1
|
Li P, Liu P, Zang D, Li C, Wang C, Zhu Y, Liu M, Lu L, Wu X, Nie H. Genome-Wide Identification and Expression Analysis of the BTB Gene Superfamily Provides Insight into Sex Determination and Early Gonadal Development of Alligator sinensis. Int J Mol Sci 2024; 25:10771. [PMID: 39409099 PMCID: PMC11477308 DOI: 10.3390/ijms251910771] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2024] [Revised: 09/29/2024] [Accepted: 10/05/2024] [Indexed: 10/20/2024] Open
Abstract
The BTB gene superfamily is widely distributed among higher eukaryotes and plays a significant role in numerous biological processes. However, there is limited knowledge about the structure and function of BTB genes in the critically endangered species Alligator sinensis, which is endemic to China. A total of 170 BTB genes were identified from the A. sinensis genome, classified into 13 families, and unevenly distributed across 16 chromosomes. Analysis of gene duplication events yielded eight pairs of tandem duplication genes and six pairs of segmental duplication genes. Phylogenetics shows that the AsBTB genes are evolutionarily conserved. The cis-regulatory elements in the AsBTB family promoter region reveal their involvement in multiple biological processes. Protein interaction network analysis indicates that the protein interactions of the AsBTB genes are centered around CLU-3, mainly participating in the regulation of biological processes through the ubiquitination pathway. The expression profile and protein interaction network analysis of AsBTB genes during sex differentiation and early gonadal development indicate that AsBTB genes are widely expressed in this process and involves numerous genes and pathways for regulation. This study provides a basis for further investigation of the role of the BTB gene in sex differentiation and gonadal development in A. sinensis.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiaobing Wu
- The Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Science, Anhui Normal University, Wuhu 241000, China; (P.L.); (P.L.); (D.Z.); (C.L.); (C.W.); (Y.Z.); (M.L.); (L.L.)
| | - Haitao Nie
- The Anhui Provincial Key Laboratory of Biodiversity Conservation and Ecological Security in the Yangtze River Basin, College of Life Science, Anhui Normal University, Wuhu 241000, China; (P.L.); (P.L.); (D.Z.); (C.L.); (C.W.); (Y.Z.); (M.L.); (L.L.)
| |
Collapse
|
2
|
Guo X, Prajapati R, Chun J, Byun I, Gebis KK, Wang YZ, Ling K, Dalton C, Blair JA, Hamidianjahromi A, Bachmann G, Rigo F, Jafar-Nejad P, Savas JN, Lee MJ, Sreedharan J, Kalb RG. Reduction of RAD23A extends lifespan and mitigates pathology in TDP-43 mice. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.10.612226. [PMID: 39314471 PMCID: PMC11419047 DOI: 10.1101/2024.09.10.612226] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Protein misfolding and aggregation are cardinal features of neurodegenerative disease (NDD) and they contribute to pathophysiology by both loss-of-function (LOF) and gain-of-function (GOF) mechanisms. This is well exemplified by TDP-43 which aggregates and mislocalizes in several NDDs. The depletion of nuclear TDP-43 leads to reduction in its normal function in RNA metabolism and the cytoplasmic accumulation of TDP-43 leads to aberrant protein homeostasis. A modifier screen found that loss of rad23 suppressed TDP-43 pathology in invertebrate and tissue culture models. Here we show in a mouse model of TDP-43 pathology that genetic or antisense oligonucleotide (ASO)-mediated reduction in rad23a confers benefits on survival and behavior, histological hallmarks of disease and reduction of mislocalized and aggregated TDP-43. This results in improved function of the ubiquitin-proteasome system (UPS) and correction of transcriptomic alterations evoked by pathologic TDP-43. RAD23A-dependent remodeling of the insoluble proteome appears to be a key event driving pathology in this model. As TDP-43 pathology is prevalent in both familial and sporadic NDD, targeting RAD23A may have therapeutic potential.
Collapse
Affiliation(s)
- Xueshui Guo
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Ravindra Prajapati
- Maurice Wohl Clinical Neuroscience Research Institute, King's College London, London, United Kingdom
| | - Jiyeon Chun
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Insuk Byun
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Kamil K Gebis
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Yi-Zhi Wang
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, CA, United States
| | - Casey Dalton
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Jeff A Blair
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Anahid Hamidianjahromi
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, CA, United States
| | | | - Jeffrey N Savas
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul, Korea
| | - Jemeen Sreedharan
- Maurice Wohl Clinical Neuroscience Research Institute, King's College London, London, United Kingdom
| | - Robert G Kalb
- Department of Neurology, Northwestern University School of Medicine, Chicago, IL, United States
| |
Collapse
|
3
|
Hamazaki J, Murata S. Relationships between protein degradation, cellular senescence, and organismal aging. J Biochem 2024; 175:473-480. [PMID: 38348509 PMCID: PMC11058314 DOI: 10.1093/jb/mvae016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2024] [Revised: 02/01/2024] [Accepted: 02/07/2024] [Indexed: 05/01/2024] Open
Abstract
Aging is a major risk factor for many diseases. Recent studies have shown that age-related disruption of proteostasis leads to the accumulation of abnormal proteins and that dysfunction of the two major intracellular proteolytic pathways, the ubiquitin-proteasome pathway, and the autophagy-lysosome pathway, is largely responsible for this process. Conversely, it has been shown that activation of these proteolytic pathways may contribute to lifespan extension and suppression of pathological conditions, making it a promising intervention for anti-aging. This review provides an overview of the important role of intracellular protein degradation in aging and summarizes how the disruption of proteostasis is involved in age-related diseases.
Collapse
Affiliation(s)
- Jun Hamazaki
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| | - Shigeo Murata
- Laboratory of Protein Metabolism, Graduate School of Pharmaceutical Sciences, the University of Tokyo, 7-3-1, Hongo, Bunkyo-ku, Tokyo 1130033, Japan
| |
Collapse
|
4
|
Tan JYK, Chew LY, Juhász G, Yu F. Interplay between autophagy and CncC regulates dendrite pruning in Drosophila. Proc Natl Acad Sci U S A 2024; 121:e2310740121. [PMID: 38408233 PMCID: PMC10927499 DOI: 10.1073/pnas.2310740121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2023] [Accepted: 01/26/2024] [Indexed: 02/28/2024] Open
Abstract
Autophagy is essential for the turnover of damaged organelles and long-lived proteins. It is responsible for many biological processes such as maintaining brain functions and aging. Impaired autophagy is often linked to neurodevelopmental and neurodegenerative diseases in humans. However, the role of autophagy in neuronal pruning during development remains poorly understood. Here, we report that autophagy regulates dendrite-specific pruning of ddaC sensory neurons in parallel to local caspase activation. Impaired autophagy causes the formation of ubiquitinated protein aggregates in ddaC neurons, dependent on the autophagic receptor Ref(2)P. Furthermore, the metabolic regulator AMP-activated protein kinase and the insulin-target of rapamycin pathway act upstream to regulate autophagy during dendrite pruning. Importantly, autophagy is required to activate the transcription factor CncC (Cap "n" collar isoform C), thereby promoting dendrite pruning. Conversely, CncC also indirectly affects autophagic activity via proteasomal degradation, as impaired CncC results in the inhibition of autophagy through sequestration of Atg8a into ubiquitinated protein aggregates. Thus, this study demonstrates the important role of autophagy in activating CncC prior to dendrite pruning, and further reveals an interplay between autophagy and CncC in neuronal pruning.
