1
|
Ren J, Pei Q, Dong H, Wei X, Li L, Duan H, Zhang G, Zhang A. Tripartite motif 25 inhibits protein aggregate degradation during PRRSV infection by suppressing p62-mediated autophagy. J Virol 2024:e0143724. [PMID: 39480084 DOI: 10.1128/jvi.01437-24] [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: 08/19/2024] [Accepted: 10/06/2024] [Indexed: 11/02/2024] Open
Abstract
Viral infection causes endoplasmic reticulum stress and protein metabolism disorder, influencing protein aggregates formation or degradation that originate from misfolded proteins. The mechanism by which host proteins are involved in the above process remains largely unknown. The present study found that porcine reproductive and respiratory syndrome virus (PRRSV) infection promoted the degradation of intracellular ubiquitinated protein aggregates via activating autophagy. The host cell E3 ligase tripartite motif-containing (TRIM)25 promoted the recruitment and aggregation of polyubiquitinated proteins and impeded their degradation caused by PRRSV. TRIM25 interacted with ubiquitinated aggregates and was part of the aggregates complex. Next, the present study investigated the mechanisms by which TRIM25 inhibited the degradation of protein aggregates, and it was found that TRIM25 interacted with both Kelch-like ECH-associated protein 1 (KEAP1) and nuclear factor E2-related factor 2 (Nrf2), facilitated the nuclear translocation of Nrf2 by targeting KEAP1 for K48-linked ubiquitination and proteasome degradation, and activated Nrf2-mediated p62 expression. Further studies indicated that TRIM25 interacted with p62 and promoted its K63-linked ubiquitination via its E3 ligase activity and thus caused impairment of its oligomerization, aggregation, and recruitment for the autophagic protein LC3, leading to the suppression of autophagy activation. Besides, TRIM25 also suppressed the p62-mediated recruitment of ubiquitinated aggregates. Activation of autophagy decreased the accumulation of protein aggregates caused by TRIM25 overexpression, and inhibition of autophagy decreased the degradation of protein aggregates caused by TRIM25 knockdown. The current results also showed that TRIM25 inhibited PRRSV replication by inhibiting the KEAP1-Nrf2-p62 axis-mediated autophagy. Taken together, the present findings showed that the PRRSV replication restriction factor TRIM25 inhibited the degradation of ubiquitinated protein aggregates during viral infection by suppressing p62-mediated autophagy.IMPORTANCESequestration of protein aggregates and their subsequent degradation prevents proteostasis imbalance and cytotoxicity. The mechanisms controlling the turnover of protein aggregates during viral infection are mostly unknown. The present study found that porcine reproductive and respiratory syndrome virus (PRRSV) infection promoted the autophagic degradation of ubiquitinated protein aggregates, whereas tripartite motif-containing (TRIM)25 reversed this process. It was also found that TRIM25 promoted the expression of p62 by activating the Kelch-like ECH-associated protein 1 (KEAP1) and nuclear factor E2-related factor 2 (Nrf2) pathway and simultaneously prevented the oligomerization of p62 by promoting its K63-linked ubiquitination, thus suppressing its recruitment of the autophagic adaptor protein LC3 and ubiquitinated aggregates, leading to the inhibition of PRRSV-induced autophagy activation and the autophagic degradation of protein aggregates. The present study identified a new mechanism of protein aggregate turnover during viral infection and provided new insights for understanding the pathogenic mechanism of PRRSV.
Collapse
Affiliation(s)
- Jiahui Ren
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Qiming Pei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Haoxin Dong
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Xuedan Wei
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Liangliang Li
- College of Agronomy, Liaocheng University, Liaocheng, China
| | - Hong Duan
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
| | - Gaiping Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
| | - Angke Zhang
- College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- International Joint Research Center of National Animal Immunology, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, College of Veterinary Medicine, Henan Agricultural University, Zhengzhou, China
- Longhu Laboratory of Advanced Immunology, Zhengzhou, China
- Ministry of Education Key Laboratory for Animal Pathogens and Biosafety, Henan Agricultural University, Zhengzhou, China
| |
Collapse
|
2
|
Hamano S, Noguchi T, Asai Y, Ito R, Komatsu R, Sato T, Inoue A, Maruyama T, Kudo TA, Hirata Y, Shindo S, Uchida Y, Hwang GW, Matsuzawa A. Aggregability of the SQSTM1/p62-based aggresome-like induced structures determines the sensitivity to parthanatos. Cell Death Discov 2024; 10:74. [PMID: 38346947 PMCID: PMC10861449 DOI: 10.1038/s41420-024-01838-2] [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: 11/24/2023] [Revised: 01/28/2024] [Accepted: 01/30/2024] [Indexed: 02/15/2024] Open
Abstract
Overactivation of poly (ADP-ribose) polymerase-1 (PARP-1) triggers a noncanonical form of programmed cell death (PCD) called parthanatos, yet the mechanisms of its induction are not fully understood. We have recently demonstrated that the aggresome-like induced structures (ALIS) composed of the autophagy receptor SQSTM1/p62 and K48-linked polyubiquitinated proteins (p62-based ALIS) mediate parthanatos. In this study, we identified the D1 dopamine receptor agonist YM435 as a unique parthanatos inhibitor that acts as the disaggregating agent for the p62-based ALIS. We found that YM435 structurally reduces aggregability of the ALIS, and then increases its hydrophilicity and liquidity, which prevents parthanatos. Moreover, dopamine and L-DOPA, a dopamine precursor, also prevented parthanatos by reducing the aggregability of the ALIS. Together, these observations suggest that aggregability of the p62-based ALIS determines the sensitivity to parthanatos, and the pharmacological properties of YM435 that reduces the aggregability may be suitable for therapeutic drugs for parthanatos-related diseases such as neurodegenerative diseases.
Collapse
Affiliation(s)
- Shuhei Hamano
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| | - Yukino Asai
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryo Ito
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Ryuto Komatsu
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tetsu Sato
- Division of Membrane Transport and Drug Targeting, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Aya Inoue
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tomoe Maruyama
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Tada-Aki Kudo
- Division of Oral Physiology, Graduate School of Dentistry, Tohoku University, Sendai, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Sawako Shindo
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
- Department of Environmental Toxicology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Yasuo Uchida
- Department of Molecular Systems Pharmaceutics, Graduate School of Biomedical and Health Sciences, Hiroshima University, Hiroshima, Japan
| | - Gi-Wook Hwang
- Laboratory of Environmental and Health Sciences, Faculty of Pharmaceutical Sciences, Tohoku Medical and Pharmaceutical University, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
3
|
Shi X, Zhang Q, Chang M, Zhang Y, Zhao M, Yang B, Li P, Zhang Y. Ferroptosis is involved in passive Heymann nephritis in rats. Heliyon 2023; 9:e21050. [PMID: 37886789 PMCID: PMC10597846 DOI: 10.1016/j.heliyon.2023.e21050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2023] [Revised: 10/04/2023] [Accepted: 10/13/2023] [Indexed: 10/28/2023] Open
Abstract
Ferroptosis is found to be involved in some experimental models of kidney diseases, but its role in membrane nephropathy (MN) is still unclear. The purpose of this study is to explore whether ferroptosis occurred in MN, and the role of ferritinophagy. In this study, passive Heymann nephritis (PHN) rats were induced by single tail vein injection of anti-Fx1A serum, and normal rats were used as control. The changes of 24 h urinary protein, serum biochemical parameters, renal pathological damage, iron content, lipid peroxidation parameters, ferroptosis markers, and ferritinophagy markers were evaluated in the two groups. Compared with the control group, PHN rats showed obvious proteinuria, hypoproteinemia, and hyperlipidemia. Besides, more severe renal pathological damage and higher Fe2+ levels were observed in PHN rats, and the levels of malondialdehyde (MDA) increased significantly, while the levels of superoxide Dismutase (SOD) and glutathione (GSH) decreased. In addition, the expression of glutathione peroxidase 4 (GPX4) in renal tissues of PHN rats decreased significantly, while the expression of transferrin receptor (TFR) and acyl-CoA synthetase long-chain family member 4 (ACSL4) increased. The expression of microtubule associated protein 1 light chain 3 (LC3) II/LC3I and nuclear receptor coactivator 4 (NCOA4) increased significantly. Therefore, our study shows that ferroptosis is involved in the pathological damage of MN, and companied by activation of ferritinophagy.
Collapse
Affiliation(s)
- Xiujie Shi
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Qi Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Meiying Chang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yifan Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Beijing University of Chinese Medicine, Beijing, 100029, China
| | - MingMing Zhao
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Bin Yang
- Department of Pathology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Peng Li
- Experimental Research Center, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
| | - Yu Zhang
- Department of Nephrology, Xiyuan Hospital, China Academy of Chinese Medical Sciences, Beijing, 100091, China
- Xin-Huangpu Joint Innovation Institute of Chinese Medicine, Guangzhou, 510535, China
| |
Collapse
|
4
|
Cazzaro S, Zhao X, Zhao VK, Kim YK, Woo JAA. Slingshot homolog-1 amplifies mitochondrial abnormalities by distinctly impairing health and clearance of mitochondria. Hum Mol Genet 2023; 32:1660-1672. [PMID: 36637427 PMCID: PMC10162431 DOI: 10.1093/hmg/ddad006] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2022] [Revised: 12/23/2022] [Accepted: 01/10/2023] [Indexed: 01/14/2023] Open
Abstract
Accumulating toxic protein assemblies, including Aβ and tau, and dysfunctional mitochondria are associated with synaptic and neuronal loss in Alzheimer's disease (AD). Such accumulations are thought to be owing to clearance defects in the autophagy-lysosome pathway. Mitochondrial dysfunction is evident in AD brains and animal models at multiple levels, such as mitochondrial genomic mutations, disrupted bioenergetics, deregulated mitochondrial dynamics and impaired clearance of damaged mitochondria (mitophagy). Slingshot homolog-1 (SSH1) is a phosphatase activated by oxidative stress, high intracellular levels of Ca2+ and Aβ42 oligomers (Aβ42O), known for its function to dephosphorylate/activate cofilin through the N-terminal region. SSH1-mediated cofilin dephosphorylation results in Ab42O-induced severing of F-actin and translocation of cofilin to mitochondria, which promotes mitochondria-mediated apoptosis, synaptic loss and synaptic deficits. On the other hand, SSH1-mediated dephosphorylation/deactivation of the autophagy-cargo receptor p62 (SQSTM1), through its C-terminal region, inhibits p62 autophagy flux. However, the interplay between these two different activities of SSH1 in Aβ42O-induced mitochondrial toxicity remains unclear. In this study, we assessed the role of endogenous SSH1 and different regions of SSH1 in regulating mitochondrial health, mitochondrial respiration, clearance of damaged mitochondria and synaptic integrity in vitro and in vivo. Our results indicate that SSH1 suppresses mitochondrial health and respiration through the cofilin-binding N-terminal region, whereas SSH1 impairs mitophagy through a newly identified ~ 100 residue p62-binding domain in the C-terminal region. These results indicate that both N-terminal and C-terminal regions negatively impact mitochondria by distinct and independent modalities to amplify mitochondrial abnormalities, making SSH1 an excellent target to mitigate AD pathogenesis.
Collapse
Affiliation(s)
- Sara Cazzaro
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
- Department of Molecular Medicine, Byrd Alzheimer’s Center & Research Institute, USF Health Morsani College of Medicine, Tampa, FL 33613, USA
| | - Xingyu Zhao
- Department of Molecular Medicine, Byrd Alzheimer’s Center & Research Institute, USF Health Morsani College of Medicine, Tampa, FL 33613, USA
| | - Victoria K Zhao
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Yenna K Kim
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| | - Jung-A A Woo
- Department of Pathology, School of Medicine, Case Western Reserve University, Cleveland, OH 44106, USA
| |
Collapse
|
5
|
Yamada Y, Noguchi T, Suzuki M, Yamada M, Hirata Y, Matsuzawa A. Reactive sulfur species disaggregate the SQSTM1/p62-based aggresome-like induced structures via the HSP70 induction and prevent parthanatos. J Biol Chem 2023; 299:104710. [PMID: 37060999 DOI: 10.1016/j.jbc.2023.104710] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2022] [Revised: 03/28/2023] [Accepted: 03/30/2023] [Indexed: 04/17/2023] Open
Abstract
Reactive sulfur species (RSS) have emerged as key regulators of protein quality control. However, the mechanisms by which RSS contribute to cellular processes are not fully understood. In this study, we identified a novel function of RSS in preventing parthanatos, a non-apoptotic form of cell death that is induced by poly (ADP-ribose) polymerase-1 (PARP-1) and mediated by the aggresome-like induced structures (ALIS) composed of SQSTM1/p62. We found that sodium tetrasulfide (Na2S4), a donor of RSS, strongly suppressed oxidative stress-dependent ALIS formation and subsequent parthanatos. On the other hand, the inhibitors of the RSS-producing enzymes, such as 3-mercaptopyruvate sulfurtransferase (3-MST) and cystathionine γ-lyase (CSE), clearly enhanced ALIS formation and parthanatos. Interestingly, we found that Na2S4 activated heat shock factor 1 (HSF1) by promoting its dissociation from heat shock protein 90 (HSP90), leading to accelerated transcription of HSP70. Considering that the genetic deletion of HSP70 allowed the enhanced ALIS formation, these findings suggest that RSS prevent parthanatos by specifically suppressing ALIS formation through induction of HSP70. Taken together, our results demonstrate a novel mechanism by which RSS prevent cell death, as well as a novel physiological role of RSS in contributing to protein quality control through HSP70 induction, which may lead to better understanding of the bioactivity of RSS.
