]
  12. Ryu, E.J.; Harding, H.P.; Angelastro, J.M.; Vitolo, O.V.; Ron, D.; Greene, L.A. Endoplasmic reticulum stress and the unfolded protein response in cellular models of Parkinson’s disease. J. Neurosci. 2002, 22, 10690–10698. [Google Scholar] [CrossRef] [Green Version]
  13. Holtz, W.A.; O’Malley, K.L. Parkinsonian mimetics induce aspects of unfolded protein response in death of dopaminergic neurons. J. Biol. Chem. 2003, 278, 19367–19377. [Google Scholar] [CrossRef] [Green Version]
  14. Ishibashi, T.; Matsuo, N.; Ogawa, S.; Tohyama, M. RA410/Sly1 suppresses MPP+ and 6-hydroxydopamine-induced cell death in SH-SY5Y cells. Neurobiol. Dis. 2005, 18, 143–151. [Google Scholar] [CrossRef]
  15. Takahashi, R.; Imai, Y.; Hattori, N.; Mizuno, Y. Parkin and endoplasmic reticulum stress. Ann. N. Y. Acad. Sci. 2003, 991, 101–106. [Google Scholar] [CrossRef] [PubMed]
  16. Yamagishi, S.; Matsuda, S.; Hitomi, J.; Miyata, S.; Mizuno, T.; Imaizumi, K.; Katayama, T.; Tohyama, M. JAB1 participates in unfolded protein responses by association and dissociation with IRE1. Neurochem. Int. 2004, 45, 765–772. [Google Scholar] [CrossRef]
  17. Smith, W.W.; Jiang, H.; Pei, Z.; Tanaka, Y.; Morita, H.; Sawa, A.; Dawson, V.L.; Dawson, T.M.; Ross, C.A. Endoplasmic reticulum stress and mitochondrial cell death pathways mediate A53T mutant alpha-synuclein-induced toxicity. Hum. Mol. Genet. 2005, 14, 3801–3811. [Google Scholar] [CrossRef]
  18. Jiang, P.; Gan, M.; Ebrahim, A.S.; Lin, W.L.; Melrose, H.L.; Yen, S.H. ER stress response plays an important role in aggregation of α-synuclein. Mol. Neurodegener. 2010, 5, 56. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  19. Colla, E.; Jensen, P.H.; Pletnikova, O.; Troncoso, J.C.; Glabe, C.; Lee, M.K. Accumulation of toxic α-synuclein oligomer within endoplasmic reticulum occurs in α-synucleinopathy in vivo. J. Neurosci. 2012, 32, 3301–3305. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Heman-Ackah, S.M.; Manzano, R.; Hoozemans, J.J.M.; Scheper, W.; Flynn, R.; Haerty, W.; Cowley, S.A.; Bassett, A.R.; Wood, M.J.A. Alpha-synuclein induces the unfolded protein response in Parkinson’s disease SNCA triplication iPSC-derived neurons. Hum. Mol. Genet. 2017, 26, 4441–4450. [Google Scholar] [CrossRef] [Green Version]
  21. Stojkovska, I.; Wani, W.Y.; Zunke, F.; Belur, N.R.; Pavlenko, E.A.; Mwenda, N.; Sharma, K.; Francelle, L.; Mazzulli, J.R. Rescue of α-synuclein aggregation in Parkinson’s patient neurons by synergistic enhancement of ER proteostasis and protein trafficking. Neuron 2022, 110, 436–451.e11. [Google Scholar] [CrossRef]
  22. Yamamuro, A.; Yoshioka, Y.; Ogita, K.; Maeda, S. Involvement of endoplasmic reticulum stress on the cell death induced by 6-hydroxydopamine in human neuroblastoma SH-SY5Y cells. Neurochem. Res. 2006, 31, 657–664. [Google Scholar] [CrossRef]
  23. Chua, C.E.; Tang, B.L. Alpha-synuclein and Parkinson’s disease: The first roadblock. J. Cell. Mol. Med. 2006, 10, 837–846. [Google Scholar] [CrossRef]
  24. Lashuel, H.A.; Hirling, H. Rescuing defective vesicular trafficking protects against alpha-synuclein toxicity in cellular and animal models of Parkinson’s disease. ACS Chem. Biol. 2006, 1, 420–424. [Google Scholar] [CrossRef] [Green Version]
  25. Olivares, D.; Huang, X.; Branden, L.; Greig, N.H.; Rogers, J.T. Physiological and pathological role of alpha-synuclein in Parkinson’s disease through iron mediated oxidative stress; the role of a putative iron-responsive element. Int. J. Mol. Sci. 2009, 10, 1226–1260. [Google Scholar] [CrossRef] [Green Version]
  26. Hoozemans, J.J.; van Haastert, E.S.; Eikelenboom, P.; de Vos, R.A.; Rozemuller, J.M.; Scheper, W. Activation of the unfolded protein response in Parkinson’s disease. Biochem. Biophys. Res. Commun. 2007, 354, 707–711. [Google Scholar] [CrossRef]
  27. Wang, H.Q.; Takahashi, R. Expanding insights on the involvement of endoplasmic reticulum stress in Parkinson’s disease. Antioxid. Redox Signal. 2007, 9, 553–561. [Google Scholar] [CrossRef] [PubMed]
  28. Hosoi, T.; Ozawa, K. Endoplasmic reticulum stress in disease: Mechanisms and therapeutic opportunities. Clin. Sci. 2009, 118, 19–29. [Google Scholar] [CrossRef] [Green Version]