Collapse
Affiliation(s)
- Jue Yu Kelly Tan
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore117543, Singapore
| | - Liang Yuh Chew
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore117543, Singapore
| | - Gábor Juhász
- Department of Anatomy, Cell and Developmental Biology, Eötvös Loránd University, BudapestH-1117, Hungary
- Institute of Genetics, Biological Research Centre, SzegedH-6726, Hungary
| | - Fengwei Yu
- Temasek Life Sciences Laboratory, National University of Singapore, Singapore117604, Singapore
- Department of Biological Sciences, National University of Singapore, Singapore117543, Singapore
| |
Collapse
|
5
|
Gong X, Huang M, Chen L. NRF1 mitigates motor dysfunction and dopamine neuron degeneration in mice with Parkinson's disease by promoting GLRX m 6 A methylation through upregulation of METTL3 transcription. CNS Neurosci Ther 2024; 30:e14441. [PMID: 37735974 PMCID: PMC10916419 DOI: 10.1111/cns.14441] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2023] [Revised: 06/29/2023] [Accepted: 08/16/2023] [Indexed: 09/23/2023] Open
Abstract
OBJECTIVE The feature of Parkinson's disease (PD) is the heavy dopaminergic neuron loss of substantia nigra pars compacta (SNpc), while glutaredoxin (GLRX) has been discovered to modulate the death of dopaminergic neurons. In this context, this study was implemented to uncover the impact of GRX1 on motor dysfunction and dopamine neuron degeneration in PD mice and its potential mechanism. METHODS A PD mouse model was established via injection with 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) into mice. After gain- and loss-of-function assays in mice, motor coordination was assessed using rotarod, pole, and open-field tests, and neurodegeneration in mouse SNpc tissues was determined using immunohistochemistry of tyrosine hydroxylase and Nissl staining. NRF1, methyltransferase-like 3 (METTL3), and GLRX expression in SNpc tissues were evaluated using qRT-PCR, Western blot, and immunohistochemistry. The N6-methyladenosine (m6 A) levels of GLRX mRNA were examined using MeRIP. The relationship among NRF1, METTL3, and GLRX was determined by RIP, ChIP, and dual luciferase assays. RESULTS Low GLRX, METTL3, and NRF1 expression were observed in MPTP-induced mice, accompanied by decreased m6 A modification level of GLRX mRNA. GLRX overexpression alleviated motor dysfunction and dopamine neuron degeneration in MPTP-induced mice. METTL3 promoted m6 A modification and IGF2BP2-dependent stability of GLRX mRNA, and NRF1 increased METTL3 expression by binding to METTL3 promoter. NRF1 overexpression increased m6 A modification of GLRX mRNA and repressed motor dysfunction and dopamine neuron degeneration in MPTP-induced mice, which was counteracted by METTL3 knockdown. CONCLUSION Conclusively, NRF1 constrained motor dysfunction and dopamine neuron degeneration in MPTP-induced PD mice by activating the METTL3/GLRX axis.
Collapse
Affiliation(s)
- Xin Gong
- Department of Neurosurgery, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaHunanP.R. China
| | - Mengyi Huang
- Department of Neurosurgery, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaHunanP.R. China
| | - Lei Chen
- Department of Neurosurgery, Hunan Provincial People's HospitalThe First Affiliated Hospital of Hunan Normal UniversityChangshaHunanP.R. China
| |
Collapse
|
6
|
Łuczyńska K, Zhang Z, Pietras T, Zhang Y, Taniguchi H. NFE2L1/Nrf1 serves as a potential therapeutical target for neurodegenerative diseases. Redox Biol 2024; 69:103003. [PMID: 38150994 PMCID: PMC10788251 DOI: 10.1016/j.redox.2023.103003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2023] [Revised: 12/15/2023] [Accepted: 12/16/2023] [Indexed: 12/29/2023] Open
Abstract
The failure of the proper protein turnover in the nervous system is mainly linked to a variety of neurodegenerative disorders. Therefore, a better understanding of key protein degradation through the ubiquitin-proteasome system is critical for effective prevention and treatment of those disorders. The proteasome expression is tightly regulated by a CNC (cap'n'collar) family of transcription factors, amongst which the nuclear factor-erythroid 2-like bZIP factor 1 (NFE2L1, also known as Nrf1, with its long isoform TCF11 and short isoform LCR-F1) has been identified as an indispensable regulator of the transcriptional expression of the ubiquitin-proteasome system. However, much less is known about how the pivotal role of NFE2L1/Nrf1, as compared to its homologous NFE2L2 (also called Nrf2), is translated to its physiological and pathophysiological functions in the nervous system insomuch as to yield its proper cytoprotective effects against neurodegenerative diseases. The potential of NFE2L1 to fulfill its unique neuronal function to serve as a novel therapeutic target for neurodegenerative diseases is explored by evaluating the hitherto established preclinical and clinical studies of Alzheimer's and Parkinson's diseases. In this review, we have also showcased a group of currently available activators of NFE2L1, along with an additional putative requirement of this CNC-bZIP factor for healthy longevity based on the experimental evidence obtained from its orthologous SKN1-A in Caenorhabditis elegans.
Collapse
Affiliation(s)
- Kamila Łuczyńska
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Poland; The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957, Warsaw, Poland
| | - Zhengwen Zhang
- Laboratory of Neuroscience, Institute of Cognitive Neuroscience and School of Pharmacy, University College London, 29-39 Brunswick Square, London, WC1N 1AX, England, United Kingdom
| | - Tadeusz Pietras
- The Second Department of Psychiatry, Institute of Psychiatry and Neurology in Warsaw, 02-957, Warsaw, Poland; Department of Clinical Pharmacology, Medical University of Lodz, 90-153, Łódź, Poland
| | - Yiguo Zhang
- Chongqing University Jiangjin Hospital, School of Medicine, Chongqing University, No. 725 Jiangzhou Avenue, Dingshan Street, Jiangjin District, Chongqing, 402260, China; The Laboratory of Cell Biochemistry and Topogenetic Regulation, College of Bioengineering & Faculty of Medical Sciences, Chongqing University, No. 174 Shazheng Street, Shapingba District, Chongqing, 400044, China.
| | - Hiroaki Taniguchi
- Institute of Genetics and Animal Biotechnology of the Polish Academy of Sciences, Jastrzebiec, 05-552, Poland.
