Chonnam Med J.  2019 Jan;55(1):8-19. 10.4068/cmj.2019.55.1.8.

Endoplasmic Reticulum Stress: Implications for Neuropsychiatric Disorders

Affiliations
  • 1Islamic International Medical College, Riphah International University, Rawalpindi, Pakistan. muneerather2@gmail.com
  • 2Government Khawaja Safdar Medical College, Sialkot, Pakistan.

Abstract

The Endoplasmic reticulum (ER), an indispensable sub-cellular component of the eukaryotic cell carries out essential functions, is critical to the survival of the organism. The chaperone proteins and the folding enzymes which are multi-domain ER effectors carry out 3-dimensional conformation of nascent polypeptides and check misfolded protein aggregation, easing the exit of functional proteins from the ER. Diverse conditions, for instance redox imbalance, alterations in ionic calcium levels, and inflammatory signaling can perturb the functioning of the ER, leading to a build-up of unfolded or misfolded proteins in the lumen. This results in ER stress, and aiming to reinstate protein homeostasis, a well conserved reaction called the unfolded protein response (UPR) is elicited. Equally, in protracted cellular stress or inadequate compensatory reaction, UPR pathway leads to cell loss. Dysfunctional ER mechanisms are responsible for neuronal degeneration in numerous human diseases, for instance Alzheimer's, Parkinson's and Huntington's diseases. In addition, mounting proof indicates that ER stress is incriminated in psychiatric diseases like major depressive disorder, bipolar disorder, and schizophrenia. Accumulating evidence suggests that pharmacological agents regulating the working of ER may have a role in diminishing advancing neuronal dysfunction in neuropsychiatric disorders. Here, new findings are examined which link the foremost mechanisms connecting ER stress and cell homeostasis. Furthermore, a supposed new pathogenic model of major neuropsychiatry disorders is provided, with ER stress proposed as the pivotal step in disease development.

Keyword

Endoplasmic Reticulum; Unfolded Protein Response; Proteostasis Deficiencies; Apoptosis; Biological Psychiatry

MeSH Terms

Apoptosis
Biological Psychiatry
Bipolar Disorder
Calcium
Depressive Disorder, Major
Endoplasmic Reticulum Stress*
Endoplasmic Reticulum*
Eukaryotic Cells
Homeostasis
Humans
Neurons
Neuropsychiatry
Oxidation-Reduction
Peptides
Proteostasis Deficiencies
Schizophrenia
Unfolded Protein Response
Calcium
Peptides

Figure

  • FIG. 1 Cellular adaptive response to endoplasmic reticulum (ER) stress. Increase in unfolded proteins can be instigated by physiological as well as disease factors. The latter may include defects in protein folding, calcium changes, redox imbalance and inflammatory signaling, resulting in the unfolded protein response (UPR). The cellular reaction to UPR involves other organelles, such as the mitochondria, and either leads to re-establishing cell homeostasis or relegating them to death. The cascades activated by UPR result in such adaptive reactions like ER associated biogenesis, ER-associated degradation (ERAD) via the ubiquitin-proteasome system and autophagy. When these regulating mechanisms are overwhelmed, stress sensors responsible for UPR commit cells to apoptosis.

  • FIG. 2 Canonical mediators of the unfolded protein response (UPR). A third of all human proteins are folded into 3-dimensional conformations in the endoplasmic reticulum (ER) before they can perform their roles in cells. If a cell is required to make a large number of proteins, or in disease states unfolded proteins may accumulate in the ER. The organism responds to this stress by triggering the UPR which is mediated by three ER stress sensors-the inositol-requiring enzyme 1 (IRE1), the activating transcription factor 6 (ATF6) and protein kinase RNA-like ER kinase (PERK). These stimulate a complex transcriptional cascade with ensuing adaptive responses which restore the folding capabilities of the ER. However, if the ER stress exceeds the capacity of this built-in quality control, cell death is incited through the up-regulation of the pro-apoptotic arm of the UPR pathway to protect the organism from the toxicity of misfolded proteins.

