1
|
Cai Y, Wang X, Xiang Y, Wang Z, Long Q, Zeng C. Codonopsis pilosula polysaccharides alleviate neuronal apoptosis induced by endoplasmic reticulum stress-activated PERK-ATF4-CHOP signaling in APP/PS1 mice. J Alzheimers Dis 2025:13872877251339484. [PMID: 40325880 DOI: 10.1177/13872877251339484] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/07/2025]
Abstract
BackgroundCodonopsis polysaccharides (CPPs) shows neuroprotective potential in Alzheimer's disease (AD) and may reduce neuronal apoptosis by modulating endoplasmic reticulum stress (ERS).ObjectiveTo investigate the protective mechanisms of CPPs against neuronal apoptosis in APP/PS1 mice, focusing on the ERS response and the PERK-ATF4-CHOP signaling pathway.MethodsAPP/PS1 mice were orally administered CPPs at different doses. Their learning and memory abilities were evaluated using the Morris water maze (MWM). The integrity of hippocampal neurons and senile plaque deposition were assessed using histopathology, immunohistochemistry, and immunofluorescence. The expression of amyloid-β (Aβ) plaques secretase protein, ERS markers, and apoptosis-related proteins was assessed using western blot analyses. The affinity of the PERK-ATF4-CHOP pathway and CPPs was analyzed and assessed using molecular docking.ResultsMWM testing revealed that CPPs improved the learning and memory abilities of APP/PS1 mice. Histopathological examination confirmed that CPPs reduced hippocampal neuronal apoptosis. Immunohistochemistry and immunofluorescence analysis showed that CPPs decreased Aβ protein expression and ERS. Western blot analysis further confirmed that CPPs reduced the expression of proteins related to Aβ synthesis; downregulated the expression of glucose-regulated protein 78 (GRP78), PERK, ATF4, CHOP, and Bcl-2 associated X protein (Bax), while upregulating the expression of B-cell lymphoma 2 (Bcl-2).ConclusionsThis study demonstrates that CPPs exert neuroprotective effects by targeting the PERK-ATF4-CHOP signaling pathway and alleviating ERS, suggesting a novel approach and potential therapeutic agent for AD treatment.
Collapse
Affiliation(s)
- Yuanqin Cai
- Key Laboratory of Yunnan Province for Research and Prevention of Neuropsychiatric Disorders with Traditional Chinese Medicine, Yunnan Provincial Department of Education, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Health Science Center, Hubei Minzu University, Enshi, Hubei, China
- Key Laboratory of Yunnan Province for Chronic Disease Prevention and Treatment with Integrated Traditional Chinese and Western Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Xi Wang
- Health Science Center, Hubei Minzu University, Enshi, Hubei, China
| | - Yang Xiang
- Key Laboratory of Yunnan Province for Research and Prevention of Neuropsychiatric Disorders with Traditional Chinese Medicine, Yunnan Provincial Department of Education, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Key Laboratory of Yunnan Province for Chronic Disease Prevention and Treatment with Integrated Traditional Chinese and Western Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Basic Medical College, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Zhenning Wang
- Key Laboratory of Yunnan Province for Research and Prevention of Neuropsychiatric Disorders with Traditional Chinese Medicine, Yunnan Provincial Department of Education, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Key Laboratory of Yunnan Province for Chronic Disease Prevention and Treatment with Integrated Traditional Chinese and Western Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Basic Medical College, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| | - Qinghua Long
- Health Science Center, Hubei Minzu University, Enshi, Hubei, China
| | - Chuhua Zeng
- Key Laboratory of Yunnan Province for Research and Prevention of Neuropsychiatric Disorders with Traditional Chinese Medicine, Yunnan Provincial Department of Education, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Key Laboratory of Yunnan Province for Chronic Disease Prevention and Treatment with Integrated Traditional Chinese and Western Medicine, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
- Basic Medical College, Yunnan University of Traditional Chinese Medicine, Kunming, Yunnan, China
| |
Collapse
|
2
|
Bravo-Jimenez MA, Sharma S, Karimi-Abdolrezaee S. The integrated stress response in neurodegenerative diseases. Mol Neurodegener 2025; 20:20. [PMID: 39972469 PMCID: PMC11837473 DOI: 10.1186/s13024-025-00811-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2024] [Accepted: 02/07/2025] [Indexed: 02/21/2025] Open
Abstract
The integrated stress response (ISR) is a conserved network in eukaryotic cells that mediates adaptive responses to diverse stressors. The ISR pathway ensures cell survival and homeostasis by regulating protein synthesis in response to internal or external stresses. In recent years, the ISR has emerged as an important regulator of the central nervous system (CNS) development, homeostasis and pathology. Dysregulation of ISR signaling has been linked to several neurodegenerative diseases. Intriguingly, while acute ISR provide neuroprotection through the activation of cell survival mechanisms, prolonged ISR can promote neurodegeneration through protein misfolding, oxidative stress, and mitochondrial dysfunction. Understanding the molecular mechanisms and dynamics of the ISR in neurodegenerative diseases aids in the development of effective therapies. Here, we will provide a timely review on the cellular and molecular mechanisms of the ISR in neurodegenerative diseases. We will highlight the current knowledge on the dual role that ISR plays as a protective or disease worsening pathway and will discuss recent advances on the therapeutic approaches that have been developed to target ISR activity in neurodegenerative diseases.
Collapse
Affiliation(s)
- Maria Astrid Bravo-Jimenez
- Department of Physiology and Pathophysiology, Multiple Sclerosis Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Children Hospital Research Institute of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Shivangi Sharma
- Department of Physiology and Pathophysiology, Multiple Sclerosis Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Children Hospital Research Institute of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada
| | - Soheila Karimi-Abdolrezaee
- Department of Physiology and Pathophysiology, Multiple Sclerosis Research Centre, Rady Faculty of Health Sciences, University of Manitoba, Children Hospital Research Institute of Manitoba, 745 Bannatyne Avenue, Winnipeg, MB, R3E 0J9, Canada.
| |
Collapse
|
3
|
Sharma K, Rai P, Tapadia MG. Impaired insulin signaling and diet-induced type 3 diabetes pathophysiology increase amyloid β expression in the Drosophila model of Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2025; 1872:119875. [PMID: 39515664 DOI: 10.1016/j.bbamcr.2024.119875] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/19/2024] [Revised: 10/25/2024] [Accepted: 10/28/2024] [Indexed: 11/16/2024]
Abstract
Compelling evidence has strongly linked unregulated sugar levels to developing Alzheimer's disease, suggesting Alzheimer's to be 'diabetes of the brain or 'type 3 diabetes. Insulin resistance contributes to the pathogenesis of Alzheimer's disease due to uncontrolled and unchecked blood glucose, though the interrelatedness between Alzheimer's disease and type 2 diabetes is debatable. Here we describe the consequences of inducing type 3 diabetes by feeding Drosophila on a high sucrose diet, which effectively mimics the pathophysiology of diabetes. A high sucrose diet increases glycogen and lipid accumulation. Inducing type 3 diabetes worsened neurodegeneration and accelerated disease progression in Drosophila expressing the Alzheimer's Familial Arctic mutation. High sucrose milieu also negatively affected locomotor ability and reduced the lifespan in the Alzheimer's disease model of Drosophila. The results showed that creating diabetic conditions by using insulin receptor (InR) knockdown in the eyes of Drosophila led to a degenerative phenotype, indicating a genetic interaction between the insulin signaling pathway and Alzheimer's disease. The expression of PERK reflects disruption in the endoplasmic reticulum homeostasis due to amyloid-β (Aβ) under a high sucrose diet. These observations demonstrated an association between type 3 diabetes and Alzheimer's disease, and that a high sucrose environment has a degenerating effect on Alzheimer's disease condition.
Collapse
Affiliation(s)
- Khushboo Sharma
- Cytogenetics lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India
| | - Pooja Rai
- Cytogenetics lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India; Department of Molecular, Cell and Cancer Biology, UMass Chan Medical School, MA 01605, USA
| | - Madhu G Tapadia
- Cytogenetics lab, Department of Zoology, Institute of Science, Banaras Hindu University, Varanasi, India.
| |
Collapse
|
4
|
Sanjari-Pour M, Faridi N, Wang P, Bathaie SZ. Protective effect of saffron carotenoids against amyloid beta-induced neurotoxicity in differentiated PC12 cells via the unfolded protein response and autophagy. Phytother Res 2024; 38:4923-4939. [PMID: 36794286 DOI: 10.1002/ptr.7773] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 01/12/2023] [Accepted: 01/29/2023] [Indexed: 02/17/2023]
Abstract
The preventive effect of saffron against Alzheimer's disease (AD) has been reported. Herein, we studied the effect of Cro and Crt, saffron carotenoids, on the cellular model of AD. The MTT assay, flow cytometry, and elevated p-JNK, p-Bcl-2, and c-PARP indicated the AβOs-induced apoptosis in differentiated PC12 cells. Then, the protective effects of Cro/Crt on dPC12 cells against AβOs were investigated in preventive and therapeutic modalities. Starvation was used as a positive control. RT-PCR and Western blot results revealed the reduced eIF2α phosphorylation and increased spliced-XBP1, Beclin1, LC3II, and p62, which indicate the AβOs-induced autophagic flux defect, autophagosome accumulation, and apoptosis. Cro and Crt inhibited the JNK-Bcl-2-Beclin1 pathway. They altered Beclin1 and LC3II and decreased p62 expressions, leading cells to survival. Cro and Crt altered the autophagic flux by different mechanisms. So, Cro increased the rate of autophagosome degradation more than Crt, while Crt increased the rate of autophagosome formation more than Cro. The application of 4μ8C and chloroquine as the inhibitors of XBP1 and autophagy, respectively, confirmed these results. So, augmentation of the survival branches of UPR and autophagy is involved and may serve as an effective strategy to prevent the progression of AβOs toxicity.
Collapse
Affiliation(s)
- Mariam Sanjari-Pour
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Nassim Faridi
- Department of Clinical Biochemistry, Faculty of Medical Sciences, Tarbiat Modares University, Tehran, Iran
| | - Ping Wang
- College of Pharmaceutical Sciences, Zhejiang University of Technology, Hangzhou, China
| | - S Zahra Bathaie
- Institute for Natural Products and Medicinal Plants, Tarbiat Modares University, Tehran, Iran
- UCLA-DOE Institute, University of California Los Angeles (UCLA), Los Angeles, California, USA
| |
Collapse
|
5
|
da Fonseca FG, Serufo ÂV, Leão TL, Lourenço KL. Viral Infections and Their Ability to Modulate Endoplasmic Reticulum Stress Response Pathways. Viruses 2024; 16:1555. [PMID: 39459886 PMCID: PMC11512299 DOI: 10.3390/v16101555] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 09/25/2024] [Accepted: 09/29/2024] [Indexed: 10/28/2024] Open
Abstract
In eukaryotic cells, the endoplasmic reticulum is particularly important in post-translational modification of proteins before they are released extracellularly or sent to another endomembrane system. The correct three-dimensional folding of most proteins occurs in the ER lumen, which has an oxidative environment that is essential for the formation of disulfide bridges, which are important in maintaining protein structure. The ER is a versatile organelle that ensures the correct structure of proteins and is essential in the synthesis of lipids and sterols, in addition to offering support in the maintenance of intracellular calcium. Consequently, the cells needed to respond to demands caused by physiological conditions and pathological disturbances in the organelle homeostasis, leading to proper functioning of the cell or even programmed cell death. Disturbances to the ER function trigger a response to the accumulation of unfolded or misfolded proteins, known as the unfolded protein response. Such disturbances include abiotic stress, pharmacological agents, and intracellular pathogens, such as viruses. When misfolded proteins accumulate in the ER, they can undergo ubiquitination and proteasomal degradation through components of the ER-associated degradation system. Once a prolonged activity of the UPR pathway occurs, indicating that homeostasis cannot be reestablished, components of this pathway induce cell death by apoptosis. Here, we discuss how viruses have evolved ways to counteract UPR responses to maximize replication. This evolutionary viral ability is important to understand cell pathology and should be taken into account when designing therapeutic interventions and vaccines.