  29. Su, L.J.; Auluck, P.K.; Outeiro, T.F.; Yeger-Lotem, E.; Kritzer, J.A.; Tardiff, D.F.; Strathearn, K.E.; Liu, F.; Cao, S.; Hamamichi, S.; et al. Compounds from an unbiased chemical screen reverse both ER-to-Golgi trafficking defects and mitochondrial dysfunction in Parkinson’s disease models. Dis. Model. Mech. 2010, 3, 194–208. [Google Scholar] [CrossRef] [Green Version]
  30. Bouman, L.; Schlierf, A.; Lutz, A.K.; Shan, J.; Deinlein, A.; Kast, J.; Galehdar, Z.; Palmisano, V.; Patenge, N.; Berg, D.; et al. Parkin is transcriptionally regulated by ATF4: Evidence for an interconnection between mitochondrial stress and ER stress. Cell Death Differ. 2011, 18, 769–782. [Google Scholar] [CrossRef] [Green Version]
  31. Bellucci, A.; Navarria, L.; Zaltieri, M.; Falarti, E.; Bodei, S.; Sigala, S.; Battistin, L.; Spillantini, M.; Missale, C.; Spano, P. Induction of the unfolded protein response by α-synuclein in experimental models of Parkinson’s disease. J. Neurochem. 2011, 116, 588–605. [Google Scholar] [CrossRef]
  32. Calì, T.; Ottolini, D.; Brini, M. Mitochondria, calcium, and endoplasmic reticulum stress in Parkinson’s disease. Biofactors 2011, 37, 228–240. [Google Scholar] [CrossRef]
  33. Doyle, K.M.; Kennedy, D.; Gorman, A.M.; Gupta, S.; Healy, S.J.; Samali, A. Unfolded proteins and endoplasmic reticulum stress in neurodegenerative disorders. J. Cell. Mol. Med. 2011, 15, 2025–2039. [Google Scholar] [CrossRef] [Green Version]
  34. Ghemrawi, R.; Khair, M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Neurodegenerative Diseases. Int. J. Mol. Sci. 2020, 21, 6127. [Google Scholar] [CrossRef] [PubMed]
  35. Costa, C.A.D.; Manaa, W.E.; Duplan, E.; Checler, F. The Endoplasmic Reticulum Stress/Unfolded Protein Response and Their Contributions to Parkinson’s Disease Physiopathology. Cells 2020, 9, 2495. [Google Scholar] [CrossRef] [PubMed]
  36. Ren, H.; Zhai, W.; Lu, X.; Wang, G. The Cross-Links of Endoplasmic Reticulum Stress, Autophagy, and Neurodegeneration in Parkinson’s Disease. Front. Aging Neurosci. 2021, 13, 691881. [Google Scholar] [CrossRef] [PubMed]
  37. Kim, S.; Kim, D.K.; Jeong, S.; Lee, J. The Common Cellular Events in the Neurodegenerative Diseases and the Associated Role of Endoplasmic Reticulum Stress. Int. J. Mol. Sci. 2022, 23, 5894. [Google Scholar] [CrossRef] [PubMed]
  38. Credle, J.J.; Forcelli, P.A.; Delannoy, M.; Oaks, A.W.; Permaul, E.; Berry, D.L.; Duka, V.; Wills, J.; Sidhu, A. α-Synuclein-mediated inhibition of ATF6 processing into COPII vesicles disrupts UPR signaling in Parkinson’s disease. Neurobiol. Dis. 2015, 76, 112–125. [Google Scholar] [CrossRef]
  39. Tsujii, S.; Ishisaka, M.; Hara, H. Modulation of endoplasmic reticulum stress in Parkinson’s disease. Eur. J. Pharmacol. 2015, 765, 154–156. [Google Scholar] [CrossRef]
  40. Baek, J.H.; Whitfield, D.; Howlett, D.; Francis, P.; Bereczki, E.; Ballard, C.; Hortobágyi, T.; Attems, J.; Aarsland, D. Unfolded protein response is activated in Lewy body dementias. Neuropathol. Appl. Neurobiol. 2016, 42, 352–365. [Google Scholar] [CrossRef] [Green Version]
  41. Gully, J.C.; Sergeyev, V.G.; Bhootada, Y.; Mendez-Gomez, H.; Meyers, C.A.; Zolotukhin, S.; Gorbatyuk, M.S.; Gorbatyuk, O.S. Up-regulation of activating transcription factor 4 induces severe loss of dopamine nigral neurons in a rat model of Parkinson’s disease. Neurosci. Lett. 2016, 627, 36–41. [Google Scholar] [CrossRef]
  42. Demmings, M.D.; Tennyson, E.C.; Petroff, G.N.; Tarnowski-Garner, H.E.; Cregan, S.P. Activating transcription factor-4 promotes neuronal death induced by Parkinson’s disease neurotoxins and α-synuclein aggregates. Cell Death Differ. 2021, 28, 1627–1643. [Google Scholar] [CrossRef]
  43. Yan, C.; Liu, J.; Gao, J.; Sun, Y.; Zhang, L.; Song, H.; Xue, L.; Zhan, L.; Gao, G.; Ke, Z.; et al. IRE1 promotes neurodegeneration through autophagy-dependent neuron death in the Drosophila model of Parkinson’s disease. Cell Death Dis. 2019, 10, 800. [Google Scholar] [CrossRef] [Green Version]
  44. Mou, Z.; Yuan, Y.H.; Zhang, Z.; Song, L.K.; Chen, N.H. Endoplasmic reticulum stress, an important factor in the development of Parkinson’s disease. Toxicol. Lett. 2020, 324, 20–29. [Google Scholar] [CrossRef] [PubMed]