| |
Collapse
|
7
|
Hatanaka A, Nakada S, Matsumoto G, Satoh K, Aketa I, Watanabe A, Hirakawa T, Tsujita T, Waku T, Kobayashi A. The transcription factor NRF1 (NFE2L1) activates aggrephagy by inducing p62 and GABARAPL1 after proteasome inhibition to maintain proteostasis. Sci Rep 2023; 13:14405. [PMID: 37658135 PMCID: PMC10474156 DOI: 10.1038/s41598-023-41492-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2023] [Accepted: 08/28/2023] [Indexed: 09/03/2023] Open
Abstract
The ubiquitin‒proteasome system (UPS) and autophagy are the two primary cellular pathways of misfolded or damaged protein degradation that maintain cellular proteostasis. When the proteasome is dysfunctional, cells compensate for impaired protein clearance by activating aggrephagy, a type of selective autophagy, to eliminate ubiquitinated protein aggregates; however, the molecular mechanisms by which impaired proteasome function activates aggrephagy remain poorly understood. Here, we demonstrate that activation of aggrephagy is transcriptionally induced by the transcription factor NRF1 (NFE2L1) in response to proteasome dysfunction. Although NRF1 has been previously shown to induce the expression of proteasome genes after proteasome inhibition (i.e., the proteasome bounce-back response), our genome-wide transcriptome analyses identified autophagy-related p62/SQSTM1 and GABARAPL1 as genes directly targeted by NRF1. Intriguingly, NRF1 was also found to be indispensable for the formation of p62-positive puncta and their colocalization with ULK1 and TBK1, which play roles in p62 activation via phosphorylation. Consistently, NRF1 knockdown substantially reduced the phosphorylation rate of Ser403 in p62. Finally, NRF1 selectively upregulated the expression of GABARAPL1, an ATG8 family gene, to induce the clearance of ubiquitinated proteins. Our findings highlight the discovery of an activation mechanism underlying NRF1-mediated aggrephagy through gene regulation when proteasome activity is impaired.
Collapse
Affiliation(s)
- Atsushi Hatanaka
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto, 610-0394, Japan
- Research Fellow of Japan Society for the Promotion of Science, Tokyo, Japan
| | - Sota Nakada
- Laboratory for Genetic Code, Department of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Gen Matsumoto
- Department of Anatomy and Neurobiology, Nagasaki University School of Medicine, Nagasaki, Japan
| | - Katsuya Satoh
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto, 610-0394, Japan
| | - Iori Aketa
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto, 610-0394, Japan
| | - Akira Watanabe
- Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Tomoaki Hirakawa
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Tadayuki Tsujita
- Laboratory of Biochemistry, Faculty of Agriculture, Saga University, Saga, Japan
- The United Graduate School of Agricultural Sciences, Kagoshima University, Kagoshima, Japan
| | - Tsuyoshi Waku
- Laboratory for Genetic Code, Department of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan
| | - Akira Kobayashi
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, 1-3 Tatara Miyakodani, Kyotanabe, Kyoto, 610-0394, Japan.
- Laboratory for Genetic Code, Department of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto, Japan.
| |
Collapse
|
8
|
Wang Y, Snell A, Dyka FM, Colvin ER, Ildefonso C, Ash JD, Lobanova ES. Overexpression of Nfe2l1 increases proteasome activity and delays vision loss in a preclinical model of human blindness. SCIENCE ADVANCES 2023; 9:eadd5479. [PMID: 37450596 PMCID: PMC10348684 DOI: 10.1126/sciadv.add5479] [Citation(s) in RCA: 3] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Accepted: 06/14/2023] [Indexed: 07/18/2023]
Abstract
Proteasomes are the central proteolytic machines that are critical for breaking down most of the damaged and abnormal proteins in human cells. Although universally applicable drugs are not yet available, the stimulation of proteasomal activity is being analyzed as a proof-of-principle strategy to increase cellular resistance to a broad range of proteotoxic stressors. These approaches have included the stimulation of proteasomes through the overexpression of individual proteasome subunits, phosphorylation, or conformational changes induced by small molecules or peptides. In contrast to these approaches, we evaluated a transcription-driven increase in the total proteasome pool to enhance the proteolytic capacity of degenerating retinal neurons. We show that overexpression of nuclear factor erythroid-2-like 1 (Nfe2l1) transcription factor stimulated proteasome biogenesis and activity, improved the clearance of the ubiquitin-proteasomal reporter, and delayed photoreceptor neuron loss in a preclinical mouse model of human blindness caused by misfolded proteins. The findings highlight Nfe2l1 as an emerging therapeutic target to treat neurodegenerative diseases linked to protein misfolding.
Collapse
Affiliation(s)
- Yixiao Wang
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Aaron Snell
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Frank M. Dyka
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Elizabeth R. Colvin
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - Cristhian Ildefonso
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
| | - John D. Ash
- Department of Ophthalmology, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Ekaterina S. Lobanova
- Department of Ophthalmology, University of Florida, Gainesville, FL 32610, USA
- Department of Pharmacology and Therapeutics, University of Florida, Gainesville, FL 32610, USA
| |
Collapse
|
9
|
Kim Y, Kim EK, Chey Y, Song MJ, Jang HH. Targeted Protein Degradation: Principles and Applications of the Proteasome. Cells 2023; 12:1846. [PMID: 37508510 PMCID: PMC10378610 DOI: 10.3390/cells12141846] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2023] [Revised: 07/10/2023] [Accepted: 07/12/2023] [Indexed: 07/30/2023] Open
Abstract
The proteasome is a multi-catalytic protease complex that is involved in protein quality control via three proteolytic activities (i.e., caspase-, trypsin-, and chymotrypsin-like activities). Most cellular proteins are selectively degraded by the proteasome via ubiquitination. Moreover, the ubiquitin-proteasome system is a critical process for maintaining protein homeostasis. Here, we briefly summarize the structure of the proteasome, its regulatory mechanisms, proteins that regulate proteasome activity, and alterations to proteasome activity found in diverse diseases, chemoresistant cells, and cancer stem cells. Finally, we describe potential therapeutic modalities that use the ubiquitin-proteasome system.
Collapse
Affiliation(s)
- Yosup Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Eun-Kyung Kim
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Yoona Chey
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Min-Jeong Song
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
| | - Ho Hee Jang
- Department of Biochemistry, College of Medicine, Gachon University, Incheon 21999, Republic of Korea
- Department of Health Sciences and Technology, Gachon Advanced Institute for Health Sciences and Technology (GAIHST), Gachon University, Incheon 21999, Republic of Korea
- Lee Gil Ya Cancer and Diabetes Institute, Gachon University, Incheon 21999, Republic of Korea
| |
Collapse
|
10
|
Choi WH, Yun Y, Byun I, Kim S, Lee S, Sim J, Levi S, Park SH, Jun J, Kleifeld O, Kim KP, Han D, Chiba T, Seok C, Kwon YT, Glickman MH, Lee MJ. ECPAS/Ecm29-mediated 26S proteasome disassembly is an adaptive response to glucose starvation. Cell Rep 2023; 42:112701. [PMID: 37384533 DOI: 10.1016/j.celrep.2023.112701] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 05/07/2023] [Accepted: 06/09/2023] [Indexed: 07/01/2023] Open
Abstract
The 26S proteasome comprises 20S catalytic and 19S regulatory complexes. Approximately half of the proteasomes in cells exist as free 20S complexes; however, our mechanistic understanding of what determines the ratio of 26S to 20S species remains incomplete. Here, we show that glucose starvation uncouples 26S holoenzymes into 20S and 19S subcomplexes. Subcomplex affinity purification and quantitative mass spectrometry reveal that Ecm29 proteasome adaptor and scaffold (ECPAS) mediates this structural remodeling. The loss of ECPAS abrogates 26S dissociation, reducing degradation of 20S proteasome substrates, including puromycylated polypeptides. In silico modeling suggests that ECPAS conformational changes commence the disassembly process. ECPAS is also essential for endoplasmic reticulum stress response and cell survival during glucose starvation. In vivo xenograft model analysis reveals elevated 20S proteasome levels in glucose-deprived tumors. Our results demonstrate that the 20S-19S disassembly is a mechanism adapting global proteolysis to physiological needs and countering proteotoxic stress.