Collapse
Affiliation(s)
- Flávio Guimarães da Fonseca
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.G.d.F.); (T.L.L.)
| | - Ângela Vieira Serufo
- CT Terapias Avançacadas e Inovadoras, CTERAPIAS, Universidade Federal de Minas Gerais, Belo Horizonte 31270-901, MG, Brazil;
| | - Thiago Lima Leão
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.G.d.F.); (T.L.L.)
| | - Karine Lima Lourenço
- Laboratório de Virologia Básica e Aplicada, Departamento de Microbiologia, Instituto de Ciências Biológicas, Universidade Federal de Minas Gerais, Av. Pres. Antônio Carlos, 6627, Pampulha, Belo Horizonte 31270-901, MG, Brazil; (F.G.d.F.); (T.L.L.)
| |
Collapse
|
6
|
Li JJ, Xin N, Yang C, Tavizon LA, Hong R, Park J, Moore TI, Tharyan RG, Antebi A, Kim HE. Unveiling the Intercompartmental Signaling Axis: Mitochondrial to ER Stress Response (MERSR) and its Impact on Proteostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2023.09.07.556674. [PMID: 38187690 PMCID: PMC10769184 DOI: 10.1101/2023.09.07.556674] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/09/2024]
Abstract
Maintaining protein homeostasis is essential for cellular health. Our previous research uncovered a cross-compartmental Mitochondrial to Cytosolic Stress Response, activated by the perturbation of mitochondrial proteostasis, which ultimately results in the improvement of proteostasis in the cytosol. Here, we found that this signaling axis also influences the unfolded protein response of the endoplasmic reticulum (UPR ER ), suggesting the presence of a Mitochondria to ER Stress Response (MERSR). During MERSR, the IRE1 branch of UPR ER is inhibited, introducing a previously unknown regulatory component of MCSR. Moreover, proteostasis is enhanced through the upregulation of the PERK-eIF2α signaling pathway, increasing phosphorylation of eIF2α and improving the ER's ability to handle proteostasis. MERSR activation in both polyglutamine and amyloid-beta peptide-expressing C. elegans disease models also led to improvement in both aggregate burden and overall disease outcome. These findings shed light on the coordination between the mitochondria and the ER in maintaining cellular proteostasis and provide further evidence for the importance of intercompartmental signaling.
Collapse
|
7
|
Wójcik P, Jastrzębski MK, Zięba A, Matosiuk D, Kaczor AA. Caspases in Alzheimer's Disease: Mechanism of Activation, Role, and Potential Treatment. Mol Neurobiol 2024; 61:4834-4853. [PMID: 38135855 PMCID: PMC11236938 DOI: 10.1007/s12035-023-03847-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 11/29/2023] [Indexed: 12/24/2023]
Abstract
With the aging of the population, treatment of conditions emerging in old age, such as neurodegenerative disorders, has become a major medical challenge. Of these, Alzheimer's disease, leading to cognitive dysfunction, is of particular interest. Neuronal loss plays an important role in the pathophysiology of this condition, and over the years, a great effort has been made to determine the role of various factors in this process. Unfortunately, until now, the exact pathomechanism of this condition remains unknown. However, the most popular theories associate AD with abnormalities in the Tau and β-amyloid (Aβ) proteins, which lead to their deposition and result in neuronal death. Neurons, like all cells, die in a variety of ways, among which pyroptosis, apoptosis, and necroptosis are associated with the activation of various caspases. It is worth mentioning that Tau and Aβ proteins are considered to be one of the caspase activators, leading to cell death. Moreover, the protease activity of caspases influences both of the previously mentioned proteins, Tau and Aβ, converting them into more toxic derivatives. Due to the variety of ways caspases impact the development of AD, drugs targeting caspases could potentially be useful in the treatment of this condition. Therefore, there is a constant need to search for novel caspase inhibitors and evaluate them in preclinical and clinical trials.
Collapse
Affiliation(s)
- Piotr Wójcik
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland.
| | - Michał K Jastrzębski
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Agata Zięba
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Dariusz Matosiuk
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland
| | - Agnieszka A Kaczor
- Department of Synthesis and Chemical Technology of Pharmaceutical Substances with Computer Modeling Laboratory, Faculty of Pharmacy, Medical University of Lublin, 4A Chodzki St., 20093, Lublin, Poland.
- School of Pharmacy, University of Eastern Finland, Yliopistonranta 1, P.O. Box 1627, 70211, Kuopio, Finland.
| |
Collapse
|
8
|
Thananthirige KPM, Chitranshi N, Basavarajappa D, Rajput R, Abbasi M, Palanivel V, Gupta VB, Paulo JA, Koronyo-Hamaoui M, Mirzaei M, Graham SL, Gupta V. Tau modulation through AAV9 therapy augments Akt/Erk survival signalling in glaucoma mitigating the retinal degenerative phenotype. Acta Neuropathol Commun 2024; 12:89. [PMID: 38845058 PMCID: PMC11158005 DOI: 10.1186/s40478-024-01804-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2023] [Accepted: 05/27/2024] [Indexed: 06/09/2024] Open
Abstract
The microtubule-associated protein Tau is a key player in various neurodegenerative conditions, including Alzheimer's disease (AD) and Tauopathies, where its hyperphosphorylation disrupts neuronal microtubular lattice stability. Glaucoma, a neurodegenerative disorder affecting the retina, leads to irreversible vision loss by damaging retinal ganglion cells and the optic nerve, often associated with increased intraocular pressure. Prior studies have indicated Tau expression and phosphorylation alterations in the retina in both AD and glaucoma, yet the causative or downstream nature of Tau protein changes in these pathologies remains unclear. This study investigates the impact of Tau protein modulation on retinal neurons under normal and experimental glaucoma conditions. Employing AAV9-mediated gene therapy for Tau overexpression and knockdown, both manipulations were found to adversely affect retinal structural and functional measures as well as neuroprotective Akt/Erk survival signalling in healthy conditions. In the experimental glaucoma model, Tau overexpression intensified inner retinal degeneration, while Tau silencing provided significant protection against these degenerative changes. These findings underscore the critical role of endogenous Tau protein levels in preserving retinal integrity and emphasize the therapeutic potential of targeting Tau in glaucoma pathology.
Collapse
Affiliation(s)
| | - Nitin Chitranshi
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Devaraj Basavarajappa
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Rashi Rajput
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Mojdeh Abbasi
- Division of Ophthalmology, Department of Biomedical and Clinical Sciences, Linköping University, 58183, Linköping, Sweden
| | - Viswanthram Palanivel
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Veer Bala Gupta
- School of Medicine, Deakin University, Melbourne, VIC, 3220, Australia
| | - Joao A Paulo
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA
| | - Maya Koronyo-Hamaoui
- Department of Neurosurgery, Cedars-Sinai Medical Center, Maxine Dunitz Neurosurgical Research Institute, 127 S. San Vicente Blvd., Los Angeles, CA, 90048, USA
- Division of Applied Cell Biology and Physiology, Departments of Neurology and Biomedical Sciences, Cedars-Sinai Medical Center, 127 S. San Vicente Blvd., Los Angeles, CA, USA
| | - Mehdi Mirzaei
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Stuart L Graham
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia
| | - Vivek Gupta
- Faculty of Medicine, Health and Human Sciences, Macquarie Medical School, Macquarie University, Sydney, NSW, 2109, Australia.
| |
Collapse
|
9
|
Martin SP, Leeman-Markowski BA. Proposed mechanisms of tau: relationships to traumatic brain injury, Alzheimer's disease, and epilepsy. Front Neurol 2024; 14:1287545. [PMID: 38249745 PMCID: PMC10797726 DOI: 10.3389/fneur.2023.1287545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2023] [Accepted: 11/30/2023] [Indexed: 01/23/2024] Open
Abstract
Traumatic brain injury (TBI), Alzheimer's disease (AD), and epilepsy share proposed mechanisms of injury, including neuronal excitotoxicity, cascade signaling, and activation of protein biomarkers such as tau. Although tau is typically present intracellularly, in tauopathies, phosphorylated (p-) and hyper-phosphorylated (hp-) tau are released extracellularly, the latter leading to decreased neuronal stability and neurofibrillary tangles (NFTs). Tau cleavage at particular sites increases susceptibility to hyper-phosphorylation, NFT formation, and eventual cell death. The relationship between tau and inflammation, however, is unknown. In this review, we present evidence for an imbalanced endoplasmic reticulum (ER) stress response and inflammatory signaling pathways resulting in atypical p-tau, hp-tau and NFT formation. Further, we propose tau as a biomarker for neuronal injury severity in TBI, AD, and epilepsy. We present a hypothesis of tau phosphorylation as an initial acute neuroprotective response to seizures/TBI. However, if the underlying seizure pathology or TBI recurrence is not effectively treated, and the pathway becomes chronically activated, we propose a "tipping point" hypothesis that identifies a transition of tau phosphorylation from neuroprotective to injurious. We outline the role of amyloid beta (Aβ) as a "last ditch effort" to revert the cell to programmed death signaling, that, when fails, transitions the mechanism from injurious to neurodegenerative. Lastly, we discuss targets along these pathways for therapeutic intervention in AD, TBI, and epilepsy.
Collapse
Affiliation(s)
- Samantha P. Martin
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- New York University Grossman School of Medicine, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| | - Beth A. Leeman-Markowski
- Comprehensive Epilepsy Center, New York University Langone Health, New York, NY, United States
- Department of Neurology, New York University Langone Health, New York, NY, United States
- VA New York Harbor Healthcare System, New York, NY, United States
| |
Collapse
|
10
|
Hicks D, Giresh K, Wrischnik LA, Weiser DC. The PPP1R15 Family of eIF2-alpha Phosphatase Targeting Subunits (GADD34 and CReP). Int J Mol Sci 2023; 24:17321. [PMID: 38139150 PMCID: PMC10743859 DOI: 10.3390/ijms242417321] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2023] [Revised: 12/01/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023] Open
Abstract
The vertebrate PPP1R15 family consists of the proteins GADD34 (growth arrest and DNA damage-inducible protein 34, the product of the PPP1R15A gene) and CReP (constitutive repressor of eIF2α phosphorylation, the product of the PPP1R15B gene), both of which function as targeting/regulatory subunits for protein phosphatase 1 (PP1) by regulating subcellular localization, modulating substrate specificity and assembling complexes with target proteins. The primary cellular function of these proteins is to facilitate the dephosphorylation of eukaryotic initiation factor 2-alpha (eIF2α) by PP1 during cell stress. In this review, we will provide a comprehensive overview of the cellular function, biochemistry and pharmacology of GADD34 and CReP, starting with a brief introduction of eIF2α phosphorylation via the integrated protein response (ISR). We discuss the roles GADD34 and CReP play as feedback inhibitors of the unfolded protein response (UPR) and highlight the critical function they serve as inhibitors of the PERK-dependent branch, which is particularly important since it can mediate cell survival or cell death, depending on how long the stressful stimuli lasts, and GADD34 and CReP play key roles in fine-tuning this cellular decision. We briefly discuss the roles of GADD34 and CReP homologs in model systems and then focus on what we have learned about their function from knockout mice and human patients, followed by a brief review of several diseases in which GADD34 and CReP have been implicated, including cancer, diabetes and especially neurodegenerative disease. Because of the potential importance of GADD34 and CReP in aspects of human health and disease, we will discuss several pharmacological inhibitors of GADD34 and/or CReP that show promise as treatments and the controversies as to their mechanism of action. This review will finish with a discussion of the biochemical properties of GADD34 and CReP, their regulation and the additional interacting partners that may provide insight into the roles these proteins may play in other cellular pathways. We will conclude with a brief outline of critical areas for future study.
Collapse
Affiliation(s)
- Danielle Hicks
- Department of Science, Mathematics and Engineering, Modesto Junior College, Modesto, CA 95350, USA
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Krithika Giresh
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Lisa A. Wrischnik
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| | - Douglas C. Weiser
- Department of Biological Sciences, University of the Pacific, Stockton, CA 95211, USA
| |
Collapse
|
11
|
Pinky, Neha, Salman M, Kumar P, Khan MA, Jamal A, Parvez S. Age-related pathophysiological alterations in molecular stress markers and key modulators of hypoxia. Ageing Res Rev 2023; 90:102022. [PMID: 37490963 DOI: 10.1016/j.arr.2023.102022] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2023] [Revised: 06/30/2023] [Accepted: 07/21/2023] [Indexed: 07/27/2023]
Abstract
Alzheimer's disease (AD) is characterized by an adverse cellular environment and pathological alterations in distinct brain regions. The development is triggered or facilitated by a condition such as hypoxia or ischemia, or inflammation and is associated with disruptions of fundamental cellular functions, including metabolic and ion homeostasis. Increasing evidence suggests that hypoxia may affect many pathological aspects of AD, including oxidative stress, mitochondrial dysfunction, ER stress, amyloidogenic processing of APP, and Aβ accumulation, which may collectively result in neurodegeneration. Further investigation into the relationship between hypoxia and AD may provide an avenue for the effective preservation and pharmacological treatment of this neurodegenerative disease. This review summarizes the effects of normoxia and hypoxia on AD pathogenesis and discusses the underlying mechanisms. Regulation of HIF-1α and the role of its key players, including P53, VEGF, and GLUT1, are also discussed.