  45. Elvira, R.; Cha, S.J.; Noh, G.M.; Kim, K.; Han, J. PERK-Mediated eIF2α Phosphorylation Contributes to The Protection of Dopaminergic Neurons from Chronic Heat Stress in Drosophila. Int. J. Mol. Sci. 2020, 21, 845. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  46. Shacham, T.; Patel, C.; Lederkremer, G.Z. PERK Pathway and Neurodegenerative Disease: To Inhibit or to Activate? Biomolecules 2021, 11, 354. [Google Scholar] [CrossRef]
  47. Knopman, D.S.; Amieva, H.; Petersen, R.C.; Chételat, G.; Holtzman, D.M.; Hyman, B.T.; Nixon, R.A.; Jones, D.T. Alzheimer disease. Nat. Rev. Dis. Prim. 2021, 7, 33. [Google Scholar] [CrossRef]
  48. Mattson, M.P.; Guo, Q. Cell and molecular neurobiology of presenilins: A role for the endoplasmic reticulum in the pathogenesis of Alzheimer’s disease? J. Neurosci. Res. 1997, 50, 505–513. [Google Scholar] [CrossRef]
  49. Mattson, M.P.; Guo, Q.; Furukawa, K.; Pedersen, W.A. Presenilins, the endoplasmic reticulum, and neuronal apoptosis in Alzheimer’s disease. J. Neurochem. 1998, 70, 1–14. [Google Scholar] [CrossRef] [PubMed]
  50. Katayama, T.; Imaizumi, K.; Sato, N.; Miyoshi, K.; Kudo, T.; Hitomi, J.; Morihara, T.; Yoneda, T.; Gomi, F.; Mori, Y.; et al. Presenilin-1 mutations downregulate the signalling pathway of the unfolded-protein response. Nat. Cell Biol. 1999, 1, 479–485. [Google Scholar] [CrossRef]
  51. Sato, N.; Urano, F.; Yoon Leem, J.; Kim, S.H.; Li, M.; Donoviel, D.; Bernstein, A.; Lee, A.S.; Ron, D.; Veselits, M.L.; et al. Upregulation of BiP and CHOP by the unfolded-protein response is independent of presenilin expression. Nat. Cell Biol. 2000, 2, 863–870. [Google Scholar] [CrossRef]
  52. Hoozemans, J.J.; Veerhuis, R.; Van Haastert, E.S.; Rozemuller, J.M.; Baas, F.; Eikelenboom, P.; Scheper, W. The unfolded protein response is activated in Alzheimer’s disease. Acta Neuropathol. 2005, 110, 165–172. [Google Scholar] [CrossRef]
  53. Salminen, A.; Kauppinen, A.; Suuronen, T.; Kaarniranta, K.; Ojala, J. ER stress in Alzheimer’s disease: A novel neuronal trigger for inflammation and Alzheimer’s pathology. J. Neuroinflamm. 2009, 6, 41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  54. Casas-Tinto, S.; Zhang, Y.; Sanchez-Garcia, J.; Gomez-Velazquez, M.; Rincon-Limas, D.E.; Fernandez-Funez, P. The ER stress factor XBP1s prevents amyloid-beta neurotoxicity. Hum. Mol. Genet. 2011, 20, 2144–2160. [Google Scholar] [CrossRef] [Green Version]
  55. Ho, Y.S.; Yang, X.; Lau, J.C.; Hung, C.H.; Wuwongse, S.; Zhang, Q.; Wang, J.; Baum, L.; So, K.F.; Chang, R.C. Endoplasmic reticulum stress induces tau pathology and forms a vicious cycle: Implication in Alzheimer’s disease pathogenesis. J. Alzheimers Dis. 2012, 28, 839–854. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  56. Roussel, B.D.; Kruppa, A.J.; Miranda, E.; Crowther, D.C.; Lomas, D.A.; Marciniak, S.J. Endoplasmic reticulum dysfunction in neurological disease. Lancet Neurol. 2013, 12, 105–118. [Google Scholar] [CrossRef] [Green Version]
  57. Kondo, T.; Asai, M.; Tsukita, K.; Kutoku, Y.; Ohsawa, Y.; Sunada, Y.; Imamura, K.; Egawa, N.; Yahata, N.; Okita, K.; et al. Modeling Alzheimer’s disease with iPSCs reveals stress phenotypes associated with intracellular Aβ and differential drug responsiveness. Cell Stem Cell 2013, 12, 487–496. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  58. Kam, T.I.; Song, S.; Gwon, Y.; Park, H.; Yan, J.J.; Im, I.; Choi, J.W.; Choi, T.Y.; Kim, J.; Song, D.K.; et al. FcγRIIb mediates amyloid-β neurotoxicity and memory impairment in Alzheimer’s disease. J. Clin. Investig. 2013, 123, 2791–2802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  59. Clarke, J.R.; Lyra e Silva, N.M.; Figueiredo, C.P.; Frozza, R.L.; Ledo, J.H.; Beckman, D.; Katashima, C.K.; Razolli, D.; Carvalho, B.M.; Frazão, R.; et al. Alzheimer-associated Aβ oligomers impact the central nervous system to induce peripheral metabolic deregulation. EMBO Mol. Med. 2015, 7, 190–210. [Google Scholar] [CrossRef]
  60. Sprenkle, N.T.; Sims, S.G.; Sánchez, C.L.; Meares, G.P. Endoplasmic reticulum stress and inflammation in the central nervous system. Mol. Neurodegener. 2017, 12, 42. [Google Scholar] [CrossRef] [Green Version]
  61. Cheng, K.C.; Chiang, H.C. XBP1 and PERK Have Distinct Roles in Aβ-Induced Pathology. Mol. Neurobiol. 2018, 55, 7523–7532. [Google Scholar] [CrossRef]
  62. Pereira, C.F.; Santos, A.E.; Moreira, P.I.; Pereira, A.C.; Sousa, F.J.; Cardoso, S.M.; Cruz, M.T. Is Alzheimer’s disease an inflammasomopathy? Ageing Res. Rev. 2019, 56, 100966. [Google Scholar] [CrossRef] [PubMed]