  • FIG. 3 Molecular mechanisms of endoplasmic reticulum stress response. Build-up of unfolded proteins in the ER lumen signals the unfolded protein response. The activated stress sensors —protein kinase RNA-like ER kinase (PERK), inositol-requiring enzyme 1 (IRE1) and activating transcription factor 6 (ATF6) - stimulate diverse cascades seeking to reinstate cell homeostasis or assign it to death. Here, the molecular events that occur in this process are explained in a simplified manner. In brief, IRE1 and ATF6 increase levels of XBP1 which is a diverse transcription factor and leads to increased expression of chaperones and other proteins involved in protein folding in ER. PERK, by virtue of phosphorylating elongation factor 2α (elf2α) puts a hold on translation, enabling ER to recover its protein folding capabilities. However, the expression of ATF4 is also increased which causes activation of CHOP and programmed cell death. The latter is also facilitated by c-Jun N-terminal kinase (JNK), a transcription factor stimulated by activated ATF6. CHOP: CCAAT/enhancer-binding protein homologous protein, ER: endoplasmic reticulum, ERAD: endoplasmic reticulum-associated protein degradation.

  • FIG. 4 A cellular pathogenic model of disease development in major psychiatric disorders. Major psychiatric disorders including mood disorders, psychotic disorders and neurodegenerative disorders are characterized by increased endoplasmic reticulum (ER) stress. ER chaperones act as first line of defense against unfolded or misfolded proteins, but accumulation of non-natively conformed polypeptides in the ER lumen activates other compensatory mechanisms. ER and mitochondria work in close unison through the mitochondria-associated ER membranes, stimulating the mitochondrial unfolded protein response (UPR mt). However, in cellular stress increased generation of reactive oxygen species (ROS) and build up of intracellular calcium ions further exacerbates ER/mt stress. During proteostasis, unfolded/misfolded proteins are degraded via the ubiquitin-proteasome system but when the latter gets saturated, toxic oligomers or larger aggregates accumulate, which can then be got rid of through lysosomal autophagic mechanisms. Since lysosomal dysfunction is present in many neuropsychiatric diseases, accumulated protein aggregates acting as danger associated molecular patterns cause activation of the inflammasome leading to inflammatory-immune signaling and ultimately increased apoptosis of neurons. This cascade is responsible for increased damage to neurons and its further elucidation can result in the development of novel therapeutic agents for otherwise recalcitrant neuropsychiatric diseases.