Collapse
Affiliation(s)
- Pinky
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Neha
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Mohd Salman
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Pratika Kumar
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| | - Mohammad Ahmed Khan
- Department of Pharmacology, School of Pharmaceutical Education and Research, Jamia Hamdard, New Delhi, India.
| | - Azfar Jamal
- Department of Biology, College of Science, Al-Zulfi-, Majmaah University, Al-Majmaah 11952, Saudi Arabia; Health and Basic Science Research Centre, Majmaah University, Al-Majmaah 11952, Saudi Arabia.
| | - Suhel Parvez
- Department of Toxicology, School of Chemical and Life Sciences, Jamia Hamdard, New Delhi 110062, India.
| |
Collapse
|
12
|
Waigi EW, Webb RC, Moss MA, Uline MJ, McCarthy CG, Wenceslau CF. Soluble and insoluble protein aggregates, endoplasmic reticulum stress, and vascular dysfunction in Alzheimer's disease and cardiovascular diseases. GeroScience 2023; 45:1411-1438. [PMID: 36823398 PMCID: PMC10400528 DOI: 10.1007/s11357-023-00748-y] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2022] [Accepted: 01/28/2023] [Indexed: 02/25/2023] Open
Abstract
Dementia refers to a particular group of symptoms characterized by difficulties with memory, language, problem-solving, and other thinking skills that affect a person's ability to perform everyday activities. Alzheimer's disease (AD) is the most common form of dementia, affecting about 6.2 million Americans aged 65 years and older. Likewise, cardiovascular diseases (CVDs) are a major cause of disability and premature death, impacting 126.9 million adults in the USA, a number that increases with age. Consequently, CVDs and cardiovascular risk factors are associated with an increased risk of AD and cognitive impairment. They share important age-related cardiometabolic and lifestyle risk factors, that make them among the leading causes of death. Additionally, there are several premises and hypotheses about the mechanisms underlying the association between AD and CVD. Although AD and CVD may be considered deleterious to health, the study of their combination constitutes a clinical challenge, and investigations to understand the mechanistic pathways for the cause-effect and/or shared pathology between these two disease constellations remains an active area of research. AD pathology is propagated by the amyloid β (Aβ) peptides. These peptides give rise to small, toxic, and soluble Aβ oligomers (SPOs) that are nonfibrillar, and it is their levels that show a robust correlation with the extent of cognitive impairment. This review will elucidate the interplay between the effects of accumulating SPOs in AD and CVDs, the resulting ER stress response, and their role in vascular dysfunction. We will also address the potential underlying mechanisms, including the possibility that SPOs are among the causes of vascular injury in CVD associated with cognitive decline. By revealing common mechanistic underpinnings of AD and CVD, we hope that novel experimental therapeutics can be designed to reduce the burden of these devastating diseases. Graphical abstract Alzheimer's disease (AD) pathology leads to the release of Aβ peptides, and their accumulation in the peripheral organs has varying effects on various components of the cardiovascular system including endoplasmic reticulum (ER) stress and vascular damage. Image created with BioRender.com.
Collapse
Affiliation(s)
- Emily W Waigi
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
| | - R Clinton Webb
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Melissa A Moss
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Mark J Uline
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
- Department of Chemical Engineering, University of South Carolina, Columbia, SC, USA
| | - Cameron G McCarthy
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA
| | - Camilla Ferreira Wenceslau
- Cardiovascular Translational Research Cententer (CTRC), Department of Cell Biology and Anatomy, University of South Carolina School of Medicine, Columbia, SC, USA.
- Biomedical Engineering Program, Univeristy of South Carolina, Columbia, SC, USA.
| |
Collapse
|
13
|
Park J, Gong JH, Chen Y, Nghiem THT, Chandrawanshi S, Hwang E, Yang CH, Kim BS, Park JW, Ryter SW, Ahn B, Joe Y, Chung HT, Yu R. Activation of ROS-PERK-TFEB by Filbertone Ameliorates Neurodegenerative Diseases via Enhancing the Autophagy-Lysosomal Pathway. J Nutr Biochem 2023; 118:109325. [PMID: 36958418 DOI: 10.1016/j.jnutbio.2023.109325] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 03/16/2023] [Accepted: 03/17/2023] [Indexed: 03/25/2023]
Abstract
The molecular mechanisms underlying the pathogenesis of neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease (PD), and Huntington's disease remain enigmatic, resulting in an unmet need for therapeutics development. Here, we suggest that filbertone, a key flavor compound found in the fruits of hazel trees of the genus Corylus, can ameliorate PD via lowering the abundance of aggregated α-synuclein. We previously reported that inhibition of hypothalamic inflammation by filbertone is mediated by suppression of nuclear factor kappa-B (NF-κB). Here, we report that filbertone activates PERK through mitochondrial ROS (mtROS) production, resulting in the increased nuclear translocation of transcription factor-EB (TFEB) in SH-SY5Y human neuroblastoma cells. TFEB activation by filbertone promotes the autophagy-lysosomal pathway (ALP), which in turn alleviates the accumulation of α-synuclein. We also demonstrate that filbertone prevented the loss of dopaminergic neurons in the substantia nigra and striatum of mice on high-fat diet (HFD). Filbertone treatment also reduced HFD-induced α-synuclein accumulation through upregulation of the ALP pathway. In addition, filbertone improved behavioral abnormalities (i.e., latency time to fall and decrease of running distance) in the MPTP-induced PD murine model. In conclusion, filbertone may show promise as a potential therapeutic for neurodegenerative disease.
Collapse
Affiliation(s)
- Jeongmin Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jeong Heon Gong
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Yubing Chen
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Thu-Hang Thi Nghiem
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Sonam Chandrawanshi
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Eunyeong Hwang
- College of Korean Medicine, Daegu Haany University, Daegu 42158, Korea
| | - Chae Ha Yang
- College of Korean Medicine, Daegu Haany University, Daegu 42158, Korea
| | - Byung-Sam Kim
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Jeong Woo Park
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | | | - Byungyong Ahn
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Yeonsoo Joe
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea
| | - Hun Taeg Chung
- Department of Biological Sciences, University of Ulsan, Ulsan, 44610, Republic of Korea.
| | - Rina Yu
- Department of Food Science and Nutrition, University of Ulsan, Ulsan, 44610, Republic of Korea.
| |
Collapse
|
14
|
Vx-809, a CFTR Corrector, Acts through a General Mechanism of Protein Folding and on the Inflammatory Process. Int J Mol Sci 2023; 24:ijms24044252. [PMID: 36835664 PMCID: PMC9965627 DOI: 10.3390/ijms24044252] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2023] [Revised: 02/08/2023] [Accepted: 02/17/2023] [Indexed: 02/25/2023] Open
Abstract
Correct protein folding is the basis of cellular well-being; thus, accumulation of misfolded proteins within the endoplasmic reticulum (ER) leads to an imbalance of homeostasis that causes stress to the ER. Various studies have shown that protein misfolding is a significant factor in the etiology of many human diseases, including cancer, diabetes, and cystic fibrosis. Misfolded protein accumulation in the ER triggers a sophisticated signal transduction pathway, the unfolded protein response (UPR), which is controlled by three proteins, resident in ER: IRE1α, PERK, and ATF6. Briefly, when ER stress is irreversible, IRE1α induces the activation of pro-inflammatory proteins; PERK phosphorylates eIF2α which induces ATF4 transcription, while ATF6 activates genes encoding ER chaperones. Reticular stress causes an alteration of the calcium homeostasis, which is released from the ER and taken up by the mitochondria, leading to an increase in the oxygen radical species production, and consequently, to oxidative stress. Accumulation of intracellular calcium, in combination with lethal ROS levels, has been associated with an increase of pro-inflammatory protein expression and the initiation of the inflammatory process. Lumacaftor (Vx-809) is a common corrector used in cystic fibrosis treatment which enhances the folding of mutated F508del-CFTR, one of the most prevalent impaired proteins underlying the disease, promoting a higher localization of the mutant protein on the cell membrane. Here, we demonstrate that this drug reduces the ER stress and, consequently, the inflammation that is caused by such events. Thus, this molecule is a promising drug to treat several pathologies that present an etiopathogenesis due to the accumulation of protein aggregates that lead to chronic reticular stress.
Collapse
|
15
|
Hamsici S, Gunay G, Acar H. Controllable membrane damage by tunable peptide aggregation with albumin. AIChE J 2022; 68:e17893. [PMID: 36816052 PMCID: PMC9937546 DOI: 10.1002/aic.17893] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2022] [Accepted: 08/24/2022] [Indexed: 11/09/2022]
Abstract
Aggregation of otherwise soluble proteins into amyloid structures is a hallmark of many disorders, such as Alzheimer's and Parkinson's disease. There is an increasing evidence that the small aggregations, instead of ordered fibrillar aggregates, are the main structures causing toxicity. However, the studies on the small aggregation phase are limited due to the variety of structures and the complexity of the physiological environment. Here, we showed an engineered co-assembling oppositely charged amyloid-like peptide pair ([II]) as a simple tool to establish methodologies to study the mechanism and kinetics of aggregation and relate its aggregation to toxicity. The toxicity mechanism of [II] is through cell membrane damage and stress, shown with YAP and eIF2α, as in the amyloid protein-initiated diseases. Albumin is demonstrated as an extrinsic and physiologically relevant molecule in controlling the aggregation lag time and toxicity of [II]. This study represents a molecular engineering strategy to create simplistic molecular tools for establishing methodologies to study the aggregation process and kinetics of amyloid-like proteins in various conditions. Understanding the nature of protein aggregation kinetics and linking them to their biological functions through engineered peptides paves the way for future designs and drug development applications.
Collapse
Affiliation(s)
- Seren Hamsici
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| | - Gokhan Gunay
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
| | - Handan Acar
- Stephenson School of Biomedical Engineering, University of Oklahoma, Norman, Oklahoma, USA
- Stephenson Cancer Center, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma, USA
| |
Collapse
|
16
|
Zhao J, Hai S, Chen J, Ma L, Rahman SU, Zhao C, Feng S, Li Y, Wu J, Wang X. Zearalenone Induces Apoptosis in Porcine Endometrial Stromal Cells through JNK Signaling Pathway Based on Endoplasmic Reticulum Stress. Toxins (Basel) 2022; 14:toxins14110758. [PMID: 36356008 PMCID: PMC9694026 DOI: 10.3390/toxins14110758] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2022] [Revised: 10/28/2022] [Accepted: 10/29/2022] [Indexed: 11/06/2022] Open
Abstract
Zearalenone (ZEA) is an estrogen-like mycotoxin characterized mainly by reproductive toxicity, to which pigs are particularly sensitive. The aim of this study was to investigate the molecular mechanism of ZEA-induced apoptosis in porcine endometrial stromal cells (ESCs) by activating the JNK signaling pathway through endoplasmic reticulum stress (ERS). In this study, ESCs were exposed to ZEA, with the ERS inhibitor sodium 4-Phenylbutyrate (4-PBA) as a reference. The results showed that ZEA could damage cell structures, induce endoplasmic reticulum swelling and fragmentation, and decreased the ratio of live cells to dead cells significantly. In addition, ZEA could increase reactive oxygen species and Ca2+ levels; upregulate the expression of GRP78, CHOP, PERK, ASK1 and JNK; activate JNK phosphorylation and its high expression in the nucleus; upregulate the expression Caspase 3 and Caspase 9; and increase the Bax/Bcl-2 ratio, resulting in increased apoptosis. After 3 h of 4-PBA-pretreatment, ZEA was added for mixed culture, which showed that the inhibition of ERS could reduce the cytotoxicity of ZEA toward ESCs. Compared with the ZEA group, ERS inhibition increased cell viability; downregulated the expression of GRP78, CHOP, PERK, ASK1 and JNK; and decreased the nuclear level of p-JNK. The Bax/Bcl-2 ratio and the expression of Caspase 3 and Caspase 9 were downregulated, significantly alleviating apoptosis. These results demonstrate that ZEA can alter the morphology of ESCs, destroy their ultrastructure, and activate the JNK signaling via the ERS pathway, leading to apoptosis.