  63. Uddin, M.S.; Tewari, D.; Sharma, G.; Kabir, M.T.; Barreto, G.E.; Bin-Jumah, M.N.; Perveen, A.; Abdel-Daim, M.M.; Ashraf, G.M. Molecular Mechanisms of ER Stress and UPR in the Pathogenesis of Alzheimer’s Disease. Mol. Neurobiol. 2020, 57, 2902–2919. [Google Scholar] [CrossRef]
  64. Ajoolabady, A.; Lindholm, D.; Ren, J.; Pratico, D. ER stress and UPR in Alzheimer’s disease: Mechanisms, pathogenesis, treatments. Cell Death Dis. 2022, 13, 706. [Google Scholar] [CrossRef]
  65. Uddin, M.S.; Yu, W.S.; Lim, L.W. Exploring ER stress response in cellular aging and neuroinflammation in Alzheimer’s disease. Ageing Res. Rev. 2021, 70, 101417. [Google Scholar] [CrossRef]
  66. Bruijn, L.I.; Miller, T.M.; Cleveland, D.W. Unraveling the mechanisms involved in motor neuron degeneration in ALS. Annu. Rev. Neurosci. 2004, 27, 723–749. [Google Scholar] [CrossRef] [Green Version]
  67. Hilton, J.B.; White, A.R.; Crouch, P.J. Metal-deficient SOD1 in amyotrophic lateral sclerosis. J. Mol. Med. 2015, 93, 481–487. [Google Scholar] [CrossRef] [Green Version]
  68. Schweingruber, C.; Hedlund, E. The Cell Autonomous and Non-Cell Autonomous Aspects of Neuronal Vulnerability and Resilience in Amyotrophic Lateral Sclerosis. Biology 2022, 11, 1191. [Google Scholar] [CrossRef]
  69. Leblond, C.S.; Kaneb, H.M.; Dion, P.A.; Rouleau, G.A. Dissection of genetic factors associated with amyotrophic lateral sclerosis. Exp. Neurol. 2014, 262, 91–101. [Google Scholar] [CrossRef] [PubMed]
  70. Van Damme, P.; Robberecht, W.; Van Den Bosch, L. Modelling amyotrophic lateral sclerosis: Progress and possibilities. Dis. Model. Mech. 2017, 10, 537–549. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Tobisawa, S.; Hozumi, Y.; Arawaka, S.; Koyama, S.; Wada, M.; Nagai, M.; Aoki, M.; Itoyama, Y.; Goto, K.; Kato, T. Mutant SOD1 linked to familial amyotrophic lateral sclerosis, but not wild-type SOD1, induces ER stress in COS7 cells and transgenic mice. Biochem. Biophys. Res. Commun. 2003, 303, 496–503. [Google Scholar] [CrossRef]
  72. Wootz, H.; Hansson, I.; Korhonen, L.; Näpänkangas, U.; Lindholm, D. Caspase-12 cleavage and increased oxidative stress during motoneuron degeneration in transgenic mouse model of ALS. Biochem. Biophys. Res. Commun. 2004, 322, 281–286. [Google Scholar] [CrossRef] [PubMed]
  73. Nagata, T.; Ilieva, H.; Murakami, T.; Shiote, M.; Narai, H.; Ohta, Y.; Hayashi, T.; Shoji, M.; Abe, K. Increased ER stress during motor neuron degeneration in a transgenic mouse model of amyotrophic lateral sclerosis. Neurol. Res. 2007, 29, 767–771. [Google Scholar] [CrossRef] [PubMed]
  74. Ilieva, E.V.; Ayala, V.; Jové, M.; Dalfó, E.; Cacabelos, D.; Povedano, M.; Bellmunt, M.J.; Ferrer, I.; Pamplona, R.; Portero-Otín, M. Oxidative and endoplasmic reticulum stress interplay in sporadic amyotrophic lateral sclerosis. Brain 2007, 130 Pt 12, 3111–3123. [Google Scholar] [CrossRef] [PubMed]
  75. Kieran, D.; Woods, I.; Villunger, A.; Strasser, A.; Prehn, J.H. Deletion of the BH3-only protein puma protects motoneurons from ER stress-induced apoptosis and delays motoneuron loss in ALS mice. Proc. Natl. Acad. Sci. USA 2007, 104, 20606–20611. [Google Scholar] [CrossRef] [Green Version]
  76. Oh, Y.K.; Shin, K.S.; Yuan, J.; Kang, S.J. Superoxide dismutase 1 mutants related to amyotrophic lateral sclerosis induce endoplasmic stress in neuro2a cells. J. Neurochem. 2008, 104, 993–1005. [Google Scholar] [CrossRef]
  77. Nishitoh, H.; Kadowaki, H.; Nagai, A.; Maruyama, T.; Yokota, T.; Fukutomi, H.; Noguchi, T.; Matsuzawa, A.; Takeda, K.; Ichijo, H. ALS-linked mutant SOD1 induces ER stress- and ASK1-dependent motor neuron death by targeting Derlin-1. Genes Dev. 2008, 22, 1451–1464. [Google Scholar] [CrossRef] [Green Version]
  78. Suzuki, H.; Kanekura, K.; Levine, T.P.; Kohno, K.; Olkkonen, V.M.; Aiso, S.; Matsuoka, M. ALS-linked P56S-VAPB, an aggregated loss-of-function mutant of VAPB, predisposes motor neurons to ER stress-related death by inducing aggregation of co-expressed wild-type VAPB. J. Neurochem. 2009, 108, 973–985. [Google Scholar] [CrossRef]
  79. Kanekura, K.; Suzuki, H.; Aiso, S.; Matsuoka, M. ER stress and unfolded protein response in amyotrophic lateral sclerosis. Mol. Neurobiol. 2009, 39, 81–89. [Google Scholar] [CrossRef]