Reference

1. Fu XL, Gao DS. Endoplasmic reticulum proteins quality control and the unfolded protein response: the regulative mechanism of organisms against stress injuries. Biofactors. 2014; 40:569–585.
Article
2. Jovaisaite V, Mouchiroud L, Auwerx J. The mitochondrial unfolded protein response, a conserved stress response pathway with implications in health and disease. J Exp Biol. 2014; 217:137–143.
Article
3. van Oosten-Hawle P, Morimoto RI. Transcellular chaperone signaling: an organismal strategy for integrated cell stress responses. J Exp Biol. 2014; 217:129–136.
Article
4. Inagi R, Ishimoto Y, Nangaku M. Proteostasis in endoplasmic reticulum--new mechanisms in kidney disease. Nat Rev Nephrol. 2014; 10:369–378.
Article
5. Freeman OJ, Mallucci GR. The UPR and synaptic dysfunction in neurodegeneration. Brain Res. 2016; 1648:530–537.
Article
6. Scheper W, Hoozemans JJ. The unfolded protein response in neurodegenerative diseases: a neuropathological perspective. Acta Neuropathol. 2015; 130:315–331.
Article
7. Zhu G, Lee AS. Role of the unfolded protein response, GRP78 and GRP94 in organ homeostasis. J Cell Physiol. 2015; 230:1413–1420.
Article
8. Jiang D, Niwa M, Koong AC. Targeting the IRE1α-XBP1 branch of the unfolded protein response in human diseases. Semin Cancer Biol. 2015; 33:48–56.
Article
9. Galehdar Z, Swan P, Fuerth B, Callaghan SM, Park DS, Cregan SP. Neuronal apoptosis induced by endoplasmic reticulum stress is regulated by ATF4-CHOP-mediated induction of the Bcl-2 homology 3-only member PUMA. J Neurosci. 2010; 30:16938–16948.
Article
10. Jung J, Michalak M, Agellon LB. Endoplasmic reticulum malfunction in the nervous system. Front Neurosci. 2017; 11:220.
Article
11. Concha NO, Smallwood A, Bonnette W, Totoritis R, Zhang G, Federowicz K, et al. Long-range inhibitor-induced conformational regulation of human IRE1α endoribonuclease activity. Mol Pharmacol. 2015; 88:1011–1023.
Article
12. Yang J, Liu H, Li L, Liu H, Shi W, Yuan X, et al. Structural insights into IRE1 functions in the unfolded protein response. Curr Med Chem. 2016; 23:4706–4716.
Article
13. Trinh MA, Kaphzan H, Wek RC, Pierre P, Cavener DR, Klann E. Brain-specific disruption of the eIF2α kinase PERK decreases ATF4 expression and impairs behavioral flexibility. Cell Rep. 2012; 1:676–688.
Article
14. Lu M, Lawrence DA, Marsters S, Acosta-Alvear D, Kimmig P, Mendez AS, et al. Opposing unfolded-protein-response signals converge on death receptor 5 to control apoptosis. Science. 2014; 345:98–101.
Article
15. Credle JJ, Forcelli PA, Delannoy M, Oaks AW, Permaul E, Berry DL, et al. α-Synuclein-mediated inhibition of ATF6 processing into COPII vesicles disrupts UPR signaling in Parkinson's disease. Neurobiol Dis. 2015; 76:112–125.
Article
16. Ishikawa T, Okada T, Ishikawa-Fujiwara T, Todo T, Kamei Y, Shigenobu S, et al. ATF6α/β-mediated adjustment of ER chaperone levels is essential for development of the notochord in medaka fish. Mol Biol Cell. 2013; 24:1387–1395.
Article
17. Genereux JC, Qu S, Zhou M, Ryno LM, Wang S, Shoulders MD, et al. Unfolded protein response-induced ERdj3 secretion links ER stress to extracellular proteostasis. EMBO J. 2015; 34:4–19.
18. Chen JJ, Genereux JC, Qu S, Hulleman JD, Shoulders MD, Wiseman RL. ATF6 activation reduces the secretion and extracellular aggregation of destabilized variants of an amyloidogenic protein. Chem Biol. 2014; 21:1564–1574.
Article
19. Plate L, Cooley CB, Chen JJ, Paxman RJ, Gallagher CM, Madoux F, et al. Small molecule proteostasis regulators that reprogram the ER to reduce extracellular protein aggregation. Elife. 2016; 5:e15550.