Collapse
Affiliation(s)
- Jie Zhao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Sirao Hai
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Jiawen Chen
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Li Ma
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Sajid Ur Rahman
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Department of Food Science and Engineering, School of Agriculture and Biology, Shanghai Jiao Tong University, Shanghai 200240, China
| | - Chang Zhao
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Shibin Feng
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
| | - Yu Li
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
| | - Jinjie Wu
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
| | - Xichun Wang
- College of Animal Science and Technology, Anhui Agricultural University, Hefei 230036, China
- Anhui Province Engineering Laboratory for Animal Food Quality and Bio-Safety, Hefei 230036, China
- Correspondence:
| |
Collapse
|
17
|
ER stress and UPR in Alzheimer's disease: mechanisms, pathogenesis, treatments. Cell Death Dis 2022; 13:706. [PMID: 35970828 PMCID: PMC9378716 DOI: 10.1038/s41419-022-05153-5] [Citation(s) in RCA: 122] [Impact Index Per Article: 40.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2022] [Revised: 07/26/2022] [Accepted: 07/29/2022] [Indexed: 01/21/2023]
Abstract
Alzheimer's disease (AD) is a devastating neurodegenerative disorder characterized by gradual loss of memory and cognitive function, which constitutes a heavy burden on the healthcare system globally. Current therapeutics to interfere with the underlying disease process in AD is still under development. Although many efforts have centered on the toxic forms of Aβ to effectively tackle AD, considering the unsatisfactory results so far it is vital to examine other targets and therapeutic approaches as well. The endoplasmic reticulum (ER) stress refers to the build-up of unfolded or misfolded proteins within the ER, thus, perturbing the ER and cellular homeostasis. Emerging evidence indicates that ER stress contributes to the onset and development of AD. A thorough elucidation of ER stress machinery in AD pathology may help to open up new therapeutic avenues in the management of this devastating condition to relieve the cognitive dementia symptoms. Herein, we aim at deciphering the unique role of ER stress in AD pathogenesis, reviewing key findings, and existing controversy in an attempt to summarize plausible therapeutic interventions in the management of AD pathophysiology.
Collapse
|
18
|
Samluk L, Ostapczuk P, Dziembowska M. Long-term mitochondrial stress induces early steps of Tau aggregation by increasing reactive oxygen species levels and affecting cellular proteostasis. Mol Biol Cell 2022; 33:ar67. [PMID: 35446108 PMCID: PMC9635289 DOI: 10.1091/mbc.e21-11-0553] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Accumulating evidence indicates that mitochondrial dysfunction is involved in the pathogenesis of neurodegenerative diseases. Both of these conditions are often associated with an increase in protein aggregation. However, still unknown are the specific defects of mitochondrial biology that play a critical role in the development of Alzheimer’s disease, in which Tau protein aggregates are observed in the brains of some patients. Here, we report that long-term mitochondrial stress triggered Tau dimerization, which is the first step of protein aggregation. Mitochondrial dysfunction was induced in HEK293T cells that received prolonged treatment with rotenone and in HEK293T cells with the knockout of NDUFA11 protein. To monitor changes in Tau protein aggregation, we took advantage of the bimolecular fluorescence complementation assay using HEK293T cells that were transfected with plasmids that encoded Tau. Inhibition of the ISR with ISRIB induced Tau dimerization, whereas ISR activation with salubrinal, guanabenz, and sephin1 partially reversed this process. Cells that were treated with ROS scavengers, N-acetyl-l-cysteine or MitoQ, significantly reduced the amount of ROS and Tau dimerization, indicating the involvement of oxidative stress in Tau aggregation. Our results indicate that long-term mitochondrial stress may induce early steps of Tau protein aggregation by affecting oxidative balance and cellular proteostasis.
Collapse
Affiliation(s)
- Lukasz Samluk
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland
| | - Piotr Ostapczuk
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland
| | - Magdalena Dziembowska
- Centre of New Technologies, University of Warsaw, S. Banacha 2c, 02-097 Warsaw, Poland
| |
Collapse
|
19
|
Chaudhry ZL, Gamal M, Ferhati I, Warda M, Ahmed BY. ER Stress in COVID-19 and Parkinson’s Disease: In Vitro and In Silico Evidences. Brain Sci 2022; 12:brainsci12040507. [PMID: 35448038 PMCID: PMC9025812 DOI: 10.3390/brainsci12040507] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 03/21/2022] [Accepted: 04/12/2022] [Indexed: 01/08/2023] Open
Abstract
The outbreak of COVID-19 caused by severe acute respiratory syndrome coronavirus 2 (SARS-CoV-2) signifies a serious worldwide concern to public health. Both transcriptome and proteome of SARS-CoV-2-infected cells synergize the progression of infection in host, which may exacerbate symptoms and/or progression of other chronic diseases such as Parkinson’s disease (PD). Oxidative stress is a well-known cause of endoplasmic reticulum (ER) stress observed in both SARS-CoV-2 and PD. In the current study, we aimed to explore the influence of PKR-like ER kinase (PERK) stress pathway under SARS-CoV-2-mediated infection and in human cell model of PD. Furthermore, we investigated whether they are interconnected and if the ER stress inhibitors could inhibit cell death and provide cellular protection. To achieve this aim, we have incorporated in silico analysis obtained from gene set enrichment analysis (GSEA), a literature review and laboratory data. The neurotoxin, 6-hydroxy dopamine (6OHDA), was used to mimic the biochemical and neuropathological characteristics of PD by inducing oxidative stress in dopamine-containing neurons differentiated from ReNVM cell line (dDCNs). Furthermore, we explored if ER stress influences activation of caspases-2, -4 and -8 in SARS-CoV-2 and in stressed dDCNs. Our laboratory data using Western blot, immunocytochemistry and 3-(4,5-dimethylthiazolyl-2)-2,5-diphenyltetrazolium bromide (MTT) analyses indicated that 6OHDA-induced toxicity triggered activation of caspases-2, -4 and -8 in dDCNs. Under SARS-CoV-2 infection of different cell types, GSEA revealed cell-specific sensitivities to oxidative and ER stresses. Cardiomyocytes and type II alveolar epithelial-like cells were more vulnerable to oxidative stress than neural cells. On the other side, only cardiomyocytes activated the unfolded protein response, however, the PERK pathway was operative in both cardiomyocytes and neural cells. In addition, caspase-4 activation by a SARS-CoV-2 was observed via in silico analyses. These results demonstrate that the ER stress pathway under oxidative stress in SARS-CoV-2 and PD are interconnected using diverse pathways. Furthermore, our results using the ER stress inhibitor and caspase specific inhibitors provided cellular protection suggesting that the use of specific inhibitors can provide effective therapeutic approaches for the treatment of COVID-19 and PD.
Collapse
Affiliation(s)
- Zahara L. Chaudhry
- Institute of Biomedical & Environmental Science and Technology, School of Life Sciences, Faculty of Creative Arts, Technologies & Science, University Square, University of Bedfordshire, Luton LU1 3JU, UK; (Z.L.C.); (I.F.)
| | - Mahmoud Gamal
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; (M.G.); (M.W.)
| | - Ingrid Ferhati
- Institute of Biomedical & Environmental Science and Technology, School of Life Sciences, Faculty of Creative Arts, Technologies & Science, University Square, University of Bedfordshire, Luton LU1 3JU, UK; (Z.L.C.); (I.F.)
| | - Mohamad Warda
- Department of Biochemistry and Molecular Biology, Faculty of Veterinary Medicine, Cairo University, Giza 12211, Egypt; (M.G.); (M.W.)
| | - Bushra Y. Ahmed
- Institute of Biomedical & Environmental Science and Technology, School of Life Sciences, Faculty of Creative Arts, Technologies & Science, University Square, University of Bedfordshire, Luton LU1 3JU, UK; (Z.L.C.); (I.F.)
- Correspondence:
| |
Collapse
|
20
|
Wodrich APK, Scott AW, Shukla AK, Harris BT, Giniger E. The Unfolded Protein Responses in Health, Aging, and Neurodegeneration: Recent Advances and Future Considerations. Front Mol Neurosci 2022; 15:831116. [PMID: 35283733 PMCID: PMC8914544 DOI: 10.3389/fnmol.2022.831116] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2021] [Accepted: 01/26/2022] [Indexed: 12/11/2022] Open
Abstract
Aging and age-related neurodegeneration are both associated with the accumulation of unfolded and abnormally folded proteins, highlighting the importance of protein homeostasis (termed proteostasis) in maintaining organismal health. To this end, two cellular compartments with essential protein folding functions, the endoplasmic reticulum (ER) and the mitochondria, are equipped with unique protein stress responses, known as the ER unfolded protein response (UPR ER ) and the mitochondrial UPR (UPR mt ), respectively. These organellar UPRs play roles in shaping the cellular responses to proteostatic stress that occurs in aging and age-related neurodegeneration. The loss of adaptive UPR ER and UPR mt signaling potency with age contributes to a feed-forward cycle of increasing protein stress and cellular dysfunction. Likewise, UPR ER and UPR mt signaling is often altered in age-related neurodegenerative diseases; however, whether these changes counteract or contribute to the disease pathology appears to be context dependent. Intriguingly, altering organellar UPR signaling in animal models can reduce the pathological consequences of aging and neurodegeneration which has prompted clinical investigations of UPR signaling modulators as therapeutics. Here, we review the physiology of both the UPR ER and the UPR mt , discuss how UPR ER and UPR mt signaling changes in the context of aging and neurodegeneration, and highlight therapeutic strategies targeting the UPR ER and UPR mt that may improve human health.
Collapse
Affiliation(s)
- Andrew P. K. Wodrich
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
- Interdisciplinary Program in Neuroscience, Georgetown University, Washington, DC, United States
- College of Medicine, University of Kentucky, Lexington, KY, United States
| | - Andrew W. Scott
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Arvind Kumar Shukla
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| | - Brent T. Harris
- Department of Pathology, Georgetown University, Washington, DC, United States
- Department of Neurology, Georgetown University, Washington, DC, United States
| | - Edward Giniger
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD, United States
| |
Collapse
|
21
|
Liu Y, Ding R, Xu Z, Xue Y, Zhang D, Zhang Y, Li W, Li X. Roles and Mechanisms of the Protein Quality Control System in Alzheimer's Disease. Int J Mol Sci 2021; 23:345. [PMID: 35008771 PMCID: PMC8745298 DOI: 10.3390/ijms23010345] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/26/2021] [Accepted: 12/27/2021] [Indexed: 12/11/2022] Open
Abstract
Alzheimer's disease (AD) is characterized by the deposition of senile plaques (SPs) and the formation of neurofibrillary tangles (NTFs), as well as neuronal dysfunctions in the brain, but in fact, patients have shown a sustained disease progression for at least 10 to 15 years before these pathologic biomarkers can be detected. Consequently, as the most common chronic neurological disease in the elderly, the challenge of AD treatment is that it is short of effective biomarkers for early diagnosis. The protein quality control system is a collection of cellular pathways that can recognize damaged proteins and thereby modulate their turnover. Abundant evidence indicates that the accumulation of abnormal proteins in AD is closely related to the dysfunction of the protein quality control system. In particular, it is the synthesis, degradation, and removal of essential biological components that have already changed in the early stage of AD, which further encourages us to pay more attention to the protein quality control system. The review mainly focuses on the endoplasmic reticulum system (ERS), autophagy-lysosome system (ALS) and the ubiquitin-proteasome system (UPS), and deeply discusses the relationship between the protein quality control system and the abnormal proteins of AD, which can not only help us to understand how and why the complex regulatory system becomes malfunctional during AD progression, but also provide more novel therapeutic strategies to prevent the development of AD.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | - Xing Li
- Department of Nutrition and Food Hygiene, College of Public Health, Zhengzhou University, Zhengzhou 450001, China; (Y.L.); (R.D.); (Z.X.); (Y.X.); (D.Z.); (Y.Z.); (W.L.)
| |
Collapse
|
22
|
Zhang M, Wang Y, Wong RMS, Yung KKL, Li R. Fine particulate matter induces endoplasmic reticulum stress-mediated apoptosis in human SH-SY5Y cells. Neurotoxicology 2021; 88:187-195. [PMID: 34813867 DOI: 10.1016/j.neuro.2021.11.012] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 11/10/2021] [Accepted: 11/18/2021] [Indexed: 02/08/2023]
Abstract
Exposure to ambient fine particulate matter (PM2.5) may contribute to brain injury, however, the molecular mechanisms have not yet been fully described. In this study, the human SH-SY5Y cells were treated with PM2.5 with different concentrations (0, 25, 100, and 250 μg/mL) for 24 h to investigate the cell apoptosis mediated by endoplasmic reticulum (ER) stress. The ratio of apoptosis, Ca2+ level, biomarkers of ER stress and apoptosis were determined. The results revealed that PM2.5 triggered the increase of apoptosis ratio and cellular Ca2+ levels. Compared with control, the expression of GRP78 and phosphorylation of IER1α and p38 were enhanced significantly in the cells under the conditions of PM2.5 exposure for activating ER stress signals. Besides, the key genes (CHOP/DR5/Caspase8/Caspase12) in ER stress-induced apoptosis signals were up-regulated after the PM2.5 treatment compared to the control. The results suggested PM2.5 induced apoptosis in SH-SY5Y cells by the stimulation of ER stress, which may be the potential mechanism of neurological diseases incurred by PM2.5.