  80. Saxena, S.; Cabuy, E.; Caroni, P. A role for motoneuron subtype-selective ER stress in disease manifestations of FALS mice. Nat. Neurosci. 2009, 12, 627–636. [Google Scholar] [CrossRef]
  81. Hetz, C.; Thielen, P.; Matus, S.; Nassif, M.; Court, F.; Kiffin, R.; Martinez, G.; Cuervo, A.M.; Brown, R.H.; Glimcher, L.H. XBP-1 deficiency in the nervous system protects against amyotrophic lateral sclerosis by increasing autophagy. Genes Dev. 2009, 23, 2294–2306. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  82. Matus, S.; Nassif, M.; Glimcher, L.H.; Hetz, C. XBP-1 deficiency in the nervous system reveals a homeostatic switch to activate autophagy. Autophagy 2009, 5, 1226–1228. [Google Scholar] [CrossRef] [Green Version]
  83. Ito, Y.; Yamada, M.; Tanaka, H.; Aida, K.; Tsuruma, K.; Shimazawa, M.; Hozumi, I.; Inuzuka, T.; Takahashi, H.; Hara, H. Involvement of CHOP, an ER-stress apoptotic mediator, in both human sporadic ALS and ALS model mice. Neurobiol. Dis. 2009, 36, 470–476. [Google Scholar] [CrossRef]
  84. Medinas, D.B.; Rozas, P.; Martínez Traub, F.; Woehlbier, U.; Brown, R.H.; Bosco, D.A.; Hetz, C. Endoplasmic reticulum stress leads to accumulation of wild-type SOD1 aggregates associated with sporadic amyotrophic lateral sclerosis. Proc. Natl. Acad. Sci. USA 2018, 115, 8209–8214. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  85. Prusiner, S.B. Prions. Proc. Natl. Acad. Sci. USA 1998, 95, 13363–13383. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  86. Aguzzi, A.; Heppner, F.L. Pathogenesis of prion diseases: A progress report. Cell Death Differ. 2000, 7, 889–902. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  87. Soto, C.; Castilla, J. The controversial protein-only hypothesis of prion propagation. Nat. Med. 2004, 10, S63–S67. [Google Scholar] [CrossRef] [PubMed]
  88. Hetz, C.; Russelakis-Carneiro, M.; Maundrell, K.; Castilla, J.; Soto, C. Caspase-12 and endoplasmic reticulum stress mediate neurotoxicity of pathological prion protein. EMBO J. 2003, 22, 5435–5445. [Google Scholar] [CrossRef] [Green Version]
  89. Hetz, C.; Russelakis-Carneiro, M.; Wälchli, S.; Carboni, S.; Vial-Knecht, E.; Maundrell, K.; Castilla, J.; Soto, C. The disulfide isomerase Grp58 is a protective factor against prion neurotoxicity. J. Neurosci. 2005, 25, 2793–2802. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Rane, N.S.; Kang, S.W.; Chakrabarti, O.; Feigenbaum, L.; Hegde, R.S. Reduced translocation of nascent prion protein during ER stress contributes to neurodegeneration. Dev. Cell 2008, 15, 359–370. [Google Scholar] [CrossRef] [Green Version]
  91. Park, K.W.; Eun Kim, G.; Morales, R.; Moda, F.; Moreno-Gonzalez, I.; Concha-Marambio, L.; Lee, A.S.; Hetz, C.; Soto, C. The Endoplasmic Reticulum Chaperone GRP78/BiP Modulates Prion Propagation in vitro and in vivo. Sci. Rep. 2017, 7, 44723. [Google Scholar] [CrossRef] [Green Version]
  92. Otero, A.; Betancor, M.; Eraña, H.; Fernández Borges, N.; Lucas, J.J.; Badiola, J.J.; Castilla, J.; Bolea, R. Prion-Associated Neurodegeneration Causes Both Endoplasmic Reticulum Stress and Proteasome Impairment in a Murine Model of Spontaneous Disease. Int. J. Mol. Sci. 2021, 22, 465. [Google Scholar] [CrossRef]
  93. Zoghbi, H.Y.; Orr, H.T. Glutamine repeats and neurodegeneration. Annu. Rev. Neurosci. 2000, 23, 217–247. [Google Scholar] [CrossRef] [PubMed]
  94. Lipinski, M.M.; Yuan, J. Mechanisms of cell death in polyglutamine expansion diseases. Curr. Opin. Pharmacol. 2004, 4, 85–90. [Google Scholar] [CrossRef] [PubMed]
  95. Nishitoh, H.; Matsuzawa, A.; Tobiume, K.; Saegusa, K.; Takeda, K.; Inoue, K.; Hori, S.; Kakizuka, A.; Ichijo, H. ASK1 is essential for endoplasmic reticulum stress-induced neuronal cell death triggered by expanded polyglutamine repeats. Genes Dev. 2002, 16, 1345–1355. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  96. Reijonen, S.; Putkonen, N.; Nørremølle, A.; Lindholm, D.; Korhonen, L. Inhibition of endoplasmic reticulum stress counteracts neuronal cell death and protein aggregation caused by N-terminal mutant huntingtin proteins. Exp. Cell Res. 2008, 314, 950–960. [Google Scholar] [CrossRef]