Collapse
Affiliation(s)
- Yutaro Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan
| | - Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan
| | - Midori Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan
| | - Mayuka Yamada
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, 980-8578, Sendai, Japan.
| |
Collapse
|
6
|
Baytas O, Kauer JA, Morrow EM. Loss of mitochondrial enzyme GPT2 causes early neurodegeneration in locus coeruleus. Neurobiol Dis 2022; 173:105831. [PMID: 35908744 PMCID: PMC9669404 DOI: 10.1016/j.nbd.2022.105831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2022] [Revised: 07/06/2022] [Accepted: 07/20/2022] [Indexed: 12/02/2022] Open
Abstract
Locus coeruleus (LC) is among the first brain areas to degenerate in Alzheimer’s disease and Parkinson’s disease; however, the underlying causes for the vulnerability of LC neurons are not well defined. Here we report a novel mechanism of degeneration of LC neurons caused by loss of the mitochondrial enzyme glutamate pyruvate transaminase 2 (GPT2). GPT2 Deficiency is a newly-recognized childhood neurometabolic disorder. The GPT2 enzyme regulates cell growth through replenishment of tricarboxylic acid (TCA) cycle intermediates and modulation of amino acid metabolism. In Gpt2-null mice, we observe an early loss of tyrosine hydroxylase (TH)-positive neurons in LC and reduced soma size at postnatal day 18. Gpt2-null LC shows selective positive Fluoro-Jade C staining. Neuron loss is accompanied by selective, prominent microgliosis and astrogliosis in LC. We observe reduced noradrenergic projections to and norepinephrine levels in hippocampus and spinal cord. Whole cell recordings in Gpt2-null LC slices show reduced soma size and abnormal action potentials with altered firing kinetics. Strikingly, we observe early decreases in phosphorylated S6 in Gpt2-null LC, preceding prominent p62 aggregation, increased LC3B-II to LC3B-I ratio, and neuronal loss. These data are consistent with a possible mechanism involving deficiency in protein synthesis and cell growth, associated subsequently with abnormal autophagy and neurodegeneration. As compared to the few genetic animal models with LC degeneration, loss of LC neurons in Gpt2-null mice is developmentally the earliest. Early neuron loss in LC in a model of human neurometabolic disease provides important clues regarding the metabolic vulnerability of LC and may lead to new therapeutic targets.
Collapse
Affiliation(s)
- Ozan Baytas
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI 02912, USA; Neuroscience Graduate Program, Brown University, Providence, RI 02912, USA
| | - Julie A Kauer
- Department of Psychiatry and Behavioral Sciences, Stanford University School of Medicine, Stanford, CA 94035, USA
| | - Eric M Morrow
- Department of Molecular Biology, Cell Biology and Biochemistry, Brown University, Providence, RI 02912, USA; Center for Translational Neuroscience, Carney Institute for Brain Science and Brown Institute for Translational Science, Brown University, Providence, RI 02912, USA.
| |
Collapse
|
7
|
Palminha NM, Dos Santos Souza C, Griffin J, Liao C, Ferraiuolo L, El-Khamisy SF. Defective repair of topoisomerase I induced chromosomal damage in Huntington's disease. Cell Mol Life Sci 2022; 79:160. [PMID: 35224690 PMCID: PMC8882575 DOI: 10.1007/s00018-022-04204-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2021] [Revised: 02/06/2022] [Accepted: 02/08/2022] [Indexed: 11/30/2022]
Abstract
Topoisomerase1 (TOP1)-mediated chromosomal breaks are endogenous sources of DNA damage that affect neuronal genome stability. Whether TOP1 DNA breaks are sources of genomic instability in Huntington's disease (HD) is unknown. Here, we report defective 53BP1 recruitment in multiple HD cell models, including striatal neurons derived from HD patients. Defective 53BP1 recruitment is due to reduced H2A ubiquitination caused by the limited RNF168 activity. The reduced availability of RNF168 is caused by an increased interaction with p62, a protein involved in selective autophagy. Depletion of p62 or disruption of the interaction between RNAF168 and p62 was sufficient to restore 53BP1 enrichment and subsequent DNA repair in HD models, providing new opportunities for therapeutic interventions. These findings are reminiscent to what was described for p62 accumulation caused by C9orf72 expansion in ALS/FTD and suggest a common mechanism by which protein aggregation perturb DNA repair signaling.
Collapse
Affiliation(s)
- Nelma M Palminha
- School of Biosciences, Firth Court, Healthy Lifespan and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Cleide Dos Santos Souza
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Jon Griffin
- School of Biosciences, Firth Court, Healthy Lifespan and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Chunyan Liao
- School of Biosciences, Firth Court, Healthy Lifespan and Neuroscience Institute, University of Sheffield, Sheffield, UK
| | - Laura Ferraiuolo
- Sheffield Institute for Translational Neuroscience (SITraN), University of Sheffield, Sheffield, UK
| | - Sherif F El-Khamisy
- School of Biosciences, Firth Court, Healthy Lifespan and Neuroscience Institute, University of Sheffield, Sheffield, UK.
- Institute of Cancer Therapeutics, University of Bradford, Bradford, UK.
| |
Collapse
|
8
|
Cui M, Yoshimori T, Nakamura S. Autophagy system as a potential therapeutic target for neurodegenerative diseases. Neurochem Int 2022; 155:105308. [PMID: 35181396 DOI: 10.1016/j.neuint.2022.105308] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2021] [Revised: 01/17/2022] [Accepted: 02/13/2022] [Indexed: 12/19/2022]
Abstract
Autophagy is an evolutionally conserved process by which cytoplasmic contents including protein aggregates and damaged organelles such as mitochondria and lysosomes, are sequestered by double-membrane structure, autophagosomes, and delivered to the lysosomes for degradation. Recently, considerable efforts have been made to reveal the role of autophagy in neurodegenerative diseases like Alzheimer's disease, Parkinson's disease and Huntington's disease. Impairment of autophagy aggravates the accumulation of misfolded protein and damaged organelles in neurons, while sufficient autophagic activity reduces such accumulation in nervous system and ameliorates the pathology. Here we summarize recent progress regarding the role of autophagy in several neurodegenerative diseases and the potential autophagy-associated therapies for them.
Collapse
Affiliation(s)
- Mengying Cui
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Tamotsu Yoshimori
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan.
| | - Shuhei Nakamura
- Department of Genetics, Graduate School of Medicine, Osaka University, Osaka, Japan; Department of Intracellular Membrane Dynamics, Graduate School of Frontier Biosciences, Osaka University, Osaka, Japan; Institute for Advanced Co-Creation Studies, Osaka University, Osaka, Japan.
| |
Collapse
|
9
|
Davidson JM, Chung RS, Lee A. The converging roles of sequestosome-1/p62 in the molecular pathways of amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD). Neurobiol Dis 2022; 166:105653. [PMID: 35143965 DOI: 10.1016/j.nbd.2022.105653] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 01/18/2022] [Accepted: 02/03/2022] [Indexed: 01/03/2023] Open
Abstract
Investigations into the pathogenetic mechanisms underlying amyotrophic lateral sclerosis (ALS) and frontotemporal dementia (FTD) have provided significant insight into the disease. At the cellular level, ALS and FTD are classified as proteinopathies, which is motor neuron degeneration and death characterized by pathological protein aggregates or dysregulated proteostasis. At both the clinical and molecular level there are common signaling pathways dysregulated across the ALS and FTD spectrum (ALS/FTD). Sequestosome-1/p62 is a multifunctional scaffold protein with roles in several signaling pathways including proteostasis, protein degradation via the ubiquitin proteasome system and autophagy, the antioxidant response, inflammatory response, and apoptosis. Notably these pathways are dysregulated in ALS and FTD. Mutations in the functional domains of p62 provide links to the pathogenetic mechanisms of p62 and dyshomeostasis of p62 levels is noted in several types of ALS and FTD. We present here that the dysregulated ALS and FTD signaling pathways are linked, with p62 converging the molecular mechanisms. This review summarizes the current literature on the complex role of p62 in the pathogenesis across the ALS/FTD spectrum. The focus is on the underlying convergent molecular mechanisms of ALS and FTD-associated proteins and pathways that dysregulate p62 levels or are dysregulated by p62, with emphasis on how p62 is implicated across the ALS/FTD spectrum.
Collapse
Affiliation(s)
- Jennilee M Davidson
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| | - Roger S Chung
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| | - Albert Lee
- Centre for Motor Neuron Disease Research, Macquarie Medical School, Faculty of Medicine, Health and Human Sciences, Macquarie University, 2 Technology Place, NSW 2109, Australia..
| |
Collapse
|
10
|
The caspase-6-p62 axis modulates p62 droplets based autophagy in a dominant-negative manner. Cell Death Differ 2021; 29:1211-1227. [PMID: 34862482 PMCID: PMC9178044 DOI: 10.1038/s41418-021-00912-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2021] [Revised: 11/22/2021] [Accepted: 11/22/2021] [Indexed: 12/15/2022] Open
Abstract
SQSTM1/p62, as a major autophagy receptor, forms droplets that are critical for cargo recognition, nucleation, and clearance. p62 droplets also function as liquid assembly platforms to allow the formation of autophagosomes at their surfaces. It is unknown how p62-droplet formation is regulated under physiological or pathological conditions. Here, we report that p62-droplet formation is selectively blocked by inflammatory toxicity, which induces cleavage of p62 by caspase-6 at a novel cleavage site D256, a conserved site across human, mouse, rat, and zebrafish. The N-terminal cleavage product is relatively stable, whereas the C-terminal product appears undetectable. Using a variety of cellular models, we show that the p62 N-terminal caspase-6 cleavage product (p62-N) plays a dominant-negative role to block p62-droplet formation. In vitro p62 phase separation assays confirm this observation. Dominant-negative regulation of p62-droplet formation by caspase-6 cleavage attenuates p62 droplets dependent autophagosome formation. Our study suggests a novel pathway to modulate autophagy through the caspase-6–p62 axis under certain stress stimuli.
Collapse
|
11
|
Macroautophagy and Mitophagy in Neurodegenerative Disorders: Focus on Therapeutic Interventions. Biomedicines 2021; 9:biomedicines9111625. [PMID: 34829854 PMCID: PMC8615936 DOI: 10.3390/biomedicines9111625] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2021] [Revised: 11/01/2021] [Accepted: 11/03/2021] [Indexed: 02/06/2023] Open
Abstract
Macroautophagy, a quality control mechanism, is an evolutionarily conserved pathway of lysosomal degradation of protein aggregates, pathogens, and damaged organelles. As part of its vital homeostatic role, macroautophagy deregulation is associated with various human disorders, including neurodegenerative diseases. There are several lines of evidence that associate protein misfolding and mitochondrial dysfunction in the etiology of Alzheimer’s, Parkinson’s, and Huntington’s diseases. Macroautophagy has been implicated in the degradation of different protein aggregates such as Aβ, tau, alpha-synuclein (α-syn), and mutant huntingtin (mHtt) and in the clearance of dysfunctional mitochondria. Taking these into consideration, targeting autophagy might represent an effective therapeutic strategy to eliminate protein aggregates and to improve mitochondrial function in these disorders. The present review describes our current understanding on the role of macroautophagy in neurodegenerative disorders and focuses on possible strategies for its therapeutic modulation.
Collapse
|
12
|
Demasi M, Augusto O, Bechara EJH, Bicev RN, Cerqueira FM, da Cunha FM, Denicola A, Gomes F, Miyamoto S, Netto LES, Randall LM, Stevani CV, Thomson L. Oxidative Modification of Proteins: From Damage to Catalysis, Signaling, and Beyond. Antioxid Redox Signal 2021; 35:1016-1080. [PMID: 33726509 DOI: 10.1089/ars.2020.8176] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The systematic investigation of oxidative modification of proteins by reactive oxygen species started in 1980. Later, it was shown that reactive nitrogen species could also modify proteins. Some protein oxidative modifications promote loss of protein function, cleavage or aggregation, and some result in proteo-toxicity and cellular homeostasis disruption. Recent Advances: Previously, protein oxidation was associated exclusively to damage. However, not all oxidative modifications are necessarily associated with damage, as with Met and Cys protein residue oxidation. In these cases, redox state changes can alter protein structure, catalytic function, and signaling processes in response to metabolic and/or environmental alterations. This review aims to integrate the present knowledge on redox modifications of proteins with their fate and role in redox signaling and human pathological conditions. Critical Issues: It is hypothesized that protein oxidation participates in the development and progression of many pathological conditions. However, no quantitative data have been correlated with specific oxidized proteins or the progression or severity of pathological conditions. Hence, the comprehension of the mechanisms underlying these modifications, their importance in human pathologies, and the fate of the modified proteins is of clinical relevance. Future Directions: We discuss new tools to cope with protein oxidation and suggest new approaches for integrating knowledge about protein oxidation and redox processes with human pathophysiological conditions. Antioxid. Redox Signal. 35, 1016-1080.