Collapse
Affiliation(s)
- Mei Zhang
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China
| | - Ying Wang
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China
| | - Ricky M S Wong
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China; Department of Chemistry, Hong Kong Baptist University, Hong Kong, China
| | - Ken Kin Lam Yung
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China; Department of Biology, Hong Kong Baptist University, Hong Kong, China; Golden Meditech Center for NeuroRegeneration Sciences, Hong Kong Baptist University, Hong Kong, China.
| | - Ruijin Li
- Institute of Environmental Science, Shanxi University, Taiyuan, PR China.
| |
Collapse
|
23
|
Wu L, He S, Ye W, Shen J, Zhao K, Zhang Y, Zhang R, Wei J, Cao S, Chen K, Le R, Xi C, Kou X, Zhao Y, Wang H, Kang L, Gao S. Surf4 facilitates reprogramming by activating the cellular response to endoplasmic reticulum stress. Cell Prolif 2021; 54:e13133. [PMID: 34585448 PMCID: PMC8560622 DOI: 10.1111/cpr.13133] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 09/13/2021] [Accepted: 09/15/2021] [Indexed: 12/21/2022] Open
Abstract
OBJECTIVES Maternal factors that are enriched in oocytes have attracted great interest as possible key factors in somatic cell reprogramming. We found that surfeit locus protein 4 (Surf4), a maternal factor, can facilitate the generation of induced pluripotent stem cells (iPSCs) previously, but the mechanism remains elusive. MATERIALS AND METHODS In this study, we investigated the function and mechanism of Surf4 in somatic cell reprogramming using a secondary reprogramming system. Alkaline phosphatase (AP) staining, qPCR and immunofluorescence (IF) staining of expression of related markers were used to evaluate efficiency of iPSCs derived from mouse embryonic fibroblasts. Embryoid body and teratoma formation assays were performed to evaluate the differentiation ability of the iPSC lines. RNA-seq, qPCR and western blot analysis were applied to validate the downstream targets of Surf4. RESULTS Surf4 can significantly facilitate the generation of iPSCs in a proliferation-independent manner. When co-expressed with Oct4, Sox2, Klf4 and c-Myc (OSKM), Surf4 can activate the response to endoplasmic reticulum (ER) stress at the early stage of reprogramming. We further demonstrated that Hspa5, a major ER chaperone, and the active spliced form of Xbp1 (sXbp1), a major mediator of ER stress, can mimic the effects of Surf4 on somatic cell reprogramming. Concordantly, blocking the unfolded protein response compromises the effect of Surf4 on reprogramming. CONCLUSIONS Surf4 promotes somatic cell reprogramming by activating the response to ER stress.
Collapse
Affiliation(s)
- Li Wu
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shengxiang He
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Anhui Toneker Biotechnology Co., Ltd., Jinzhai, China
| | - Wen Ye
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Jiacheng Shen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Kun Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanping Zhang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Ran Zhang
- Anhui Toneker Biotechnology Co., Ltd., Jinzhai, China
| | - Junhao Wei
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Shuyuan Cao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Kang Chen
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Rongrong Le
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Chenxiang Xi
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Xiaochen Kou
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Yanhong Zhao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Hong Wang
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| | - Lan Kang
- Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Shanghai Key Laboratory of Signaling and Disease Research, School of Life Sciences and Technology, Institute for Regenerative Medicine, Shanghai East Hospital, Tongji University, Shanghai, China
| | - Shaorong Gao
- Shanghai Key Laboratory of Maternal Fetal Medicine, Clinical and Translational Research Center of Shanghai First Maternity and Infant Hospital, School of Life Sciences and Technology, Tongji University, Shanghai, China.,Frontier Science Center for Stem Cell Research, School of Life Sciences and Technology, Tongji University, Shanghai, China
| |
Collapse
|
24
|
Tan ZX, Dong F, Wu LY, Feng YS, Zhang F. The Beneficial Role of Exercise on Treating Alzheimer's Disease by Inhibiting β-Amyloid Peptide. Mol Neurobiol 2021; 58:5890-5906. [PMID: 34415486 DOI: 10.1007/s12035-021-02514-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 08/03/2021] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is associated with a very large burden on global healthcare systems. Thus, it is imperative to find effective treatments of the disease. One feature of AD is the accumulation of neurotoxic β-amyloid peptide (Aβ). Aβ induces multiple pathological processes that are deleterious to nerve cells. Despite the development of medications that target the reduction of Aβ to treat AD, none has proven to be effective to date. Non-pharmacological interventions, such as physical exercise, are also being studied. The benefits of exercise on AD are widely recognized. Experimental and clinical studies have been performed to verify the role that exercise plays in reducing Aβ deposition to alleviate AD. This paper reviewed the various mechanisms involved in the exercise-induced reduction of Aβ, including the regulation of amyloid precursor protein cleaved proteases, the glymphatic system, brain-blood transport proteins, degrading enzymes and autophagy, which is beneficial to promote exercise therapy as a means of prevention and treatment of AD and indicates that exercise may provide new therapeutic targets for the treatment of AD.
Collapse
Affiliation(s)
- Zi-Xuan Tan
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Fang Dong
- Department of Clinical Laboratory Medicine, The Third Hospital of Hebei Medical University, Shijiazhuang, 050051, People's Republic of China
| | - Lin-Yu Wu
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Ya-Shuo Feng
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China
| | - Feng Zhang
- Department of Rehabilitation Medicine, The Third Hospital of Hebei Medical University, No. 139 Ziqiang Road, Shijiazhuang, Hebei, 050051, People's Republic of China. .,Hebei Key Laboratory of Critical Disease Mechanism and Intervention, Shijiazhuang, 050051, People's Republic of China.
| |
Collapse
|
25
|
Qu X, Wu S, Gao J, Qin Z, Zhou Z, Liu J. Weighted gene co expression network analysis (WGCNA) with key pathways and hub-genes related to micro RNAs in ischemic stroke. IET Syst Biol 2021; 15:93-100. [PMID: 33880887 PMCID: PMC8675812 DOI: 10.1049/syb2.12016] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 02/22/2021] [Accepted: 03/08/2021] [Indexed: 11/19/2022] Open
Abstract
Ischemic stroke (IS) is one of the major causes of death and disability worldwide. However, the specific mechanism of gene interplay and the biological function in IS are not clear. Therefore, more research into IS is necessary. Dataset GSE110993 including 20 ischemic stroke (IS) and 20 control specimens are used to establish both groups and the raw RNA‐seq data were analysed. Weighted gene co‐expression network analysis (WGCNA) was used to screen the key micro‐RNA modules. The centrality of key genes were determined by module membership (mm) and gene significance (GS). The key pathways were identified by enrichment analysis with Kyoto Protocol Gene and Genome Encyclopedia (KEGG), and the key genes were validated by protein‐protein interactions network. Result: Upon investigation, 1185 up‐ and down‐regulated genes were gathered and distributed into three modules in response to their degree of correlation to clinical traits of IS, among which the turquoise module show a trait‐correlation of 0.77. The top 140 genes were further identified by GS and MM. KEGG analysis showed two pathways may evolve in the progress of IS. Discussion: CXCL12 and EIF2a may be important biomarkers for the accurate diagnosis and treatment in IS.
Collapse
Affiliation(s)
- Xiang Qu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| | - Shuang Wu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| | - Jinggui Gao
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| | - Zhenxiu Qin
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| | - Zhenhua Zhou
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| | - Jingli Liu
- Department of Neurology, The First Affiliated Hospital of Guangxi Medical University, Shuangyong Road, Nanning, Guangxi, China
| |
Collapse
|
26
|
Abstract
Unfolded protein response (UPR) is an evolutionarily conserved pathway triggered during perturbation of endoplasmic reticulum (ER) homeostasis in response to the accumulation of unfolded/misfolded proteins under various stress conditions like viral infection, diseased states etc. It is an adaptive signalling cascade with the main purpose of relieving the stress from the ER, which may otherwise lead to the initiation of cell death via apoptosis. ER stress if prolonged, contribute to the aetiology of various diseases like cancer, type II diabetes, neurodegenerative diseases, viral infections etc. Understanding the role of UPR in disease progression will help design pharmacological drugs targeting the sensors of signalling cascade acting as potential therapeutic agents against various diseases. The current review aims at highlighting the relevance of different pathways of UPR in disease progression and control, including the available pharmaceutical interventions responsible for ameliorating diseased state via modulating UPR pathways.
Collapse
|
27
|
Endoplasmic Reticulum Stress Is Involved in Glucocorticoid-Induced Apoptosis in PC12 Cells. ACTA ACUST UNITED AC 2021; 2021:5565671. [PMID: 33628710 PMCID: PMC7895572 DOI: 10.1155/2021/5565671] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2021] [Revised: 01/28/2021] [Accepted: 02/03/2021] [Indexed: 02/05/2023]
Abstract
Objective The present study selected PC12 cells to construct a neuronal injury model induced by glucocorticoids (GC) in vitro, aiming to explore whether the endoplasmic reticulum stress (ERS) PKR-like endoplasmic reticulum kinase (PERK)-activating transcription factor 4 (ATF4)-C/EBP-homologous protein (CHOP) and inositol requirement 1 (IRE1)-apoptosis signal regulating kinase 1 (ASK1)-C-Jun amino-terminal kinase (JNK) signaling pathways are associated with the neuronal injury process induced by GC and provide morphological evidence. Methods Cell models with different doses and different durations of GC exposure were established. The viability of PC12 cells was detected by the CCK-8 assay, and the apoptosis rate of PC12 cells was detected by the flow cytometry assay. The expression of microtubule-associated protein 2 (Map2); glucocorticoids receptor (GR); cellular oncogene fos (C-fos); and ERS-related proteins, glucose-regulated protein 78 (GRP78), p-PERK, p-IRE1, ATF4, ASK1, JNK, and CHOP, was observed by immunofluorescence staining. Results The results of immunofluorescence staining showed that PC12 cells abundantly expressed Map2 and GR. The CCK-8 assay revealed that high-concentration GC exposure significantly inhibited the cell viability of PC12 cells. The flow cytometry assay indicated that high-concentration GC exposure significantly increased the apoptosis rate of PC12 cells. Immunofluorescence staining showed that GC exposure significantly increased the expression of C-fos, GRP78, p-PERK, p-IRE1, ATF4, ASK1, JNK, and CHOP. Treatment with ERS inhibitor 4-phenylbutyric acid (4-PBA) and GR inhibitor RU38486 attenuated related damage and downregulated the expression of the abovementioned proteins. Conclusion High-concentration GC exposure can significantly inhibit the viability of PC12 cells and induce apoptosis. PERK-ATF4-CHOP and IRE1-ASK1-JNK pathways are involved in the above damage process.
Collapse
|
28
|
Molecular Dysfunctions of Mitochondria-Associated Membranes (MAMs) in Alzheimer's Disease. Int J Mol Sci 2020; 21:ijms21249521. [PMID: 33327665 PMCID: PMC7765134 DOI: 10.3390/ijms21249521] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/04/2020] [Accepted: 12/08/2020] [Indexed: 02/07/2023] Open
Abstract
Alzheimer’s disease (AD) is a multifactorial neurodegenerative pathology characterized by a progressive decline of cognitive functions. Alteration of various signaling cascades affecting distinct subcellular compartment functions and their communication likely contribute to AD progression. Among others, the alteration of the physical association between the endoplasmic reticulum (ER) and mitochondria, also referred as mitochondria-associated membranes (MAMs), impacts various cellular housekeeping functions such as phospholipids-, glucose-, cholesterol-, and fatty-acid-metabolism, as well as calcium signaling, which are all altered in AD. Our review describes the physical and functional proteome crosstalk between the ER and mitochondria and highlights the contribution of distinct molecular components of MAMs to mitochondrial and ER dysfunctions in AD progression. We also discuss potential strategies targeting MAMs to improve mitochondria and ER functions in AD.