Collapse
Affiliation(s)
- Won Hoon Choi
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Yejin Yun
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Insuk Byun
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Sumin Kim
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Seho Lee
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Jiho Sim
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Shahar Levi
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Seo Hyeong Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| | - Jeongmoo Jun
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Oded Kleifeld
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel
| | - Kwang Pyo Kim
- Department of Applied Chemistry, Institute of Natural Science, Global Center for Pharmaceutical Ingredient Materials, Kyung Hee University, Yongin 17104, Korea
| | - Dohyun Han
- Proteomics Core Facility, Biomedical Research Institute, Seoul National University Hospital, Seoul 03080, Korea
| | - Tomoki Chiba
- Graduate School of Life and Environmental Sciences, University of Tsukuba, Ibaraki 305-8577, Japan
| | - Chaok Seok
- Department of Chemistry, Seoul National University, Seoul 08826, Korea
| | - Yong Tae Kwon
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea; Ischemic/Hypoxic Disease Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Michael H Glickman
- Department of Biology, Technion-Israel Institute of Technology, Haifa 32000, Israel.
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea; Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea; Ischemic/Hypoxic Disease Institute, Convergence Research Center for Dementia, Seoul National University College of Medicine, Seoul 03080, Korea.
| |
Collapse
|
11
|
Chang X, Liu J, Wang Y, Guan X, Liu R. Mitochondrial disorder and treatment of ischemic cardiomyopathy: Potential and advantages of Chinese herbal medicine. Biomed Pharmacother 2023; 159:114171. [PMID: 36641924 DOI: 10.1016/j.biopha.2022.114171] [Citation(s) in RCA: 15] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 12/22/2022] [Accepted: 12/28/2022] [Indexed: 01/14/2023] Open
Abstract
Mitochondrial dysfunction is the main cause of damage to the pathological mechanism of ischemic cardiomyopathy. In addition, mitochondrial dysfunction can also affect the homeostasis of cardiomyocytes or endothelial cell dysfunction, leading to a vicious cycle of mitochondrial oxidative stress. And mitochondrial dysfunction is also an important pathological basis for ischemic cardiomyopathy and reperfusion injury after myocardial infarction or end-stage coronary heart disease. Therefore, mitochondria can be used as therapeutic targets against myocardial ischemia injury, and the regulation of mitochondrial morphology, function and structure is a key and important way of targeting mitochondrial quality control therapeutic mechanisms. Mitochondrial quality control includes mechanisms such as mitophagy, mitochondrial dynamics (mitochondrial fusion/fission), mitochondrial biosynthesis, and mitochondrial unfolded protein responses. Among them, the increase of mitochondrial fragmentation caused by mitochondrial pathological fission is the initial factor. The protective mitochondrial fusion can strengthen the interaction and synthesis of paired mitochondria and promote mitochondrial biosynthesis. In ischemia or hypoxia, pathological mitochondrial fission can promote the formation of mitochondrial fragments, fragmented mitochondria can lead to damaged mitochondrial DNA production, which can lead to mitochondrial biosynthesis dysfunction, insufficient mitochondrial ATP production, and mitochondrial ROS. Burst growth or loss of mitochondrial membrane potential. This eventually leads to the accumulation of damaged mitochondria. Then, under the leadership of mitophagy, damaged mitochondria can complete the mitochondrial degradation process through mitophagy, and transport the morphologically and structurally damaged mitochondria to lysosomes for degradation. But once the pathological mitochondrial fission increases, the damaged mitochondria increases, which may activate the pathway of cardiomyocyte death. Although laboratory studies have found that a variety of mitochondrial-targeted drugs can reduce myocardial ischemia and protect cardiomyocytes, there are still few drugs that have successfully passed clinical trials. In this review, we describe the role of MQS in ischemia/hypoxia-induced cardiomyocyte physiopathology and elucidate the relevant mechanisms of mitochondrial dysfunction in ischemic cardiomyopathy. In addition, we also further explained the advantages of natural products in improving mitochondrial dysfunction and protecting myocardial cells from the perspective of pharmacological mechanism, and explained its related mechanisms. Potential targeted therapies that can be used to improve MQS under ischemia/hypoxia are discussed, aiming to accelerate the development of cardioprotective drugs targeting mitochondrial dysfunction.
Collapse
Affiliation(s)
- Xing Chang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Jinfeng Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Yanli Wang
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Xuanke Guan
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China
| | - Ruxiu Liu
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, 100053, China.
| |
Collapse
|
12
|
Harada Y, Ohkawa Y, Maeda K, Taniguchi N. Glycan quality control in and out of the endoplasmic reticulum of mammalian cells. FEBS J 2022; 289:7147-7162. [PMID: 34492158 DOI: 10.1111/febs.16185] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2021] [Revised: 08/23/2021] [Accepted: 09/06/2021] [Indexed: 01/13/2023]
Abstract
The endoplasmic reticulum (ER) is equipped with multiple quality control systems (QCS) that are necessary for shaping the glycoproteome of eukaryotic cells. These systems facilitate the productive folding of glycoproteins, eliminate defective products, and function as effectors to evoke cellular signaling in response to various cellular stresses. These ER functions largely depend on glycans, which contain sugar-based codes that, when needed, function to recruit carbohydrate-binding proteins that determine the fate of glycoproteins. To ensure their functionality, the biosynthesis of such glycans is therefore strictly monitored by a system that selectively degrades structurally defective glycans before adding them to proteins. This system, which is referred to as the glycan QCS, serves as a mechanism to reduce the risk of abnormal glycosylation under conditions where glycan biosynthesis is genetically or metabolically stalled. On the other hand, glycan QCS increases the risk of global hypoglycosylation by limiting glycan availability, which can lead to protein misfolding and the activation of unfolded protein response to maintaining cell viability or to initiate cell death programs. This review summarizes the current state of our knowledge of the mechanisms underlying glycan QCS in mammals and its physiological and pathological roles in embryogenesis, tumor progression, and congenital disorders associated with abnormal glycosylation.