Collapse
Affiliation(s)
- Marilene Demasi
- Laboratório de Bioquímica e Biofísica, Instituto Butantan, São Paulo, Brazil
| | - Ohara Augusto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Etelvino J H Bechara
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Renata N Bicev
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Fernanda M Cerqueira
- CENTD, Centre of Excellence in New Target Discovery, Instituto Butantan, São Paulo, Brazil
| | - Fernanda M da Cunha
- Departamento de Bioquímica, Escola Paulista de Medicina, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Ana Denicola
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Sayuri Miyamoto
- Departamento de Bioquímica and Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, Brazil
| | - Lía M Randall
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| | - Cassius V Stevani
- Departamento de Química Fundamental, Instituto de Química, Universidade de São Paulo, São Paulo, Brazil
| | - Leonor Thomson
- Laboratorios Fisicoquímica Biológica-Enzimología, Facultad de Ciencias, Instituto de Química Biológica, Universidad de la República, Montevideo, Uruguay
| |
Collapse
|
13
|
Somlapura M, Gottschalk B, Lahiri P, Kufferath I, Pabst D, Rülicke T, Graier WF, Denk H, Zatloukal K. Different Roles of p62 (SQSTM1) Isoforms in Keratin-Related Protein Aggregation. Int J Mol Sci 2021; 22:ijms22126227. [PMID: 34207662 PMCID: PMC8227998 DOI: 10.3390/ijms22126227] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2021] [Revised: 06/01/2021] [Accepted: 06/05/2021] [Indexed: 12/03/2022] Open
Abstract
p62/Sequestosome-1 (p62) is a multifunctional adaptor protein and is also a constant component of disease-associated protein aggregates, including Mallory–Denk bodies (MDBs), in steatohepatitis and hepatocellular carcinoma. We investigated the interaction of the two human p62 isoforms, p62-H1 (full-length isoform) and p62-H2 (partly devoid of PB1 domain), with keratins 8 and 18, the major components of MDBs. In human liver, p62-H2 is expressed two-fold higher compared to p62-H1 at the mRNA level and is present in slightly but not significantly higher concentrations at the protein level. Co-transfection studies in CHO-K1 cells, PLC/PRF/5 cells as well as p62− total-knockout and wild-type mouse fibroblasts revealed marked differences in the cytoplasmic distribution and aggregation behavior of the two p62 isoforms. Transfection-induced overexpression of p62-H2 generated large cytoplasmic aggregates in PLC/PRF/5 and CHO-K1 cells that mostly co-localized with transfected keratins resembling MDBs or (transfection without keratins) intracytoplasmic hyaline bodies. In fibroblasts, however, transfected p62-H2 was predominantly diffusely distributed in the cytoplasm. Aggregation of p62-H2 and p62ΔSH2 as well as the interaction with K8 (but not with K18) involves acquisition of cross-β-sheet conformation as revealed by staining with luminescent conjugated oligothiophenes. These results indicate the importance of considering p62 isoforms in protein aggregation disease.
Collapse
Affiliation(s)
- Meghana Somlapura
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (M.S.); (I.K.); (D.P.); (H.D.)
| | - Benjamin Gottschalk
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (B.G.); (W.F.G.)
| | - Pooja Lahiri
- School of Medical Science and Technology, Indian Institute of Technology Kharagpur, Kharagpur 721302, India;
| | - Iris Kufferath
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (M.S.); (I.K.); (D.P.); (H.D.)
| | - Daniela Pabst
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (M.S.); (I.K.); (D.P.); (H.D.)
| | - Thomas Rülicke
- Department of Biomedical Sciences, University of Veterinary Medicine Vienna, 1210 Vienna, Austria;
| | - Wolfgang F. Graier
- Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Molecular Biology and Biochemistry, Medical University of Graz, 8010 Graz, Austria; (B.G.); (W.F.G.)
| | - Helmut Denk
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (M.S.); (I.K.); (D.P.); (H.D.)
| | - Kurt Zatloukal
- Diagnostic and Research Center for Molecular Biomedicine, Institute of Pathology, Medical University of Graz, 8010 Graz, Austria; (M.S.); (I.K.); (D.P.); (H.D.)
- Correspondence: ; Tel.: +43-(0)316-385-71731
| |
Collapse
|
14
|
Sunchu B, Riordan RT, Yu Z, Almog I, Dimas-Munoz J, Drake AC, Perez VI. Aggresome-Like Formation Promotes Resistance to Proteotoxicity in Cells from Long-Lived Species. J Gerontol A Biol Sci Med Sci 2020; 75:1439-1447. [PMID: 32515471 PMCID: PMC7357592 DOI: 10.1093/gerona/glaa069] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Indexed: 11/13/2022] Open
Abstract
The capacity of cells to maintain proteostasis declines with age, causing rapid accumulation of damaged proteins and protein aggregates, which plays an important role in age-related disease etiology. While our group and others have identified that proteostasis is enhanced in long-lived species, there are no data on whether this leads to better resistance to proteotoxicity. We compared the sensitivity of cells from long- (naked mole rat [NMR]) and short- (Mouse) lived species to proteotoxicity, by measuring the survival of fibroblasts under polyglutamine (polyQ) toxicity, a well-established model of protein aggregation. Additionally, to evaluate the contribution of proteostatic mechanisms to proteotoxicity resistance, we down-regulated a key protein of each mechanism (autophagy-ATG5; ubiquitin-proteasome-PSMD14; and chaperones-HSP27) in NMR fibroblasts. Furthermore, we analyzed the formation and subcellular localization of inclusions in long- and short-lived species. Here, we show that fibroblasts from long-lived species are more resistant to proteotoxicity than their short-lived counterparts. Surprisingly, this does not occur because the NMR cells have less polyQ82 protein aggregates, but rather they have an enhanced capacity to handle misfolded proteins and form protective perinuclear and aggresome-like inclusions. All three proteostatic mechanisms contribute to this resistance to polyQ toxicity but autophagy has the greatest effect. Overall, our data suggest that the resistance to proteotoxicity observed in long-lived species is not due to a lower level of protein aggregates but rather to enhanced handling of the protein aggregates through the formation of aggresome-like inclusions, a well-recognized protective mechanism against proteotoxicty.
Collapse
Affiliation(s)
- Bharath Sunchu
- Linus Pauling Institute, Oregon State University, Corvallis
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Ruben T Riordan
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Zhen Yu
- Linus Pauling Institute, Oregon State University, Corvallis
| | - Ido Almog
- Linus Pauling Institute, Oregon State University, Corvallis
| | - Jovita Dimas-Munoz
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Andrew C Drake
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| | - Viviana I Perez
- Linus Pauling Institute, Oregon State University, Corvallis
- Department of Biochemistry and Biophysics, Oregon State University, Corvallis
| |
Collapse
|
15
|
Yamanaka T, Nishiyama R, Shimogori T, Nukina N. Proteomics-Based Approach Identifies Altered ER Domain Properties by ALS-Linked VAPB Mutation. Sci Rep 2020; 10:7610. [PMID: 32376919 PMCID: PMC7203144 DOI: 10.1038/s41598-020-64517-z] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2019] [Accepted: 04/17/2020] [Indexed: 12/13/2022] Open
Abstract
An ER transmembrane protein, vesicle-associated membrane protein-associated protein B (VAPB), binds to several organelle-resident membrane proteins to mediate ER-organelle tethering. Mutation in amyotrophic lateral sclerosis (ALS) induces protein misfolding and aggregation, leading to ER disorganization. Gain or loss of function is suggested for VAPB mutation, however comprehensive study focusing on VAPB-ER domain has yet been performed. We here conducted proteomic characterization of the ER containing VAPB and its ALS-linked P56S mutant. For this purpose, we first optimized the proteomics of different ER domains immuno-isolated from cultured cells, and identified ER sheet- and tubule-specific proteomes. By using these as references, we found that VAPB-ER proteome had intermediate ER domain properties but its tubular property was specifically decreased by its mutation. Biochemical, immunofluorescence and proximity ligation assays suggested this was mediated by delocalization of VAPB from ER tubules. The VAPB-ER proteomics further suggested reduced incorporation of multiple proteins located in different organelles, which was confirmed by proximity ligation assay. Taken together, our proteomics-based approach indicates altered ER domain properties and impaired ER-organelle tethering by VAPB mutation.
Collapse
Affiliation(s)
- Tomoyuki Yamanaka
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, Kyoto, Japan.
| | - Risa Nishiyama
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, Kyoto, Japan
| | - Tomomi Shimogori
- Molecular Mechanisms of Brain Development, RIKEN Center for Brain Science, Saitama, 351-0198, Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, Kyoto, Japan.
| |
Collapse
|
16
|
Lin DS, Ho CS, Huang YW, Wu TY, Lee TH, Huang ZD, Wang TJ, Yang SJ, Chiang MF. Impairment of Proteasome and Autophagy Underlying the Pathogenesis of Leukodystrophy. Cells 2020; 9:E1124. [PMID: 32370022 PMCID: PMC7290671 DOI: 10.3390/cells9051124] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2020] [Revised: 04/24/2020] [Accepted: 04/28/2020] [Indexed: 01/01/2023] Open
Abstract
Impairment of the ubiquitin-proteasome-system (UPS) and autophagy causing cytoplasmic aggregation of ubiquitin andp62 have been implicated in the pathogenesis of most neurodegenerative disorders, yet, they have not been fully elucidated in leukodystrophies. The relationship among impairment of UPS, autophagy, and globoid cell leukodystrophy (GLD), one of the most common demyelinating leukodystrophies, is clarified in this study. We examined the ubiquitin and autophagy markers in the brains of twitcher mice, a murine model of infantile GLD, and in human oligodendrocytes incubated with psychosine. Immunohistochemical examinations showed spatiotemporal accumulation of ubiquitin- and p62-aggregates mainly in the white matter of brain and spinal cord at disease progression. Western blot analysis demonstrated a significant accumulation of ubiquitin, p62, and LC3-II in insoluble fraction in parallel with progressive demyelination and neuroinflammation in twitcher brains. In vitro study validated a dose- and time-dependent cytotoxicity of psychosine upon autophagy and UPS machinery. Inhibition of autophagy and UPS exacerbated the accumulation of insoluble ubiquitin, p62, and LC3-II proteins mediated by psychosine cytotoxicity as well as increased cytoplasmic deposition of ubiquitin- and p62-aggregates, and accumulation of autophagosomes and autolysosomes. Further, the subsequent accumulation of reactive oxygen species and reduction of mitochondrial respiration led to cell death. Our studies validate the impairment of proteasome and autophagy underlying the pathogenesis of GLD. These findings provide a novel insight into pathogenesis of GLD and suggest a specific pathomechanism as an ideal target for therapeutic approaches.
Collapse
Affiliation(s)
- Dar-Shong Lin
- Department of Pediatrics, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Department of Medicine and Institute of Biomedical Sciences, Mackay Medical College, New Taipei 25245, Taiwan
| | - Che-Sheng Ho
- Department of Pediatric Neurology, Mackay Memorial Hospital, Taipei 10449, Taiwan;
| | - Yu-Wen Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (T.-Y.W.); (T.-H.L.); (Z.-D.H.); (S.-J.Y.)
| | - Tsu-Yen Wu
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (T.-Y.W.); (T.-H.L.); (Z.-D.H.); (S.-J.Y.)
| | - Tsung-Han Lee
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (T.-Y.W.); (T.-H.L.); (Z.-D.H.); (S.-J.Y.)
| | - Zo-Darr Huang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (T.-Y.W.); (T.-H.L.); (Z.-D.H.); (S.-J.Y.)
| | - Tuan-Jen Wang
- Department of Laboratory Medicine, Mackay Memorial Hospital, Taipei 10449, Taiwan;
| | - Shun-Jie Yang
- Department of Medical Research, Mackay Memorial Hospital, Taipei 10449, Taiwan; (Y.-W.H.); (T.-Y.W.); (T.-H.L.); (Z.-D.H.); (S.-J.Y.)
| | - Ming-Fu Chiang
- Department of Neurosurgery, Mackay Memorial Hospital, Taipei 10449, Taiwan
- Mackay Medicine, Nursing and Management College, Taipei 11260, Taiwan
- Graduate Institute of Injury Prevention and Control, Taipei Medical University, Taipei 11031, Taiwan
| |
Collapse
|
17
|
Homma T, Fujii J. Emerging connections between oxidative stress, defective proteolysis, and metabolic diseases. Free Radic Res 2020; 54:931-946. [DOI: 10.1080/10715762.2020.1734588] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata, Japan
| |
Collapse
|
18
|
López-Pérez Ó, Toivonen JM, Otero A, Solanas L, Zaragoza P, Badiola JJ, Osta R, Bolea R, Martín-Burriel I. Impairment of autophagy in scrapie-infected transgenic mice at the clinical stage. J Transl Med 2020; 100:52-63. [PMID: 31477795 DOI: 10.1038/s41374-019-0312-z] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2019] [Revised: 07/23/2019] [Accepted: 07/25/2019] [Indexed: 12/24/2022] Open
Abstract
Autophagy appears to play a role in the etiology and progress of misfolded protein disorders. Although this process is dysregulated in prion diseases, it is unknown whether this impairment is a cause or a consequence of prion neuropathology. The study of autophagy during the progress of the disease could elucidate its role. For this purpose, we have investigated its regulation at different stages of the disease in Tg338 mice, a transgenic murine model that overexpresses the highly susceptible ovine VRQ prion protein allele. Mice were intracerebrally inoculated with mouse-adapted classical scrapie and euthanized at the preclinical and clinical stages of the disease. Regulation of autophagy was investigated analyzing the distribution of LC3-B and p62 proteins by immunohistochemistry. Moreover, the expression of genes involved in autophagy regulation was quantified by real-time PCR. LC3-B and p62 proteins were downregulated and upregulated, respectively, in the central nervous system of infected mice with clinical signs of scrapie. Accumulation of p62 correlated with scrapie-related lesions, suggesting an impairment of autophagy in highly prion-affected areas. In addition, Gas5 (growth arrest-specific 5), Atg5 (autophagy-related 5), and Fbxw7 (F-box and WD repeat domain containing 7) transcripts were downregulated in mesencephalon and cervical spinal cord of the same group of animals. The impairment of autophagic machinery seems to be part of the pathological process of scrapie, but only during the late stage of prion infection. Similarities between Tg338 mice and the natural ovine disease make them a reliable in vivo model to study prion infection and autophagy side by side.