  97. Duennwald, M.L.; Lindquist, S. Impaired ERAD and ER stress are early and specific events in polyglutamine toxicity. Genes Dev. 2008, 22, 3308–3319. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Leitman, J.; Ulrich Hartl, F.; Lederkremer, G.Z. Soluble forms of polyQ-expanded huntingtin rather than large aggregates cause endoplasmic reticulum stress. Nat. Commun. 2013, 4, 2753. [Google Scholar] [CrossRef] [Green Version]
  99. Platt, F.M.; d’Azzo, A.; Davidson, B.L.; Neufeld, E.F.; Tifft, C.J. Lysosomal storage diseases. Nat. Rev. Dis. Primers 2018, 4, 27. [Google Scholar] [CrossRef]
  100. Southwood, C.M.; Garbern, J.; Jiang, W.; Gow, A. The unfolded protein response modulates disease severity in Pelizaeus-Merzbacher disease. Neuron 2002, 36, 585–596. [Google Scholar] [CrossRef] [Green Version]
  101. Pelled, D.; Lloyd-Evans, E.; Riebeling, C.; Jeyakumar, M.; Platt, F.M.; Futerman, A.H. Inhibition of calcium uptake via the sarco/endoplasmic reticulum Ca2+-ATPase in a mouse model of Sandhoff disease and prevention by treatment with N-butyldeoxynojirimycin. J. Biol. Chem. 2003, 278, 29496–29501. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  102. Tessitore, A.; Martin, M.D.; Sano, R.; Ma, Y.; Mann, L.; Ingrassia, A.; Laywell, E.D.; Steindler, D.A.; Hendershot, L.M.; d’Azzo, A. GM1-ganglioside-mediated activation of the unfolded protein response causes neuronal death in a neurodegenerative gangliosidosis. Mol. Cell 2004, 15, 753–766. [Google Scholar] [CrossRef] [PubMed]
  103. Thelen, M.; Damme, M.; Schweizer, M.; Hagel, C.; Wong, A.M.; Cooper, J.D.; Braulke, T.; Galliciotti, G. Disruption of the autophagy-lysosome pathway is involved in neuropathology of the nclf mouse model of neuronal ceroid lipofuscinosis. PLoS ONE 2012, 7, e35493. [Google Scholar] [CrossRef]
  104. Virgolini, M.J.; Feliziani, C.; Cambiasso, M.J.; Lopez, P.H.; Bollo, M. Neurite atrophy and apoptosis mediated by PERK signaling after accumulation of GM2-ganglioside. Biochim. Biophys. Acta Mol. Cell Res. 2019, 1866, 225–239. [Google Scholar] [CrossRef] [PubMed]
  105. Flores-Obando, R.E.; Freidin, M.M.; Hernández, A.I.; Abrams, C.K. Activation of the unfolded protein response by Connexin47 mutations associated with Pelizaeus-Merzbacher-like disease. Mol. Cell. Neurosci. 2022, 120, 103716. [Google Scholar] [CrossRef]
  106. Tamatani, M.; Matsuyama, T.; Yamaguchi, A.; Mitsuda, N.; Tsukamoto, Y.; Taniguchi, M.; Che, Y.H.; Ozawa, K.; Hori, O.; Nishimura, H.; et al. ORP150 protects against hypoxia/ischemia-induced neuronal death. Nat. Med. 2001, 7, 317–323. [Google Scholar] [CrossRef]
  107. Bando, Y.; Katayama, T.; Kasai, K.; Taniguchi, M.; Tamatani, M.; Tohyama, M. GRP94 (94 kDa glucose-regulated protein) suppresses ischemic neuronal cell death against ischemia/reperfusion injury. Eur. J. Neurosci. 2003, 18, 829–840. [Google Scholar] [CrossRef]
  108. Larner, S.F.; Hayes, R.L.; McKinsey, D.M.; Pike, B.R.; Wang, K.K. Increased expression and processing of caspase-12 after traumatic brain injury in rats. J. Neurochem. 2004, 88, 78–90. [Google Scholar] [CrossRef]
  109. Tajiri, S.; Oyadomari, S.; Yano, S.; Morioka, M.; Gotoh, T.; Hamada, J.I.; Ushio, Y.; Mori, M. Ischemia-induced neuronal cell death is mediated by the endoplasmic reticulum stress pathway involving CHOP. Cell Death Differ. 2004, 11, 403–415. [Google Scholar] [CrossRef]
  110. Hayashi, T.; Saito, A.; Okuno, S.; Ferrand-Drake, M.; Dodd, R.L.; Chan, P.H. Damage to the endoplasmic reticulum and activation of apoptotic machinery by oxidative stress in ischemic neurons. J. Cereb. Blood Flow Metab. 2005, 25, 41–53. [Google Scholar] [CrossRef] [Green Version]
  111. Bonnefont-Rousselot, D.; Collin, F.; Jore, D.; Gardès-Albert, M. Reaction mechanism of melatonin oxidation by reactive oxygen species in vitro. J. Pineal Res. 2011, 50, 328–335. [Google Scholar] [CrossRef] [PubMed]
  112. Galano, A.; Tan, D.X.; Reiter, R.J. Melatonin as a natural ally against oxidative stress: A physicochemical examination. J. Pineal Res. 2011, 51, 1–16. [Google Scholar] [CrossRef] [PubMed]
  113. Kim, S.H.; Lee, S.M. Cytoprotective effects of melatonin against necrosis and apoptosis induced by ischemia/reperfusion injury in rat liver. J. Pineal Res. 2008, 44, 165–171. [Google Scholar] [CrossRef] [PubMed]
  114. Muñoz-Casares, F.C.; Padillo, F.J.; Briceño, J.; Collado, J.A.; Muñoz-Castañeda, J.R.; Ortega, R.; Cruz, A.; Túnez, I.; Montilla, P.; Pera, C.; et al. Melatonin reduces apoptosis and necrosis induced by ischemia/reperfusion injury of the pancreas. J. Pineal Res. 2006, 40, 195–203. [Google Scholar] [CrossRef] [PubMed]