Collapse
Affiliation(s)
- Yoichiro Harada
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Yuki Ohkawa
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Kento Maeda
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| | - Naoyuki Taniguchi
- Department of Glyco-Oncology and Medical Biochemistry, Osaka International Cancer Institute, Osaka, Japan
| |
Collapse
|
13
|
Activation of the membrane-bound Nrf1 transcription factor by USP19, a ubiquitin-specific protease C-terminally anchored in the endoplasmic reticulum. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2022; 1869:119299. [PMID: 35613680 DOI: 10.1016/j.bbamcr.2022.119299] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/04/2021] [Revised: 05/12/2022] [Accepted: 05/16/2022] [Indexed: 11/23/2022]
Abstract
The membrane-bound transcription factor Nrf1 (encoded by Nfe2l1) is activated by sensing glucose deprivation, cholesterol abundance, proteasomal inhibition and oxidative stress and then mediates distinct signaling responses to maintain cellular homeostasis. Herein, we found that Nrf1 stability and transactivity are both enhanced by USP19, a ubiquitin-specific protease tail-anchored in the endoplasmic reticulum (ER) through its C-terminal transmembrane domain. Further experiments revealed that USP19 directly interacts with Nrf1 in proximity to the ER and topologically acts as a deubiquitinating enzyme to remove ubiquitin moieties from this protein, which allow it to circumvent potential proteasomal degradation. This USP19-mediated effect takes place only after Nrf1 is retro-translocated by p97 out of the ER membrane to dislocate the cytoplasmic side. Conversely, knockout of USP19 causes significant decreases in the abundance of Nrf1 and the entrance of its active isoform into the nucleus, which result in the downregulation of its target proteasomal subunits and a modest reduction in USP19-/--derived tumor growth in xenograft mice when compared with wild-type controls. Altogether, these results demonstrate that USP19 serves as a novel mechanistic modulator of Nrf1, but not Nrf2, thereby enabling Nrf1 to be rescued from the putative ubiquitin-directed ER-associated degradation pathway. In turn, our additional experimental evidence has revealed that transcriptional expression of endogenous USP19 and its promoter-driven reporter genes is differentially regulated by Nrf2, as well by Nrf1, at distinct layers within a complex hierarchical regulatory network.
Collapse
|
14
|
Dysfunction of the energy sensor NFE2L1 triggers uncontrollable AMPK signaling and glucose metabolism reprogramming. Cell Death Dis 2022; 13:501. [PMID: 35614059 PMCID: PMC9133051 DOI: 10.1038/s41419-022-04917-3] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2021] [Revised: 04/28/2022] [Accepted: 05/05/2022] [Indexed: 12/14/2022]
Abstract
The antioxidant transcription factor NFE2L1 (also called Nrf1) acts as a core regulator of redox signaling and metabolism homeostasis, and thus, its dysfunction results in multiple systemic metabolic diseases. However, the molecular mechanism(s) by which NFE2L1 regulates glycose and lipid metabolism remains elusive. Here, we found that loss of NFE2L1 in human HepG2 cells led to a lethal phenotype upon glucose deprivation and NFE2L1 deficiency could affect the uptake of glucose. Further experiments revealed that glycosylation of NFE2L1 enabled it to sense the energy state. These results indicated that NFE2L1 can serve as a dual sensor and regulator of glucose homeostasis. The transcriptome, metabolome, and seahorse data further revealed that disruption of NFE2L1 could reprogram glucose metabolism to aggravate the Warburg effect in NFE2L1-silenced hepatoma cells, concomitant with mitochondrial damage. Co-expression and Co-immunoprecipitation experiments demonstrated that NFE2L1 could directly interact and inhibit AMPK. Collectively, NFE2L1 functioned as an energy sensor and negatively regulated AMPK signaling through directly interacting with AMPK. The novel NFE2L1/AMPK signaling pathway delineate the mechanism underlying of NFE2L1-related metabolic diseases and highlight the crosstalk between redox homeostasis and metabolism homeostasis.
Collapse
|
15
|
Abstract
The mechanistic target of rapamycin (mTOR) regulates numerous extracellular and intracellular signals involved in the maintenan-ce of cellular homeostasis and cell growth. mTOR also functions as an endogenous inhibitor of autophagy. Under nutrient-rich conditions, mTOR complex 1 (mTORC1) phosphorylates the ULK1 complex, preventing its activation and subsequent autophagosome formation, while inhibition of mTORC1 using either rapamycin or nutrient deprivation induces autophagy. Autophagy and proteasomal proteolysis provide amino acids necessary for protein translation. Although the connection between mTORC1 and autophagy is well characterized, the association of mTORC1 inhibition with proteasome biogenesis and activity has not been fully elucidated yet. Proteasomes are long-lived cellular organelles. Their spatiotemporal rather than homeostatic regulation could be another adaptive cellular mechanism to respond to starvation. Here, we reviewed several published reports and the latest research from our group to examine the connection between mTORC1 and proteasome. We have also investigated and described the effect of mTORC1 inhibition on proteasome activity using purified proteasomes. Since mTORC1 inhibitors are currently evaluated as treatments for several human diseases, a better understanding of the link between mTORC1 activity and proteasome function is of utmost importance.
Collapse
Affiliation(s)
- Seo Hyeong Park
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won Hoon Choi
- BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Min Jae Lee
- Department of Biochemistry and Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
| |
Collapse
|
16
|
Na HJ, Abramowitz LK, Hanover JA. Cytosolic O-GlcNAcylation and PNG1 maintain Drosophila gut homeostasis by regulating proliferation and apoptosis. PLoS Genet 2022; 18:e1010128. [PMID: 35294432 PMCID: PMC8959174 DOI: 10.1371/journal.pgen.1010128] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2021] [Revised: 03/28/2022] [Accepted: 03/02/2022] [Indexed: 11/29/2022] Open
Abstract
Tissue homeostasis requires a delicate balance between stem cell self-renewal, proliferation, and differentiation. Essential to this process is glycosylation, with both intra-and extra-cellular glycosylation being required for stem cell homeostasis. However, it remains unknown how intracellular glycosylation, O-GlcNAcylation, interfaces with cellular components of the extracellular glycosylation machinery, like the cytosolic N-glycanase NGLY1. In this study, we utilize the Drosophila gut and uncover a pathway in which O-GlcNAcylation cooperates with the NGLY1 homologue PNG1 to regulate proliferation in intestinal stem cells (ISCs) and apoptosis in differentiated enterocytes. Further, the CncC antioxidant signaling pathway and ENGase, an enzyme involved in the processing of free oligosaccharides in the cytosol, interact with O-GlcNAc and PNG1 through regulation of protein aggregates to contribute to gut maintenance. These findings reveal a complex coordinated regulation between O-GlcNAcylation and the cytosolic glycanase PNG1 critical to balancing proliferation and apoptosis to maintain gut homeostasis. Understanding glycosylation is of great importance as changes in both intra- and extra-cellular glycosylation have been well documented in diseases. However, it remains relatively unclear how intracellular glycosylation, O-GlcNAcylation, interfaces with the cellular machinery of extracellular glycosylation. In this study, we seek to understand how coordination between a cytosolic N-glycanase, NGLY1, and intracellular glycosylation, is utilized during stem cell proliferation. Using the Drosophila gut as a model system, our data showed O-GlcNAcylation and the Drosophila homologue of NGY1, PNG1, have key roles in both progenitor and differentiated cells that contribute to tissue homeostasis. Further, the CncC antioxidant signaling pathway and ENGase, an enzyme involved in the processing of free oligosaccharides in the cytosol, interact with O-GlcNAc and PNG1 through regulation of protein aggregates to contribute to gut maintenance. Our study suggests that alterations in O-GlcNAc and NGLY1 could be future targets utilized as therapeutics for nutrient-sensitive stem cell-derived diseases such as cancer.