Collapse
Affiliation(s)
- Óscar López-Pérez
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain.,Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain
| | - Janne Markus Toivonen
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Alicia Otero
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain
| | - Laura Solanas
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain
| | - Pilar Zaragoza
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Juan José Badiola
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain
| | - Rosario Osta
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Rosa Bolea
- Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain
| | - Inmaculada Martín-Burriel
- Laboratorio de Genética Bioquímica (LAGENBIO), Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain. .,Centro de Encefalopatías y Enfermedades Transmisibles Emergentes, Universidad de Zaragoza, IA2, IIS Aragón, 50013, Zaragoza, Spain.
| |
Collapse
|
19
|
Valionyte E, Yang Y, Roberts SL, Kelly J, Lu B, Luo S. Lowering Mutant Huntingtin Levels and Toxicity: Autophagy-Endolysosome Pathways in Huntington's Disease. J Mol Biol 2019; 432:2673-2691. [PMID: 31786267 DOI: 10.1016/j.jmb.2019.11.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2019] [Revised: 11/04/2019] [Accepted: 11/19/2019] [Indexed: 02/06/2023]
Abstract
Huntington's disease (HD) is a monogenetic neurodegenerative disease, which serves as a model of neurodegeneration with protein aggregation. Autophagy has been suggested to possess a great value to tackle protein aggregation toxicity and neurodegenerative diseases. Current studies suggest that autophagy-endolysosomal pathways are critical for HD pathology. Here we review recent advancement in the studies of autophagy and selective autophagy relating HD. Restoration of autophagy flux and enhancement of selective removal of mutant huntingtin/disease-causing protein would be effective approaches towards tackling HD as well as other similar neurodegenerative disorders.
Collapse
Affiliation(s)
- Evelina Valionyte
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth PL6 8BU, UK
| | - Yi Yang
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth PL6 8BU, UK
| | - Sheridan L Roberts
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth PL6 8BU, UK
| | - Jack Kelly
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth PL6 8BU, UK
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology, Collaborative Innovation Center for Brain Science, School of Life Sciences, Fudan University, Shanghai 200438, China
| | - Shouqing Luo
- Peninsula Schools of Medicine and Dentistry, Institute of Translational and Stratified Medicine, University of Plymouth, Research Way, Plymouth PL6 8BU, UK.
| |
Collapse
|
20
|
Cellular Responses to Proteasome Inhibition: Molecular Mechanisms and Beyond. Int J Mol Sci 2019; 20:ijms20143379. [PMID: 31295808 PMCID: PMC6678303 DOI: 10.3390/ijms20143379] [Citation(s) in RCA: 29] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Revised: 06/26/2019] [Accepted: 07/01/2019] [Indexed: 02/06/2023] Open
Abstract
Proteasome inhibitors have been actively tested as potential anticancer drugs and in the treatment of inflammatory and autoimmune diseases. Unfortunately, cells adapt to survive in the presence of proteasome inhibitors activating a variety of cell responses that explain why these therapies have not fulfilled their expected results. In addition, all proteasome inhibitors tested and approved by the FDA have caused a variety of side effects in humans. Here, we describe the different types of proteasome complexes found within cells and the variety of regulators proteins that can modulate their activities, including those that are upregulated in the context of inflammatory processes. We also summarize the adaptive cellular responses activated during proteasome inhibition with special emphasis on the activation of the Autophagic-Lysosomal Pathway (ALP), proteaphagy, p62/SQSTM1 enriched-inclusion bodies, and proteasome biogenesis dependent on Nrf1 and Nrf2 transcription factors. Moreover, we discuss the role of IRE1 and PERK sensors in ALP activation during ER stress and the involvement of two deubiquitinases, Rpn11 and USP14, in these processes. Finally, we discuss the aspects that should be currently considered in the development of novel strategies that use proteasome activity as a therapeutic target for the treatment of human diseases.
Collapse
|
21
|
Homma T, Fujii J. An SOD1 deficiency aggravates proteasome inhibitor bortezomib-induced testicular damage in mice. Biochim Biophys Acta Gen Subj 2019; 1863:1108-1115. [PMID: 30974160 DOI: 10.1016/j.bbagen.2019.04.005] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2019] [Revised: 03/23/2019] [Accepted: 04/05/2019] [Indexed: 10/27/2022]
Abstract
Proteasomes play a key role in maintaining cellular homeostasis by the proteolytic removal of proteins, including ubiquitinated proteins and/or oxidatively-damaged proteins. The proteasome inhibitor bortezomib (BTZ) has been reported to exert testicular toxicity in mice. In the current study, we treated SOD1-knockout (KO) mice with BTZ and investigated the issue of whether oxidative stress is involved in the development of testicular toxicity. The BTZ treatment significantly increased superoxide production and cell death in the testes of SOD1-KO mice compared to wild-type (WT) mice. We also found that high levels of both ubiquitinated proteins and p62 accumulated and underwent aggregation in the seminiferous tubules of BTZ-injected SOD1-KO mice. Furthermore, the proteolytic activities of proteasomes were significantly decreased in the testes of BTZ-injected SOD1-KO mice compared to their WT counterparts. These results suggest that a combination of oxidative stress caused by an SOD1 deficiency and proteasome inhibition by BTZ accelerates the impairment of proteasomes, which results in severe testicular damage in SOD1-KO mice.
Collapse
Affiliation(s)
- Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan.
| | - Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata 990-9585, Japan
| |
Collapse
|
22
|
Autophagy as a Homeostatic Mechanism in Response to Stress Conditions in the Central Nervous System. Mol Neurobiol 2019; 56:6594-6608. [DOI: 10.1007/s12035-019-1546-x] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 03/12/2019] [Indexed: 12/11/2022]
|
23
|
Acute expression of the transcription factor Nrf2 after treatment with quinolinic acid is not induced by oxidative stress in the rat striatum. Neurotoxicology 2019; 73:120-131. [PMID: 30876764 DOI: 10.1016/j.neuro.2019.03.003] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2018] [Revised: 02/26/2019] [Accepted: 03/10/2019] [Indexed: 11/20/2022]
Abstract
Quinolinic acid (QUIN) is an excitotoxic and pro-oxidant molecule used in the study of neurodegenerative disorders because it reproduces certain biochemical characteristics present in these diseases. The use of antioxidant molecules in the QUIN model reduces cellular damage through the nuclear factor erythroid 2-related to factor 2 (Nrf2) pathway. The Nrf2 transcription factor is considered the master regulator of antioxidant genes expression, and its activation occurs by an increase in the reactive oxygen species (ROS) levels or in the presence of electrophilic compounds. However, Nrf2 activation also occurs in an oxidative stress-independent process caused by the disruption of the Keap1-Nrf2 complex by the direct interaction of Keap1 with certain proteins, such as DPP3 and p62. The aim of this study was to evaluate the effect of QUIN on Nrf2 activation over short periods of time. QUIN administration increased Nrf2 activation at 30 min in the striatum without increasing ROS production or modifying the redox cellular state. Moreover, QUIN increased Keap1 and Nrf2 nuclear levels and increased the protein-protein interaction between Keap1 and DPP3 and Keap1 and p62 30 min after QUIN administration. Finally, we found that Nrf2 activation primarily occurs in striatal neurons. Our results show that QUIN administration in vivo stimulates Nrf2 expression and activation in the absence of oxidative stress primarily in neurons and increases the interaction of p62 and DPP3 with Keap1, which could participate in Nrf2 activation.
Collapse
|
24
|
van Echten-Deckert G, Alam S. Sphingolipid metabolism - an ambiguous regulator of autophagy in the brain. Biol Chem 2019; 399:837-850. [PMID: 29908127 DOI: 10.1515/hsz-2018-0237] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Accepted: 05/25/2018] [Indexed: 01/12/2023]
Abstract
In mammals, the brain exhibits the highest lipid content in the body next to adipose tissue. Complex sphingolipids are characteristic compounds of neuronal membranes. Vital neural functions including information flux and transduction occur along these membranes. It is therefore not surprising that neuronal function and survival is dependent on the metabolism of these lipids. Autophagy is a critical factor for the survival of post-mitotic neurons. On the one hand, it fulfils homeostatic and waste-recycling functions and on the other hand, it constitutes an effective strategy to eliminate harmful proteins that cause neuronal death. A growing number of experimental data indicate that several sphingolipids as well as enzymes catalyzing their metabolic transformations efficiently but very differently affect neuronal autophagy and hence survival. This review attempts to elucidate the roles and mechanisms of sphingolipid metabolism with regard to the regulation of autophagy and its consequences for brain physiology and pathology.
Collapse
Affiliation(s)
- Gerhild van Echten-Deckert
- LIMES Institute, Unit Membrane Biology and Lipid Biochemistry, Kekulé-Institute of the University Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| | - Shah Alam
- LIMES Institute, Unit Membrane Biology and Lipid Biochemistry, Kekulé-Institute of the University Bonn, Gerhard-Domagk-Str. 1, D-53121 Bonn, Germany
| |
Collapse
|
25
|
Xu C, Chen X, Sheng WB, Yang P. Trehalose restores functional autophagy suppressed by high glucose. Reprod Toxicol 2019; 85:51-58. [PMID: 30769031 DOI: 10.1016/j.reprotox.2019.02.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2018] [Revised: 01/17/2019] [Accepted: 02/09/2019] [Indexed: 12/21/2022]
Abstract
Autophagy is required for neurulation, and autophagy activators with minimal toxicity, such as the natural compound trehalose, a nonreducing disaccharide, possess high therapeutic value. To determine whether trehalose directly induces autophagy, FITC-labeled trehalose was used for tracing its presence in autophagosome complexes. Trehalose was as potent as rapamycin and starvation in inducing de novo autophagosome formation and increasing autophagosome flux in GFP-LC3 reporter cells and C17.2 neural stem cells. Trehalose effectively reversed high glucose-suppressed autophagy and reduced p62 protein expression. Trehalose abolished the disruption of autophagosome complexes under high glucose conditions in vitro and maternal diabetes in vivo. Autophagosomes induced by trehalose were functionally active, forming mitophagy and reticulophagy in removing damaged cellular organelles in neuroepithelial cells exposed to maternal diabetes. Thus, trehalose directly participated in functional autophagosome generation by incorporating itself into autophagosomes. These findings provide the mechanistic basis for the use of trehalose in preventing disruptive autophagy-associated pathogenesis.
Collapse
Affiliation(s)
- Cheng Xu
- Department of Obstetrics, Gynecology & Reproductive Sciences, Baltimore, MD, USA
| | - Xi Chen
- Department of Obstetrics, Gynecology & Reproductive Sciences, Baltimore, MD, USA
| | - Wei-Bin Sheng
- Department of Obstetrics, Gynecology & Reproductive Sciences, Baltimore, MD, USA
| | - Peixin Yang
- Department of Obstetrics, Gynecology & Reproductive Sciences, Baltimore, MD, USA; Department of Biochemistry & Molecular Biology, University of Maryland School of Medicine, Baltimore, Maryland, USA.
| |
Collapse
|
26
|
Li Y, Xue Y, Xu X, Wang G, Liu Y, Wu H, Li W, Wang Y, Chen Z, Zhang W, Zhu Y, Ji W, Xu T, Liu L, Chen Q. A mitochondrial FUNDC1/HSC70 interaction organizes the proteostatic stress response at the risk of cell morbidity. EMBO J 2018; 38:embj.201798786. [PMID: 30591555 DOI: 10.15252/embj.201798786] [Citation(s) in RCA: 67] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 08/25/2018] [Accepted: 10/04/2018] [Indexed: 01/22/2023] Open
Abstract
Both protein quality and mitochondrial quality are vital for the cellular activity, and impaired proteostasis and mitochondrial dysfunction are common etiologies of aging and age-related disorders. Here, we report that the mitochondrial outer membrane protein FUNDC1 interacts with the chaperone HSC70 to promote the mitochondrial translocation of unfolded cytosolic proteins for degradation by LONP1 or for formation of non-aggresomal mitochondrion-associated protein aggregates (MAPAs) upon proteasome inhibition in cultured human cells. Integrative approaches including csCLEM, Apex, and biochemical analysis reveal that MAPAs contain ubiquitinated cytosolic proteins, autophagy receptor p62, and mitochondrial proteins. MAPAs are segregated from mitochondria in a FIS1-dependent manner and can subsequently be degraded via autophagy. Although the FUNDC1/HSC70 pathway promotes the degradation of unfolded cytosolic proteins, excessive accumulation of unfolded proteins on the mitochondria prior to MAPA formation impairs mitochondrial integrity and activates AMPK, leading to cellular senescence. We suggest that human mitochondria organize cellular proteostatic response at the risk of their own malfunction and cell lethality.