  115. Tuñón, M.J.; San-Miguel, B.; Crespo, I.; Laliena, A.; Vallejo, D.; Álvarez, M.; Prieto, J.; González-Gallego, J. Melatonin treatment reduces endoplasmic reticulum stress and modulates the unfolded protein response in rabbits with lethal fulminant hepatitis of viral origin. J Pineal Res. 2013, 55, 221–228. [Google Scholar] [CrossRef] [PubMed]
  116. San-Miguel, B.; Crespo, I.; Vallejo, D.; Álvarez, M.; Prieto, J.; González-Gallego, J.; Tuñón, M.J. Melatonin modulates the autophagic response in acute liver failure induced by the rabbit hemorrhagic disease virus. J. Pineal Res. 2014, 56, 313–321. [Google Scholar] [CrossRef]
  117. Fernández, A.; Ordóñez, R.; Reiter, R.J.; González-Gallego, J.; Mauriz, J.L. Melatonin and endoplasmic reticulum stress: Relation to autophagy and apoptosis. J. Pineal Res. 2015, 59, 292–307. [Google Scholar] [CrossRef]
  118. Motilva, V.; García-Mauriño, S.; Talero, E.; Illanes, M. New paradigms in chronic intestinal inflammation and colon cancer: Role of melatonin. J. Pineal Res. 2011, 51, 44–60. [Google Scholar] [CrossRef] [PubMed]
  119. Naziroğlu, M.; Tokat, S.; Demirci, S. Role of melatonin on electromagnetic radiation-induced oxidative stress and Ca2+ signaling molecular pathways in breast cancer. J. Recept. Signal Transduct. 2012, 32, 290–297. [Google Scholar] [CrossRef]
  120. Hosseinzadeh, A.; Kamrava, S.K.; Joghataei, M.T.; Darabi, R.; Shakeri-Zadeh, A.; Shahriari, M.; Reiter, R.J.; Ghaznavi, H.; Mehrzadi, S. Apoptosis signaling pathways in osteoarthritis and possible protective role of melatonin. J. Pineal Res. 2016, 61, 411–425. [Google Scholar] [CrossRef] [Green Version]
  121. Mortezaee, K. Human hepatocellular carcinoma: Protection by melatonin. J. Cell. Physiol. 2018, 233, 6486–6508. [Google Scholar] [CrossRef] [PubMed]
  122. Yang, Y.; Cheung, H.H.; Zhang, C.; Wu, J.; Chan, W.Y. Melatonin as Potential Targets for Delaying Ovarian Aging. Curr. Drug Targets 2019, 20, 16–28. [Google Scholar] [CrossRef] [PubMed]
  123. Fu, Z.; Jiao, Y.; Wang, J.; Zhang, Y.; Shen, M.; Reiter, R.J.; Xi, Q.; Chen, Y. Cardioprotective Role of Melatonin in Acute Myocardial Infarction. Front. Physiol. 2020, 11, 366. [Google Scholar] [CrossRef]
  124. Huang, K.; Luo, X.; Zhong, Y.; Deng, L.; Feng, J. New insights into the role of melatonin in diabetic cardiomyopathy. Pharmacol. Res. Perspect. 2022, 10, e00904. [Google Scholar] [CrossRef]
  125. Carloni, S.; Albertini, M.C.; Galluzzi, L.; Buonocore, G.; Proietti, F.; Balduini, W. Melatonin reduces endoplasmic reticulum stress and preserves sirtuin 1 expression in neuronal cells of newborn rats after hypoxia-ischemia. J. Pineal Res. 2014, 57, 192–199. [Google Scholar] [CrossRef]
  126. Wongprayoon, P.; Govitrapong, P. Melatonin Protects SH-SY5Y Neuronal Cells Against Methamphetamine-Induced Endoplasmic Reticulum Stress and Apoptotic Cell Death. Neurotox. Res. 2017, 31, 1–10. [Google Scholar] [CrossRef]
  127. Tungkum, W.; Jumnongprakhon, P.; Tocharus, C.; Govitrapong, P.; Tocharus, J. Melatonin suppresses methamphetamine-triggered endoplasmic reticulum stress in C6 cells glioma cell lines. J. Toxicol. Sci. 2017, 42, 63–71. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  128. Feng, D.; Wang, B.; Wang, L.; Abraham, N.; Tao, K.; Huang, L.; Shi, W.; Dong, Y.; Qu, Y. Pre-ischemia melatonin treatment alleviated acute neuronal injury after ischemic stroke by inhibiting endoplasmic reticulum stress-dependent autophagy via PERK and IRE1 signalings. J. Pineal Res. 2017, 62, e12395. [Google Scholar] [CrossRef]
  129. Song, J.; Kim, O.Y. Melatonin Modulates Neuronal Cell Death Induced by Endoplasmic Reticulum Stress under Insulin Resistance Condition. Nutrients 2017, 9, 593. [Google Scholar] [CrossRef] [Green Version]