Article
20. Arnould T, Michel S, Renard P. Mitochondria retrograde signaling and the UPR mt: where are we in mammals? Int J Mol Sci. 2015; 16:18224–18251.
Article
21. Schulz AM, Haynes CM. UPR(mt)-mediated cytoprotection and organismal aging. Biochim Biophys Acta. 2015; 1847:1448–1456.
Article
22. Nargund AM, Pellegrino MW, Fiorese CJ, Baker BM, Haynes CM. Mitochondrial import efficiency of ATFS-1 regulates mitochondrial UPR activation. Science. 2012; 337:587–590.
Article
23. Fiorese CJ, Schulz AM, Lin YF, Rosin N, Pellegrino MW, Haynes CM. The transcription factor ATF5 mediates a mammalian mitochondrial UPR. Curr Biol. 2016; 26:2037–2043.
Article
24. Rainbolt TK, Saunders JM, Wiseman RL. YME1L degradation reduces mitochondrial proteolytic capacity during oxidative stress. EMBO Rep. 2015; 16:97–106.
Article
25. Gorman GS, Pfeffer G, Griffin H, Blakely EL, Kurzawa-Akanbi M, Gabriel J, et al. Clonal expansion of secondary mitochondrial DNA deletions associated with spinocerebellar ataxia type 28. JAMA Neurol. 2015; 72:106–111.
Article
26. Milisav I, Šuput D, Ribarič S. Unfolded protein response and macroautophagy in Alzheimer's, Parkinson's and prion diseases. Molecules. 2015; 20:22718–22756.
Article
27. Heneka MT, Carson MJ, El Khoury J, Landreth GE, Brosseron F, Feinstein DL, et al. Neuroinflammation in Alzheimer's disease. Lancet Neurol. 2015; 14:388–405.
Article
28. Duran-Aniotz C, Cornejo VH, Espinoza S, Ardiles ÁO, Medinas DB, Salazar C, et al. IRE1 signaling exacerbates Alzheimer's disease pathogenesis. Acta Neuropathol. 2017; 134:489–506.
Article
29. Gourmaud S, Paquet C, Dumurgier J, Pace C, Bouras C, Gray F, et al. Increased levels of cerebrospinal fluid JNK3 associated with amyloid pathology: links to cognitive decline. J Psychiatry Neurosci. 2015; 40:151–161.
Article
30. Rozpedek W, Markiewicz L, Diehl JA, Pytel D, Majsterek I. Unfolded protein response and PERK kinase as a new therapeutic target in the pathogenesis of Alzheimer's disease. Curr Med Chem. 2015; 22:3169–3184.
Article
31. Wang P, Ding K. Proteoglycans and glycosaminoglycans in misfolded proteins formation in Alzheimer's disease. Protein Pept Lett. 2014; 21:1048–1056.
Article
32. Genereux JC, Wiseman RL. Regulating extracellular proteostasis capacity through the unfolded protein response. Prion. 2015; 9:10–21.
Article
33. Chen JJ, Genereux JC, Wiseman RL. Endoplasmic reticulum quality control and systemic amyloid disease: impacting protein stability from the inside out. IUBMB Life. 2015; 67:404–413.
Article
34. Halliday M, Radford H, Mallucci GR. Prions: generation and spread versus neurotoxicity. J Biol Chem. 2014; 289:19862–19868.
Article
35. Riboldi G, Nizzardo M, Simone C, Falcone M, Bresolin N, Comi GP, et al. ALS genetic modifiers that increase survival of SOD1 mice and are suitable for therapeutic development. Prog Neurobiol. 2011; 95:133–148.
Article
36. Valdés P, Mercado G, Vidal RL, Molina C, Parsons G, Court FA, et al. Control of dopaminergic neuron survival by the unfolded protein response transcription factor XBP1. Proc Natl Acad Sci U S A. 2014; 111:6804–6809.
Article
37. Colla E, Jensen PH, Pletnikova O, Troncoso JC, Glabe C, Lee MK. Accumulation of toxic α-synuclein oligomer within endoplasmic reticulum occurs in α-synucleinopathy in vivo. J Neurosci. 2012; 32:3301–3305.
Article
38. Valenzuela V, Collyer E, Armentano D, Parsons GB, Court FA, Hetz C. Activation of the unfolded protein response enhances motor recovery after spinal cord injury. Cell Death Dis. 2012; 3:e272.