Collapse
Affiliation(s)
- Yanjun Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yanhong Xue
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Xiaojun Xu
- College of Life Science and Technology, HuaZhong University of Science and Technology, Wuhan, Hubei, China
| | - Guopeng Wang
- School of Life Sciences, Peking University, Beijing, China
| | - Yiqun Liu
- School of Life Sciences, Peking University, Beijing, China
| | - Hao Wu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Wenhui Li
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yueying Wang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Ziheng Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Weilin Zhang
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Yushan Zhu
- College of Life Sciences, Nankai University, Tianjin, China
| | - Wei Ji
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China
| | - Tao Xu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Science and Technology, HuaZhong University of Science and Technology, Wuhan, Hubei, China
| | - Lei Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China
| | - Quan Chen
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, China .,College of Life Sciences, Nankai University, Tianjin, China.,University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
27
|
Noguchi T, Suzuki M, Mutoh N, Hirata Y, Tsuchida M, Miyagawa S, Hwang GW, Aoki J, Matsuzawa A. Nuclear-accumulated SQSTM1/p62-based ALIS act as microdomains sensing cellular stresses and triggering oxidative stress-induced parthanatos. Cell Death Dis 2018; 9:1193. [PMID: 30546061 PMCID: PMC6294141 DOI: 10.1038/s41419-018-1245-y] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2018] [Revised: 11/13/2018] [Accepted: 11/27/2018] [Indexed: 12/25/2022]
Abstract
Aggresome-like induced structures (ALIS) have been described as ubiquitinated protein-containing aggresomes transiently formed in response to various stresses. In this study, we provide evidence that ALIS composed of SQSTM1/p62 act as a key determinant of oxidative stress-induced parthanatos, which is newly discovered and distinct from regular programmed cell death. Interestingly, we first found that chemical stresses induced by particular chemical drugs, such as several cephalosporin antibiotics, cause oxidative stress-mediated parthanatos, accompanied by the ALIS formation. Blocking the ALIS formation potently suppressed the parthanatos, and p62 knockout cells exhibited the attenuated ALIS formation and high resistance to parthanatos. Moreover, we also found that the redox-sensing activity of p62 is required for nuclear accumulation of the p62-based ALIS, resulting in the induction of parthanatos. Together, our results demonstrate unexpected functions of p62 and ALIS as cell death mediators sensing oxidative stress, and thus uncover a novel mechanism whereby p62 mediates parthanatos.
Collapse
Affiliation(s)
- Takuya Noguchi
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Midori Suzuki
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Natsumi Mutoh
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Yusuke Hirata
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Mei Tsuchida
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Sayoko Miyagawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Gi-Wook Hwang
- Laboratory of Molecular and Biochemical Toxicology, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Junken Aoki
- Laboratory of Molecular and Cellular Biochemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan
| | - Atsushi Matsuzawa
- Laboratory of Health Chemistry, Graduate School of Pharmaceutical Sciences, Tohoku University, Aoba-6-3, Aramaki, Aoba-ku, Sendai, 980-8578, Japan.
| |
Collapse
|
28
|
Jena KK, Kolapalli SP, Mehto S, Nath P, Das B, Sahoo PK, Ahad A, Syed GH, Raghav SK, Senapati S, Chauhan S, Chauhan S. TRIM16 controls assembly and degradation of protein aggregates by modulating the p62-NRF2 axis and autophagy. EMBO J 2018; 37:embj.201798358. [PMID: 30143514 PMCID: PMC6138442 DOI: 10.15252/embj.201798358] [Citation(s) in RCA: 87] [Impact Index Per Article: 14.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2017] [Revised: 07/18/2018] [Accepted: 07/26/2018] [Indexed: 12/20/2022] Open
Abstract
Sequestration of protein aggregates in inclusion bodies and their subsequent degradation prevents proteostasis imbalance, cytotoxicity, and proteinopathies. The underlying molecular mechanisms controlling the turnover of protein aggregates are mostly uncharacterized. Herein, we show that a TRIM family protein, TRIM16, governs the process of stress-induced biogenesis and degradation of protein aggregates. TRIM16 facilitates protein aggregate formation by positively regulating the p62-NRF2 axis. We show that TRIM16 is an integral part of the p62-KEAP1-NRF2 complex and utilizes multiple mechanisms for stabilizing NRF2. Under oxidative and proteotoxic stress conditions, TRIM16 activates ubiquitin pathway genes and p62 via NRF2, leading to ubiquitination of misfolded proteins and formation of protein aggregates. We further show that TRIM16 acts as a scaffold protein and, by interacting with p62, ULK1, ATG16L1, and LC3B, facilitates autophagic degradation of protein aggregates. Thus, TRIM16 streamlines the process of stress-induced aggregate clearance and protects cells against oxidative/proteotoxic stress-induced toxicity in vitro and in vivo Taken together, this work identifies a new mechanism of protein aggregate turnover, which could be relevant in protein aggregation-associated diseases such as neurodegeneration.
Collapse
Affiliation(s)
- Kautilya Kumar Jena
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India.,School of Biotechnology, KIIT University, Bhubaneswar, India
| | | | - Subhash Mehto
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Parej Nath
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India.,School of Biotechnology, KIIT University, Bhubaneswar, India
| | - Biswajit Das
- Tumor Microenvironment and Animal Models, Institute of Life Sciences, Bhubaneswar, India.,Manipal University, Manipal, India
| | - Pradyumna Kumar Sahoo
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| | - Abdul Ahad
- Immuno-Genomics and Systems Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Gulam Hussain Syed
- Molecular Virology and Infectious Diseases, Institute of Life Sciences, Bhubaneswar, India
| | - Sunil K Raghav
- Immuno-Genomics and Systems Biology, Institute of Life Sciences, Bhubaneswar, India
| | - Shantibhusan Senapati
- Tumor Microenvironment and Animal Models, Institute of Life Sciences, Bhubaneswar, India
| | - Swati Chauhan
- Translational Research, Institute of Life Sciences, Bhubaneswar, India
| | - Santosh Chauhan
- Cell Biology and Infectious Diseases Unit, Institute of Life Sciences, Bhubaneswar, India
| |
Collapse
|
29
|
Kivinen N. The role of autophagy in age-related macular degeneration. Acta Ophthalmol 2018; 96 Suppl A110:1-50. [PMID: 29633521 DOI: 10.1111/aos.13753] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Affiliation(s)
- Niko Kivinen
- Department of Ophthalmology; University of Eastern Finland; Kuopio Finland
| |
Collapse
|
30
|
Xiao Y, Chen X, Huang S, Li G, Mo M, Zhang L, Chen C, Guo W, Zhou M, Wu Z, Cen L, Long S, Li S, Yang X, Qu S, Pei Z, Xu P. Iron promotes α-synuclein aggregation and transmission by inhibiting TFEB-mediated autophagosome-lysosome fusion. J Neurochem 2018; 145:34-50. [PMID: 29364516 DOI: 10.1111/jnc.14312] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Revised: 01/15/2018] [Accepted: 01/17/2018] [Indexed: 12/21/2022]
Affiliation(s)
- Yousheng Xiao
- Department of Neurology; The First Affiliated Hospital of Guangxi Medical University; Nanning China
- Department of Neurology; National Key Clinical; Department and Key Discipline of Neurology; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Xiang Chen
- Department of Neurology; National Key Clinical; Department and Key Discipline of Neurology; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Shuxuan Huang
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Guihua Li
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Mingshu Mo
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Li Zhang
- Geriatric Neurology Department; Nanjing Brain Hospital; Nanjing Medical University; Nanjing China
| | - Chaojun Chen
- Department of Neurology; Guangzhou Chinese Medical Integrated Hospital (Huadu); Guangzhou China
| | - Wenyuan Guo
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Miaomiao Zhou
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Zhuohua Wu
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| | - Luan Cen
- Department of Neurology; The First Affiliated Hospital of Guangxi Medical University; Nanning China
| | - Simei Long
- Department of Neurology; National Key Clinical; Department and Key Discipline of Neurology; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Shaomin Li
- Ann Romney Center for Neurologic Disease; Brigham and Women's Hospital; Harvard Medical School; Boston MA USA
| | - Xinling Yang
- Department of Neurology; The Second Affiliated Hospital of Xinjiang Medical University; Urumqi China
| | - Shaogang Qu
- Clinical Medicine Research Center; Shunde Hospital; Southern Medical University; Foshan China
| | - Zhong Pei
- Department of Neurology; National Key Clinical; Department and Key Discipline of Neurology; The First Affiliated Hospital of Sun Yat-sen University; Guangzhou China
| | - Pingyi Xu
- Department of Neurology; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
- Key Laboratory of Respiratory Disease; The First Affiliated Hospital of Guangzhou Medical University; Guangzhou China
| |
Collapse
|
31
|
Zaffagnini G, Savova A, Danieli A, Romanov J, Tremel S, Ebner M, Peterbauer T, Sztacho M, Trapannone R, Tarafder AK, Sachse C, Martens S. p62 filaments capture and present ubiquitinated cargos for autophagy. EMBO J 2018; 37:embj.201798308. [PMID: 29343546 PMCID: PMC5830917 DOI: 10.15252/embj.201798308] [Citation(s) in RCA: 228] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2017] [Revised: 01/03/2018] [Accepted: 01/03/2018] [Indexed: 12/14/2022] Open
Abstract
The removal of misfolded, ubiquitinated proteins is an essential part of the protein quality control. The ubiquitin-proteasome system (UPS) and autophagy are two interconnected pathways that mediate the degradation of such proteins. During autophagy, ubiquitinated proteins are clustered in a p62-dependent manner and are subsequently engulfed by autophagosomes. However, the nature of the protein substrates targeted for autophagy is unclear. Here, we developed a reconstituted system using purified components and show that p62 and ubiquitinated proteins spontaneously coalesce into larger clusters. Efficient cluster formation requires substrates modified with at least two ubiquitin chains longer than three moieties and is based on p62 filaments cross-linked by the substrates. The reaction is inhibited by free ubiquitin, K48-, and K63-linked ubiquitin chains, as well as by the autophagosomal marker LC3B, suggesting a tight cross talk with general proteostasis and autophagosome formation. Our study provides mechanistic insights on how substrates are channeled into autophagy.
Collapse
Affiliation(s)
- Gabriele Zaffagnini
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Adriana Savova
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Alberto Danieli
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Julia Romanov
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Shirley Tremel
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Michael Ebner
- Department of Structural and Computational Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Thomas Peterbauer
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Martin Sztacho
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Riccardo Trapannone
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| | - Abul K Tarafder
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Carsten Sachse
- Structural and Computational Biology Unit, European Molecular Biology Laboratory, Heidelberg, Germany
| | - Sascha Martens
- Department of Biochemistry and Cell Biology, Max F. Perutz Laboratories (MFPL), Vienna Biocenter (VBC), University of Vienna, Vienna, Austria
| |
Collapse
|
32
|
Luis-Ravelo D, Estévez-Silva H, Barroso-Chinea P, Afonso-Oramas D, Salas-Hernández J, Rodríguez-Núñez J, Acevedo-Arozena A, Marcellino D, González-Hernández T. Pramipexole reduces soluble mutant huntingtin and protects striatal neurons through dopamine D3 receptors in a genetic model of Huntington's disease. Exp Neurol 2018; 299:137-147. [DOI: 10.1016/j.expneurol.2017.10.019] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/29/2017] [Accepted: 10/19/2017] [Indexed: 12/26/2022]
|
33
|
Huang N, Erie C, Lu ML, Wei J. Aberrant subcellular localization of SQSTM1/p62 contributes to increased vulnerability to proteotoxic stress recovery in Huntington's disease. Mol Cell Neurosci 2017; 88:43-52. [PMID: 29246658 DOI: 10.1016/j.mcn.2017.12.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2017] [Revised: 12/08/2017] [Accepted: 12/10/2017] [Indexed: 10/18/2022] Open
Abstract
Proteotoxic stress plays an important role in the pathogenesis of Huntington's disease (HD). Autophagy is proposed as a compensatory mechanism to remove protein aggregates under proteotoxic stress by up-regulating p62 expression. In the present study, we investigated the molecular action of p62 to proteotoxic stress in HD cells. Using two different HD cellular models, STHdhQ7 and STHdhQ111 cells derived from wild type and HD knock-in mice and human fibroblasts from healthy and HD patients, we found that HD cells are more vulnerable to cell death under proteotoxic stress and during stress recovery. We further showed that P62 was up-regulated in both STHdhQ7 and STHdhQ111 cells in response to the stress with distinct subcellular localization patterns. While dispersed p62 puncti were found in STHdhQ7 cells, p62 bodies were initially present in the lysosomes and accumulated to the juxtanuclear regions of STHdhQ111 cells as MG132 incubation continued. Unlike in STHdhQ7 cells, p62 puncti were not associated with K48-linked polyubiquitinated protein aggregates or proteasomal components in STHdhQ111. Interestingly, addition of cysteine during MG132 incubation rescued cell death in STHdhQ111 cells caused by stress recovery and altered the subcellular distribution of p62. Our data suggest that aberrant positioning of p62 affects the proteasomal clearance of protein aggregates and may contribute to the increased vulnerability to proteotoxic stress-induced cell death in HD cells.