Collapse
|
29
|
Ghemrawi R, Khair M. Endoplasmic Reticulum Stress and Unfolded Protein Response in Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E6127. [PMID: 32854418 PMCID: PMC7503386 DOI: 10.3390/ijms21176127] [Citation(s) in RCA: 245] [Impact Index Per Article: 49.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2020] [Revised: 08/14/2020] [Accepted: 08/20/2020] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is an important organelle involved in protein quality control and cellular homeostasis. The accumulation of unfolded proteins leads to an ER stress, followed by an adaptive response via the activation of the unfolded protein response (UPR), PKR-like ER kinase (PERK), inositol-requiring transmembrane kinase/endoribonuclease 1α (IRE1α) and activating transcription factor 6 (ATF6) pathways. However, prolonged cell stress activates apoptosis signaling leading to cell death. Neuronal cells are particularly sensitive to protein misfolding, consequently ER and UPR dysfunctions were found to be involved in many neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and prions diseases, among others characterized by the accumulation and aggregation of misfolded proteins. Pharmacological UPR modulation in affected tissues may contribute to the treatment and prevention of neurodegeneration. The association between ER stress, UPR and neuropathology is well established. In this review, we provide up-to-date evidence of UPR activation in neurodegenerative disorders followed by therapeutic strategies targeting the UPR and ameliorating the toxic effects of protein unfolding and aggregation.
Collapse
Affiliation(s)
- Rose Ghemrawi
- College of Pharmacy, Al Ain University, Abu Dhabi 112612, UAE
| | - Mostafa Khair
- Core Technology Platforms, New York University Abu Dhabi, Abu Dhabi 129188, UAE;
| |
Collapse
|
30
|
Murray HC, Dieriks BV, Swanson MEV, Anekal PV, Turner C, Faull RLM, Belluscio L, Koretsky A, Curtis MA. The unfolded protein response is activated in the olfactory system in Alzheimer's disease. Acta Neuropathol Commun 2020; 8:109. [PMID: 32665027 PMCID: PMC7362534 DOI: 10.1186/s40478-020-00986-7] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2020] [Accepted: 07/03/2020] [Indexed: 01/04/2023] Open
Abstract
Olfactory dysfunction is an early and prevalent symptom of Alzheimer’s disease (AD) and the olfactory bulb is a nexus of beta-amyloid plaque and tau neurofibrillary tangle (NFT) pathology during early AD progression. To mitigate the accumulation of misfolded proteins, an endoplasmic reticulum stress response called the unfolded protein response (UPR) occurs in the AD hippocampus. However, chronic UPR activation can lead to apoptosis and the upregulation of beta-amyloid and tau production. Therefore, UPR activation in the olfactory system could be one of the first changes in AD. In this study, we investigated whether two proteins that signal UPR activation are expressed in the olfactory system of AD cases with low or high amounts of aggregate pathology. We used immunohistochemistry to label two markers of UPR activation (p-PERK and p-eIF2α) concomitantly with neuronal markers (NeuN and PGP9.5) and pathology markers (beta-amyloid and tau) in the olfactory bulb, piriform cortex, entorhinal cortex and the CA1 region of the hippocampus in AD and normal cases. We show that UPR activation, as indicated by p-PERK and p-eIF2α expression, is significantly increased throughout the olfactory system in AD cases with low (Braak stage III-IV) and high-level (Braak stage V-VI) pathology. We further show that UPR activation occurs in the mitral cells and in the anterior olfactory nucleus of the olfactory bulb where tau and amyloid pathology is abundant. However, UPR activation is not present in neurons when they contain NFTs and only rarely occurs in neurons containing diffuse tau aggregates. We conclude that UPR activation is prevalent in all regions of the olfactory system and support previous findings suggesting that UPR activation likely precedes NFT formation. Our data indicate that chronic UPR activation in the olfactory system might contribute to the olfactory dysfunction that occurs early in the pathogenesis of AD.
Collapse
|
31
|
Liu XQ, Deng YX, Dai Z, Hu T, Cai WW, Liu HF, Li H, Zhu WL, Li BY, Wang Q, Zhang SJ. Sodium tanshinone IIA sulfonate protects against Aβ1–42-induced cellular toxicity by modulating Aβ-degrading enzymes in HT22 cells. Int J Biol Macromol 2020; 151:47-55. [DOI: 10.1016/j.ijbiomac.2020.02.040] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 02/01/2020] [Accepted: 02/05/2020] [Indexed: 02/07/2023]
|
32
|
He Z, Wang M, Zhao Q, Li X, Liu P, Ren B, Wu C, Du X, Li N, Liu Q. Bis(ethylmaltolato)oxidovanadium (IV) mitigates neuronal apoptosis resulted from amyloid-beta induced endoplasmic reticulum stress through activating peroxisome proliferator-activated receptor γ. J Inorg Biochem 2020; 208:111073. [PMID: 32466853 DOI: 10.1016/j.jinorgbio.2020.111073] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 03/14/2020] [Accepted: 03/19/2020] [Indexed: 01/14/2023]
Abstract
Neuronal apoptosis caused by amyloid-beta (Aβ) overproduction is one of the most important pathological features in Alzheimer's disease (AD). Endoplasmic reticulum (ER) stress induced by Aβ overload plays a critical role in this process. Bis(ethylmaltolato)oxidovanadium (IV) (BEOV), a vanadium compound which had been regarded as peroxisome proliferator-activated receptor γ (PPARγ) agonist, was reported to exert an antagonistic effect on ER stress. In this study, we tested whether BEOV could ameliorate the Aβ-induced neuronal apoptosis by inhibiting ER stress. It was observed that BEOV treatment ameliorated both tunicamycin-induced and/or Aβ-induced ER stress and neurotoxicity in a dose-dependent manner through downgrading ER stress-associated and apoptosis-associated proteins in primary hippocampal neurons. Consistent with in vitro results, BEOV also reduced ER stress and inhibited neuronal apoptosis in hippocampi and cortexes of transgenic AD model mice. Moreover, by adopting GW9662 and salubrinal, the inhibitor of PPARγ and hyperphosphorylated eukaryotic translation initiation factor 2α, respectively, we further confirmed that BEOV alleviated Aβ-induced ER stress and neuronal apoptosis in primary hippocampal neurons by activating PPARγ. Taken together, these results provided scientific evidences to support the concept that BEOV ameliorates Aβ-induced ER stress and neuronal apoptosis through activating PPARγ.
Collapse
Affiliation(s)
- Zhijun He
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China; College of optoelectronic engineering, Shenzhen university, Shenzhen, Guangdong 518060, China
| | - Menghuan Wang
- School of Basic Medical Sciences, Shenzhen University, Shenzhen, Guangdong 518055, China
| | - Qionghui Zhao
- Shenzhen Food Inspection Center of CIQ, Shenzhen, Guangdong 518055, China
| | - Xiaoqian Li
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China
| | - Pengan Liu
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China
| | - Bingyu Ren
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China
| | - Chong Wu
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China
| | - Xiubo Du
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China
| | - Nan Li
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China; Shenzhen Bay Laboratory, Shenzhen 518055, China.
| | - Qiong Liu
- College of life sciences and oceanography, Shenzhen university, Shenzhen, Guangdong 518055, China; Shenzhen-Hong Kong Institute of Brain Science-Shenzhen Fundamental Research Institutions, China.
| |
Collapse
|
33
|
Rozpędek-Kamińska W, Siwecka N, Wawrzynkiewicz A, Wojtczak R, Pytel D, Diehl JA, Majsterek I. The PERK-Dependent Molecular Mechanisms as a Novel Therapeutic Target for Neurodegenerative Diseases. Int J Mol Sci 2020; 21:E2108. [PMID: 32204380 PMCID: PMC7139310 DOI: 10.3390/ijms21062108] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2020] [Revised: 03/13/2020] [Accepted: 03/17/2020] [Indexed: 12/11/2022] Open
Abstract
Higher prevalence of neurodegenerative diseases is strictly connected with progressive aging of the world population. Interestingly, a broad range of age-related, neurodegenerative diseases is characterized by a common pathological mechanism-accumulation of misfolded and unfolded proteins within the cells. Under certain circumstances, such protein aggregates may evoke endoplasmic reticulum (ER) stress conditions and subsequent activation of the unfolded protein response (UPR) signaling pathways via the protein kinase RNA-like endoplasmic reticulum kinase (PERK)-dependent manner. Under mild to moderate ER stress, UPR has a pro-adaptive role. However, severe or long-termed ER stress conditions directly evoke shift of the UPR toward its pro-apoptotic branch, which is considered to be a possible cause of neurodegeneration. To this day, there is no effective cure for Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD), or prion disease. Currently available treatment approaches for these diseases are only symptomatic and cannot affect the disease progression. Treatment strategies, currently under detailed research, include inhibition of the PERK-dependent UPR signaling branches. The newest data have reported that the use of small-molecule inhibitors of the PERK-mediated signaling branches may contribute to the development of a novel, ground-breaking therapeutic approach for neurodegeneration. In this review, we critically describe all the aspects associated with such targeted therapy against neurodegenerative proteopathies.
Collapse
Affiliation(s)
- Wioletta Rozpędek-Kamińska
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Natalia Siwecka
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Adam Wawrzynkiewicz
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Radosław Wojtczak
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| | - Dariusz Pytel
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; (D.P.); (J.A.D.)
| | - J. Alan Diehl
- Department of Biochemistry and Molecular Biology, Hollings Cancer Center, Medical University of South Carolina, Charleston, SC 29425, USA; (D.P.); (J.A.D.)
| | - Ireneusz Majsterek
- Department of Clinical Chemistry and Biochemistry, Medical University of Lodz, 90-419 Lodz, Poland; (W.R.-K.); (N.S.); (A.W.); (R.W.)
| |
Collapse
|
34
|
Akhter Y, Nabi J, Hamid H, Tabassum N, Pottoo FH, Sharma A. Protein Quality Control in Neurodegeneration and Neuroprotection. QUALITY CONTROL OF CELLULAR PROTEIN IN NEURODEGENERATIVE DISORDERS 2020. [DOI: 10.4018/978-1-7998-1317-0.ch001] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Proteostasis is essential for regulating the integrity of the proteome. Disruption of proteostasis under some rigorous conditions leads to the aggregation and accumulation of misfolded toxic proteins, which plays a central role in the pathogenesis of protein conformational disorders. The protein quality control (PQC) system serves as a multi-level security system to shield cells from abnormal proteins. The intrinsic PQC systems maintaining proteostasis include the ubiquitin-proteasome system (UPS), chaperon-mediated autophagy (CMA), and autophagy-lysosome pathway (ALP) that serve to target misfolded proteins for unfolding, refolding, or degradation. Alterations of PQC systems in neurons have been implicated in the pathogenesis of various neurodegenerative disorders. This chapter provides an overview of PQC pathways to set a framework for discussion of the role of PQC in neurodegenerative disorders. Additionally, various pharmacological approaches targeting PQC are summarized.
Collapse
Affiliation(s)
- Yasmeena Akhter
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Jahangir Nabi
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Hinna Hamid
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Nahida Tabassum
- Department of Pharmaceutical Sciences (Pharmacology Division), Faculty of Applied Sciences and Technology, University of Kashmir, Srinagar, India
| | - Faheem Hyder Pottoo
- Department of Pharmaology, College of Clinical Pharmacy, Imam Abdulrahman Bin Faisal University, Saudi Arabia
| | - Aashish Sharma
- Centre for Research in Medical Devices (CURAM), National University of Ireland, Ireland & School of Medical and Allied Sciences, GD Goenka University, Gurgaon, India
| |
Collapse
|
35
|
β-Amyloid Peptide: the Cell Compartment Multi-faceted Interaction in Alzheimer's Disease. Neurotox Res 2019; 37:250-263. [PMID: 31811589 DOI: 10.1007/s12640-019-00116-9] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/14/2019] [Revised: 08/29/2019] [Accepted: 09/16/2019] [Indexed: 12/17/2022]
Abstract
Alzheimer's disease (AD) is the most widespread form of dementia, characterized by memory loss and reduction of cognitive functions that strongly interfere with normal daily life. Numerous evidences show that aggregates of the amyloid beta peptide, formed by 39 to 42 amino acid residues (Aβ39-43), from soluble small oligomers to large fibrils are characteristic markers of this pathology. However, AD is a complex disease and its neurodegenerative molecular mechanism is not yet fully understood. Growing evidence suggests a link between Aβ polymorphic nature, oligomers and fibrils, and specific mechanisms of neurodegeneration. The Aβ variable nature and its multiplicity of interactions with different proteins and organelles reflect the complexity of this pathology. In this review, we analyze the effects of the interaction between Aβ peptide and different cellular compartments in relation to the different kinds and sizes of amyloid aggregates. In particular, Aβ interaction with different cell structures such as the plasma membrane, mitochondria, lysosomes, nucleus, and endoplasmic reticulum is discussed. Further, we analyze the Aβ peptide ability to modify the structure and function of the target organelle, inducing alteration of its physiological role thus contributing to the pathological event. Dysfunction of cellular components terminating with the activation of the cellular death mechanism and subsequent neurodegeneration is also taken into consideration.