Article
39. Martínez G, Vidal RL, Mardones P, Serrano FG, Ardiles AO, Wirth C, et al. Regulation of memory formation by the transcription factor XBP1. Cell Rep. 2016; 14:1382–1394.
Article
40. Gautam M, Jara JH, Sekerkova G, Yasvoina MV, Martina M, Özdinler PH. Absence of alsin function leads to corticospinal motor neuron vulnerability via novel disease mechanisms. Hum Mol Genet. 2016; 25:1074–1087.
Article
41. Liu EY, Russ J, Wu K, Neal D, Suh E, McNally AG, et al. C9orf72 hypermethylation protects against repeat expansion-associated pathology in ALS/FTD. Acta Neuropathol. 2014; 128:525–541.
Article
42. Carrara M, Sigurdardottir A, Bertolotti A. Decoding the selectivity of eIF2α holophosphatases and PPP1R15A inhibitors. Nat Struct Mol Biol. 2017; 24:708–716.
Article
43. Hyrskyluoto A, Bruelle C, Lundh SH, Do HT, Kivinen J, Rappou E, et al. Ubiquitin-specific protease-14 reduces cellular aggregates and protects against mutant huntingtin-induced cell degeneration: involvement of the proteasome and ER stress-activated kinase IRE1α. Hum Mol Genet. 2014; 23:5928–5939.
Article
44. Ederle H, Dormann D. TDP-43 and FUS en route from the nucleus to the cytoplasm. FEBS Lett. 2017; 591:1489–1507.
Article
45. Anuncibay-Soto B, Pérez-Rodríguez D, Santos-Galdiano M, Font E, Regueiro-Purriños M, Fernández-López A. Post-ischemic salubrinal treatment results in a neuroprotective role in global cerebral ischemia. J Neurochem. 2016; 138:295–306.
Article
46. Liu GL, Wang KY, Guo H, Zhao SJ, Shen Y, Zhao YB. Inositol-requiring protein 1α signaling pathway is activated in the temporal cortex of patients with mesial temporal lobe epilepsy. Neurol Sci. 2013; 34:357–364.
Article
47. Bengesser SA, Fuchs R, Lackner N, Birner A, Reininghaus B, Meier-Allard N, et al. Endoplasmic reticulum stress and bipolar disorder - almost forgotten therapeutic drug targets in the unfolded protein response pathway revisited. CNS Neurol Disord Drug Targets. 2016; 15:403–413.
Article
48. Tsai SY, Pokrass MJ, Klauer NR, De Credico NE, Su TP. Sigma-1 receptor chaperones in neurodegenerative and psychiatric disorders. Expert Opin Ther Targets. 2014; 18:1461–1476.
Article
49. So J, Warsh JJ, Li PP. Impaired endoplasmic reticulum stress response in B-lymphoblasts from patients with bipolar-I disorder. Biol Psychiatry. 2007; 62:141–147.
Article
50. Pfaffenseller B, Wollenhaupt-Aguiar B, Fries GR, Colpo GD, Burque RK, Bristot G, et al. Impaired endoplasmic reticulum stress response in bipolar disorder: cellular evidence of illness progression. Int J Neuropsychopharmacol. 2014; 17:1453–1463.
Article
51. Hayashi A, Kasahara T, Kametani M, Toyota T, Yoshikawa T, Kato T. Aberrant endoplasmic reticulum stress response in lymphoblastoid cells from patients with bipolar disorder. Int J Neuropsychopharmacol. 2009; 12:33–43.
Article
52. Cheng D, Zhang K, Zhen G, Xue Z. The -116C/G polymorphism in XBP1 gene is associated with psychiatric illness in Asian population: a meta-analysis. Am J Med Genet B Neuropsychiatr Genet. 2014; 165B:665–672.
53. Breen MS, White CH, Shekhtman T, Lin K, Looney D, Woelk CH, et al. Lithium-responsive genes and gene networks in bipolar disorder patient-derived lymphoblastoid cell lines. Pharmacogenomics J. 2016; 16:446–453.
Article
54. Kakiuchi C, Ishigaki S, Oslowski CM, Fonseca SG, Kato T, Urano F. Valproate, a mood stabilizer, induces WFS1 expression and modulates its interaction with ER stress protein GRP94. PLoS One. 2009; 4:e4134.
Article
55. Durcan TM, Fon EA. The three ‘P’s of mitophagy: PARKIN, PINK1, and post-translational modifications. Genes Dev. 2015; 29:989–999.