Collapse
Affiliation(s)
- Ningjing Huang
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton 33431, FL, USA; Department of Neurology, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai 200071, China
| | - Christine Erie
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton 33431, FL, USA
| | - Michael L Lu
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton 33431, FL, USA
| | - Jianning Wei
- Department of Biomedical Science, Charles E. Schmidt College of Medicine, Florida Atlantic University, Boca Raton 33431, FL, USA.
| |
Collapse
|
34
|
Vodicka P, Chase K, Iuliano M, Valentine DT, Sapp E, Lu B, Kegel-Gleason KB, Sena-Esteves M, Aronin N, DiFiglia M. Effects of Exogenous NUB1 Expression in the Striatum of HDQ175/Q7 Mice. J Huntingtons Dis 2017; 5:163-74. [PMID: 27314618 DOI: 10.3233/jhd-160195] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
BACKGROUND Reducing mutant huntingtin (mHTT) in neurons may be a therapy for Huntington's disease (HD). Elevating NUB1 protein reduced mHTT levels in cell and fly models of HD through a proteasome dependent mechanism. OBJECTIVE To examine the effects of augmenting NUB1 in HD mouse striatum on mHTT levels. METHODS Striata of HDQ175/Q7 mice were injected at 3 months of age with recombinant AAV2/9 coding for NUB1 or GFP under the control of the neuron specific human synapsin 1 promoter and examined 6 months post-injection for levels of huntingtin, the striatal markers DARPP32 and PDE10A, the astrocyte marker GFAP, and the autophagy and mHTT aggregate marker P62 using immunolabeling of brain sections and Western blot assay of striatal subcellular fractions. RESULTS By Western blot human HD brain had only one of the two variants of NUB1 present in human control brain. In striatum of WT and HD mice NUB1 was localized in medium size neurons and enriched in the nucleus of large neurons. In the striatum of NUB1 injected HD mice, there was widespread neuronal distribution of exogenous NUB1 labeling and protein levels were ∼2.5-fold endogenous levels. DARPP32 and GFAP distribution and levels were unchanged but PDE10A levels were lower in crude homogenates and P62 was increased in nuclear enriched P1 fractions. Elevating NUB1 did not change levels of full-length mHTT or the number and size of mHTT (S830) positive nuclear inclusions. CONCLUSION Findings suggest that increasing NUB1 protein in striatal neurons of HDQ175/Q7 mice in vivo may be relatively safe but is ineffective in reducing mHTT. Increased NUB1 expression in HD striatum alters PDE10A and P62 which are known to be influenced by mHTT.
Collapse
Affiliation(s)
- Petr Vodicka
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Kathryn Chase
- Department of Medicine and The RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Maria Iuliano
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Dana T Valentine
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Ellen Sapp
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Boxun Lu
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA.,State Key Laboratory of Medical Neurobiology, School of Life Sciences, Fudan University, China
| | - Kimberly B Kegel-Gleason
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| | - Miguel Sena-Esteves
- Department of Neurology, Gene Therapy Center, University of Massachusetts Medical School, Worcester, MA, USA
| | - Neil Aronin
- Department of Medicine and The RNA Therapeutics Institute, University of Massachusetts Medical School, Worcester, MA, USA
| | - Marian DiFiglia
- Department of Neurology, Massachusetts General Hospital and Harvard Medical School, Charlestown, MA, USA
| |
Collapse
|
35
|
Billes V, Kovács T, Hotzi B, Manzéger A, Tagscherer K, Komlós M, Tarnóci A, Pádár Z, Erdős A, Bjelik A, Legradi A, Gulya K, Gulyás B, Vellai T. AUTEN-67 (Autophagy Enhancer-67) Hampers the Progression of Neurodegenerative Symptoms in a Drosophila model of Huntington's Disease. J Huntingtons Dis 2017; 5:133-47. [PMID: 27163946 DOI: 10.3233/jhd-150180] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
BACKGROUND Autophagy, a lysosome-mediated self-degradation process of eukaryotic cells, serves as a main route for the elimination of cellular damage [1-3]. Such damages include aggregated, oxidized or misfolded proteins whose accumulation can cause various neurodegenerative pathologies, including Huntington's disease (HD). OBJECTIVE Here we examined whether enhanced autophagic activity can alleviate neurophatological features in a Drosophila model of HD (the transgenic animals express a human mutant Huntingtin protein with a long polyglutamine repeat, 128Q). METHODS We have recently identified an autophagy-enhancing small molecule, AUTEN-67 (autophagy enhancer 67), with potent neuroprotective effects [4]. AUTEN-67 was applied to induce autophagic activity in the HD model used in this study. RESULTS We showed that AUTEN-67 treatment interferes with the progressive accumulation of ubiquitinated proteins in the brain of Drosophila transgenic for the pathological 128Q form of human Huntingtin protein. The compound significantly improved the climbing ability and moderately extended the mean life span of these flies. Furthermore, brain tissue samples from human patients diagnosed for HD displayed increased levels of the autophagy substrate SQSTM1/p62 protein, as compared with controls. CONCLUSIONS These results imply that AUTEN-67 impedes the progression of neurodegenerative symptoms characterizing HD, and that autophagy is a promising therapeutic target for treating this pathology. In humans, AUTEN-67 may have the potential to delay the onset and decrease the severity of HD.
Collapse
Affiliation(s)
- Viktor Billes
- Velgene Biotechnology Research Ltd., Szeged, Hungary
| | - Tibor Kovács
- Velgene Biotechnology Research Ltd., Szeged, Hungary.,Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Bernadette Hotzi
- Velgene Biotechnology Research Ltd., Szeged, Hungary.,Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Anna Manzéger
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | - Kinga Tagscherer
- Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| | | | - Anna Tarnóci
- Velgene Biotechnology Research Ltd., Szeged, Hungary
| | - Zsolt Pádár
- Velgene Biotechnology Research Ltd., Szeged, Hungary
| | - Attila Erdős
- Velgene Biotechnology Research Ltd., Szeged, Hungary
| | - Annamaria Bjelik
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Adam Legradi
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Károly Gulya
- Department of Cell Biology and Molecular Medicine, University of Szeged, Szeged, Hungary
| | - Balázs Gulyás
- Karolinska Institute, Department of Clinical Neuroscience, Stockholm, Sweden.,Lee Kong Chian School of Medicine, Nanyang Technological University, Singapore.,Imperial College London, Department of Medicine, Division of Brain Sciences, London, UK
| | - Tibor Vellai
- Velgene Biotechnology Research Ltd., Szeged, Hungary.,Department of Genetics, Eötvös Loránd University, Budapest, Hungary
| |
Collapse
|
36
|
Mizunoe Y, Kobayashi M, Sudo Y, Watanabe S, Yasukawa H, Natori D, Hoshino A, Negishi A, Okita N, Komatsu M, Higami Y. Trehalose protects against oxidative stress by regulating the Keap1-Nrf2 and autophagy pathways. Redox Biol 2017; 15:115-124. [PMID: 29241092 PMCID: PMC5730428 DOI: 10.1016/j.redox.2017.09.007] [Citation(s) in RCA: 148] [Impact Index Per Article: 21.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 09/09/2017] [Accepted: 09/13/2017] [Indexed: 12/19/2022] Open
Abstract
Dysfunction of autophagy, which regulates cellular homeostasis by degrading organelles and proteins, is associated with pathogenesis of various diseases such as cancer, neurodegeneration and metabolic disease. Trehalose, a naturally occurring nontoxic disaccharide found in plants, insects, microorganisms and invertebrates, but not in mammals, was reported to function as a mechanistic target of the rapamycin (mTOR)-independent inducer of autophagy. In addition, trehalose functions as an antioxidant though its underlying molecular mechanisms remain unclear. In this study, we showed that trehalose not only promoted autophagy, but also increased p62 protein expression, in an autophagy-independent manner. In addition, trehalose increased nuclear translocation of nuclear factor (erythroid-derived 2)-like 2 (Nrf2) in a p62-dependent manner and enhance expression of its downstream antioxidant factors, heme oxygenase-1 (Ho-1) and nicotinamide adenine dinucleotide phosphate quinone dehydrogenase 1 (Nqo1). Moreover, treatment with trehalose significantly reduced amount of reactive oxygen species. Collectively, these results suggested that trehalose can function as a novel activator of the p62-Keap1/Nrf2 pathway, in addition to inducing autophagy. Therefore, trehalose may be useful to treat many chronic diseases involving oxidative stress and dysfunction of autophagy.
Collapse
Affiliation(s)
- Yuhei Mizunoe
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan; Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| | - Masaki Kobayashi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan; Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| | - Yuka Sudo
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan; Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan
| | - Shukoh Watanabe
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan
| | - Hiromine Yasukawa
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan
| | - Daiki Natori
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan
| | - Ayana Hoshino
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan
| | - Arisa Negishi
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan
| | - Naoyuki Okita
- Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan; Department of Internal Medicine Research, Sasaki Institute, Sasaki Foundation, Tokyo 101-0062, Japan
| | - Masaaki Komatsu
- Department of Biochemistry, Niigata University Graduate School of Medical and Dental Sciences, Niigata 951-8510, Japan
| | - Yoshikazu Higami
- Laboratory of Molecular Pathology and Metabolic Disease, Faculty of Pharmaceutical Sciences, Tokyo University of Science, 2641 Noda, Chiba 278-8510, Japan; Translational Research Center, Research Institute of Science and Technology, Tokyo University of Science, Noda 278-8510, Japan.
| |
Collapse
|
37
|
Kino Y, Washizu C, Kurosawa M, Yamada M, Doi H, Takumi T, Adachi H, Katsuno M, Sobue G, Hicks GG, Hattori N, Shimogori T, Nukina N. FUS/TLS acts as an aggregation-dependent modifier of polyglutamine disease model mice. Sci Rep 2016; 6:35236. [PMID: 27739513 PMCID: PMC5064419 DOI: 10.1038/srep35236] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2016] [Accepted: 09/26/2016] [Indexed: 12/13/2022] Open
Abstract
FUS/TLS is an RNA/DNA-binding protein associated with neurodegenerative diseases including amyotrophic lateral sclerosis and frontotemporal lobar degeneration. Previously, we found that a prion-like domain in the N-terminus of FUS/TLS mediates co-aggregation between FUS/TLS and mutant huntingtin, the gene product of Huntington's disease (HD). Here, we show that heterozygous knockout of FUS/TLS worsened the phenotypes of model mice of (HD, but not spinal and bulbar muscular atrophy (SBMA). This difference was correlated with the degree of pathological association between disease proteins and FUS/TLS. Co-aggregation between FUS/TLS and mutant huntingtin resulted in the depletion of free FUS/TLS protein in HD mice that was detected as a monomer in SDS-PAGE analysis. Recently, we found that FUS/TLS paralogs, TAF15 and EWS, were up-regulated in homozygous FUS/TLS knockout mice. These two proteins were up-regulated in both HD and FUS/TLS heterozygote mice, and were further elevated in HD-TLS+/- double mutant mice, consistent with the functional impairment of FUS/TLS. These results suggest that FUS/TLS sequestration by co-aggregation is a rate-limiting factor of disease phenotypes of HD and that inclusions may have an adverse aspect, rather than being simply benign or protective. In addition, our results highlight inclusions as repositories of potential modifiers of neurodegeneration.