Collapse
|
36
|
Hashimoto S, Saido TC. Critical review: involvement of endoplasmic reticulum stress in the aetiology of Alzheimer's disease. Open Biol 2019; 8:rsob.180024. [PMID: 29695619 PMCID: PMC5936719 DOI: 10.1098/rsob.180024] [Citation(s) in RCA: 94] [Impact Index Per Article: 15.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2018] [Accepted: 04/03/2018] [Indexed: 12/18/2022] Open
Abstract
The endoplasmic reticulum (ER) stress response is regarded as an important process in the aetiology of Alzheimer's disease (AD). The accumulation of pathogenic misfolded proteins and the disruption of intracellular calcium (Ca2+) signalling are considered to be fundamental mechanisms that underlie the induction of ER stress, leading to neuronal cell death. Indeed, a number of studies have proposed molecular mechanisms linking ER stress to AD pathogenesis based on results from in vitro systems and AD mouse models. However, stress responsivity was largely different between each mouse model, even though all of these models display AD-related pathologies. While several reports have shown elevated ER stress responses in amyloid precursor protein (APP) and presenilin 1 (PS1) double-transgenic (Tg) AD mouse models, we and other groups, in contrast, observed no such ER stress response in APP-single-Tg or App-knockin mice. Therefore, it is debatable whether the ER stress observed in APP and PS1 double-Tg mice is due to AD pathology. From these findings, the roles of ER stress in AD pathogenesis needs to be carefully addressed in future studies. In this review, we summarize research detailing the relationship between ER stress and AD, and analyse the results in detail.
Collapse
Affiliation(s)
- Shoko Hashimoto
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takaomi C Saido
- Laboratory for Proteolytic Neuroscience, RIKEN Center for Brain Science, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| |
Collapse
|
37
|
Xu S, Di Z, He Y, Wang R, Ma Y, Sun R, Li J, Wang T, Shen Y, Fang S, Feng L, Shen Y. Mesencephalic astrocyte-derived neurotrophic factor (MANF) protects against Aβ toxicity via attenuating Aβ-induced endoplasmic reticulum stress. J Neuroinflammation 2019; 16:35. [PMID: 30760285 PMCID: PMC6373169 DOI: 10.1186/s12974-019-1429-0] [Citation(s) in RCA: 47] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2018] [Accepted: 02/03/2019] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Extracellular accumulation of amyloid β-peptide (Aβ) is one of pathological hallmarks of Alzheimer's disease (AD) and contributes to the neuronal loss. Mesencephalic astrocyte-derived neurotrophic factor (MANF) is an endoplasmic reticulum (ER) stress-inducible neurotrophic factor. Many groups, including ours, have proved that MANF rescues neuronal loss in several neurological disorders, such as Parkinson's disease and cerebral ischemia. However, whether MANF exerts its protective effect against Aβ neurotoxicity in AD remains unknown. METHODS In the present study, the characteristic expressions of MANF in Aβ1-42-treated neuronal cells as well as in the brains of APP/PS1 transgenic mice were analyzed by immunofluorescence staining, qPCR, and Western blot. The effects of MANF overexpression, MANF knockdown, or recombination human MANF protein (rhMANF) on neuron viability, apoptosis, and the expression of ER stress-related proteins following Aβ1-42 exposure were also investigated. RESULTS The results showed the increased expressions of MANF, as well as ER stress markers immunoglobulin-binding protein (BiP) and C/EBP homologous protein (CHOP), in the brains of the APP/PS1 transgenic mice and Aβ1-42-treated neuronal cells. MANF overexpression or rhMANF treatment partially protected against Aβ1-42-induced neuronal cell death, associated with marked decrease of cleaved caspase-3, whereas MANF knockdown with siRNA aggravated Aβ1-42 cytotoxicity including caspase-3 activation. Further study demonstrated that the expressions of BiP, ATF6, phosphorylated-IRE1, XBP1s, phosphorylated-eIF2α, ATF4, and CHOP were significantly downregulated by MANF overexpression or rhMANF treatment in neuronal cells following Aβ1-42 exposure, whereas knockdown of MANF has the opposite effect. CONCLUSIONS These findings demonstrate that MANF may exert neuroprotective effects against Aβ-induced neurotoxicity through attenuating ER stress, suggesting that an applicability of MANF as a therapeutic candidate for AD.
Collapse
Affiliation(s)
- Shengchun Xu
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Zemin Di
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yufeng He
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Runjie Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yuyang Ma
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Rui Sun
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Jing Li
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China
| | - Tao Wang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Yujun Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China
| | - Shengyun Fang
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China.,Center for Biomedical Engineering and Technology, University of Maryland, Baltimore, MD, USA
| | - Lijie Feng
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China. .,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| | - Yuxian Shen
- School of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China. .,Biopharmaceutical Research Institute, Anhui Medical University, Hefei, 230032, China. .,Institute of Basic Medical Sciences, Anhui Medical University, Hefei, 230032, China.
| |
Collapse
|
38
|
Link between the unfolded protein response and dysregulation of mitochondrial bioenergetics in Alzheimer's disease. Cell Mol Life Sci 2019; 76:1419-1431. [PMID: 30683981 PMCID: PMC6420888 DOI: 10.1007/s00018-019-03009-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2018] [Revised: 12/19/2018] [Accepted: 01/09/2019] [Indexed: 12/11/2022]
Abstract
Alzheimer’s disease (AD) is a progressive neurodegenerative disorder affecting more than 47.5 million people worldwide. Metabolic impairments are common hallmarks of AD, and amyloid-β (Aβ) peptide and hyperphosphorylated tau protein—the two foremost histopathological signs of AD—have been implicated in mitochondrial dysfunction. Many neurodegenerative disorders, including AD, show excessive amounts of mis-/unfolded proteins leading to an activation of the unfolded protein response (UPR). In the present study, we aimed to characterize the link between ER stress and bioenergetics defects under normal condition (human SH-SY5Y neuroblastoma cells: control cells) or under pathological AD condition [SH-SY5Y cells overexpressing either the human amyloid precursor protein (APP) or mutant tau (P301L)]. More specifically, we measured UPR gene expression, cell viability, and bioenergetics parameters, such as ATP production and mitochondrial membrane potential (MMP) in basal condition and after an induced ER stress by thapsigargin. We detected highly activated UPR and dysregulated bioenergetics in basal condition in both AD cellular models. Strikingly, acute-induced ER stress increased the activity of the UPR in both AD cellular models, leading to up-regulation of apoptotic pathways, and further dysregulated mitochondrial function.
Collapse
|
39
|
García-González P, Cabral-Miranda F, Hetz C, Osorio F. Interplay Between the Unfolded Protein Response and Immune Function in the Development of Neurodegenerative Diseases. Front Immunol 2018; 9:2541. [PMID: 30450103 PMCID: PMC6224445 DOI: 10.3389/fimmu.2018.02541] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence suggests that the immune and nervous systems are in close interaction in health and disease conditions. Protein aggregation and proteostasis dysfunction at the level of the endoplasmic reticulum (ER) are central contributors to neurodegenerative diseases. The unfolded protein response (UPR) is the main transduction pathway that maintains protein homeostasis under conditions of protein misfolding and aggregation. Brain inflammation often coexists with the degenerative process in different brain diseases. Interestingly, besides its well-described role in neuronal fitness, the UPR has also emerged as a key regulator of ontogeny and function of several immune cell types. Nevertheless, the contribution of the UPR to brain inflammation initiated by immune cells remains largely unexplored. In this review, we provide a perspective on the potential role of ER stress signaling in brain-associated immune cells and the possible implications to neuroinflammation and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paulina García-González
- Laboratory of Immunology and Cellular Stress, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Felipe Cabral-Miranda
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Brain Health and Metabolism, FONDAP Center for Geroscience, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Brain Health and Metabolism, FONDAP Center for Geroscience, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, United States
| | - Fabiola Osorio
- Laboratory of Immunology and Cellular Stress, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
40
|
PDGFR and IGF-1R Inhibitors Induce a G2/M Arrest and Subsequent Cell Death in Human Glioblastoma Cell Lines. Cells 2018; 7:cells7090131. [PMID: 30200644 PMCID: PMC6162497 DOI: 10.3390/cells7090131] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 08/25/2018] [Accepted: 08/27/2018] [Indexed: 12/19/2022] Open
Abstract
Glioblastomas are highly resistant to radiation and chemotherapy. Currently, there are no effective therapies for this type of tumor. Signaling mechanisms initiated by PDGFR and IGF-1R are important in glioblastoma, and inhibition of the signal transduction pathways initiated by these receptors could be a useful alternative strategy for glioblastoma treatment. We have studied the effects of the PDGFR inhibitor JNJ-10198409 (JNJ) and the IGF-1R inhibitor picropodophyllin (PPP) in glioblastoma cell lines as well as in primary cultures derived from patients affected by this type of tumor. JNJ and PPP treatment blocked PDGFR and IGF-1R signaling respectively and reduced Akt and Erk 1/2 phosphorylation. Both inhibitors diminished cell proliferation, inducing a G2/M block of the cell cycle. Cell death induced by JNJ was caspase-dependent, Annexin-V positive and caused PARP cleavage, especially in T98 cells, suggesting an apoptotic mechanism. However, cell death induced by PPP was not completely inhibited by caspase inhibitors in all cell lines apart from LN-229 cells, indicating a caspase-independent mechanism. Several inhibitors targeted against different cell death pathways could not block this caspase-independent component, which may be a non-programmed necrotic mechanism. Apoptotic arrays performed in T98 and LN-229 cells upon JNJ and PPP treatment revealed that procaspase 3 levels were augmented by both drugs in T98 cells and only by JNJ in LN229-cells. Furthermore, XIAP and survivin levels were much higher in LN-229 cells than in T98 cells, revealing that LN-229 cells are more susceptible to undergo caspase-independent cell death mechanisms. JNJ and PPP combination was more effective than each treatment alone.
Collapse
|
41
|
Chu J, Pelletier J. Therapeutic Opportunities in Eukaryotic Translation. Cold Spring Harb Perspect Biol 2018; 10:cshperspect.a032995. [PMID: 29440069 DOI: 10.1101/cshperspect.a032995] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
The ability to block biological processes with selective small molecules provides advantages distinct from most other experimental approaches. These include rapid time to onset, swift reversibility, ability to probe activities in manners that cannot be accessed by genetic means, and the potential to be further developed as therapeutic agents. Small molecule inhibitors can also be used to alter expression and activity without affecting the stoichiometry of interacting partners. These tenets have been especially evident in the field of translation. Small molecule inhibitors were instrumental in enabling investigators to capture short-lived complexes and characterize specific steps of protein synthesis. In addition, several drugs that are the mainstay of modern antimicrobial drug therapy are potent inhibitors of prokaryotic translation. Currently, there is much interest in targeting eukaryotic translation as decades of research have revealed that deregulated protein synthesis in cancer cells represents a targetable vulnerability. In addition to being potential therapeutics, small molecules that manipulate translation have also been shown to influence cognitive processes such as memory. In this review, we focus on small molecule modulators that target the eukaryotic translation initiation apparatus and provide an update on their potential application to the treatment of disease.