Collapse
Affiliation(s)
- Hyun-jin Na
- Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - Lara K. Abramowitz
- Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
| | - John A. Hanover
- Laboratory of Cellular and Molecular Biology, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland, United States of America
- * E-mail:
| |
Collapse
|
17
|
Lee HJ, Lee DM, Seo MJ, Kang HC, Kwon SK, Choi KS. PSMD14 Targeting Triggers Paraptosis in Breast Cancer Cells by Inducing Proteasome Inhibition and Ca 2+ Imbalance. Int J Mol Sci 2022; 23:ijms23052648. [PMID: 35269789 PMCID: PMC8910635 DOI: 10.3390/ijms23052648] [Citation(s) in RCA: 10] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 02/23/2022] [Accepted: 02/25/2022] [Indexed: 02/06/2023] Open
Abstract
PSMD14, a subunit of the 19S regulatory particles of the 26S proteasome, was recently identified as a potential prognostic marker and therapeutic target in diverse human cancers. Here, we show that the silencing and pharmacological blockade of PSMD14 in MDA-MB 435S breast cancer cells induce paraptosis, a non-apoptotic cell death mode characterized by extensive vacuolation derived from the endoplasmic reticulum (ER) and mitochondria. The PSMD14 inhibitor, capzimin (CZM), inhibits proteasome activity but differs from the 20S proteasome subunit-inhibiting bortezomib (Bz) in that it does not induce aggresome formation or Nrf1 upregulation, which underlie Bz resistance in cancer cells. In addition to proteasome inhibition, the release of Ca2+ from the ER into the cytosol critically contributes to CZM-induced paraptosis. Induction of paraptosis by targeting PSMD14 may provide an attractive therapeutic strategy against cancer cells resistant to proteasome inhibitors or pro-apoptotic drugs.
Collapse
Affiliation(s)
- Hong-Jae Lee
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (H.-J.L.); (D.-M.L.); (M.-J.S.)
| | - Dong-Min Lee
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (H.-J.L.); (D.-M.L.); (M.-J.S.)
| | - Min-Ji Seo
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (H.-J.L.); (D.-M.L.); (M.-J.S.)
| | - Ho-Chul Kang
- Department of Physiology, Ajou University School of Medicine, Suwon 16499, Korea;
| | - Seok-Kyu Kwon
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul 02792, Korea;
- Division of Bio-Medical Science & Technology, KIST School, Korea University of Science & Technology (UST), Daejeon 34113, Korea
| | - Kyeong-Sook Choi
- Department of Biochemistry and Molecular Biology, Ajou University, Suwon 16499, Korea; (H.-J.L.); (D.-M.L.); (M.-J.S.)
- Correspondence: ; Tel.: +82-31-219-4552; Fax: +82-31-219-5059
| |
Collapse
|
18
|
Ishii K, Fusegi M, Mori T, Teshima K, Ninomiya N, Kohno K, Sato A, Ishida T, Miyakoshi Y, Yano T. A Redox-Silent Analogue of Tocotrienol May Break the Homeostasis of Proteasomes in Human Malignant Mesothelioma Cells by Inhibiting STAT3 and NRF1. Int J Mol Sci 2022; 23:ijms23052655. [PMID: 35269802 PMCID: PMC8910454 DOI: 10.3390/ijms23052655] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2022] [Revised: 02/25/2022] [Accepted: 02/26/2022] [Indexed: 02/04/2023] Open
Abstract
6-O-Carboxypropyl-alpha-tocotrienol (α-T3E) is a multi-target redox-silent analogue of tocotrienol that exhibits cytotoxicity against many cancer cells, including malignant mesothelioma (MM) cells. α-T3E has several molecular targets to effectively induce cytotoxicity against MM cells; however, the mechanisms underlying this cytotoxicity remain unclear. In the present study, we demonstrated that the α-T3E-dependent disruption of the homeostasis of proteasomes strongly induced endoplasmic reticulum (ER) stress, which resulted in effective cytotoxicity against MM cells. The α-T3E-dependent disruption of the homeostasis of proteasomes depended on decreases in proteasome subunits via the inactivation of signal transducer and activator of transcription 3 (STAT3) and nuclear factor erythroid 2 related factor-1 (NRF1), which inhibited protease activity, such as chymotrypsin-like activity, in proteasomes. The α-T3E-dependent inhibition of this activity also induced severe ER stress and ultimately resulted in effective cytotoxicity against MM cells with chemoresistance. The present results indicate that α-T3E acts as an effective anti-mesothelioma agent by disrupting the homeostasis of proteasomes through the simultaneous inactivation of STAT3 and NRF1.
Collapse
Affiliation(s)
- Kyota Ishii
- Laboratory of Molecular Bromacology, Graduate School of Food and Nutritional Sciences, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.I.); (M.F.)
| | - Momoka Fusegi
- Laboratory of Molecular Bromacology, Graduate School of Food and Nutritional Sciences, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.I.); (M.F.)
| | - Tatsuki Mori
- Department of Food and Life Sciences, Faculty of Food and Nutritional Sciences, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (T.M.); (K.T.); (N.N.)
| | - Kosuke Teshima
- Department of Food and Life Sciences, Faculty of Food and Nutritional Sciences, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (T.M.); (K.T.); (N.N.)
| | - Nanako Ninomiya
- Department of Food and Life Sciences, Faculty of Food and Nutritional Sciences, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (T.M.); (K.T.); (N.N.)
| | - Kakeru Kohno
- Research Institute of Life Innovation, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.K.); (A.S.); (T.I.); (Y.M.)
| | - Ayami Sato
- Research Institute of Life Innovation, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.K.); (A.S.); (T.I.); (Y.M.)
| | - Tatsuya Ishida
- Research Institute of Life Innovation, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.K.); (A.S.); (T.I.); (Y.M.)
| | - Yuichi Miyakoshi
- Research Institute of Life Innovation, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.K.); (A.S.); (T.I.); (Y.M.)
| | - Tomohiro Yano
- Research Institute of Life Innovation, Toyo University, Oura District, Gunma, Itakura Town 374-0193, Japan; (K.K.); (A.S.); (T.I.); (Y.M.)
- Correspondence: ; Tel./Fax: +81-276-82-9143
| |
Collapse
|
19
|
Kim S, Park SH, Choi WH, Lee MJ. Evaluation of Immunoproteasome-Specific Proteolytic Activity Using Fluorogenic Peptide Substrates. Immune Netw 2022; 22:e28. [PMID: 35799704 PMCID: PMC9250865 DOI: 10.4110/in.2022.22.e28] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 03/29/2022] [Accepted: 04/05/2022] [Indexed: 12/01/2022] Open
Abstract
The 26S proteasome irreversibly hydrolyzes polyubiquitylated substrates to maintain protein homeostasis; it also regulates immune responses by generating antigenic peptides. An alternative form of the 26S proteasome is the immunoproteasome, which contains substituted catalytic subunits (β1i/PSMB9, β2i/PSMB10, and β5i/PSMB8) instead of constitutively expressed counterparts (β1/PSMB6, β2/PSMB7, and β5/PSMB5). The immunoproteasome expands the peptide repertoire presented on MHC class I molecules. However, how its activity changes in this context is largely elusive, possibly due to the lack of a standardized methodology to evaluate its specific activity. Here, we describe an assay protocol that measures the immunoproteasome activity of whole-cell lysates using commercially available fluorogenic peptide substrates. Our results showed that the most accurate assessment of immunoproteasome activity could be achieved by combining β5i-targeting substrate Ac-ANW-AMC and immunoproteasome inhibitor ONX-0914. This simple and reliable protocol may contribute to future studies of immunoproteasomes and their pathophysiological roles during viral infection, inflammation, and tumorigenesis.
Collapse
Affiliation(s)
- Sumin Kim
- Department of Biochemistry & Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Seo Hyeong Park
- Department of Biochemistry & Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Won Hoon Choi
- Department of Biochemistry & Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- BK21 FOUR Biomedical Science Program, Seoul National University College of Medicine, Seoul 03080, Korea
| | - Min Jae Lee
- Department of Biochemistry & Molecular Biology, Seoul National University College of Medicine, Seoul 03080, Korea
- Department of Biomedical Sciences, Seoul National University Graduate School, Seoul 03080, Korea
| |
Collapse
|
20
|
Oleoylethanolamide Reduces Hepatic Oxidative Stress and Endoplasmic Reticulum Stress in High-Fat Diet-Fed Rats. Antioxidants (Basel) 2021; 10:antiox10081289. [PMID: 34439537 PMCID: PMC8389293 DOI: 10.3390/antiox10081289] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2021] [Revised: 08/03/2021] [Accepted: 08/11/2021] [Indexed: 12/30/2022] Open
Abstract
Long-term high-fat diet (HFD) consumption can cause weight gain and obesity, two conditions often associated with hepatic non-alcoholic fatty liver and oxidative stress. Oleoylethanolamide (OEA), a lipid compound produced by the intestine from oleic acid, has been associated with different beneficial effects in diet-induced obesity and hepatic steatosis. However, the role of OEA on hepatic oxidative stress has not been fully elucidated. In this study, we used a model of diet-induced obesity to study the possible antioxidant effect of OEA in the liver. In this model rats with free access to an HFD for 77 days developed obesity, steatosis, and hepatic oxidative stress, as compared to rats consuming a low-fat diet for the same period. Several parameters associated with oxidative stress were then measured after two weeks of OEA administration to diet-induced obese rats. We showed that OEA reduced, compared to HFD-fed rats, obesity, steatosis, and the plasma level of triacylglycerols and transaminases. Moreover, OEA decreased the amount of malondialdehyde and carbonylated proteins and restored the activity of antioxidant enzymes superoxide dismutase, catalase, and glutathione peroxidase, which decreased in the liver of HFD-fed rats. OEA had also an improving effect on parameters linked to endoplasmic reticulum stress, thus demonstrating a role in the homeostatic control of protein folding. Finally, we reported that OEA differently regulated the expression of two transcription factors involved in the control of lipid metabolism and antioxidant genes, namely nuclear factor erythroid-derived 2-related factor 1 (Nrf1) and Nrf2, thus suggesting, for the first time, new targets of the protective effect of OEA in the liver.
Collapse
|
21
|
Igarashi K, Nishizawa H, Saiki Y, Matsumoto M. The transcription factor BACH1 at the crossroads of cancer biology: From epithelial-mesenchymal transition to ferroptosis. J Biol Chem 2021; 297:101032. [PMID: 34339740 PMCID: PMC8387770 DOI: 10.1016/j.jbc.2021.101032] [Citation(s) in RCA: 43] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2021] [Revised: 07/27/2021] [Accepted: 07/29/2021] [Indexed: 02/07/2023] Open
Abstract
The progression of cancer involves not only the gradual evolution of cells by mutations in DNA but also alterations in the gene expression induced by those mutations and input from the surrounding microenvironment. Such alterations contribute to cancer cells' abilities to reprogram metabolic pathways and undergo epithelial-to-mesenchymal transition (EMT), which facilitate the survival of cancer cells and their metastasis to other organs. Recently, BTB and CNC homology 1 (BACH1), a heme-regulated transcription factor that represses genes involved in iron and heme metabolism in normal cells, was shown to shape the metabolism and metastatic potential of cancer cells. The growing list of BACH1 target genes in cancer cells reveals that BACH1 promotes metastasis by regulating various sets of genes beyond iron metabolism. BACH1 represses the expression of genes that mediate cell–cell adhesion and oxidative phosphorylation but activates the expression of genes required for glycolysis, cell motility, and matrix protein degradation. Furthermore, BACH1 represses FOXA1 gene encoding an activator of epithelial genes and activates SNAI2 encoding a repressor of epithelial genes, forming a feedforward loop of EMT. By synthesizing these observations, we propose a “two-faced BACH1 model”, which accounts for the dynamic switching between metastasis and stress resistance along with cancer progression. We discuss here the possibility that BACH1-mediated promotion of cancer also brings increased sensitivity to iron-dependent cell death (ferroptosis) through crosstalk of BACH1 target genes, imposing programmed vulnerability upon cancer cells. We also discuss the future directions of this field, including the dynamics and plasticity of EMT.
Collapse
Affiliation(s)
- Kazuhiko Igarashi
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan.
| | - Hironari Nishizawa
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Yuriko Saiki
- Department of Investigative Pathology, Tohoku University Graduate School of Medicine, Sendai, Japan
| | - Mitsuyo Matsumoto
- Department of Biochemistry, Tohoku University Graduate School of Medicine, Sendai, Japan; Center for Regulatory Epigenome and Diseases, Tohoku University Graduate School of Medicine, Sendai, Japan
| |
Collapse
|
22
|
Vriend J, Nachtigal MW. Ubiquitin Proteasome Pathway Transcriptome in Epithelial Ovarian Cancer. Cancers (Basel) 2021; 13:cancers13112659. [PMID: 34071321 PMCID: PMC8198060 DOI: 10.3390/cancers13112659] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2021] [Revised: 05/22/2021] [Accepted: 05/24/2021] [Indexed: 12/26/2022] Open
Abstract
In this article, we reviewed the transcription of genes coding for components of the ubiquitin proteasome pathway in publicly available datasets of epithelial ovarian cancer (EOC). KEGG analysis was used to identify the major pathways distinguishing EOC of low malignant potential (LMP) from invasive high-grade serous ovarian carcinomas (HGSOC), and to identify the components of the ubiquitin proteasome system that contributed to these pathways. We identified elevated transcription of several genes encoding ubiquitin conjugases associated with HGSOC. Fifty-eight genes coding for ubiquitin ligases and more than 100 genes encoding ubiquitin ligase adaptors that were differentially expressed between LMP and HGSOC were also identified. Many differentially expressed genes encoding E3 ligase adaptors were Cullin Ring Ligase (CRL) adaptors, and 64 of them belonged to the Cullin 4 DCX/DWD family of CRLs. The data suggest that CRLs play a role in HGSOC and that some of these proteins may be novel therapeutic targets. Differential expression of genes encoding deubiquitinases and proteasome subunits was also noted.
Collapse
Affiliation(s)
- Jerry Vriend
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- Correspondence: ; Tel.: +1-204-789-3732
| | - Mark W. Nachtigal
- Department of Biochemistry and Medical Genetics, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada;
- Department of Obstetrics, Gynecology & Reproductive Sciences, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada
| |
Collapse
|
23
|
Wang Z, Hou Y, Ren S, Liu Z, Zuo Z, Huang S, Wang W, Wang H, Chen Y, Xu Y, Yamamoto M, Zhang Q, Fu J, Pi J. CL316243 treatment mitigates the inflammation in white adipose tissues of juvenile adipocyte-specific Nfe2l1 knockout mice. Free Radic Biol Med 2021; 165:289-298. [PMID: 33545311 DOI: 10.1016/j.freeradbiomed.2021.01.043] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/15/2020] [Revised: 01/16/2021] [Accepted: 01/22/2021] [Indexed: 12/13/2022]
Abstract
Nuclear factor-erythroid 2-related factor 1 (NFE2L1) is a key transcription factor that regulates cellular adaptive responses to various stresses. Our previous studies revealed that adult adipocyte-specific Nfe2l1-knockout [Nfe2l1(f)-KO] mice show adipocyte hypertrophy and severe adipose inflammation, which can be worsened by rosiglitazone, a peroxisome proliferator-activated receptor γ agonist. To further assess the crucial roles of NFE2L1 in adipocytes, we investigated the effect of CL316243, a β3 adrenergic agonist that promotes lipolysis via a post-translational mechanism, on adipose inflammation in juvenile Nfe2l1(f)-KO mice. In contrast to adult mice, 4-week-old juvenile Nfe2l1(f)-KO mice displayed a normal fat distribution but reduced fasting plasma glycerol levels and elevated adipocyte hypertrophy and macrophage infiltration in inguinal and gonadal WAT. In addition, Nfe2l1(f)-KO mice had decreased expression of multiple lipolytic genes and reduced lipolytic activity in WAT. While 7 days of CL316243 treatment showed no significant effect on adipose inflammation in Nfe2l1-Floxed control mice, the same treatment dramatically alleviated macrophage infiltration and mRNA expression of inflammation and pyroptosis-related genes in WAT of Nfe2l1(f)-KO mice. Together with previous findings in adult mice, the current study highlights that NFE2L1 plays a fundamental regulatory role in lipolytic gene expression and thus might be an important target to improve adipose plasticity and lipid homeostasis.
Collapse
Affiliation(s)
- Zhendi Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Yongyong Hou
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Suping Ren
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Zhiyuan Liu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Zhuo Zuo
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Sicui Huang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Wanqi Wang
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Huihui Wang
- Group of Chronic Disease and Environmental Genomics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Yanyan Chen
- The First Affiliated Hospital, China Medical University, No. 155 Nanjing North Road, Heping Area, Shenyang, 110001, PR China
| | - Yuanyuan Xu
- Group of Chronic Disease and Environmental Genomics, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, No 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China
| | - Masayuki Yamamoto
- Department of Medical Biochemistry, Tohoku University Graduate School of Medicine, Sendai, 980-8575, Japan
| | - Qiang Zhang
- Department of Environmental Health, Rollins School of Public Health, Emory University, Atlanta, GA, 30322, USA
| | - Jingqi Fu
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China.
| | - Jingbo Pi
- Program of Environmental Toxicology, School of Public Health, China Medical University, No. 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China; Key Laboratory of Liaoning Province on Toxic and Biological Effects of Arsenic, China Medical University, No 77 Puhe Road, Shenyang North New Area, Shenyang, 110122, PR China.
| |
Collapse
|
24
|
Çetin G, Klafack S, Studencka-Turski M, Krüger E, Ebstein F. The Ubiquitin-Proteasome System in Immune Cells. Biomolecules 2021; 11:biom11010060. [PMID: 33466553 PMCID: PMC7824874 DOI: 10.3390/biom11010060] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/16/2020] [Accepted: 12/22/2020] [Indexed: 12/11/2022] Open
Abstract
The ubiquitin–proteasome system (UPS) is the major intracellular and non-lysosomal protein degradation system. Thanks to its unique capacity of eliminating old, damaged, misfolded, and/or regulatory proteins in a highly specific manner, the UPS is virtually involved in almost all aspects of eukaryotic life. The critical importance of the UPS is particularly visible in immune cells which undergo a rapid and profound functional remodelling upon pathogen recognition. Innate and/or adaptive immune activation is indeed characterized by a number of substantial changes impacting various cellular processes including protein homeostasis, signal transduction, cell proliferation, and antigen processing which are all tightly regulated by the UPS. In this review, we summarize and discuss recent progress in our understanding of the molecular mechanisms by which the UPS contributes to the generation of an adequate immune response. In this regard, we also discuss the consequences of UPS dysfunction and its role in the pathogenesis of recently described immune disorders including cancer and auto-inflammatory diseases.
Collapse
|
25
|
Kobayashi A. Roles of NRF3 in the Hallmarks of Cancer: Proteasomal Inactivation of Tumor Suppressors. Cancers (Basel) 2020; 12:cancers12092681. [PMID: 32962187 PMCID: PMC7563620 DOI: 10.3390/cancers12092681] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 09/15/2020] [Accepted: 09/17/2020] [Indexed: 01/18/2023] Open
Abstract
Simple Summary This review summarizes recent advances in our understanding of the physiological roles of the NFE2-related factor 2 (NRF2)-related transcription factor NRF3 in cancer. NRF3 confers cells with six so-called “hallmarks of cancer” through upregulating gene expression of specific target genes, leading to tumorigenesis and cancer malignancy. These driver gene-like functions of NRF3 in cancer are distinct from those of NRF2. Abstract The physiological roles of the NRF2-related transcription factor NRF3 (NFE2L3) have remained unknown for decades. The remarkable development of human cancer genome databases has led to strong suggestions that NRF3 has functional significance in cancer; specifically, high NRF3 mRNA levels are induced in many cancer types, such as colorectal cancer and pancreatic adenocarcinoma, and are associated with poor prognosis. On the basis of this information, the involvement of NRF3 in tumorigenesis and cancer malignancy has been recently proposed. NRF3 confers cancer cells with selective growth advantages by enhancing 20S proteasome assembly through induction of the chaperone gene proteasome maturation protein (POMP) and consequently promoting degradation of the tumor suppressors p53 and retinoblastoma (Rb) in a ubiquitin-independent manner. This new finding offers insight into the proteasomal but not the genetic inactivation mechanism of tumor suppressors. Moreover, NRF3 promotes cancer malignancy-related processes, including metastasis and angiogenesis. Finally, the molecular mechanisms underlying NRF3 activation have been elucidated, and this knowledge is expected to provide many insights that are useful for the development of anticancer drugs that attenuate NRF3 transcriptional activity. Collectively, the evidence indicates that NRF3 confers cells with six so-called “hallmarks of cancer”, implying that it exhibits cancer driver gene-like function. This review describes recent research advances regarding the newly discovered addiction of cancer cells to NRF3 compared to NRF2.
Collapse
Affiliation(s)
- Akira Kobayashi
- Laboratory for Genetic Code, Graduate School of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan; ; Tel.: +81-774-65-6273
- Department of Medical Life Systems, Faculty of Life and Medical Sciences, Doshisha University, Kyotanabe, Kyoto 610-0394, Japan
| |
Collapse
|