Collapse
Affiliation(s)
- Yoshihiro Kino
- CREST(Core Research for Evolutionary Science and Technology), JST, Saitama, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Laboratory for Structural Neuropathology , Brain Science Institute, RIKEN, Saitama, Japan.,Laboratory for Molecular Mechanisms of Thalamus Development, Brain Science Institute, RIKEN, Saitama, Japan.,Department of Bioinformatics and Molecular Neuropathology, Meiji Pharmaceutical University, Tokyo, Japan
| | - Chika Washizu
- Laboratory for Structural Neuropathology , Brain Science Institute, RIKEN, Saitama, Japan
| | - Masaru Kurosawa
- CREST(Core Research for Evolutionary Science and Technology), JST, Saitama, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Laboratory for Structural Neuropathology , Brain Science Institute, RIKEN, Saitama, Japan.,Laboratory for Molecular Mechanisms of Thalamus Development, Brain Science Institute, RIKEN, Saitama, Japan
| | - Mizuki Yamada
- Laboratory for Structural Neuropathology , Brain Science Institute, RIKEN, Saitama, Japan
| | - Hiroshi Doi
- Department of Clinical Neurology and Stroke Medicine, Graduate School of Medicine, Yokohama City University, Yokohama, Japan
| | - Toru Takumi
- CREST(Core Research for Evolutionary Science and Technology), JST, Saitama, Japan.,Laboratory for Mental Biology, Brain Science Institute, RIKEN, Saitama, Japan.,Graduate School of Biomedical Sciences, Hiroshima University, Hiroshima, Japan
| | - Hiroaki Adachi
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Geoffrey G Hicks
- Manitoba Institute of Cell Biology, University of Manitoba, Winnipeg, Canada
| | - Nobutaka Hattori
- Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan
| | - Tomomi Shimogori
- Laboratory for Molecular Mechanisms of Thalamus Development, Brain Science Institute, RIKEN, Saitama, Japan
| | - Nobuyuki Nukina
- CREST(Core Research for Evolutionary Science and Technology), JST, Saitama, Japan.,Department of Neuroscience for Neurodegenerative Disorders, Juntendo University Graduate School of Medicine, Tokyo, Japan.,Laboratory for Structural Neuropathology , Brain Science Institute, RIKEN, Saitama, Japan.,Laboratory for Molecular Mechanisms of Thalamus Development, Brain Science Institute, RIKEN, Saitama, Japan.,Laboratory of Structural Neuropathology, Doshisha University Graduate School of Brain Science, Kyoto, Japan
| |
Collapse
|
38
|
Ntsapi C, Loos B. Caloric restriction and the precision-control of autophagy: A strategy for delaying neurodegenerative disease progression. Exp Gerontol 2016; 83:97-111. [DOI: 10.1016/j.exger.2016.07.014] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Revised: 07/18/2016] [Accepted: 07/25/2016] [Indexed: 01/07/2023]
|
39
|
Cecarini V, Bonfili L, Cuccioloni M, Mozzicafreddo M, Angeletti M, Keller JN, Eleuteri AM. The fine-tuning of proteolytic pathways in Alzheimer's disease. Cell Mol Life Sci 2016; 73:3433-51. [PMID: 27120560 PMCID: PMC11108445 DOI: 10.1007/s00018-016-2238-6] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2015] [Revised: 03/31/2016] [Accepted: 04/21/2016] [Indexed: 11/28/2022]
Abstract
Several integrated proteolytic systems contribute to the maintenance of cellular homeostasis through the continuous removal of misfolded, aggregated or oxidized proteins and damaged organelles. Among these systems, the proteasome and autophagy play the major role in protein quality control, which is a fundamental issue in non-proliferative cells such as neurons. Disturbances in the functionality of these two pathways are frequently observed in neurodegenerative diseases, like Alzheimer's disease, and reflect the accumulation of protease-resistant, deleterious protein aggregates. In this review, we explored the sophisticated crosstalk between the ubiquitin-proteasome system and autophagy in the removal of the harmful structures that characterize Alzheimer's disease neurons. We also dissected the role of the numerous shuttle factors and chaperones that, directly or indirectly interacting with ubiquitin and LC3, are used for cargo selection and delivery to one pathway or the other.
Collapse
Affiliation(s)
- Valentina Cecarini
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy.
| | - Laura Bonfili
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Massimiliano Cuccioloni
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Matteo Mozzicafreddo
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Mauro Angeletti
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| | - Jeffrey N Keller
- Pennington Biomedical Research Centre, Louisiana State University System, Baton Rouge, LA, 70808, USA
| | - Anna Maria Eleuteri
- Department of Biosciences and Veterinary Medicine, School of Biosciences and Veterinary Medicine, University of Camerino, Via Gentile III da Varano, 62032, Camerino, Italy
| |
Collapse
|
40
|
Johansson I, Monsen VT, Pettersen K, Mildenberger J, Misund K, Kaarniranta K, Schønberg S, Bjørkøy G. The marine n-3 PUFA DHA evokes cytoprotection against oxidative stress and protein misfolding by inducing autophagy and NFE2L2 in human retinal pigment epithelial cells. Autophagy 2016; 11:1636-51. [PMID: 26237736 PMCID: PMC4590664 DOI: 10.1080/15548627.2015.1061170] [Citation(s) in RCA: 82] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Accumulation and aggregation of misfolded proteins is a hallmark of several diseases collectively known as proteinopathies. Autophagy has a cytoprotective role in diseases associated with protein aggregates. Age-related macular degeneration (AMD) is the most common neurodegenerative eye disease that evokes blindness in elderly. AMD is characterized by degeneration of retinal pigment epithelial (RPE) cells and leads to loss of photoreceptor cells and central vision. The initial phase associates with accumulation of intracellular lipofuscin and extracellular deposits called drusen. Epidemiological studies have suggested an inverse correlation between dietary intake of marine n-3 polyunsaturated fatty acids (PUFAs) and the risk of developing neurodegenerative diseases, including AMD. However, the disease-preventive mechanism(s) mobilized by n-3 PUFAs is not completely understood. In human retinal pigment epithelial cells we find that physiologically relevant doses of the n-3 PUFA docosahexaenoic acid (DHA) induce a transient increase in cellular reactive oxygen species (ROS) levels that activates the oxidative stress response regulator NFE2L2/NRF2 (nuclear factor, erythroid derived 2, like 2). Simultaneously, there is a transient increase in intracellular protein aggregates containing SQSTM1/p62 (sequestosome 1) and an increase in autophagy. Pretreatment with DHA rescues the cells from cell cycle arrest induced by misfolded proteins or oxidative stress. Cells with a downregulated oxidative stress response, or autophagy, respond with reduced cell growth and survival after DHA supplementation. These results suggest that DHA both induces endogenous antioxidants and mobilizes selective autophagy of misfolded proteins. Both mechanisms could be relevant to reduce the risk of developing aggregate-associate diseases such as AMD.
Collapse
Affiliation(s)
- Ida Johansson
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Vivi Talstad Monsen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Pettersen
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Jennifer Mildenberger
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway.,d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kristine Misund
- d Department of Cancer Research and Molecular Medicine ; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway.,e KG Jebsen Center for Myeloma Research; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Kai Kaarniranta
- f Department of Ophthalmology ; Institute of Clinical Medicine; University of Eastern Finland ; Kuopio , Finland.,g Department of Ophthalmology ; Kuopio University Hospital ; Kuopio , Finland
| | - Svanhild Schønberg
- a Department of Laboratory Medicine ; Children's and Women's Health; Faculty of Medicine; Norwegian University of Science and Technology ; Trondheim , Norway
| | - Geir Bjørkøy
- b Department of Technology ; University College of Sør-Trøndelag ; Trondheim , Norway.,c Centre of Molecular Inflammation Research and Department of Cancer Research and Molecular Medicine ; Norwegian University of Science and Technology ; Trondheim , Norway
| |
Collapse
|
41
|
Atypical Huntington's disease with the clinical presentation of behavioural variant of frontotemporal dementia. J Neural Transm (Vienna) 2016; 123:1423-1433. [PMID: 27287334 DOI: 10.1007/s00702-016-1579-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2016] [Accepted: 05/24/2016] [Indexed: 10/21/2022]
Abstract
Huntington's disease is an incurable, adult-onset, autosomal dominant inherited disorder caused by an expanded trinucleotide repeat (CAG). In this study, we describe a Huntington's disease patient displaying clinical symptoms of the behavioural variant of frontotemporal dementia in the absence of tremor and ataxia. The clinical onset was at the age of 36 years and the disease progressed slowly (18 years). Genetic testing revealed expanded trinucleotide CAG repeats in the Huntingtin gene, together with a Glu318Gly polymorphism in presenilin 1. Neuropathological assessment revealed extensive amyloid β (Aβ) aggregates in all cortical regions. No inclusions displaying hyperphosphorylated tau or phosphorylated transactive response DNA-binding protein 43 (TDP43) were found. A high number of p62 (sequestosome 1) immunopositive intranuclear inclusions were seen mainly in the cortex, while subcortical areas were affected to a lesser extent. Confocal microscopy revealed that the majority of p62 intranuclear lesions co-localised with the fused-in-sarcoma protein (FUS) immunostaining. The morphology of the inclusions resembled intranuclear aggregates in Huntington's disease. The presented proband suffered from Huntington's disease showed atypical distribution of FUS positive intranuclear aggregates in the cortical areas with concomitant Alzheimer's disease pathology.
Collapse
|
42
|
Liebl MP, Hoppe T. It's all about talking: two-way communication between proteasomal and lysosomal degradation pathways via ubiquitin. Am J Physiol Cell Physiol 2016; 311:C166-78. [PMID: 27225656 DOI: 10.1152/ajpcell.00074.2016] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Selective degradation of proteins requires a fine-tuned coordination of the two major proteolytic pathways, the ubiquitin-proteasome system (UPS) and autophagy. Substrate selection and proteolytic activity are defined by a plethora of regulatory cofactors influencing each other. Both proteolytic pathways are initiated by ubiquitylation to mark substrate proteins for degradation, although the size and/or topology of the modification are different. In this context E3 ubiquitin ligases, ensuring the covalent attachment of activated ubiquitin to the substrate, are of special importance. The regulation of E3 ligase activity, competition between different E3 ligases for binding E2 conjugation enzymes and substrates, as well as their interplay with deubiquitylating enzymes (DUBs) represent key events in the cross talk between the UPS and autophagy. The coordination between both degradation routes is further influenced by heat shock factors and ubiquitin-binding proteins (UBPs) such as p97, p62, or optineurin. Mutations in enzymes and ubiquitin-binding proteins or a general decline of both proteolytic systems during aging result in accumulation of damaged and aggregated proteins. Thus further mechanistic understanding of how UPS and autophagy communicate might allow therapeutic intervention especially against age-related diseases.
Collapse
Affiliation(s)
- Martina P Liebl
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Thorsten Hoppe
- Institute for Genetics and Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, Cologne, Germany
| |
Collapse
|
43
|
Dafinca R, Scaber J, Ababneh N, Lalic T, Weir G, Christian H, Vowles J, Douglas AGL, Fletcher-Jones A, Browne C, Nakanishi M, Turner MR, Wade-Martins R, Cowley SA, Talbot K. C9orf72 Hexanucleotide Expansions Are Associated with Altered Endoplasmic Reticulum Calcium Homeostasis and Stress Granule Formation in Induced Pluripotent Stem Cell-Derived Neurons from Patients with Amyotrophic Lateral Sclerosis and Frontotemporal Dementia. Stem Cells 2016; 34:2063-78. [PMID: 27097283 PMCID: PMC4979662 DOI: 10.1002/stem.2388] [Citation(s) in RCA: 179] [Impact Index Per Article: 22.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 02/26/2016] [Accepted: 03/19/2016] [Indexed: 12/13/2022]
Abstract
An expanded hexanucleotide repeat in a noncoding region of the C9orf72 gene is a major cause of amyotrophic lateral sclerosis (ALS), accounting for up to 40% of familial cases and 7% of sporadic ALS in European populations. We have generated induced pluripotent stem cells (iPSCs) from fibroblasts of patients carrying C9orf72 hexanucleotide expansions, differentiated these to functional motor and cortical neurons, and performed an extensive phenotypic characterization. In C9orf72 iPSC‐derived motor neurons, decreased cell survival is correlated with dysfunction in Ca2+ homeostasis, reduced levels of the antiapoptotic protein Bcl‐2, increased endoplasmic reticulum (ER) stress, and reduced mitochondrial membrane potential. Furthermore, C9orf72 motor neurons, and also cortical neurons, show evidence of abnormal protein aggregation and stress granule formation. This study is an extensive characterization of iPSC‐derived motor neurons as cellular models of ALS carrying C9orf72 hexanucleotide repeats, which describes a novel pathogenic link between C9orf72 mutations, dysregulation of calcium signaling, and altered proteostasis and provides a potential pharmacological target for the treatment of ALS and the related neurodegenerative disease frontotemporal dementia. Stem Cells2016;34:2063–2078
Collapse
Affiliation(s)
| | | | | | | | | | | | - Jane Vowles
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | | | | - Cathy Browne
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | - Mahito Nakanishi
- Research Center for Stem Cell Engineering, National Institute of Advanced Industrial Science and Technology (AIST), Tsukuba, Ibaraki, Japan
| | | | | | - Sally A Cowley
- James Martin Stem Cell Facility, Sir William Dunn School of Pathology, University of Oxford, Oxford, United Kingdom
| | | |
Collapse
|
44
|
Valdés A, Artemenko KA, Bergquist J, García-Cañas V, Cifuentes A. Comprehensive Proteomic Study of the Antiproliferative Activity of a Polyphenol-Enriched Rosemary Extract on Colon Cancer Cells Using Nanoliquid Chromatography–Orbitrap MS/MS. J Proteome Res 2016; 15:1971-85. [DOI: 10.1021/acs.jproteome.6b00154] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Affiliation(s)
- Alberto Valdés
- Laboratory
of Foodomics, Institute of Food Science Research (CIAL, CSIC), Calle Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Konstantin A. Artemenko
- Analytical
Chemistry, Department of Chemistry-BMC and SciLifeLab, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden
| | - Jonas Bergquist
- Analytical
Chemistry, Department of Chemistry-BMC and SciLifeLab, Uppsala University, Husargatan 3, 75124 Uppsala, Sweden
| | - Virginia García-Cañas
- Laboratory
of Foodomics, Institute of Food Science Research (CIAL, CSIC), Calle Nicolás Cabrera 9, 28049 Madrid, Spain
| | - Alejandro Cifuentes
- Laboratory
of Foodomics, Institute of Food Science Research (CIAL, CSIC), Calle Nicolás Cabrera 9, 28049 Madrid, Spain
| |
Collapse
|
45
|
Bersuker K, Brandeis M, Kopito RR. Protein misfolding specifies recruitment to cytoplasmic inclusion bodies. J Cell Biol 2016; 213:229-41. [PMID: 27114501 PMCID: PMC5084276 DOI: 10.1083/jcb.201511024] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2015] [Accepted: 03/21/2016] [Indexed: 12/17/2022] Open
Abstract
Inclusion bodies (IBs) containing aggregated disease-associated proteins and polyubiquitin (poly-Ub) conjugates are universal histopathological features of neurodegenerative diseases. Ub has been proposed to target proteins to IBs for degradation via autophagy, but the mechanisms that govern recruitment of ubiquitylated proteins to IBs are not well understood. In this paper, we use conditionally destabilized reporters that undergo misfolding and ubiquitylation upon removal of a stabilizing ligand to examine the role of Ub conjugation in targeting proteins to IBs that are composed of an N-terminal fragment of mutant huntingtin, the causative protein of Huntington's disease. We show that reporters are excluded from IBs in the presence of the stabilizing ligand but are recruited to IBs after ligand washout. However, we find that Ub conjugation is not necessary to target reporters to IBs. We also report that forced Ub conjugation by the Ub fusion degradation pathway is not sufficient for recruitment to IBs. Finally, we find that reporters and Ub conjugates are stable at IBs. These data indicate that compromised folding states, rather than conjugation to Ub, can specify recruitment to IBs.