Collapse
Affiliation(s)
- Jennifer Chu
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada
| | - Jerry Pelletier
- Department of Biochemistry, McGill University, Montreal, Quebec H3G 1Y6, Canada.,Department of Oncology, McGill University, Montreal, Quebec H3G 1Y6, Canada.,Rosalind and Morris Goodman Cancer Research Center, McGill University, Montreal, Quebec H3G 1Y6, Canada
| |
Collapse
|
42
|
Lee DY, Hong SH, Kim B, Lee DS, Yu K, Lee KS. Neuropeptide Y mitigates ER stress–induced neuronal cell death by activating the PI3K–XBP1 pathway. Eur J Cell Biol 2018; 97:339-348. [DOI: 10.1016/j.ejcb.2018.04.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2018] [Revised: 04/04/2018] [Accepted: 04/05/2018] [Indexed: 01/17/2023] Open
|
43
|
Targeting the Endoplasmic Reticulum Unfolded Protein Response to Counteract the Oxidative Stress-Induced Endothelial Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4946289. [PMID: 29725497 PMCID: PMC5872601 DOI: 10.1155/2018/4946289] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/18/2018] [Indexed: 12/22/2022]
Abstract
In endothelial cells, the tight control of the redox environment is essential for the maintenance of vascular homeostasis. The imbalance between ROS production and antioxidant response can induce endothelial dysfunction, the initial event of many cardiovascular diseases. Recent studies have revealed that the endoplasmic reticulum could be a new player in the promotion of the pro- or antioxidative pathways and that in such a modulation, the unfolded protein response (UPR) pathways play an essential role. The UPR consists of a set of conserved signalling pathways evolved to restore the proteostasis during protein misfolding within the endoplasmic reticulum. Although the first outcome of the UPR pathways is the promotion of an adaptive response, the persistent activation of UPR leads to increased oxidative stress and cell death. This molecular switch has been correlated to the onset or to the exacerbation of the endothelial dysfunction in cardiovascular diseases. In this review, we highlight the multiple chances of the UPR to induce or ameliorate oxidative disturbances and propose the UPR pathways as a new therapeutic target for the clinical management of endothelial dysfunction.
Collapse
|
44
|
Niu X, Zhang J, Ling C, Bai M, Peng Y, Sun S, Li Y, Zhang Z. Polysaccharide from Angelica sinensis protects H9c2 cells against oxidative injury and endoplasmic reticulum stress by activating the ATF6 pathway. J Int Med Res 2018. [PMID: 29517941 PMCID: PMC5991254 DOI: 10.1177/0300060518758863] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022] Open
Abstract
Objectives Angelica sinensis exerts various pharmacological effects, such as antioxidant and anti-apoptotic activity. This study aimed to investigate the active ingredients in A. sinensis with antioxidant properties and whether A. sinensis polysaccharide (ASP) protects H9c2 cells against oxidative and endoplasmic reticulum (ER) stress. Methods The ingredients of A. sinensis and their targets and related pathways were determined using web-based databases. Markers of oxidative stress, cell viability, apoptosis, and ER stress-related signalling pathways were measured in H9c2 cells treated with hydrogen peroxide (H2O2) and ASP. Results The ingredient–pathway–disease network showed that A. sinensis exerted protective effects against oxidative injury through its various active ingredients on regulation of multiple pathways. Subsequent experiments showed that ASP pretreatment significantly decreased H2O2-induced cytotoxicity and apoptosis in H9c2 cells. ASP pretreatment inhibited H2O2-induced reactive oxygen species generation, lactic dehydrogenase release, and malondialdehyde production. ASP exerted beneficial effects by inducing activating transcription factor 6 (ATF6) and increasing ATF6 target protein levels, which in turn attenuated ER stress and increased antioxidant activity. Conclusions Our findings indicate that ASP, a major water-soluble component of A. sinensis, exerts protective effects against H2O2-induced injury in H9c2 cells by activating the ATF6 pathway, thus ameliorating ER and oxidative stress.
Collapse
Affiliation(s)
- Xiaowei Niu
- 1 The First School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu, China
| | | | - Chun Ling
- 3 The First People's Hospital of Chuzhou, Chuzhou, Anhui, China
| | - Ming Bai
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| | - Yu Peng
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| | - Shaobo Sun
- 6 Key Lab of Prevention and Treatment for Chronic Disease, Traditional Chinese Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Yingdong Li
- 6 Key Lab of Prevention and Treatment for Chronic Disease, Traditional Chinese Medicine of Gansu Province, Lanzhou, Gansu, China
| | - Zheng Zhang
- 4 Department of Cardiology, the First Hospital of Lanzhou University, Lanzhou, Gansu, China.,5 Gansu Key Laboratory of Cardiovascular Disease, Lanzhou, Gansu, China
| |
Collapse
|
45
|
Liu XJ, Wei J, Shang YH, Huang HC, Lao FX. Modulation of AβPP and GSK3β by Endoplasmic Reticulum Stress and Involvement in Alzheimer's Disease. J Alzheimers Dis 2018; 57:1157-1170. [PMID: 28339396 DOI: 10.3233/jad-161111] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Alzheimer's disease (AD) is a dementia disease with neuronal loss and synaptic impairment. This impairment is caused, at least partly, by the generation of two main AD hallmarks, namely the hyperphosphorylated tau protein comprising neurofibrillary tangles and senile plaques containing amyloid-β (Aβ) peptides. The amyloid-β protein precursor (AβPP) and glycogen synthase kinase-3β (GSK3β) are two main proteins associated with AD and are closely correlated with these hallmarks. Recently, both of the proteins were reported to be modulated by endoplasmic reticulum stress (ERS) and are involved in the pathogenesis of AD. The mechanism of ERS plus the modulation of AβPP processing and GSK3β activity by ERS in AD are summarized and explored in this review.
Collapse
Affiliation(s)
- Xin-Jun Liu
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Jun Wei
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Ying-Hui Shang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Han-Chang Huang
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| | - Feng-Xue Lao
- Beijing Key Laboratory of Bioactive Substances and Functional Foods, Beijing Union University, Beijing, P.R. China.,College of Arts and Science of Beijing Union University, Beijing, P.R. China
| |
Collapse
|
46
|
Kim EJ, Grant GR, Bowman AS, Haider N, Gudiseva HV, Chavali VRM. Complete Transcriptome Profiling of Normal and Age-Related Macular Degeneration Eye Tissues Reveals Dysregulation of Anti-Sense Transcription. Sci Rep 2018; 8:3040. [PMID: 29445097 PMCID: PMC5813239 DOI: 10.1038/s41598-018-21104-7] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Accepted: 01/30/2018] [Indexed: 11/21/2022] Open
Abstract
Age-related macular degeneration (AMD) predominantly affects the retina and retinal pigment epithelium in the posterior eye. While there are numerous studies investigating the non-coding transcriptome of retina and RPE, few significant differences between AMD and normal tissues have been reported. Strand specific RNA sequencing of both peripheral retina (PR) and RPE-Choroid-Sclera (PRCS), in both AMD and matched normal controls were generated. The transcriptome analysis reveals a highly significant and consistent impact on anti-sense transcription as well as moderate changes in the regulation of non-coding (sense) RNA. Hundreds of genes that do not express anti-sense transcripts in normal PR and PRCS demonstrate significant anti-sense expression in AMD in all patient samples. Several pathways are highly enriched in the upregulated anti-sense transcripts-in particular the EIF2 signaling pathway. These results call for a deeper exploration into anti-sense and noncoding RNA regulation in AMD and their potential as therapeutic targets.
Collapse
Affiliation(s)
- Eun Ji Kim
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Gregory R Grant
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Department of Genetics, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anita S Bowman
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Functional Genomics Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Naqi Haider
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
- Functional Genomics Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Harini V Gudiseva
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA
| | - Venkata Ramana Murthy Chavali
- Department of Ophthalmology, University of Pennsylvania School of Medicine, Philadelphia, Pennsylvania, USA.
- Functional Genomics Lab, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
47
|
XBP1 and PERK Have Distinct Roles in Aβ-Induced Pathology. Mol Neurobiol 2018; 55:7523-7532. [PMID: 29427089 DOI: 10.1007/s12035-018-0942-y] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/25/2017] [Accepted: 01/28/2018] [Indexed: 10/18/2022]
Abstract
Endoplasmic reticulum (ER) stress triggers multiple cellular signals to restore cellular function or induce proapoptosis that is altered in the brains of patients with Alzheimer's disease (AD). However, the role of ER stress in β-amyloid (Aβ)-induced AD pathology remains elusive, and data obtained from different animal models and under different experimental conditions are sometimes controversial. The current study conducted in vivo genetic experiments to systematically examine the distinct role of each ER stress effector during disease progression. Our results indicated that inositol-requiring enzyme 1 was activated before protein kinase RNA-like endoplasmic reticulum kinase (PERK) activation in Aβ42 transgenic flies. Proteasome activity played a key role in this sequential activation. Furthermore, our study separated learning deficits from early degeneration in Aβ-induced impairment by demonstrating that X-box binding protein 1 overexpression at an early stage reversed Aβ-induced early death without affecting learning performance in the Aβ42 transgenic flies. PERK activation was determined to only enhance Aβ-induced learning deficits. Moreover, proteasome overactivation was determined to delay PERK activation and improve learning deficits. Altogether, the findings of this study demonstrate the complex roles of ER stress during Aβ pathogenesis and the possibility of using different ER stress effectors as reporters to indicate the status of disease progression.
Collapse
|
48
|
Alzheimer's disease pathology and the unfolded protein response: prospective pathways and therapeutic targets. Behav Pharmacol 2018; 28:161-178. [PMID: 28252521 DOI: 10.1097/fbp.0000000000000299] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
Many vital interdependent cellular functions including proteostasis, lipogenesis and Ca homeostasis are executed by the endoplasmic reticulum (ER). Exogenous insults can impair ER performance: this must be rapidly corrected or cell death will ensue. Protective adaptations can boost the functional capacity of the ER and form the basis of the unfolded protein response (UPR). Activated in response to the accumulation of misfolded proteins, the UPR can halt protein translation while increasing protein-handling chaperones and the degradation of erroneous proteins through a conserved three-tier molecular cascade. However, prolonged activation of the UPR can result in the maladaptation of the system, resulting in the activation of inflammatory and apoptotic effectors. Recently, UPR and its involvement in neurodegenerative disease has attracted much interest and numerous potentially 'drugable' points of crosstalk are now emerging. Here, we summarize the functions of the ER and UPR, and highlight evidence for its potential role in the pathogenesis of Alzheimer's disease, before discussing several key targets with therapeutic potential.
Collapse
|
49
|
Aβ1-42 induces cell damage via RAGE-dependent endoplasmic reticulum stress in bEnd.3 cells. Exp Cell Res 2018; 362:83-89. [DOI: 10.1016/j.yexcr.2017.11.005] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 11/01/2017] [Accepted: 11/04/2017] [Indexed: 12/27/2022]
|
50
|
Gerakis Y, Hetz C. Emerging roles of ER stress in the etiology and pathogenesis of Alzheimer's disease. FEBS J 2017; 285:995-1011. [PMID: 29148236 DOI: 10.1111/febs.14332] [Citation(s) in RCA: 181] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2017] [Revised: 10/03/2017] [Accepted: 11/13/2017] [Indexed: 12/12/2022]
Abstract
Alzheimer's disease (AD) is a progressive neurodegenerative disease characterized by synaptic dysfunction and accumulation of abnormal aggregates formed by amyloid-β peptides or phosphorylated tau proteins. Accumulating evidence suggests that alterations in the buffering capacity of the proteostasis network are a salient feature of AD. The endoplasmic reticulum (ER) is the main compartment involved in protein folding and secretion and is drastically affected in AD neurons. ER stress triggers the activation of the unfolded protein response (UPR), a signal transduction pathway that enforces adaptive programs to recover homeostasis or trigger apoptosis of irreversibly damaged cells. Experimental manipulation of specific UPR signaling modules in preclinical models of AD has revealed a key role of this pathway in regulating protein misfolding and neurodegeneration. Recent studies suggest that the UPR also influences synaptic plasticity and memory through ER stress-independent mechanisms. Consequently, targeting of the UPR in AD is emerging as an interesting therapeutic approach to modify the two pillars of AD, protein misfolding and synaptic failure. Here, we review the functional role of ER stress signaling in AD, discussing the complex involvement of the pathway in controlling neuronal survival, the amyloid cascade, neurodegeneration and synaptic function. Recent intervention efforts to target the UPR with pharmacological and gene therapy strategies are also discussed.
Collapse
Affiliation(s)
- Yannis Gerakis
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile.,Center for Geroscience, Brain Health and Metabolism, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, USA.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA.,Cellular and Molecular Biology Program, Institute of Biomedical Sciences, University of Chile, Santiago, Chile
| |
Collapse
|