Article
56. Merenlender-Wagner A, Malishkevich A, Shemer Z, Udawela M, Gibbons A, Scarr E, et al. Autophagy has a key role in the pathophysiology of schizophrenia. Mol Psychiatry. 2015; 20:126–132.
Article
57. Toker L, Agam G. Mitochondrial dysfunction in psychiatric morbidity: current evidence and therapeutic prospects. Neuropsychiatr Dis Treat. 2015; 11:2441–2447.
58. Penke B, Bogár F, Fülöp L. Protein folding and misfolding, endoplasmic reticulum stress in neurodegenerative diseases: in trace of novel drug targets. Curr Protein Pept Sci. 2016; 17:169–182.
Article
59. Engin F, Hotamisligil GS. Restoring endoplasmic reticulum function by chemical chaperones: an emerging therapeutic approach for metabolic diseases. Diabetes Obes Metab. 2010; 12:Suppl 2. 108–115.
Article
60. Ricobaraza A, Cuadrado-Tejedor M, Marco S, Pérez-Otaño I, García-Osta A. Phenylbutyrate rescues dendritic spine loss associated with memory deficits in a mouse model of Alzheimer disease. Hippocampus. 2012; 22:1040–1050.
Article
61. Nunes AF, Amaral JD, Lo AC, Fonseca MB, Viana RJ, Callaerts-Vegh Z, et al. TUDCA, a bile acid, attenuates amyloid precursor protein processing and amyloid-β deposition in APP/PS1 mice. Mol Neurobiol. 2012; 45:440–454.
Article
62. Wei H, Kim SJ, Zhang Z, Tsai PC, Wisniewski KE, Mukherjee AB. ER and oxidative stresses are common mediators of apoptosis in both neurodegenerative and non-neurodegenerative lysosomal storage disorders and are alleviated by chemical chaperones. Hum Mol Genet. 2008; 17:469–477.
Article
63. Peng J, Liang G, Inan S, Wu Z, Joseph DJ, Meng Q, et al. Dantrolene ameliorates cognitive decline and neuropathology in Alzheimer triple transgenic mice. Neurosci Lett. 2012; 516:274–279.
Article
64. Oseki KT, Monteforte PT, Pereira GJ, Hirata H, Ureshino RP, Bincoletto C, et al. Apoptosis induced by Aβ25-35 peptide is Ca(2+)-IP3 signaling-dependent in murine astrocytes. Eur J Neurosci. 2014; 40:2471–2478.
Article
65. Gong T, Wang Q, Lin Z, Chen ML, Sun GZ. Endoplasmic reticulum (ER) stress inhibitor salubrinal protects against ceramide-induced SH-SY5Y cell death. Biochem Biophys Res Commun. 2012; 427:461–465.
Article
66. Patel S, Sharma D, Kalia K, Tiwari V. Crosstalk between endoplasmic reticulum stress and oxidative stress in schizophrenia: the dawn of new therapeutic approaches. Neurosci Biobehav Rev. 2017; 83:589–603.
Article
67. Inokuchi Y, Nakajima Y, Shimazawa M, Kurita T, Kubo M, Saito A, et al. Effect of an inducer of BiP, a molecular chaperone, on endoplasmic reticulum (ER) stress-induced retinal cell death. Invest Ophthalmol Vis Sci. 2009; 50:334–344.
Article
68. Shen YE, Wang Y, Yu GC, Liu C, Zhang ZY, Zhang LM. Effects of edaravone on amyloid-β precursor protein processing in SY5Y-APP695 cells. Neurotox Res. 2013; 24:139–147.
Article
69. Fan J, Long H, Li Y, Liu Y, Zhou W, Li Q, et al. Edaravone protects against glutamate-induced PERK/EIF2α/ATF4 integrated stress response and activation of caspase-12. Brain Res. 2013; 1519:1–8.
Article
70. Park GB, Kim YS, Lee HK, Song H, Cho DH, Lee WJ, et al. Endoplasmic reticulum stress-mediated apoptosis of EBV-transformed B cells by cross-linking of CD70 is dependent upon generation of reactive oxygen species and activation of p38 MAPK and JNK pathway. J Immunol. 2010; 185:7274–7284.
Article
Full Text Links
  • CMJ
Actions
Cited
CITED
export Copy
Close
Share
  • Twitter
  • Facebook
Similar articles
Copyright © 2024 by Korean Association of Medical Journal Editors. All rights reserved.     E-mail: koreamed@kamje.or.kr