Collapse
Affiliation(s)
- Kirill Bersuker
- Department of Biology, Stanford University, Stanford, CA 94305
| | - Michael Brandeis
- Department of Genetics, The Hebrew University of Jerusalem, Jerusalem 91904, Israel
| | - Ron R Kopito
- Department of Biology, Stanford University, Stanford, CA 94305
| |
Collapse
|
46
|
Mancias JD, Kimmelman AC. Mechanisms of Selective Autophagy in Normal Physiology and Cancer. J Mol Biol 2016; 428:1659-80. [PMID: 26953261 DOI: 10.1016/j.jmb.2016.02.027] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 02/25/2016] [Accepted: 02/25/2016] [Indexed: 01/04/2023]
Abstract
Selective autophagy is critical for regulating cellular homeostasis by mediating lysosomal turnover of a wide variety of substrates including proteins, aggregates, organelles, and pathogens via a growing class of molecules termed selective autophagy receptors. The molecular mechanisms of selective autophagy receptor action and regulation are complex. Selective autophagy receptors link their bound cargo to the autophagosomal membrane by interacting with lipidated ATG8 proteins (LC3/GABARAP) that are intimately associated with the autophagosome membrane. The cargo signals that selective autophagy receptors recognize are diverse but their recognition can be broadly grouped into two classes, ubiquitin-dependent cargo recognition versus ubiquitin-independent. The roles of post-translational modification of selective autophagy receptors in regulating these pathways in response to stimuli are an active area of research. Here we will review recent advances in the identification of selective autophagy receptors and their regulatory mechanisms. Given its importance in maintaining cellular homeostasis, disruption of autophagy can lead to disease including neurodegeneration and cancer. The role of autophagy in cancer is complex as autophagy can mediate promotion or inhibition of tumorigenesis. Here we will also review the importance of autophagy in cancer with a specific focus on the role of selective autophagy receptors.
Collapse
Affiliation(s)
- Joseph D Mancias
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA; Department of Cell Biology, Harvard Medical School, Boston, MA 02115, USA.
| | - Alec C Kimmelman
- Division of Genomic Stability and DNA Repair, Department of Radiation Oncology, Dana-Farber Cancer Institute, Boston, MA 02215, USA.
| |
Collapse
|
47
|
Kurosawa M, Matsumoto G, Sumikura H, Hatsuta H, Murayama S, Sakurai T, Shimogori T, Hattori N, Nukina N. Serine 403-phosphorylated p62/SQSTM1 immunoreactivity in inclusions of neurodegenerative diseases. Neurosci Res 2016; 103:64-70. [DOI: 10.1016/j.neures.2015.08.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2015] [Revised: 07/14/2015] [Accepted: 08/10/2015] [Indexed: 10/23/2022]
|
48
|
Navarro-Yepes J, Anandhan A, Bradley E, Bohovych I, Yarabe B, de Jong A, Ovaa H, Zhou Y, Khalimonchuk O, Quintanilla-Vega B, Franco R. Inhibition of Protein Ubiquitination by Paraquat and 1-Methyl-4-Phenylpyridinium Impairs Ubiquitin-Dependent Protein Degradation Pathways. Mol Neurobiol 2015; 53:5229-51. [PMID: 26409479 DOI: 10.1007/s12035-015-9414-9] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2015] [Accepted: 09/01/2015] [Indexed: 12/21/2022]
Abstract
Intracytoplasmic inclusions of protein aggregates in dopaminergic cells (Lewy bodies) are the pathological hallmark of Parkinson's disease (PD). Ubiquitin (Ub), alpha (α)-synuclein, p62/sequestosome 1, and oxidized proteins are the major components of Lewy bodies. However, the mechanisms involved in the impairment of misfolded/oxidized protein degradation pathways in PD are still unclear. PD is linked to mitochondrial dysfunction and environmental pesticide exposure. In this work, we evaluated the effects of the pesticide paraquat (PQ) and the mitochondrial toxin 1-methyl-4-phenylpyridinium (MPP(+)) on Ub-dependent protein degradation pathways. No increase in the accumulation of Ub-bound proteins or aggregates was observed in dopaminergic cells (SK-N-SH) treated with PQ or MPP(+), or in mice chronically exposed to PQ. PQ decreased Ub protein content, but not its mRNA transcription. Protein synthesis inhibition with cycloheximide depleted Ub levels and potentiated PQ-induced cell death. The inhibition of proteasomal activity by PQ was found to be a late event in cell death progression and had neither effect on the toxicity of either MPP(+) or PQ, nor on the accumulation of oxidized sulfenylated, sulfonylated (DJ-1/PARK7 and peroxiredoxins), and carbonylated proteins induced by PQ. PQ- and MPP(+)-induced Ub protein depletion prompted the dimerization/inactivation of the Ub-binding protein p62 that regulates the clearance of ubiquitinated proteins by autophagy. We confirmed that PQ and MPP(+) impaired autophagy flux and that the blockage of autophagy by the overexpression of a dominant-negative form of the autophagy protein 5 (dnAtg5) stimulated their toxicity, but there was no additional effect upon inhibition of the proteasome. PQ induced an increase in the accumulation of α-synuclein in dopaminergic cells and membrane-associated foci in yeast cells. Our results demonstrate that the inhibition of protein ubiquitination by PQ and MPP(+) is involved in the dysfunction of Ub-dependent protein degradation pathways.
Collapse
Affiliation(s)
- Juliana Navarro-Yepes
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, 114 VBS 0905, Lincoln, NE, 68583, USA.,Department of Toxicology, CINVESTAV-IPN, IPN No. 2508, Colonia Zacatenco, Mexico City, D.F., 07360, Mexico
| | - Annadurai Anandhan
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA.,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, 114 VBS 0905, Lincoln, NE, 68583, USA
| | - Erin Bradley
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Iryna Bohovych
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA.,Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Bo Yarabe
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Annemieke de Jong
- Division of Cell Biology II, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - Huib Ovaa
- Division of Cell Biology II, The Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - You Zhou
- Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Oleh Khalimonchuk
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA.,Department of Biochemistry, University of Nebraska-Lincoln, Lincoln, NE, USA
| | - Betzabet Quintanilla-Vega
- Department of Toxicology, CINVESTAV-IPN, IPN No. 2508, Colonia Zacatenco, Mexico City, D.F., 07360, Mexico.
| | - Rodrigo Franco
- Redox Biology Center, University of Nebraska-Lincoln, Lincoln, NE, USA. .,School of Veterinary Medicine and Biomedical Sciences, University of Nebraska-Lincoln, 114 VBS 0905, Lincoln, NE, 68583, USA.
| |
Collapse
|
49
|
Jeong JK, Park SY. Neuroprotective effect of cellular prion protein (PrPC) is related with activation of alpha7 nicotinic acetylcholine receptor (α7nAchR)-mediated autophagy flux. Oncotarget 2015; 6:24660-74. [PMID: 26295309 PMCID: PMC4694786 DOI: 10.18632/oncotarget.4953] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/28/2015] [Indexed: 01/08/2023] Open
Abstract
Activation of the alpha7 nicotinic acetylcholine receptor (α7nAchR) is regulated by prion protein (PrPC) expression and has a neuroprotective effect by modulating autophagic flux. In this study, we hypothesized that PrPC may regulate α7nAchR activation and that may prevent prion-related neurodegenerative diseases by regulating autophagic flux. PrP(106-126) treatment decreased α7nAchR expression and activation of autophagic flux. In addition, the α7nAchR activator PNU-282987 enhanced autophagic flux and protected neuron cells against PrP(106-126)-induced apoptosis. However, activation of autophagy and the protective effects of PNU-282987 were inhibited in PrPC knockout hippocampal neuron cells. In addition, PrPC knockout hippocampal neuron cells showed decreased α7nAchR expression levels. Adenoviral overexpression of PrPC in PrPC knockout hippocampal neuron cells resulted in activation of autophagic flux and inhibition of prion peptide-mediated cell death via α7nAchR activation. This is the first report demonstrating that activation of α7nAchR-mediated autophagic flux is regulated by PrPC, and that activation of α7nAchR regulated by PrPC expression may play a pivotal role in protection of neuron cells against prion peptide-induced neuron cell death by autophagy. These results suggest that α7nAchR-mediated autophagic flux may be involved in the pathogenesis of prion-related diseases and may be a therapeutic target for prion-related neurodegenerative diseases.
Collapse
Affiliation(s)
- Jae-Kyo Jeong
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju, Korea
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, Korea
| | - Sang-Youel Park
- Biosafety Research Institute, College of Veterinary Medicine, Chonbuk National University, Jeonju, Korea
- Department of Bioactive Material Sciences and Research Center of Bioactive Materials, Chonbuk National University, Jeonju, Korea
| |
Collapse
|
50
|
del Caño-Espinel M, Acebes JR, Sanchez D, Ganfornina MD. Lazarillo-related Lipocalins confer long-term protection against type I Spinocerebellar Ataxia degeneration contributing to optimize selective autophagy. Mol Neurodegener 2015; 10:11. [PMID: 25888134 PMCID: PMC4374295 DOI: 10.1186/s13024-015-0009-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2014] [Accepted: 03/02/2015] [Indexed: 12/22/2022] Open
Abstract
Background A diverse set of neurodegenerative disorders are caused by abnormal extensions of polyglutamine (poly-Q) stretches in various, functionally unrelated proteins. A common feature of these diseases is altered proteostasis. Autophagy induction is part of the endogenous response to poly-Q protein expression. However, if autophagy is not resolved properly, clearance of toxic proteins or aggregates cannot occur effectively. Likewise, excessive autophagy induction can cause autophagic stress and neurodegeneration. The Lipocalins ApoD, Glial Lazarillo (GLaz) and Neural Lazarillo (NLaz) are neuroprotectors upon oxidative stress or aging. In this work we test whether these Lipocalins also protect against poly-Q-triggered deterioration of protein quality control systems. Results Using a Drosophila retinal degeneration model of Type-1 Spinocerebellar Ataxia (SCA1) combined with genetic manipulation of NLaz and GLaz expression, we demonstrate that both Lipocalins protect against SCA1 neurodegeneration. They are part of the endogenous transcriptional response to SCA1, and their effect is non-additive, suggesting participation in a similar mechanism. GLaz beneficial effects persist throughout aging, and appears when expressed by degenerating neurons or by retinal support and glial cells. GLaz gain-of-function reduces cell death and the extent of ubiquitinated proteins accumulation, and decreases the expression of Atg8a/LC3, p62 mRNA and protein levels, and GstS1 induction. Over-expression of GLaz is able to reduce p62 and ubiquitinated proteins levels when rapamycin-dependent and SCA1-dependent inductions of autophagy are combined. In the absence of neurodegeneration, GLaz loss-of-function increases Atg8a/LC3 mRNA and p62 protein levels without altering p62 mRNA levels. Knocking-down autophagy, by interfering with Atg8a or p62 expression or by expressing dominant-negative Atg1/ULK1 or Atg4a transgenes, rescues SCA1-dependent neurodegeneration in a similar extent to the protective effect of GLaz. Further GLaz-dependent improvement is concealed. Conclusions This work shows for the first time that a Lipocalin rescues neurons from pathogenic SCA1 degeneration by optimizing clearance of aggregation-prone proteins. GLaz modulates key autophagy genes and lipid-peroxide clearance responsive genes. Down-regulation of selective autophagy causes similar and non-additive rescuing effects. These data suggest that SCA1 neurodegeneration concurs with autophagic stress, and places Lazarillo-related Lipocalins as valuable players in the endogenous protection against the two major contributors to aging and neurodegeneration: ROS-dependent damage and proteostasis deterioration. Electronic supplementary material The online version of this article (doi:10.1186/s13024-015-0009-8) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Manuela del Caño-Espinel
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Judith R Acebes
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Diego Sanchez
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| | - Maria D Ganfornina
- Instituto de Biología y Genética Molecular-Departamento de Bioquímica y Biología Molecular y Fisiología, Universidad de Valladolid-CSIC, c/ Sanz y Forés 3, 47003, Valladolid, Spain.
| |
Collapse
|