  130. Xue, F.; Shi, C.; Chen, Q.; Hang, W.; Xia, L.; Wu, Y.; Tao, S.Z.; Zhou, J.; Shi, A.; Chen, J. Melatonin Mediates Protective Effects against Kainic Acid-Induced Neuronal Death through Safeguarding ER Stress and Mitochondrial Disturbance. Front. Mol. Neurosci. 2017, 10, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Shi, C.; Zeng, J.; Li, Z.; Chen, Q.; Hang, W.; Xia, L.; Wu, Y.; Chen, J.; Shi, A. Melatonin Mitigates Kainic Acid-Induced Neuronal Tau Hyperphosphorylation and Memory Deficits through Alleviating ER Stress. Front. Mol. Neurosci. 2018, 11, 5. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  132. Lin, Y.W.; Chen, T.Y.; Hung, C.Y.; Tai, S.H.; Huang, S.Y.; Chang, C.C.; Hung, H.Y.; Lee, E.J. Melatonin protects brain against ischemia/reperfusion injury by attenuating endoplasmic reticulum stress. Int. J. Mol. Med. 2018, 42, 182–192. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  133. Xu, W.; Lu, X.; Zheng, J.; Li, T.; Gao, L.; Lenahan, C.; Shao, A.; Zhang, J.; Yu, J. Melatonin Protects Against Neuronal Apoptosis via Suppression of the ATF6/CHOP Pathway in a Rat Model of Intracerebral Hemorrhage. Front. Neurosci. 2018, 12, 638. [Google Scholar] [CrossRef] [Green Version]
  134. Thangwong, P.; Jearjaroen, P.; Govitrapong, P.; Tocharus, C.; Tocharus, J. Melatonin improves cognitive function by suppressing endoplasmic reticulum stress and promoting synaptic plasticity during chronic cerebral hypoperfusion in rats. Biochem. Pharmacol. 2022, 198, 114980. [Google Scholar] [CrossRef] [PubMed]
  135. Ling, Z.Q.; Tian, Q.; Wang, L.; Fu, Z.Q.; Wang, X.C.; Wang, Q.; Wang, J.Z. Constant illumination induces Alzheimer-like damages with endoplasmic reticulum involvement and the protection of melatonin. J. Alzheimers Dis. 2009, 16, 287–300. [Google Scholar] [CrossRef]
  136. Lee, J.H.; Yoon, Y.M.; Han, Y.S.; Jung, S.K.; Lee, S.H. Melatonin protects mesenchymal stem cells from autophagy-mediated death under ischaemic ER-stress conditions by increasing prion protein expression. Cell Prolif. 2019, 52, e12545. [Google Scholar] [CrossRef] [Green Version]
  137. Sharma, R.; Reiter, R.J.; Ma, Q. Melatonin: A hypothesis regarding its use to treat Wilson disease. Med. Hypotheses 2019, 133, 109408. [Google Scholar] [CrossRef]
  138. Promyo, K.; Iqbal, F.; Chaidee, N.; Chetsawang, B. Aluminum chloride-induced amyloid β accumulation and endoplasmic reticulum stress in rat brain are averted by melatonin. Food Chem. Toxicol. 2020, 146, 111829. [Google Scholar] [CrossRef]
Figure 1. The number of articles related to ER stress among articles on neurodegenerative diseases published in PubMed from 1990 to 23 November 2022.
Figure 1. The number of articles related to ER stress among articles on neurodegenerative diseases published in PubMed from 1990 to 23 November 2022.
Ijms 24 02381 g001
Figure 2. The number of articles related to neurodegenerative diseases and ER stress investigated by PubMed by 23 November 2022.
Figure 2. The number of articles related to neurodegenerative diseases and ER stress investigated by PubMed by 23 November 2022.
Ijms 24 02381 g002
Figure 3. The number of articles related to ER stress in PD published in PubMed from 2000 to 23 November 2022.
Figure 3. The number of articles related to ER stress in PD published in PubMed from 2000 to 23 November 2022.
Ijms 24 02381 g003
Figure 4. The number of articles related to ER stress in AD published in PubMed from 1997 to 23 November 2022.
Figure 4. The number of articles related to ER stress in AD published in PubMed from 1997 to 23 November 2022.
Ijms 24 02381 g004
Figure 5. The number of articles related to ER stress in ALS published in PubMed from 2003 to 23 November 2022.
Figure 5. The number of articles related to ER stress in ALS published in PubMed from 2003 to 23 November 2022.
Ijms 24 02381 g005
Figure 6. The number of articles related to ER stress in TSE published in PubMed from 1999 to 23 November 2022.
Figure 6. The number of articles related to ER stress in TSE published in PubMed from 1999 to 23 November 2022.
Ijms 24 02381 g006
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Yoo, Y.-M.; Joo, S.S. Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress. Int. J. Mol. Sci. 2023, 24, 2381. https://doi.org/10.3390/ijms24032381

AMA Style

Yoo Y-M, Joo SS. Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress. International Journal of Molecular Sciences. 2023; 24(3):2381. https://doi.org/10.3390/ijms24032381

Chicago/Turabian Style

Yoo, Yeong-Min, and Seong Soo Joo. 2023. "Melatonin Can Modulate Neurodegenerative Diseases by Regulating Endoplasmic Reticulum Stress" International Journal of Molecular Sciences 24, no. 3: 2381. https://doi.org/10.3390/ijms24032381

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop