1
|
Wu Y, Zhu BT. Role of protein disulfide isomerase in mediating sulfasalazine-induced ferroptosis in HT22 cells: The PDI-NOS-NO-ROS/lipid-ROS cascade. Arch Biochem Biophys 2025; 768:110366. [PMID: 40023379 DOI: 10.1016/j.abb.2025.110366] [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: 11/03/2024] [Revised: 02/24/2025] [Accepted: 02/26/2025] [Indexed: 03/04/2025]
Abstract
Ferroptosis is a form of regulated cell death resulting from excessive lipid peroxidation. Sulfasalazine (SAS), an anti-inflammatory drug, can induce ferroptosis through inhibiting the system Xc- and triggering glutathione depletion. SAS has attracted considerable interest in recent years because of its potential for repurposing as an anticancer agent. Our recent studies have shown that protein disulfide isomerase (PDI) is an upstream mediator of chemically-induced ferroptosis through catalyzing the dimerization of nitric oxide synthase (NOS) and NO accumulation in cultured HT22 hippocampal neuronal cells. The present study aims to investigate SAS-induced ferroptotic cell death in HT22 cells with a focus on determining the role of PDI in mediating SAS-induced ferroptosis. We find that SAS induces ferroptotic cell death in HT22 cells, which is accompanied by a time-dependent sequential increase in the accumulation of cellular NO, ROS and lipid-ROS. We find that treatment of HT22 cells with SAS activates PDI-mediated iNOS activation (dimerization) and NO accumulation. In addition, SAS also strongly upregulates iNOS protein levels in HT22 cells. PDI knockdown or pharmacological inhibition of PDI's activity each suppresses SAS-induced iNOS dimerization, which is associated with abrogation of SAS-induced accumulation of NO, ROS and lipid-ROS, and a strong protection against ferroptotic cell death. On the other hand, PDI activation through the use of a TrxR1 inhibitor can strongly sensitize cells to SAS-induced ferroptosis. Together, these experimental observations demonstrate a crucial role of PDI in SAS-induced ferroptosis in a cell culture model through the activation of the PDI → NOS → NO → ROS/lipid-ROS pathway. Insights gained from this study also provide effective strategies to selectively sensitizing human cancer cells to SAS-induced ferroptosis, such as through the use of NO-releasing agents or TrxR1 inhibitors.
Collapse
Affiliation(s)
- Yufei Wu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 518172, China; Shenzhen Bay Laboratory, Shenzhen, 518172, China.
| |
Collapse
|
2
|
Liu Y, Sun Y, Chen A, Chen J, Zhu T, Wang S, Qiao W, Zhou D, Zhang X, Chen S, Shi Y, Yang Y, Wang J, Wu L, Fan L. Involvement of disulfidptosis in the pathophysiology of autism spectrum disorder. Life Sci 2025; 369:123531. [PMID: 40054734 DOI: 10.1016/j.lfs.2025.123531] [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: 12/28/2024] [Revised: 02/22/2025] [Accepted: 03/03/2025] [Indexed: 03/30/2025]
Abstract
Autism Spectrum Disorder (ASD) is a complex neurodevelopmental disorder, with oxidative stress recognized as a key pathogenic mechanisms. Oxidative stress disrupts intracellular dynamic- thiol/disulfide homeostasis (DTDH), potentially leading to disulfidptosis, a newly identified cell death mechanism. While studies suggest a link between DTDH and ASD, direct evidence implicating disulfidptosis in ASD pathogenesis remains limited. In this study, Mendelian randomization analysis revealed a significant causal association between disulfidptosis-related sulfhydryl oxidase 1 and 2 and ASD (OR1 = 0.883, OR2 = 0.924, p < 0.05). A positive correlation between protein disulfide-isomerase and cognitive performance (OR = 1.021, p < 0.01) further supported the role of disulfidptosis in ASD. Seven disulfidptosis-related genes (TIMP1, STAT3, VWA1, ADA, IL5, PF4, and TXNDC12) were identified and linked to immune cell alterations. A TF-miRNA-mRNA regulatory network and a predictive model (AUC = 0.759) were constructed and external validation datasets (AUC = 0.811). Immune infiltration analysis demonstrated altered expression of naive B cells and three other types of immune cells in ASD children. Animal experiments further validated the differential expression of key genes, highlighting their relevance to ASD pathogenesis. Animal experiments found that BTBR mice exhibit glucose starvation and NADPH depletion, with the specific indicator Slc7a11 being highly expressed. Silencing Slc7a11 can improve core ASD impairments in BTBR mice. CONCLUSION: This study establishes the first mechanistic link between disulfidptosis and ASD, identifies seven key genes and their regulatory network, and develops a predictive model with clinical utility. Animal experiments further confirmed the strong association between disulfidpotosis and ASD phenotypes. These findings offer novel therapeutic targets for modulating oxidative stress in ASD.
Collapse
Affiliation(s)
- Yutong Liu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yaqi Sun
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Anjie Chen
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Jiaqi Chen
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Tikang Zhu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Shuting Wang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Wanying Qiao
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Ding Zhou
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Xirui Zhang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Shuangshuang Chen
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yaxin Shi
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Yuan Yang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China
| | - Jia Wang
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China; Key Laboratory of Children development and genetic research, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin 150081, China
| | - Lijie Wu
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China; Key Laboratory of Children development and genetic research, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin 150081, China
| | - Lili Fan
- Department of Children's and Adolescent Health, Public Health College, Harbin Medical University, Harbin 150081, China; Key Laboratory of Children development and genetic research, Harbin Medical University, Harbin 150081, Heilongjiang Province, China; Key Laboratory of Precision nutrition and health, Ministry of Education, Harbin Medical University, Harbin 150081, China.
| |
Collapse
|
3
|
Lee R, Lee WY, Kim DW, Park HJ. Artemisinin alleviates cisplatin-induced damage in GC-1 spermatogonia through ER stress mechanisms. Heliyon 2025; 11:e42579. [PMID: 40034267 PMCID: PMC11874544 DOI: 10.1016/j.heliyon.2025.e42579] [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: 10/29/2024] [Revised: 02/05/2025] [Accepted: 02/07/2025] [Indexed: 03/05/2025] Open
Abstract
Artemisinin, a compound derived from Artemisia annua, is primarily utilized for malaria treatment. Its mechanism of action involves the rapid and effective inhibition of protein synthesis in malarial parasites. Recently, artemisinin has garnered extensive research attention for its anticancer, antioxidant, and anti-inflammatory properties, as well as its potential role as an adjuvant in cancer treatment. Cisplatin is a commonly used anticancer agent; however, its therapeutic benefits are accompanied by side effects that negatively impact male reproductive function. In this study, the mechanism of the protective effect of artemisinin against cisplatin-induced cytotoxicity was investigated. Type B mouse spermatogonia (GC-1 spg cells), derived from mouse testes, were treated with various concentrations of artemisinin (10-200 μM) to identify the optimal concentration for promoting cell proliferation. Cisplatin induced antiproliferative effects and apoptotic cell death in GC-1 spg cells, whereas the combination of cisplatin and artemisinin restored cell proliferation and reduced apoptosis. Treatment with cisplatin resulted in elevated levels of endoplasmic reticulum (ER) stress-related factors, such as Bip/GRP78, PDI, and Ero1-la, in GC-1 spg cells, while the combination with artemisinin effectively inhibited and reduced these levels. Additionally, cisplatin increased inflammatory markers, including COX2, iNOS, and NF-κB, which were subsequently decreased by artemisinin. This study evaluates artemisinin, a naturally derived compound, as a potential mitigator of side effects on male germ cells during cisplatin-based anticancer treatment. In conclusion, these findings suggest that artemisinin may serve as a supplement or functional agent in cancer therapy.
Collapse
Affiliation(s)
- Ran Lee
- Department of Livestock, Korea National University of Agriculture and Fisheries, Jeonbuk, 54874, Republic of Korea
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si, 26339, Republic of Korea
| | - Won-Yong Lee
- Department of Livestock, Korea National University of Agriculture and Fisheries, Jeonbuk, 54874, Republic of Korea
| | - Dong-Wook Kim
- Department of Livestock, Korea National University of Agriculture and Fisheries, Jeonbuk, 54874, Republic of Korea
| | - Hyun-Jung Park
- Department of Animal Biotechnology, College of Life Science, Sangji University, Wonju-si, 26339, Republic of Korea
| |
Collapse
|
4
|
Zhang S, Zhang M, Ma W, Wang L, Yu B. Knockdown of ERO1L attenuates tumor growth, migration and invasion in lung adenocarcinoma through Wnt/β‑catenin pathway. Biotechnol Genet Eng Rev 2024; 40:1910-1923. [PMID: 37014092 DOI: 10.1080/02648725.2023.2197325] [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: 02/21/2023] [Accepted: 03/24/2023] [Indexed: 04/05/2023]
Abstract
Recent studies confirm the critical roles of endoplasmic reticulum oxidoreductase 1 alpha (ERO1L) in malignant behavior of various cancers. Nevertheless, what function ERO1L plays in lung adenocarcinoma (LUAD) remains uncovered. The expressions and clinical significance of ERO1L in LUAD were investigated using the TCGA dataset. The ERO1L levels were examined by RT-qPCR. The LUAD cell proliferation was valued using colony formation as well as CCK-8 assays. The invasion and migration abilities of LUAD cells were detected through Transwell in addition to wound healing assays. The effects of ERO1L on LUAD cell apoptosis were determined by flow cytometric analysis. Moreover, we also established mouse xenograft models of LUAD cells to confirm the functions of ERO1L in vivo. The ERO1L levels in tumors were identified by immunohistochemistry. Western blot was used for the detection of the levels of Wnt/βcatenin signaling-related proteins. The TCGA database revealed that ERO1L expressions were higher in LUAD tissues than those in non-cancerous tissues. ERO1L overexpression was related to poorer overall survival of LUAD patients. In addition, ERO1L silence suppresses LUAD cell clone formation, proliferation, migration as well as invasion but induces apoptosis. Moreover, we also verified that ERO1L silence could promote LUAD growth in vivo. Based on the mechanism analysis, ERO1L was confirmed to regulate LUAD development via Wnt/βcatenin cascade signal. ERO1L, the expression of which was increased in LUAD tissues, functioned as an oncogene. ERO1L silence significantly attenuated LUAD tumorigenesis, likely via inhibition of Wnt/βcatenin signaling, indicating that ERO1L could be exploited as a promising biomarker in LUAD treatment.
Collapse
Affiliation(s)
- Shupeng Zhang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Meng Zhang
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Weichang Ma
- Department of Thyroid and Breast Surgery, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Linan Wang
- Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| | - Bing Yu
- Department of Respiratory Medicine, Yantai Affiliated Hospital of Binzhou Medical University, Yantai, Shandong, China
| |
Collapse
|
5
|
Huang X, Hou MJ, Zhu BT. Protection of HT22 neuronal cells against chemically-induced ferroptosis by catechol estrogens: protein disulfide isomerase as a mechanistic target. Sci Rep 2024; 14:23988. [PMID: 39402104 PMCID: PMC11473836 DOI: 10.1038/s41598-024-74742-5] [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: 05/15/2024] [Accepted: 09/30/2024] [Indexed: 10/17/2024] Open
Abstract
Ferroptosis is a form of regulated cell death, characterized by excessive iron-dependent lipid peroxidation. Biochemically, ferroptosis can be selectively induced by erastin through glutathione depletion or through inhibition of glutathione peroxidase 4 by RSL3, which leads to accumulation of cytotoxic lipid reactive oxygen species (ROS). Protein disulfide isomerase (PDI) was recently shown to mediate erastin/RSL3-induced ferroptosis and thus also become a new target for protection against chemically-induced ferroptosis. The present study aims to identify endogenous compounds that can protect against erastin/RSL3-induced ferroptotic cell death. We find that 2-hydroxyestrone, 2-hydroxyestradiol, 4-hydroxyestrone and 4-hydroxyestradiol, four major endogenous catechol estrogens, are effective inhibitors of PDI, and can strongly protect against chemically-induced ferroptotic cell death in cultured HT22 mouse hippocampal neuronal cells. The CETSA assay showed that these catechol estrogens can bind to PDI in live cells. PDI knockdown attenuates the protective effect of these catechol estrogens against chemically-induced ferroptosis. Mechanistically, inhibition of PDI's catalytic activity by catechol estrogens abrogates erastin/RSL3-induced dimerization of nitric oxide synthase, thereby preventing the subsequent accumulation of cellular nitric oxide, ROS and lipid-ROS, and ultimately ferroptotic cell death. In addition, joint treatment of cells with catechol estrogens also abrogates erastin/RSL3-induced upregulation of nitric oxide synthase protein levels, which also contributes to the cytoprotective effect of the catechol estrogens. In conclusion, the present study demonstrates that the catechol estrogens are protectors of HT22 neuronal cells against chemically-induced ferroptosis, and inhibition of PDI's catalytic activity by these estrogens contributes to a novel, estrogen receptor-independent mechanism of cytoprotection.
Collapse
Affiliation(s)
- Xuanqi Huang
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Ming-Jie Hou
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China
| | - Bao Ting Zhu
- Shenzhen Key Laboratory of Steroid Drug Discovery and Development, School of Medicine, The Chinese University of Hong Kong, Shenzhen, 2001 Longxiang Road, Longgang District, Shenzhen, 518172, China.
- Shenzhen Bay Laboratory, Shenzhen, 518055, China.
| |
Collapse
|
6
|
Yeewa R, Sangphukieo A, Jantaree P, Wongkummool W, Yamsri T, Poompouang S, Chaiyawat P, Lo Piccolo L, Jantrapirom S. ERO1A inhibition mitigates neuronal ER stress and ameliorates UBQLN2 ALS phenotypes in Drosophila melanogaster. Prog Neurobiol 2024; 242:102674. [PMID: 39395630 DOI: 10.1016/j.pneurobio.2024.102674] [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: 05/05/2024] [Revised: 09/25/2024] [Accepted: 10/06/2024] [Indexed: 10/14/2024]
Abstract
Modulating the ER stress pathway holds therapeutic promise for neurodegenerative diseases; however, identifying optimal targets remains challenging. In this study, we conducted an unbiased screening to systematically search for commonly up-regulated proteins in ER stress-related neurodegenerative conditions, with endoplasmic reticulum oxidoreductase 1 alpha (ERO1A) emerging as a significant hit. Further experiments conducted in the model organism Drosophila melanogaster demonstrated that elevated levels of Drosophila ERO1A (ERO1L) were indeed detrimental to neurons. Conversely, genetic suppression or pharmacological inhibition of ERO1L activity provided neuroprotection under ER stress and extended the lifespan of flies. To translate these findings, we performed a genetic modifier screening and underscored significant neuroprotective effects against UBQLN2ALS pathology. Additionally, administration of the chemical probe inhibitor of ERO1A, known as EN460, enhanced locomotive functions and neuromuscular junction (NMJ) morphology in Drosophila UBQLN2ALS model. Mechanistically, targeting ERO1L during environmental or pathological ER stress mitigated proteotoxic stress by lowering either the PERK or IRE1 branches of the unfolded protein response (UPR). These findings suggest ERO1A as a promising therapeutic target in UBQLN2ALS and other ER stress-related conditions.
Collapse
Affiliation(s)
- Ranchana Yeewa
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Apiwat Sangphukieo
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Phatcharida Jantaree
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Wasinee Wongkummool
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Titaree Yamsri
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Siwat Poompouang
- Drosophila Centre for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Parunya Chaiyawat
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand
| | - Luca Lo Piccolo
- Center of Multidisciplinary Technology for Advanced Medicine (CMUTEAM), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| | - Salinee Jantrapirom
- Drosophila Centre for Human Diseases and Drug Discovery (DHD), Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand; Department of Pharmacology, Faculty of Medicine, Chiang Mai University, Chiang Mai, Thailand.
| |
Collapse
|
7
|
Zhang C, Fu Y, Zheng W, Chang F, Shen Y, Niu J, Wang Y, Ma X. Enhancing the Antibody Production Efficiency of Chinese Hamster Ovary Cells through Improvement of Disulfide Bond Folding Ability and Apoptosis Resistance. Cells 2024; 13:1481. [PMID: 39273052 PMCID: PMC11394227 DOI: 10.3390/cells13171481] [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: 07/02/2024] [Revised: 08/30/2024] [Accepted: 09/02/2024] [Indexed: 09/15/2024] Open
Abstract
The complex structure of monoclonal antibodies (mAbs) expressed in Chinese hamster ovary (CHO) cells may result in the accumulation of unfolded proteins, triggering endoplasmic reticulum (ER) stress and an unfolded protein response (UPR). If the protein folding ability cannot maintain ER homeostasis, the cell will shut down protein translation and ultimately induce apoptosis. We co-overexpressed HsQSOX1b and survivin proteins in the antibody-producing cell line CHO-PAb to obtain a new cell line, CHO-PAb-QS. Compared with CHO-PAb cells, the survival time of CHO-PAb-QS cells in batch culture was extended by 2 days, and the antibody accumulation and productivity were increased by 52% and 45%, respectively. The proportion of (HC-LC)2 was approximately doubled in the CHO-PAb-QS cells, which adapted to the accelerated disulfide bond folding capacity by upregulating the UPR's strength and increasing the ER content. The results of the apoptosis assays indicated that the CHO-PAb-QS cell line exhibited more excellent resistance to apoptosis induced by ER stress. Finally, CHO-PAb-QS cells exhibited mild oxidative stress but did not significantly alter the redox status. This study demonstrated that strategies based on HsQSOX1b and survivin co-overexpression could facilitate protein disulfide bond folding and anti-apoptosis ability, enhancing antibody production efficiency in CHO cell lines.
Collapse
Affiliation(s)
- Chen Zhang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Yunhui Fu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Wenyun Zheng
- Shanghai Key Laboratory of New Drug Design, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China
| | - Feng Chang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Yue Shen
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Jinping Niu
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Yangmin Wang
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| | - Xingyuan Ma
- State Key Laboratory of Bioreactor Engineering, School of Biotechnology, East China University of Science and Technology, Shanghai 200237, China; (C.Z.)
| |
Collapse
|
8
|
Casas-Martinez JC, Samali A, McDonagh B. Redox regulation of UPR signalling and mitochondrial ER contact sites. Cell Mol Life Sci 2024; 81:250. [PMID: 38847861 PMCID: PMC11335286 DOI: 10.1007/s00018-024-05286-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 04/11/2024] [Accepted: 05/18/2024] [Indexed: 06/13/2024]
Abstract
Mitochondria and the endoplasmic reticulum (ER) have a synergistic relationship and are key regulatory hubs in maintaining cell homeostasis. Communication between these organelles is mediated by mitochondria ER contact sites (MERCS), allowing the exchange of material and information, modulating calcium homeostasis, redox signalling, lipid transfer and the regulation of mitochondrial dynamics. MERCS are dynamic structures that allow cells to respond to changes in the intracellular environment under normal homeostatic conditions, while their assembly/disassembly are affected by pathophysiological conditions such as ageing and disease. Disruption of protein folding in the ER lumen can activate the Unfolded Protein Response (UPR), promoting the remodelling of ER membranes and MERCS formation. The UPR stress receptor kinases PERK and IRE1, are located at or close to MERCS. UPR signalling can be adaptive or maladaptive, depending on whether the disruption in protein folding or ER stress is transient or sustained. Adaptive UPR signalling via MERCS can increase mitochondrial calcium import, metabolism and dynamics, while maladaptive UPR signalling can result in excessive calcium import and activation of apoptotic pathways. Targeting UPR signalling and the assembly of MERCS is an attractive therapeutic approach for a range of age-related conditions such as neurodegeneration and sarcopenia. This review highlights the emerging evidence related to the role of redox mediated UPR activation in orchestrating inter-organelle communication between the ER and mitochondria, and ultimately the determination of cell function and fate.
Collapse
Affiliation(s)
- Jose C Casas-Martinez
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland
- Apoptosis Research Centre, University of Galway, Galway, Ireland
| | - Afshin Samali
- Apoptosis Research Centre, University of Galway, Galway, Ireland
- School of Biological and Chemical Sciences, University of Galway, Galway, Ireland
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, University of Galway, Galway, Ireland.
- Apoptosis Research Centre, University of Galway, Galway, Ireland.
| |
Collapse
|
9
|
Zhao LX, Zou SP, Shen Q, Xue YP, Zheng YG. Enhancing the expression of the unspecific peroxygenase in Komagataella phaffii through a combination strategy. Appl Microbiol Biotechnol 2024; 108:320. [PMID: 38709366 DOI: 10.1007/s00253-024-13166-7] [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: 02/17/2024] [Revised: 04/19/2024] [Accepted: 05/01/2024] [Indexed: 05/07/2024]
Abstract
The unspecific peroxygenase (UPO) from Cyclocybe aegerita (AaeUPO) can selectively oxidize C-H bonds using hydrogen peroxide as an oxygen donor without cofactors, which has drawn significant industrial attention. Many studies have made efforts to enhance the overall activity of AaeUPO expressed in Komagataella phaffii by employing strategies such as enzyme-directed evolution, utilizing appropriate promoters, and screening secretion peptides. Building upon these previous studies, the objective of this study was to further enhance the expression of a mutant of AaeUPO with improved activity (PaDa-I) by increasing the gene copy number, co-expressing chaperones, and optimizing culture conditions. Our results demonstrated that a strain carrying approximately three copies of expression cassettes and co-expressing the protein disulfide isomerase showed an approximately 10.7-fold increase in volumetric enzyme activity, using the 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) as the substrate. After optimizing the culture conditions, the volumetric enzyme activity of this strain further increased by approximately 48.7%, reaching 117.3 U/mL. Additionally, the purified catalytic domain of PaDa-I displayed regioselective hydroxylation of R-2-phenoxypropionic acid. The results of this study may facilitate the industrial application of UPOs. KEY POINTS: • The secretion of the catalytic domain of PaDa-I can be significantly enhanced through increasing gene copy numbers and co-expressing of protein disulfide isomerase. • After optimizing the culture conditions, the volumetric enzyme activity can reach 117.3 U/mL, using the 2,2'-azino-bis(3-ethylbenzothiazoline-6-sulfonic acid) as the substrate. • The R-2-phenoxypropionic acid can undergo the specific hydroxylation reaction catalyzed by catalytic domain of PaDa-I, resulting in the formation of R-2-(4-hydroxyphenoxy)propionic acid.
Collapse
Affiliation(s)
- Li-Xiang Zhao
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Shu-Ping Zou
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Qi Shen
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| | - Ya-Ping Xue
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China.
| | - Yu-Guo Zheng
- Key Laboratory of Bioorganic Synthesis of Zhejiang Province, College of Biotechnology and Bioengineering, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
- Engineering Research Center of Bioconversion and Biopurification of Ministry of Education, Zhejiang University of Technology, Hangzhou, 310014, People's Republic of China
| |
Collapse
|
10
|
Sanyasi C, Balakrishnan SS, Chinnasamy T, Venugopalan N, Kandavelu P, Batra-Safferling R, Muthuvel SK. Insights on the dynamic behavior of protein disulfide isomerase in the solution environment through the SAXS technique. In Silico Pharmacol 2024; 12:23. [PMID: 38584776 PMCID: PMC10997565 DOI: 10.1007/s40203-024-00198-0] [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: 10/25/2023] [Accepted: 02/17/2024] [Indexed: 04/09/2024] Open
Abstract
The dynamic behavior of Protein Disulfide Isomerase (PDI) in an aqueous solution environment under physiologically active pH has been experimentally verified in this study using Small Angle X-ray Scattering (SAXS) technique. The structural mechanism of dimerization for full-length PDI molecules and co-complex with two renowned substrates has been comprehensively discussed. The structure models obtained from the SAXS data of PDI purified from bovine liver display behavior duality between unaccompanied-enzyme and after engaged with substrates. The analysis of SAXS data revealed that PDI exists as a homo-dimer in the solution environment, and substrate induction provoked its segregation into monomer to enable the enzyme to interact systematically with incoming clients. Supplementary Information The online version contains supplementary material available at 10.1007/s40203-024-00198-0.
Collapse
Affiliation(s)
- Chandrasekar Sanyasi
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, 605014 India
| | - Susmida Seni Balakrishnan
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, 605014 India
| | - Thirunavukkarasu Chinnasamy
- Department of Biochemistry and Molecular Biology, School of Life Sciences, Pondicherry University, Pondicherry, 605014 India
| | - Nagarajan Venugopalan
- GMCA Structural Biology Facility, X-Ray Science Division, Argonne National Laboratory, Argonne, IL USA
| | - Palani Kandavelu
- SER-CAT and The Department of Biochemistry and Molecular Biology, University of Georgia, Athens, GA 30601 USA
| | - Renu Batra-Safferling
- Institute of Complex Systems (ICS-6: Structural Biochemistry), Forschungszentrum Jülich, 52425 Jülich, Germany
| | - Suresh Kumar Muthuvel
- Department of Bioinformatics, School of Life Sciences, Pondicherry University, Pondicherry, 605014 India
| |
Collapse
|
11
|
Gaspar RS, Laurindo FRM. Sulfenylation: an emerging element of the protein disulfide isomerase code for thrombosis. J Thromb Haemost 2023; 21:2054-2057. [PMID: 37468176 DOI: 10.1016/j.jtha.2023.04.020] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2023] [Accepted: 04/19/2023] [Indexed: 07/21/2023]
Affiliation(s)
- Renato Simões Gaspar
- Laboratorio de Biologia Vascular, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco Rafael Martins Laurindo
- Laboratorio de Biologia Vascular, Instituto do Coracao, Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil.
| |
Collapse
|
12
|
Holendova B, Plecita-Hlavata L. Cysteine residues in signal transduction and its relevance in pancreatic beta cells. Front Endocrinol (Lausanne) 2023; 14:1221520. [PMID: 37455926 PMCID: PMC10339824 DOI: 10.3389/fendo.2023.1221520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Accepted: 06/13/2023] [Indexed: 07/18/2023] Open
Abstract
Cysteine is one of the least abundant but most conserved amino acid residues in proteins, playing a role in their structure, metal binding, catalysis, and redox chemistry. Thiols present in cysteines can be modified by post-translational modifications like sulfenylation, acylation, or glutathionylation, regulating protein activity and function and serving as signals. Their modification depends on their position in the structure, surrounding amino acids, solvent accessibility, pH, etc. The most studied modifications are the redox modifications by reactive oxygen, nitrogen, and sulfur species, leading to reversible changes that serve as cell signals or irreversible changes indicating oxidative stress and cell damage. Selected antioxidants undergoing reversible oxidative modifications like peroxiredoxin-thioredoxin system are involved in a redox-relay signaling that can propagate to target proteins. Cysteine thiols can also be modified by acyl moieties' addition (derived from lipid metabolism), resulting in protein functional modification or changes in protein anchoring in the membrane. In this review, we update the current knowledge on cysteine modifications and their consequences in pancreatic β-cells. Because β-cells exhibit well-balanced redox homeostasis, the redox modifications of cysteines here serve primarily for signaling purposes. Similarly, lipid metabolism provides regulatory intermediates that have been shown to be necessary in addition to redox modifications for proper β-cell function and, in particular, for efficient insulin secretion. On the contrary, the excess of reactive oxygen, nitrogen, and sulfur species and the imbalance of lipids under pathological conditions cause irreversible changes and contribute to oxidative stress leading to cell failure and the development of type 2 diabetes.
Collapse
Affiliation(s)
| | - Lydie Plecita-Hlavata
- Laboratory of Pancreatic Islet Research, Institute of Physiology, Czech Academy of Sciences, Prague, Czechia
| |
Collapse
|
13
|
Chetot T, Serfaty X, Carret L, Kriznik A, Sophie-Rahuel-Clermont, Grand L, Jacolot M, Popowycz F, Benoit E, Lambert V, Lattard V. Splice variants of protein disulfide isomerase - identification, distribution and functional characterization in the rat. Biochim Biophys Acta Gen Subj 2023; 1867:130280. [PMID: 36423740 DOI: 10.1016/j.bbagen.2022.130280] [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: 03/13/2022] [Revised: 10/28/2022] [Accepted: 11/13/2022] [Indexed: 11/23/2022]
Abstract
BACKGROUND Protein Disulfide Isomerase (PDI) enzyme is an emerging therapeutic target in oncology and hematology. Although PDI reductase activity has been studied with isolated fragments of the protein, natural structural variations affecting reductase activity have not been addressed. METHODS In this study, we discovered four coding splice variants of the Pdi pre-mRNA in rats. In vitro Michaelis constants and apparent maximum steady-state rate constants after purification and distribution in different rat tissues were determined. RESULTS The consensus sequence was found to be the most expressed splice variant while the second most expressed variant represents 15 to 35% of total Pdi mRNA. The third variant shows a quasi-null expression profile and the fourth was not quantifiable. The consensus sequence splice variant and the second splice variant are widely expressed (transcription level) in the liver and even more present in males. Measurements of the reductase activity of recombinant PDI indicate that the consensus sequence and third splice variant are fully active variants. The second most expressed variant, differing by a lack of signal peptide, was found active but less than the consensus sequence. GENERAL SIGNIFICANCE Our work emphasizes the importance of taking splice variants into account when studying PDI-like proteins to understand the full biological functionalities of PDI.
Collapse
Affiliation(s)
- Thomas Chetot
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France
| | - Xavier Serfaty
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France
| | - Léna Carret
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France
| | | | | | - Lucie Grand
- Univ Lyon, INSA Lyon, Université Lyon 1, CNRS, CPE Lyon, UMR 5246, ICBMS, 69621 Villeurbanne Cedex, France
| | - Maïwenn Jacolot
- Univ Lyon, INSA Lyon, Université Lyon 1, CNRS, CPE Lyon, UMR 5246, ICBMS, 69621 Villeurbanne Cedex, France
| | - Florence Popowycz
- Univ Lyon, INSA Lyon, Université Lyon 1, CNRS, CPE Lyon, UMR 5246, ICBMS, 69621 Villeurbanne Cedex, France
| | - Etienne Benoit
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France
| | - Véronique Lambert
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France
| | - Virginie Lattard
- USC 1233 RS2GP, VetAgro Sup, INRAe, Université de Lyon, 69280 Marcy l'étoile, France.
| |
Collapse
|
14
|
Mishra S, Raval M, Kachhawaha AS, Tiwari BS, Tiwari AK. Aging: Epigenetic modifications. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2023; 197:171-209. [PMID: 37019592 DOI: 10.1016/bs.pmbts.2023.02.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/15/2023]
Abstract
Aging is one of the most complex and irreversible health conditions characterized by continuous decline in physical/mental activities that eventually poses an increased risk of several diseases and ultimately death. These conditions cannot be ignored by anyone but there are evidences that suggest that exercise, healthy diet and good routines may delay the Aging process significantly. Several studies have demonstrated that Epigenetics plays a key role in Aging and Aging-associated diseases through methylation of DNA, histone modification and non-coding RNA (ncRNA). Comprehension and relevant alterations in these epigenetic modifications can lead to new therapeutic avenues of age-delaying contrivances. These processes affect gene transcription, DNA replication and DNA repair, comprehending epigenetics as a key factor in understanding Aging and developing new avenues for delaying Aging, clinical advancements in ameliorating aging-related diseases and rejuvenating health. In the present article, we have described and advocated the epigenetic role in Aging and associated diseases.
Collapse
|
15
|
Tripathi R, Gupta R, Sahu M, Srivastava D, Das A, Ambasta RK, Kumar P. Free radical biology in neurological manifestations: mechanisms to therapeutics interventions. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2022; 29:62160-62207. [PMID: 34617231 DOI: 10.1007/s11356-021-16693-2] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Accepted: 09/20/2021] [Indexed: 06/13/2023]
Abstract
Recent advancements and growing attention about free radicals (ROS) and redox signaling enable the scientific fraternity to consider their involvement in the pathophysiology of inflammatory diseases, metabolic disorders, and neurological defects. Free radicals increase the concentration of reactive oxygen and nitrogen species in the biological system through different endogenous sources and thus increased the overall oxidative stress. An increase in oxidative stress causes cell death through different signaling mechanisms such as mitochondrial impairment, cell-cycle arrest, DNA damage response, inflammation, negative regulation of protein, and lipid peroxidation. Thus, an appropriate balance between free radicals and antioxidants becomes crucial to maintain physiological function. Since the 1brain requires high oxygen for its functioning, it is highly vulnerable to free radical generation and enhanced ROS in the brain adversely affects axonal regeneration and synaptic plasticity, which results in neuronal cell death. In addition, increased ROS in the brain alters various signaling pathways such as apoptosis, autophagy, inflammation and microglial activation, DNA damage response, and cell-cycle arrest, leading to memory and learning defects. Mounting evidence suggests the potential involvement of micro-RNAs, circular-RNAs, natural and dietary compounds, synthetic inhibitors, and heat-shock proteins as therapeutic agents to combat neurological diseases. Herein, we explain the mechanism of free radical generation and its role in mitochondrial, protein, and lipid peroxidation biology. Further, we discuss the negative role of free radicals in synaptic plasticity and axonal regeneration through the modulation of various signaling molecules and also in the involvement of free radicals in various neurological diseases and their potential therapeutic approaches. The primary cause of free radical generation is drug overdosing, industrial air pollution, toxic heavy metals, ionizing radiation, smoking, alcohol, pesticides, and ultraviolet radiation. Excessive generation of free radicals inside the cell R1Q1 increases reactive oxygen and nitrogen species, which causes oxidative damage. An increase in oxidative damage alters different cellular pathways and processes such as mitochondrial impairment, DNA damage response, cell cycle arrest, and inflammatory response, leading to pathogenesis and progression of neurodegenerative disease other neurological defects.
Collapse
Affiliation(s)
- Rahul Tripathi
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rohan Gupta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Mehar Sahu
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Devesh Srivastava
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Ankita Das
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Rashmi K Ambasta
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India
| | - Pravir Kumar
- Molecular Neuroscience and Functional Genomics Laboratory, Delhi Technological University (Formerly Delhi College of Engineering), Delhi, India.
- , Delhi, India.
- Molecular Neuroscience and Functional Genomics Laboratory, Shahbad Daulatpur, Bawana Road, Delhi, 110042, India.
| |
Collapse
|
16
|
Sorrentino I, Galli M, Medraño-Fernandez I, Sitia R. Transfer of H 2O 2 from Mitochondria to the endoplasmic reticulum via Aquaporin-11. Redox Biol 2022; 55:102410. [PMID: 35863264 PMCID: PMC9304643 DOI: 10.1016/j.redox.2022.102410] [Citation(s) in RCA: 22] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2022] [Revised: 07/12/2022] [Accepted: 07/12/2022] [Indexed: 01/20/2023] Open
Abstract
Some aquaporins (AQPs) can transport H2O2 across membranes, allowing redox signals to proceed in and between cells. Unlike other peroxiporins, human AQP11 is an endoplasmic reticulum (ER)-resident that can conduit H2O2 to the cytosol. Here, we show that silencing Ero1α, an ER flavoenzyme that generates abundant H2O2 during oxidative folding, causes a paradoxical increase in luminal H2O2 levels. The simultaneous AQP11 downregulation prevents this increase, implying that H2O2 reaches the ER from an external source(s). Pharmacological inhibition of the electron transport chain reveals that Ero1α downregulation activates superoxide production by complex III. In the intermembrane space, superoxide dismutase 1 generates H2O2 that enters the ER channeled by AQP11. Meanwhile, the number of ER-mitochondria contact sites increases as well, irrespective of AQP11 expression. Taken together, our findings identify a novel interorganellar redox response that is activated upon Ero1α downregulation and transfers H2O2 from mitochondria to the ER via AQP11.
Collapse
Affiliation(s)
- Ilaria Sorrentino
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Mauro Galli
- Department of Medical Biology, Medical University of Białystok, 15222, Białystok, Poland
| | - Iria Medraño-Fernandez
- Department of Bioengineering and Aerospace Engineering, University Carlos III of Madrid, 28911, Madrid, Spain.
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS), Ospedale San Raffaele, Università Vita-Salute San Raffaele, 20132, Milan, Italy.
| |
Collapse
|
17
|
Fan F, Zhang Q, Zhang Y, Huang G, Liang X, Wang CC, Wang L, Lu D. Two protein disulfide isomerase subgroups work synergistically in catalyzing oxidative protein folding. PLANT PHYSIOLOGY 2022; 188:241-254. [PMID: 34609517 PMCID: PMC8774737 DOI: 10.1093/plphys/kiab457] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 08/31/2021] [Indexed: 05/13/2023]
Abstract
Disulfide bonds play essential roles in the folding of secretory and plasma membrane proteins in the endoplasmic reticulum (ER). In eukaryotes, protein disulfide isomerase (PDI) is an enzyme catalyzing the disulfide bond formation and isomerization in substrates. The Arabidopsis (Arabidopsis thaliana) genome encodes diverse PDIs including structurally distinct subgroups PDI-L and PDI-M/S. It remains unclear how these AtPDIs function to catalyze the correct disulfide formation. We found that one Arabidopsis ER oxidoreductin-1 (Ero1), AtERO1, can interact with multiple PDIs. PDI-L members AtPDI2/5/6 mainly serve as an isomerase, while PDI-M/S members AtPDI9/10/11 are more efficient in accepting oxidizing equivalents from AtERO1 and catalyzing disulfide bond formation. Accordingly, the pdi9/10/11 triple mutant exhibited much stronger inhibition than pdi1/2/5/6 quadruple mutant under dithiothreitol treatment, which caused disruption of disulfide bonds in plant proteins. Furthermore, AtPDI2/5 work synergistically with PDI-M/S members in relaying disulfide bonds from AtERO1 to substrates. Our findings reveal the distinct but overlapping roles played by two structurally different AtPDI subgroups in oxidative protein folding in the ER.
Collapse
Affiliation(s)
- Fenggui Fan
- State Key Laboratory of Plant Genomics, Center for Agricultural Resources Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Shijiazhuang 050021, China
- Key Laboratory of Resource Biology and Biotechnology in Western China, Ministry of Education & College of Life Sciences, Northwest University, Xi'an, 710069, China
| | - Qiao Zhang
- Hebei Collaboration Innovation Center for Cell Signaling, Hebei Normal University, Shijiazhuang 050024, China
| | - Yini Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Guozhong Huang
- State Key Laboratory of Plant Genomics, Center for Agricultural Resources Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Shijiazhuang 050021, China
| | - Xuelian Liang
- Hebei Collaboration Innovation Center for Cell Signaling, Hebei Normal University, Shijiazhuang 050024, China
| | - Chih-chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| | - Dongping Lu
- State Key Laboratory of Plant Genomics, Center for Agricultural Resources Research, Institute of Genetics and Developmental Biology, Chinese Academy of Sciences, Shijiazhuang 050021, China
- Hebei Collaboration Innovation Center for Cell Signaling, Hebei Normal University, Shijiazhuang 050024, China
- Author for communication:
| |
Collapse
|
18
|
Lennicke C, Cochemé HM. Redox metabolism: ROS as specific molecular regulators of cell signaling and function. Mol Cell 2021; 81:3691-3707. [PMID: 34547234 DOI: 10.1016/j.molcel.2021.08.018] [Citation(s) in RCA: 526] [Impact Index Per Article: 131.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/02/2021] [Accepted: 08/12/2021] [Indexed: 12/12/2022]
Abstract
Redox reactions are intrinsically linked to energy metabolism. Therefore, redox processes are indispensable for organismal physiology and life itself. The term reactive oxygen species (ROS) describes a set of distinct molecular oxygen derivatives produced during normal aerobic metabolism. Multiple ROS-generating and ROS-eliminating systems actively maintain the intracellular redox state, which serves to mediate redox signaling and regulate cellular functions. ROS, in particular hydrogen peroxide (H2O2), are able to reversibly oxidize critical, redox-sensitive cysteine residues on target proteins. These oxidative post-translational modifications (PTMs) can control the biological activity of numerous enzymes and transcription factors (TFs), as well as their cellular localization or interactions with binding partners. In this review, we describe the diverse roles of redox regulation in the context of physiological cellular metabolism and provide insights into the pathophysiology of diseases when redox homeostasis is dysregulated.
Collapse
Affiliation(s)
- Claudia Lennicke
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK
| | - Helena M Cochemé
- MRC London Institute of Medical Sciences, Du Cane Road, London W12 0NN, UK; Institute of Clinical Sciences, Imperial College London, Hammersmith Hospital Campus, Du Cane Road, London W12 0NN, UK.
| |
Collapse
|
19
|
Peterson AL, Siddiqui G, Sloan EK, Creek DJ. β-Adrenoceptor regulation of metabolism in U937 derived macrophages. Mol Omics 2021; 17:583-595. [PMID: 34105576 DOI: 10.1039/d1mo00057h] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Macrophages have important roles in the immune system including clearing pathogens and wound healing. Metabolic phenotypes in macrophages have been associated with functional phenotypes, where pro-inflammatory macrophages have an increased rate of glycolysis and anti-inflammatory macrophages primarily use oxidative phosphorylation. β-adrenoceptor (βAR) signalling in macrophages has been implicated in disease states such as cancer, atherosclerosis and rheumatoid arthritis. The impact of βAR signalling on macrophage metabolism has not been defined. Using metabolomics and proteomics, we describe the impact of βAR signalling on macrophages treated with isoprenaline. We found that βAR signalling alters proteins involved in cytoskeletal rearrangement and redox homeostasis of the cell. We showed that βAR signalling in macrophages shifts glucose metabolism from glycolysis towards the tricarboxylic acid cycle and pentose phosphate pathways. We also show that βAR signalling perturbs purine metabolism by accumulating adenylate and guanylate pools. Taken together, these results indicate that βAR signalling shifts metabolism to support redox processes and upregulates proteins involved in cytoskeletal changes, which may contribute to βAR effects on macrophage function.
Collapse
Affiliation(s)
- Amanda L Peterson
- Drug Delivery, Disposition and Dynamics Theme, Monash Institute of Pharmaceutical Science, Monash University, Parkville, Victoria 3052, Australia.
| | | | | | | |
Collapse
|
20
|
The regulation of Hypoxia-Inducible Factor-1 (HIF-1alpha) expression by Protein Disulfide Isomerase (PDI). PLoS One 2021; 16:e0246531. [PMID: 33539422 PMCID: PMC7861413 DOI: 10.1371/journal.pone.0246531] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 01/20/2021] [Indexed: 01/05/2023] Open
Abstract
Hypoxia-inducible factor-1alpha (HIF-1alpha), a transcription factor, plays a critical role in adaption to hypoxia, which is a major feature of diseases, including cancer. Protein disulfide isomerase (PDI) is up-regulated in numerous cancers and leads to cancer progression. PDI, a member of the TRX superfamily, regulates the transcriptional activities of several transcription factors. To investigate the mechanisms by which PDI affects the function of HIF-1alpha, the overexpression or knockdown of PDI was performed. The overexpression of PDI decreased HIF-1alpha expression in the human hepatocarcinoma cell line, Hep3B, whereas the knockdown of endogenous PDI increased its expression. NH4Cl inhibited the decrease in HIF-1alpha expression by PDI overexpression, suggesting that HIF-1alpha was degraded by the lysosomal pathway. HIF-1alpha is transferred to lysosomal membranes by heat shock cognate 70 kDa protein (HSC70). The knockdown of HSC70 abolished the decrease, and PDI facilitated the interaction between HIF-1alpha and HSC70. HIF-1alpha directly interacted with PDI. PDI exists not only in the endoplasmic reticulum (ER), but also in the cytosol. Hypoxia increased cytosolic PDI. We also investigated changes in the redox state of HIF-1alpha using PEG-maleimide, which binds to thiols synthesized from disulfide bonds by reduction. An up-shift in the HIF-1alpha band by the overexpression of PDI was detected, suggesting that PDI formed disulfide bond in HIF-1alpha. HIF-1alpha oxidized by PDI was not degraded in HSC70-knockdown cells, indicating that the formation of disulfide bond in HIF-1alpha was important for decreases in HIF-1alpha expression. To the best of our knowledge, this is the first study to show the regulation of the expression and redox state of HIF-1alpha by PDI. We also demonstrated that PDI formed disulfide bonds in HIF-1alpha 1–245 aa and decreased its expression. In conclusion, the present results showed that PDI is a novel factor regulating HIF-1alpha through lysosome-dependent degradation by changes in its redox state.
Collapse
|
21
|
Bradley KL, Stokes CA, Marciniak SJ, Parker LC, Condliffe AM. Role of unfolded proteins in lung disease. Thorax 2021; 76:92-99. [PMID: 33077618 PMCID: PMC7803888 DOI: 10.1136/thoraxjnl-2019-213738] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2019] [Revised: 09/18/2020] [Accepted: 09/20/2020] [Indexed: 01/01/2023]
Abstract
The lungs are exposed to a range of environmental toxins (including cigarette smoke, air pollution, asbestos) and pathogens (bacterial, viral and fungal), and most respiratory diseases are associated with local or systemic hypoxia. All of these adverse factors can trigger endoplasmic reticulum (ER) stress. The ER is a key intracellular site for synthesis of secretory and membrane proteins, regulating their folding, assembly into complexes, transport and degradation. Accumulation of misfolded proteins within the lumen results in ER stress, which activates the unfolded protein response (UPR). Effectors of the UPR temporarily reduce protein synthesis, while enhancing degradation of misfolded proteins and increasing the folding capacity of the ER. If successful, homeostasis is restored and protein synthesis resumes, but if ER stress persists, cell death pathways are activated. ER stress and the resulting UPR occur in a range of pulmonary insults and the outcome plays an important role in many respiratory diseases. The UPR is triggered in the airway of patients with several respiratory diseases and in corresponding experimental models. ER stress has been implicated in the initiation and progression of pulmonary fibrosis, and evidence is accumulating suggesting that ER stress occurs in obstructive lung diseases (particularly in asthma), in pulmonary infections (some viral infections and in the setting of the cystic fibrosis airway) and in lung cancer. While a number of small molecule inhibitors have been used to interrogate the role of the UPR in disease models, many of these tools have complex and off-target effects, hence additional evidence (eg, from genetic manipulation) may be required to support conclusions based on the impact of such pharmacological agents. Aberrant activation of the UPR may be linked to disease pathogenesis and progression, but at present, our understanding of the context-specific and disease-specific mechanisms linking these processes is incomplete. Despite this, the ability of the UPR to defend against ER stress and influence a range of respiratory diseases is becoming increasingly evident, and the UPR is therefore attracting attention as a prospective target for therapeutic intervention strategies.
Collapse
Affiliation(s)
- Kirsty L Bradley
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Clare A Stokes
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | | | - Lisa C Parker
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| | - Alison M Condliffe
- Department of Infection, Immunity and Cardiovascular Diseases, The University of Sheffield, Sheffield, UK
| |
Collapse
|
22
|
PDI-Regulated Disulfide Bond Formation in Protein Folding and Biomolecular Assembly. Molecules 2020; 26:molecules26010171. [PMID: 33396541 PMCID: PMC7794689 DOI: 10.3390/molecules26010171] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2020] [Revised: 12/25/2020] [Accepted: 12/28/2020] [Indexed: 02/06/2023] Open
Abstract
Disulfide bonds play a pivotal role in maintaining the natural structures of proteins to ensure their performance of normal biological functions. Moreover, biological molecular assembly, such as the gluten network, is also largely dependent on the intermolecular crosslinking via disulfide bonds. In eukaryotes, the formation and rearrangement of most intra- and intermolecular disulfide bonds in the endoplasmic reticulum (ER) are mediated by protein disulfide isomerases (PDIs), which consist of multiple thioredoxin-like domains. These domains assist correct folding of proteins, as well as effectively prevent the aggregation of misfolded ones. Protein misfolding often leads to the formation of pathological protein aggregations that cause many diseases. On the other hand, glutenin aggregation and subsequent crosslinking are required for the formation of a rheologically dominating gluten network. Herein, the mechanism of PDI-regulated disulfide bond formation is important for understanding not only protein folding and associated diseases, but also the formation of functional biomolecular assembly. This review systematically illustrated the process of human protein disulfide isomerase (hPDI) mediated disulfide bond formation and complemented this with the current mechanism of wheat protein disulfide isomerase (wPDI) catalyzed formation of gluten networks.
Collapse
|
23
|
Yang S, Zhou R, Zhang C, He S, Su Z. Mitochondria-Associated Endoplasmic Reticulum Membranes in the Pathogenesis of Type 2 Diabetes Mellitus. Front Cell Dev Biol 2020; 8:571554. [PMID: 33195204 PMCID: PMC7606698 DOI: 10.3389/fcell.2020.571554] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2020] [Accepted: 08/21/2020] [Indexed: 02/05/2023] Open
Abstract
The endoplasmic reticulum (ER) and mitochondria are essential intracellular organelles that actively communicate via temporally and spatially formed contacts called mitochondria-associated membranes (MAMs). These mitochondria-ER contacts are not only necessary for the physiological function of the organelles and their coordination with each other, but they also control the intracellular lipid exchange, calcium signaling, cell survival, and homeostasis in cellular metabolism. Growing evidence strongly supports the role of the mitochondria-ER connection in the insulin resistance of peripheral tissues, pancreatic β cell dysfunction, and the consequent development of type 2 diabetes mellitus (T2DM). In this review, we summarize current advances in the understanding of the mitochondria-ER connection and specifically focus on addressing a new perspective of the alterations in mitochondria-ER communication in insulin signaling and β cell maintenance.
Collapse
Affiliation(s)
- Shanshan Yang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Ruixue Zhou
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Caixia Zhang
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Siyuan He
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| | - Zhiguang Su
- Molecular Medicine Research Center and National Clinical Research Center for Geriatrics, West China Hospital, State Key Laboratory of Biotherapy, Sichuan University, Chengdu, China
| |
Collapse
|
24
|
ERO1L promotes IL6/sIL6R signaling and regulates MUC16 expression to promote CA125 secretion and the metastasis of lung cancer cells. Cell Death Dis 2020; 11:853. [PMID: 33056994 PMCID: PMC7560734 DOI: 10.1038/s41419-020-03067-8] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Revised: 06/23/2020] [Accepted: 06/29/2020] [Indexed: 12/24/2022]
Abstract
The abnormal secretion of CA125, a classic tumor marker, is usually related to a poor prognosis in various tumors. Thus, this study aimed to explore the potential mechanisms that promote CA125 secretion in lung cancer. By querying the database, the gene endoplasmic reticulum oxidoreductase 1L (ERO1L) was identified and chosen as the research subject. The antibody chips were used to screen the lung cancer cell supernatant and found that the most obvious secreted protein was CA125. ERO1L was found to promote the secretion of IL6R by affecting the formation of disulfide bonds. IL6R bound to IL6 and triggered the activation of the NF-κB signaling pathway. Then, NF-κB bound to the promoter of MUC16, resulting in overexpression of MUC16. The extracellular segment of MUC16 was cleaved to form CA125, while the C terminus of MUC16 promoted the EMT phenotype and the release of IL6, forming a positive feedback pathway. In conclusion, ERO1L might affect the secretion of CA125 through the IL6 signaling pathway and form a positive feedback loop to further promote the development of lung cancer. This might expand the application scope of CA125 in lung cancer.
Collapse
|
25
|
Kanemura S, Sofia EF, Hirai N, Okumura M, Kadokura H, Inaba K. Characterization of the endoplasmic reticulum-resident peroxidases GPx7 and GPx8 shows the higher oxidative activity of GPx7 and its linkage to oxidative protein folding. J Biol Chem 2020; 295:12772-12785. [PMID: 32719007 DOI: 10.1074/jbc.ra120.013607] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
Oxidative protein folding occurs primarily in the mammalian endoplasmic reticulum, enabled by a diverse network comprising more than 20 members of the protein disulfide isomerase (PDI) family and more than five PDI oxidases. Although the canonical disulfide bond formation pathway involving Ero1α and PDI has been well-studied so far, the physiological roles of the newly identified PDI oxidases, glutathione peroxidase-7 (GPx7) and -8 (GPx8), are only poorly understood. We here demonstrated that human GPx7 has much higher reactivity with H2O2 and hence greater PDI oxidation activity than human GPx8. The high reactivity of GPx7 is due to the presence of a catalytic tetrad at the redox-active site, which stabilizes the sulfenylated species generated upon the reaction with H2O2 Although it was previously postulated that GPx7 catalysis involved a highly reactive peroxidatic cysteine that can be sulfenylated by H2O2, we revealed that a resolving cysteine instead regulates the PDI oxidation activity of GPx7. We also determined that GPx7 formed complexes preferentially with PDI and P5 in H2O2-treated cells. Altogether, these results suggest that human GPx7 functions as an H2O2-dependent PDI oxidase in cells, whereas PDI oxidation may not be the central physiological role of human GPx8.
Collapse
Affiliation(s)
- Shingo Kanemura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan.,School of Science and Technology, Kwansei Gakuin University, Gakuen, Sanda, Hyogo, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki, Aza, Aoba-ku, Sendai, Miyagi, Japan
| | - Elza Firdiani Sofia
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Naoya Hirai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Masaki Okumura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki, Aza, Aoba-ku, Sendai, Miyagi, Japan
| | - Hiroshi Kadokura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
26
|
Özcelik D. Treatment of Neuroblastoma Cells with Inhibitors of Protein Disulfide Isomerase Upregulates NQO1 Activity. Chem Res Toxicol 2020; 33:2099-2107. [DOI: 10.1021/acs.chemrestox.0c00101] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Dennis Özcelik
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark
| |
Collapse
|
27
|
Narayanan D, Ma S, Özcelik D. Targeting the Redox Landscape in Cancer Therapy. Cancers (Basel) 2020; 12:cancers12071706. [PMID: 32605023 PMCID: PMC7407119 DOI: 10.3390/cancers12071706] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2020] [Revised: 06/22/2020] [Accepted: 06/25/2020] [Indexed: 12/18/2022] Open
Abstract
Reactive oxygen species (ROS) are produced predominantly by the mitochondrial electron transport chain and by NADPH oxidases in peroxisomes and in the endoplasmic reticulum. The antioxidative defense counters overproduction of ROS with detoxifying enzymes and molecular scavengers, for instance, superoxide dismutase and glutathione, in order to restore redox homeostasis. Mutations in the redox landscape can induce carcinogenesis, whereas increased ROS production can perpetuate cancer development. Moreover, cancer cells can increase production of antioxidants, leading to resistance against chemo- or radiotherapy. Research has been developing pharmaceuticals to target the redox landscape in cancer. For instance, inhibition of key players in the redox landscape aims to modulate ROS production in order to prevent tumor development or to sensitize cancer cells in radiotherapy. Besides the redox landscape of a single cell, alternative strategies take aim at the multi-cellular level. Extracellular vesicles, such as exosomes, are crucial for the development of the hypoxic tumor microenvironment, and hence are explored as target and as drug delivery systems in cancer therapy. This review summarizes the current pharmaceutical and experimental interventions of the cancer redox landscape.
Collapse
Affiliation(s)
- Dilip Narayanan
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Sana Ma
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
| | - Dennis Özcelik
- Department of Drug Design and Pharmacology, University of Copenhagen, Universitetsparken 2, 2100 Copenhagen, Denmark; (D.N.); (S.M.)
- current address: Chemistry | Biology | Pharmacy Information Center, ETH Zürich, Vladimir-Prelog-Weg 10, 8093 Zürich, Switzerland
- Correspondence:
| |
Collapse
|
28
|
Hatami S, White CW, Qi X, Buchko M, Ondrus M, Kinnear A, Himmat S, Sergi C, Nagendran J, Freed DH. Immunity and Stress Responses Are Induced During Ex Situ Heart Perfusion. Circ Heart Fail 2020; 13:e006552. [PMID: 32498623 DOI: 10.1161/circheartfailure.119.006552] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 04/23/2020] [Indexed: 12/13/2022]
Abstract
BACKGROUND Ex situ heart perfusion (ESHP) preserves the donated heart in a perfused, beating condition preventing cold storage-related ischemia and provides a platform to evaluate myocardial viability during preservation. However, myocardial function declines gradually during ESHP. Extracorporeal circulation systems are associated with the induction of systemic inflammatory and stress responses. Our aim was to evaluate the incidence of inflammation and induction of endoplasmic reticulum stress responses during an extended period of ESHP. METHODS Cardiac function, myocardial tissue injury, markers of inflammation, oxidative stress, and endoplasmic reticulum stress were assessed in healthy pig hearts, perfused for 12 hours either in nonworking mode (non-WM=7) or working mode (WM, n=6). RESULTS Cardiac function declined during ESHP but was significantly better preserved in the hearts perfused in WM (median 11-hour cardiac index/1-hour cardiac index: WM=27% versus non-WM=9.5%, P=0.022). Myocardial markers of endoplasmic reticulum stress were expressed higher in ESHP hearts compared with in vivo samples. The proinflammatory cytokines and oxidized low-density lipoprotein significantly increased in the perfusate throughout the perfusion in both perfusion groups. The left ventricular expression of the cytokines and malondialdehyde was induced in non-WM, whereas it was not different between WM and in vivo. CONCLUSIONS Myocardial function declines during ESHP regardless of perfusion mode. However, ESHP in WM may lead to superior preservation of myocardial function and viability. Both inflammation and endoplasmic reticulum stress responses are significantly induced during ESHP and may contribute to the myocardial functional decline, representing a potential therapeutic target to improve the clinical donor heart preservation.
Collapse
Affiliation(s)
- Sanaz Hatami
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | | | - Xiao Qi
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Max Buchko
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Martin Ondrus
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Alexandra Kinnear
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
| | - Sayed Himmat
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Consolato Sergi
- Laboratory Medicine and Pathology (C.S.), University of Alberta, Edmonton, Canada
| | - Jayan Nagendran
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Alberta Transplant Institute, Edmonton, Canada (J.N., D.N.F.)
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| | - Darren H Freed
- Departments of Surgery (S. Hatami, X.Q., M.B., M.O., A.K., S. Himmat, J.N., D.H.F.), University of Alberta, Edmonton, Canada
- Physiology (D.H.F.), University of Alberta, Edmonton, Canada
- Biomedical Engineering (D.H.F.), University of Alberta, Edmonton, Canada
- Alberta Transplant Institute, Edmonton, Canada (J.N., D.N.F.)
- Canadian Transplant Research Program (S. Hatami, X.Q., S. Himmat, J.N., D.H.F.)
| |
Collapse
|
29
|
Madero-Ayala PA, Mares-Alejandre RE, Ramos-Ibarra MA. A molecular dynamics approach on the Y393C variant of protein disulfide isomerase A1. Chem Biol Drug Des 2020; 96:1341-1347. [PMID: 32352225 DOI: 10.1111/cbdd.13700] [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: 01/30/2020] [Revised: 04/03/2020] [Accepted: 04/26/2020] [Indexed: 11/28/2022]
Abstract
Human protein disulfide isomerase A1 (PDIA1) shows both catalytic (i.e., oxidoreductase) and non-catalytic (i.e., chaperone) activities and plays a crucial role in the oxidative folding of proteins within the endoplasmic reticulum. PDIA1 dysregulation is a common trait in numerous pathophysiological conditions, including neurodegenerative disorders and cancerous diseases. The 1178A>G mutation of the human PDIA1-encoding gene is a non-synonymous single nucleotide polymorphism detected in patients with Cole-Carpenter syndrome type 1 (CSS1), a particularly rare bone disease. In vitro studies showed that the encoded variant (PDIA1 Y393C) exhibits limited oxidoreductase activity. To gain knowledge on the structure-function relationship, we undertook a molecular dynamics (MD) approach to examine the structural stability of PDIA1 Y393C. Results showed that significant conformational changes are the structural consequence of the amino acid substitution Tyr>Cys at position 393 of the PDIA1 protein. This structure-based study provides further knowledge about the molecular origin of CCS1.
Collapse
Affiliation(s)
- Pablo A Madero-Ayala
- Grupo de Investigación en Biotecnología y Biociencias, Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana, México
| | - Rosa E Mares-Alejandre
- Grupo de Investigación en Biotecnología y Biociencias, Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana, México
| | - Marco A Ramos-Ibarra
- Grupo de Investigación en Biotecnología y Biociencias, Facultad de Ciencias Químicas e Ingeniería, Universidad Autónoma de Baja California, Tijuana, México
| |
Collapse
|
30
|
Shergalis AG, Hu S, Bankhead A, Neamati N. Role of the ERO1-PDI interaction in oxidative protein folding and disease. Pharmacol Ther 2020; 210:107525. [PMID: 32201313 DOI: 10.1016/j.pharmthera.2020.107525] [Citation(s) in RCA: 101] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2019] [Revised: 02/04/2020] [Accepted: 02/19/2020] [Indexed: 02/06/2023]
Abstract
Protein folding in the endoplasmic reticulum is an oxidative process that relies on protein disulfide isomerase (PDI) and endoplasmic reticulum oxidase 1 (ERO1). Over 30% of proteins require the chaperone PDI to promote disulfide bond formation. PDI oxidizes cysteines in nascent polypeptides to form disulfide bonds and can also reduce and isomerize disulfide bonds. ERO1 recycles reduced PDI family member PDIA1 using a FAD cofactor to transfer electrons to oxygen. ERO1 dysfunction critically affects several diseases states. Both ERO1 and PDIA1 are overexpressed in cancers and implicated in diabetes and neurodegenerative diseases. Cancer-associated ERO1 promotes cell migration and invasion. Furthermore, the ERO1-PDIA1 interaction is critical for epithelial-to-mesenchymal transition. Co-expression analysis of ERO1A gene expression in cancer patients demonstrated that ERO1A is significantly upregulated in lung adenocarcinoma (LUAD), glioblastoma and low-grade glioma (GBMLGG), pancreatic ductal adenocarcinoma (PAAD), and kidney renal papillary cell carcinoma (KIRP) cancers. ERO1Α knockdown gene signature correlates with knockdown of cancer signaling proteins including IGF1R, supporting the search for novel, selective ERO1 inhibitors for the treatment of cancer. In this review, we explore the functions of ERO1 and PDI to support inhibition of this interaction in cancer and other diseases.
Collapse
Affiliation(s)
- Andrea G Shergalis
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States
| | - Shuai Hu
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States; Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, United States
| | - Armand Bankhead
- Department of Computational Medicine and Bioinformatics, University of Michigan Medical School, Ann Arbor, MI 48109, United States; Department of Biostatistics, University of Michigan School of Public Health, Ann Arbor, MI 48109, United States
| | - Nouri Neamati
- Department of Medicinal Chemistry, College of Pharmacy, University of Michigan, Rogel Cancer Center, Ann Arbor, MI 48109, United States.
| |
Collapse
|
31
|
Engineering a Pichia pastoris nitrilase whole cell catalyst through the increased nitrilase gene copy number and co-expressing of ER oxidoreductin 1. Appl Microbiol Biotechnol 2020; 104:2489-2500. [DOI: 10.1007/s00253-020-10422-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Revised: 01/18/2020] [Accepted: 01/26/2020] [Indexed: 12/14/2022]
|
32
|
Liu C, Hao Y, Yin F, Liu J. Geniposide Balances the Redox Signaling to Mediate Glucose-Stimulated Insulin Secretion in Pancreatic β-Cells. Diabetes Metab Syndr Obes 2020; 13:509-520. [PMID: 32158246 PMCID: PMC7049278 DOI: 10.2147/dmso.s240794] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 01/23/2020] [Indexed: 12/22/2022] Open
Abstract
PURPOSE To investigate the effect of geniposide on the biosynthesis of insulin and the expression protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductin 1 (ERO1) in the presence of low (5 mM) and high (25 mM) glucose in pancreatic β cells. METHODS The content of insulin was measured by ELISA, the number of SH groups was determined with the classical chromogenic reagent, 5,5'-dithiobis-(2-nitrobenzoic) acid (DTNB; also known as Ellman's reagent), the expressions of PDI and ERO1 were analyzed by Western blot. RESULTS Geniposide played contrary roles on the accumulation of H2O2, the ratio of GSH/GSSG and the thiol-disulfide balance in the presence of low (5 mM) and high (25 mM) glucose in rat pancreatic INS-1 cells. Geniposide also regulated the protein levels of protein disulfide isomerase (PDI) and endoplasmic reticulum oxidoreductin1 (ERO1), the two key enzymes for the production of H2O2 during the biosynthesis of insulin in INS-1 cells. CONCLUSION Geniposide affects glucose-stimulated insulin secretion by modulating the thiol-disulfide balance that is controlled by the redox signaling in pancreatic β cells.
Collapse
Affiliation(s)
- Chunyan Liu
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing400054, People’s Republic of China
| | - Yanan Hao
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing400054, People’s Republic of China
| | - Fei Yin
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing400054, People’s Republic of China
| | - Jianhui Liu
- Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Chongqing400054, People’s Republic of China
- Correspondence: Jianhui Liu; Fei Yin Chongqing Key Laboratory of Medicinal Chemistry & Molecular Pharmacology, Chongqing University of Technology, Hongguang Road 69, Ba’nan District, Chongqing400054, People’s Republic of China Tel/Fax +86-23-6256-3182 Email ;
| |
Collapse
|
33
|
Mullick M, Nayak S. Comprehending the Unfolded Protein Response as a Conduit for Improved Mesenchymal Stem Cell-Based Therapeutics. REGENERATIVE ENGINEERING AND TRANSLATIONAL MEDICINE 2019. [DOI: 10.1007/s40883-019-00143-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
34
|
The role of mitochondria-associated membranes in cellular homeostasis and diseases. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2019; 350:119-196. [PMID: 32138899 DOI: 10.1016/bs.ircmb.2019.11.002] [Citation(s) in RCA: 83] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Mitochondria and endoplasmic reticulum (ER) are fundamental in the control of cell physiology regulating several signal transduction pathways. They continuously communicate exchanging messages in their contact sites called MAMs (mitochondria-associated membranes). MAMs are specific microdomains acting as a platform for the sorting of vital and dangerous signals. In recent years increasing evidence reported that multiple scaffold proteins and regulatory factors localize to this subcellular fraction suggesting MAMs as hotspot signaling domains. In this review we describe the current knowledge about MAMs' dynamics and processes, which provided new correlations between MAMs' dysfunctions and human diseases. In fact, MAMs machinery is strictly connected with several pathologies, like neurodegeneration, diabetes and mainly cancer. These pathological events are characterized by alterations in the normal communication between ER and mitochondria, leading to deep metabolic defects that contribute to the progression of the diseases.
Collapse
|
35
|
Markiewicz E, Idowu OC. DNA damage in human skin and the capacities of natural compounds to modulate the bystander signalling. Open Biol 2019; 9:190208. [PMID: 31847786 PMCID: PMC6936251 DOI: 10.1098/rsob.190208] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2019] [Accepted: 11/19/2019] [Indexed: 12/20/2022] Open
Abstract
Human skin is a stratified organ frequently exposed to sun-generated ultraviolet radiation (UVR), which is considered one of the major factors responsible for DNA damage. Such damage can be direct, through interactions of DNA with UV photons, or indirect, mainly through enhanced production of reactive oxygen species that introduce oxidative changes to the DNA. Oxidative stress and DNA damage also associate with profound changes at the cellular and molecular level involving several cell cycle and signal transduction factors responsible for DNA repair or irreversible changes linked to ageing. Crucially, some of these factors constitute part of the signalling known for the induction of biological changes in non-irradiated, neighbouring cells and defined as the bystander effect. Network interactions with a number of natural compounds, based on their known activity towards these biomarkers in the skin, reveal the capacity to inhibit both the bystander signalling and cell cycle/DNA damage molecules while increasing expression of the anti-oxidant enzymes. Based on this information, we discuss the likely polypharmacology applications of the natural compounds and next-generation screening technologies in improving the anti-oxidant and DNA repair capacities of the skin.
Collapse
|
36
|
Xin Z, Bian X, Gao W, Wang Y, Yao Z, Shi T, Guo C. Riboflavin deficiency alters cholesterol homeostasis partly by reducing apolipoprotein B100 synthesis in HepG2 cells. INT J VITAM NUTR RES 2019; 91:204-211. [PMID: 31656126 DOI: 10.1024/0300-9831/a000610] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Riboflavin deficiency led to lower blood cholesterol level and higher content of hepatic cholesterol in rats and the mechanisms are not clarified yet. We hypothesized that riboflavin deficiency might alter cholesterol homeostasis via apolipoprotein B100, one of the important proteins in cholesterol transport. To test this hypothesis, HepG2 cells were cultured in riboflavin-deficient media for 4 days to develop riboflavin deficiency. Compared to riboflavin-sufficient cells, the mRNA (0. 37 ± 0.04 vs 1.03 ± 0.29 relative expression level, n = 3) and protein expressions of apolipoprotein B100 (intracellular: 173.7 ± 14.4 vs 254.8 ± 47.2 μg/mg protein; extracellular: 93.8 ± 31.1 vs 161.6 ± 23.9 μg/mg protein; n = 3) were significantly reduced in riboflavin-deficient cells (P < 0.05). Endoplasmic reticulum oxidoreductin 1 and protein disulfide isomerase, two enzymes involved in the oxidative folding of apolipoprotein B100, were also lower remarkably in expression at both mRNA and protein levels. Meanwhile, intracellular cholesterol was increased (256.3 ± 17.1 μM/g protein vs 181.4 ± 23.9 μM/g protein, n = 4) and extracellular cholesterol decreased (110.0 ± 23.2 μM/g protein vs 166.2 ± 34.6 μM/g protein, n = 4) significantly in riboflavin-deficient cells (P < 0.05). Very low-density lipoprotein was also diminished (29.0 ± 6.1 μM/g protein vs 67.0 ± 11.0 μM/g protein, n = 4) in the culture media (P < 0.05). These findings suggest that riboflavin deficiency alters cholesterol homeostasis partly by reducing apolipoprotein B100 synthesis in HepG2 cells.
Collapse
Affiliation(s)
- Zhonghao Xin
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Xiangyu Bian
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Weina Gao
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Yawen Wang
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Zhanxin Yao
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Tala Shi
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| | - Changjiang Guo
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin, PR China
| |
Collapse
|
37
|
Özcelik D, Pezacki JP. Small Molecule Inhibition of Protein Disulfide Isomerase in Neuroblastoma Cells Induces an Oxidative Stress Response and Apoptosis Pathways. ACS Chem Neurosci 2019; 10:4068-4075. [PMID: 31343165 DOI: 10.1021/acschemneuro.9b00301] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Protein disulfide isomerase (PDI) is a multifunctional enzyme located in the endoplasmic reticulum (ER) contributing to redox homeostasis and oxidative protein folding. PDI is associated with many diseases including neurodegenerative disorders like Alzheimer's disease and, hence, is considered a promising drug target. In this study, we investigate the abscisic acid (ABA)-derived PDI inhibitor origamicin for its neuropharmacological potential. First, we validated the function of origamicin by monitoring the inhibition of PDI's oxidoreductase activity using an in vitro enzyme activity assay. We also applied Huisgen cycloaddition chemistry (or "click chemistry") to interrogate the interaction of origamicin and PDI. Then, we evaluated the impact of origamicin on the viability of the neuroblastoma cell line SH-SY5Y. Next, we analyzed the gene expression profile of SH-SY5Y cells upon treatment with origamicin. We found 207 differentially expressed genes, including MYC. Computational analysis revealed an enrichment of genes involved in the oxidative stress response and the p53 signaling pathway. Induction of the p53 signaling pathway and downregulation of MYC are known to affect processes such as cell cycle, cellular repair, and apoptosis. Our study reveals the molecular mechanism of PDI inhibition by origamicin. Furthermore, this study provides important gene expression profiles that offer insights into the underlying mechanism of PDI inhibition and creates a valuable starting point for neuropharmacological applications of origamicin and other PDI inhibitors.
Collapse
Affiliation(s)
- Dennis Özcelik
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
| | - John Paul Pezacki
- Department of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
- Department of Biochemistry, Microbiology, and Immunology, Ottawa Institute for Systems Biology, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
38
|
Beal DM, Bastow EL, Staniforth GL, von der Haar T, Freedman RB, Tuite MF. Quantitative Analyses of the Yeast Oxidative Protein Folding Pathway In Vitro and In Vivo. Antioxid Redox Signal 2019; 31:261-274. [PMID: 30880408 PMCID: PMC6602113 DOI: 10.1089/ars.2018.7615] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 03/14/2019] [Accepted: 03/14/2019] [Indexed: 12/11/2022]
Abstract
Aims: Efficient oxidative protein folding (OPF) in the endoplasmic reticulum (ER) is a key requirement of the eukaryotic secretory pathway. In particular, protein folding linked to the formation of disulfide bonds, an activity dependent on the enzyme protein disulfide isomerase (PDI), is crucial. For the de novo formation of disulfide bonds, reduced PDI must be reoxidized by an ER-located oxidase (ERO1). Despite some knowledge of this pathway, the kinetic parameters with which these components act and the importance of specific parameters, such as PDI reoxidation by Ero1, for the overall performance of OPF in vivo remain poorly understood. Results: We established an in vitro system using purified yeast (Saccharomyces cerevisiae) PDI (Pdi1p) and ERO1 (Ero1p) to investigate OPF. This necessitated the development of a novel reduction/oxidation processing strategy to generate homogenously oxidized recombinant yeast Ero1p. This new methodology enabled the quantitative assessment of the interaction of Pdi1p and Ero1p in vitro by measuring oxygen consumption and reoxidation of reduced RNase A. The resulting quantitative data were then used to generate a simple model that can describe the oxidizing capacity of Pdi1p and Ero1p in vitro and predict the in vivo effect of modulation of the levels of these proteins. Innovation: We describe a model that can be used to explore the OPF pathway and its control in a quantitative way. Conclusion: Our study informs and provides new insights into how OPF works at a molecular level and provides a platform for the design of more efficient heterologous protein expression systems in yeast.
Collapse
Affiliation(s)
- Dave M. Beal
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Emma L. Bastow
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Gemma L. Staniforth
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Tobias von der Haar
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| | - Robert B. Freedman
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
- School of Life Sciences, Gibbet Hill Campus, University of Warwick, Coventry, United Kingdom
| | - Mick F. Tuite
- Kent Fungal Group, School of Biosciences, University of Kent, Canterbury, United Kingdom
| |
Collapse
|
39
|
Zhu R, Li X, Xu J, Barrabi C, Kekulandara D, Woods J, Chen X, Liu M. Defective endoplasmic reticulum export causes proinsulin misfolding in pancreatic β cells. Mol Cell Endocrinol 2019; 493:110470. [PMID: 31158417 PMCID: PMC6613978 DOI: 10.1016/j.mce.2019.110470] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/18/2019] [Revised: 05/30/2019] [Accepted: 05/30/2019] [Indexed: 02/06/2023]
Abstract
Endoplasmic reticulum (ER) homeostasis is essential for cell function. Increasing evidence indicates that, efficient protein ER export is important for ER homeostasis. However, the consequence of impaired ER export remains largely unknown. Herein, we found that defective ER protein transport caused by either Sar1 mutants or brefeldin A impaired proinsulin oxidative folding in the ER of β-cells. Misfolded proinsulin formed aberrant disulfide-linked dimers and high molecular weight proinsulin complexes, and induced ER stress. Limiting proinsulin load to the ER alleviated ER stress, indicating that misfolded proinsulin is a direct cause of ER stress. This study revealed significance of efficient ER export in maintaining ER protein homeostasis and native folding of proinsulin. Given the fact that proinsulin misfolding plays an important role in diabetes, this study suggests that enhancing ER export may be a potential therapeutic target to prevent/delay β-cell failure caused by proinsulin misfolding and ER stress.
Collapse
Affiliation(s)
- Ruimin Zhu
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA; Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Xin Li
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Jialu Xu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China
| | - Cesar Barrabi
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Dilini Kekulandara
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - James Woods
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA
| | - Xuequn Chen
- Department of Physiology, School of Medicine, Wayne State University, Detroit, MI, USA.
| | - Ming Liu
- Department of Endocrinology and Metabolism, Tianjin Medical University General Hospital, Tianjin 300052, China.
| |
Collapse
|
40
|
Bian X, Gao W, Wang Y, Yao Z, Xu Q, Guo C, Li B. Riboflavin deficiency affects lipid metabolism partly by reducing apolipoprotein B100 synthesis in rats. J Nutr Biochem 2019; 70:75-81. [PMID: 31176989 DOI: 10.1016/j.jnutbio.2019.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2018] [Revised: 03/12/2019] [Accepted: 04/25/2019] [Indexed: 12/18/2022]
Abstract
Lipid metabolism is dependent on riboflavin status. Apolipoprotein B100 plays an important role in lipids transportation. This study was aimed to investigate the effect of riboflavin status on lipid metabolism and explore its association with apolipoprotein B100 synthesis in vivo. Riboflavin deficiency was developed in rats by feeding riboflavin-deficient diets. Compared to the control rats, the mRNA and protein expressions of apolipoprotein B100 were significantly reduced in riboflavin-deficient rats. Endoplasmic reticulum oxidoreductin 1 (ERO1) and protein disulfide isomerase (PDI), two enzymes involved in the oxidative folding of apolipoprotein B100, were also lowered remarkably in expression at protein level. Meanwhile, total cholesterol and triglyceride levels were decreased in the plasma and increased in the liver of riboflavin-deficient rats. The plasma very low-density lipoprotein cholesterol (VLDL-c) and low-density lipoprotein cholesterol (LDL-c) were also reduced in riboflavin-deficient rats. Our findings demonstrate that riboflavin deficiency affects lipid metabolism partly by reducing apolipoprotein B100 synthesis.
Collapse
Affiliation(s)
- Xiangyu Bian
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, People's Republic of China; Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China
| | - Weina Gao
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China
| | - Yawen Wang
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China
| | - Zhanxin Yao
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China
| | - Qingao Xu
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China
| | - Changjiang Guo
- Department of Nutrition and Food Hygiene, Institute of Environmental and Operational Medicine, Tianjin 300050, People's Republic of China.
| | - Bailin Li
- College of Food Science and Technology, Shanghai Ocean University, Shanghai 201306, People's Republic of China.
| |
Collapse
|
41
|
Hayes KE, Batsomboon P, Chen WC, Johnson BD, Becker A, Eschrich S, Yang Y, Robart AR, Dudley GB, Geldenhuys WJ, Hazlehurst LA. Inhibition of the FAD containing ER oxidoreductin 1 (Ero1) protein by EN-460 as a strategy for treatment of multiple myeloma. Bioorg Med Chem 2019; 27:1479-1488. [PMID: 30850265 PMCID: PMC6554731 DOI: 10.1016/j.bmc.2019.02.016] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2018] [Revised: 01/31/2019] [Accepted: 02/08/2019] [Indexed: 11/28/2022]
Abstract
Multiple myeloma (MM) cells demonstrate high basal endoplasmic reticulum (ER) stress and are typically exquisitely sensitive to agents such as proteasome inhibitors that activate the unfolded protein response. The flavin adenosine dinucleotide (FAD) containing endoplasmic reticulum oxidoreductin enzyme (Ero1L) catalyzes de-novo disulfide bridge formation of ER resident proteins and contributes to proper protein folding. Here we show that increased Ero1L expression is prognostic of poor outcomes for MM patients relapsing on therapy. We propose that targeting protein folding via inhibition of Ero1L may represent a novel therapeutic strategy for the treatment of MM. In this report we show that treatment of MM cells with EN-460, a known inhibitor of ERO1L, was sufficient to inhibit cell proliferation and induce apoptosis. Furthermore, we show that cell death correlated in part with induction of ER stress. We also show that EN460 inhibited the enzyme activity of Ero1L, with an IC50 of 22.13 μM, consistent with previous reports. However, EN-460 was also found to inhibit other FAD-containing enzymes including MAO-A (IC50 = 7.91 μM), MAO-B (IC50 = 30.59 μM) and LSD1 (IC50 = 4.16 μM), suggesting overlap in inhibitor activity and the potential need to develop more specific inhibitors to enable pharmacological validation of ERO1L as a target for the treatment of MM. We additionally prepared and characterized azide-tagged derivatives of EN-460 as possible functional probe compounds (e.g., for photo-affinity labeling) for future target-engagement studies and further development of structure-activity relationships.
Collapse
Affiliation(s)
- Karen E Hayes
- Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, United States
| | - Paratchata Batsomboon
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States
| | - Wei-Chih Chen
- Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, United States
| | - Brennan D Johnson
- Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, United States
| | | | | | - Yan Yang
- Moffitt Cancer Center, Tampa, FL 33612, United States
| | - Aaron R Robart
- Biochemistry, School of Medicine, West Virginia University, Morgantown, WV 26506, United States
| | - Gregory B Dudley
- C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States; Cancer Center, West Virginia University, Morgantown, WV 26506, United States
| | - Werner J Geldenhuys
- Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, United States; C. Eugene Bennett Department of Chemistry, West Virginia University, Morgantown, WV 26506, United States; Cancer Center, West Virginia University, Morgantown, WV 26506, United States; Department of Neuroscience, School of Medicine, West Virginia University, Morgantown, WV 26506, United States.
| | - Lori A Hazlehurst
- Pharmaceutical Sciences, School of Pharmacy, West Virginia University, Morganton, WV 26506, United States; Cancer Center, West Virginia University, Morgantown, WV 26506, United States.
| |
Collapse
|
42
|
Özcelik D, Seto A, Rakic B, Farzam A, Supek F, Pezacki JP. Gene Expression Profiling of Endoplasmic Reticulum Stress in Hepatitis C Virus-Containing Cells Treated with an Inhibitor of Protein Disulfide Isomerases. ACS OMEGA 2018; 3:17227-17235. [PMID: 30775641 PMCID: PMC6369735 DOI: 10.1021/acsomega.8b02676] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/06/2018] [Accepted: 11/23/2018] [Indexed: 05/30/2023]
Abstract
Protein disulfide isomerases (PDIs) catalyze disulfide bond formation between protein cysteine residues during protein folding in the endoplasmic reticulum (ER) lumen and are essential for maintaining ER homoeostasis. The life cycle of the hepatitis C virus (HCV) is closely associated with the ER. Synthesis and maturation of HCV proteins occur in the ER membrane and are mediated by multiple host cell factors that include also PDI. Here, we present a study investigating the effect of PDI inhibition on Huh7 human hepatoma cells harboring an HCV subgenomic replicon using the abscisic acid-derived PDI inhibitor origamicin. Transcriptional profiling shows that origamicin changed the expression levels of genes involved in the oxidative and ER stress responses and the unfolded protein response, as indicated by the upregulation of antioxidant enzymes and chaperone proteins, the downregulation of cell-cycle proteins, and induction of apoptosis-associated genes. Our data suggest that origamicin negatively impacts HCV replication by causing an imbalance in cellular homoeostasis and induction of stress responses. These insights suggest that inhibition of PDIs by low-molecular-weight inhibitors could be a promising approach to the discovery of novel antiviral compounds.
Collapse
Affiliation(s)
- Dennis Özcelik
- Department
of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
| | - Andrew Seto
- Department
of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
| | - Bojana Rakic
- Department
of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
| | - Ali Farzam
- Department
of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
| | - Frantisek Supek
- Department
of Genetics & Neglected Diseases, Genomics
Institute of the Novartis Research Foundation, San Diego, California 92121, United States
| | - John Paul Pezacki
- Department
of Chemistry and Biomolecular Sciences, University of Ottawa, 10 Marie Curie Street, Ottawa, Ontario K1N 6N5, Canada
- Department
of Biochemistry, Microbiology, and Immunology, Ottawa Institute for Systems Biology, 451 Smyth Road, Ottawa, Ontario K1H 8M5, Canada
| |
Collapse
|
43
|
Morris G, Puri BK, Walder K, Berk M, Stubbs B, Maes M, Carvalho AF. The Endoplasmic Reticulum Stress Response in Neuroprogressive Diseases: Emerging Pathophysiological Role and Translational Implications. Mol Neurobiol 2018; 55:8765-8787. [PMID: 29594942 PMCID: PMC6208857 DOI: 10.1007/s12035-018-1028-6] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2018] [Accepted: 03/20/2018] [Indexed: 02/07/2023]
Abstract
The endoplasmic reticulum (ER) is the main cellular organelle involved in protein synthesis, assembly and secretion. Accumulating evidence shows that across several neurodegenerative and neuroprogressive diseases, ER stress ensues, which is accompanied by over-activation of the unfolded protein response (UPR). Although the UPR could initially serve adaptive purposes in conditions associated with higher cellular demands and after exposure to a range of pathophysiological insults, over time the UPR may become detrimental, thus contributing to neuroprogression. Herein, we propose that immune-inflammatory, neuro-oxidative, neuro-nitrosative, as well as mitochondrial pathways may reciprocally interact with aberrations in UPR pathways. Furthermore, ER stress may contribute to a deregulation in calcium homoeostasis. The common denominator of these pathways is a decrease in neuronal resilience, synaptic dysfunction and even cell death. This review also discusses how mechanisms related to ER stress could be explored as a source for novel therapeutic targets for neurodegenerative and neuroprogressive diseases. The design of randomised controlled trials testing compounds that target aberrant UPR-related pathways within the emerging framework of precision psychiatry is warranted.
Collapse
Affiliation(s)
- Gerwyn Morris
- Tir Na Nog, Bryn Road seaside 87, Llanelli, Wales, SA15 2LW, UK
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
| | - Basant K Puri
- Department of Medicine, Imperial College London, Hammersmith Hospital, London, England, W12 0HS, UK.
| | - Ken Walder
- The Centre for Molecular and Medical Research, School of Medicine, Deakin University, P.O. Box 291, Geelong, 3220, Australia
| | - Michael Berk
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, University of Melbourne, Melbourne, Australia
- Orygen, the National Centre of Excellence in Youth Mental Health, Parkville, Australia
- Centre for Youth Mental Health, University of Melbourne, Melbourne, Australia
- Florey Institute for Neuroscience and Mental Health, Melbourne, Australia
| | - Brendon Stubbs
- Physiotherapy Department, South London and Maudsley NHS Foundation Trust, London, UK
- Health Service and Population Research Department, Institute of Psychiatry, Psychology and Neuroscience, King's College London, London, UK
- Faculty of Health, Social Care and Education, Anglia Ruskin University, Chelmsford, UK
| | - Michael Maes
- IMPACT Strategic Research Centre, School of Medicine, Deakin University, Geelong, Australia
- Department of Psychiatry, Chulalongkorn University, Bangkok, Thailand
| | - André F Carvalho
- Department of Psychiatry, Faculty of Medicine, University of Toronto, Toronto, ON, Canada
- Centre for Addiction & Mental Health (CAMH), Toronto, ON, Canada
| |
Collapse
|
44
|
Emerging roles of endoplasmic reticulum-resident selenoproteins in the regulation of cellular stress responses and the implications for metabolic disease. Biochem J 2018; 475:1037-1057. [PMID: 29559580 DOI: 10.1042/bcj20170920] [Citation(s) in RCA: 57] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2017] [Revised: 02/12/2018] [Accepted: 02/13/2018] [Indexed: 12/25/2022]
Abstract
Chronic metabolic stress leads to cellular dysfunction, characterized by excessive reactive oxygen species, endoplasmic reticulum (ER) stress and inflammation, which has been implicated in the pathogenesis of obesity, type 2 diabetes and cardiovascular disease. The ER is gaining recognition as a key organelle in integrating cellular stress responses. ER homeostasis is tightly regulated by a complex antioxidant system, which includes the seven ER-resident selenoproteins - 15 kDa selenoprotein, type 2 iodothyronine deiodinase and selenoproteins S, N, K, M and T. Here, the findings from biochemical, cell-based and mouse studies investigating the function of ER-resident selenoproteins are reviewed. Human experimental and genetic studies are drawn upon to highlight the relevance of these selenoproteins to the pathogenesis of metabolic disease. ER-resident selenoproteins have discrete roles in the regulation of oxidative, ER and inflammatory stress responses, as well as intracellular calcium homeostasis. To date, only two of these ER-resident selenoproteins, selenoproteins S and N have been implicated in human disease. Nonetheless, the potential of all seven ER-resident selenoproteins to ameliorate metabolic dysfunction warrants further investigation.
Collapse
|
45
|
Bambino K, Zhang C, Austin C, Amarasiriwardena C, Arora M, Chu J, Sadler KC. Inorganic arsenic causes fatty liver and interacts with ethanol to cause alcoholic liver disease in zebrafish. Dis Model Mech 2018; 11:dmm.031575. [PMID: 29361514 PMCID: PMC5894941 DOI: 10.1242/dmm.031575] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2017] [Accepted: 12/07/2017] [Indexed: 12/19/2022] Open
Abstract
The rapid increase in fatty liver disease (FLD) incidence is attributed largely to genetic and lifestyle factors; however, environmental toxicants are a frequently overlooked factor that can modify the effects of more common causes of FLD. Chronic exposure to inorganic arsenic (iAs) is associated with liver disease in humans and animal models, but neither the mechanism of action nor the combinatorial interaction with other disease-causing factors has been fully investigated. Here, we examined the contribution of iAs to FLD using zebrafish and tested the interaction with ethanol to cause alcoholic liver disease (ALD). We report that zebrafish exposed to iAs throughout development developed specific phenotypes beginning at 4 days post-fertilization (dpf), including the development of FLD in over 50% of larvae by 5 dpf. Comparative transcriptomic analysis of livers from larvae exposed to either iAs or ethanol revealed the oxidative stress response and the unfolded protein response (UPR) caused by endoplasmic reticulum (ER) stress as common pathways in both these models of FLD, suggesting that they target similar cellular processes. This was confirmed by our finding that arsenic is synthetically lethal with both ethanol and a well-characterized ER-stress-inducing agent (tunicamycin), suggesting that these exposures work together through UPR activation to cause iAs toxicity. Most significantly, combined exposure to sub-toxic concentrations of iAs and ethanol potentiated the expression of UPR-associated genes, cooperated to induce FLD, reduced the expression of as3mt, which encodes an arsenic-metabolizing enzyme, and significantly increased the concentration of iAs in the liver. This demonstrates that iAs exposure is sufficient to cause FLD and that low doses of iAs can potentiate the effects of ethanol to cause liver disease. This article has an associated First Person interview with the first author of the paper. Summary: Using zebrafish, the authors show that exposure to a common environmental contaminant, inorganic arsenic, increases the risk of alcoholic liver disease.
Collapse
Affiliation(s)
- Kathryn Bambino
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chi Zhang
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| | - Christine Austin
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Chitra Amarasiriwardena
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Manish Arora
- Department of Environmental Medicine and Public Health, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Jaime Chu
- Department of Pediatrics, Division of Pediatric Hepatology, Icahn School of Medicine at Mount Sinai, New York, New York 10029, USA
| | - Kirsten C Sadler
- Program in Biology, New York University Abu Dhabi, Saadiyat Island Campus, PO Box 129188 Abu Dhabi, United Arab Emirates
| |
Collapse
|
46
|
Liu G, Wang J, Hou Y, Huang YB, Wang J, Li C, Guo S, Li L, Hu SQ. Characterization of wheat endoplasmic reticulum oxidoreductin 1 and its application in Chinese steamed bread. Food Chem 2018; 256:31-39. [PMID: 29606453 DOI: 10.1016/j.foodchem.2018.02.080] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Revised: 02/04/2018] [Accepted: 02/14/2018] [Indexed: 11/15/2022]
Abstract
This study investigated characteristics of recombinant wheat Endoplasmic Reticulum Oxidoreductin 1 (wEro1) and its influence on Chinese steamed bread (CSB) qualities. The purified wEro1 monomer, which contained two conserved redox active motif sites, bound to flavin adenine dinucleotide (FAD) cofactor with a molecular weight of ∼47 kDa. wEro1 catalyzed the reduction of both bound and free FAD, and its reduction activity of free FAD reached 7.8 U/mg. Moreover, wEro1 catalyzed the oxidation of dithiothreitol and wheat protein disulfide isomerase (wPDI). Both glutathione and the reduced ribonuclease could work as electron donors for wEro1 in catalyzing the oxidation of wPDI. Additionally, wEro1 supplementation improved the CSB qualities with an increased specific volume of CSB and decreased crumb hardness, which was attributed to water-insoluble wheat proteins increasing and gluten network strengthening. The results give an understanding of the properties and function of wEro1 to facilitate its application especially in the flour-processing industry.
Collapse
Affiliation(s)
- Guang Liu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510641, China; Sericultural & Agri-Food Research Institute, Guangdong Academy of Agricultural Sciences, Key Laboratory of Functional Foods, Ministry of Agriculture, Guangdong Key Laboratory of Agricultural Products Processing, Guangzhou 510610, China
| | - JingJing Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510641, China
| | - Yi Hou
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - Yan-Bo Huang
- State Key Laboratory of Pulp and Paper Engineering, South China University of Technology, Guangzhou, Guangdong 510640, China
| | - JiaJia Wang
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510641, China
| | - Cunzhi Li
- Department of Food Science and Engineering, Jinan University, Guangzhou 510632, Guangdong, China
| | - ShiJun Guo
- Guangzhou Panyu Polytechnic, Guangzhou 511483, China
| | - Lin Li
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510641, China
| | - Song-Qing Hu
- School of Food Science and Engineering, South China University of Technology, Guangzhou, Guangdong 510641, China; Overseas Expertise Introduction Center for Discipline Innovation of Food Nutrition and Human Health (111 Center), Guangzhou, China.
| |
Collapse
|
47
|
Nichenametla SN, Mattocks DAL, Malloy VL, Pinto JT. Sulfur amino acid restriction-induced changes in redox-sensitive proteins are associated with slow protein synthesis rates. Ann N Y Acad Sci 2018; 1418:80-94. [PMID: 29377163 DOI: 10.1111/nyas.13556] [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] [Received: 07/12/2017] [Revised: 10/02/2017] [Accepted: 10/27/2017] [Indexed: 12/29/2022]
Abstract
The mechanisms underlying life span extension by sulfur amino acid restriction (SAAR) are unclear. Cysteine and methionine are essential for the biosynthesis of proteins and glutathione (GSH), a major redox buffer in the endoplasmic reticulum (ER). We hypothesized that SAAR alters protein synthesis by modulating the redox milieu. Male F344-rats were fed control (CD: 0.86% methionine without cysteine) and SAAR diets (0.17% methionine without cysteine) for 12 weeks. Growth rates, food intake, cysteine and GSH levels, proteins associated with redox status and translation, and fractional protein synthesis rates (FSRs) were determined in liver. Despite a 40% higher food intake, growth rates for SAAR rats were 27% of those fed CD. Hepatic free cysteine in SAAR rats was 55% compared with CD rats. SAAR altered tissue distribution of GSH, as hepatic and erythrocytic levels were 56% and 196% of those in CD rats. Lower GSH levels did not induce ER stress (i.e., unchanged expression of Xbp1s , Chop, and Grp78), but activated PERK and its substrates eIF2-α and NRF2. SAAR-induced changes in translation-initiation machinery (higher p-eIF2-α and 4E-BP1, and lower eIF4G-1) resulted in slower protein synthesis rates (53% of CD). Proteins involved in the antioxidant response (NRF2, KEAP1, GCLM, and NQO1) and protein folding (PDI and ERO1-α) were increased in SAAR. Lower FSR and efficient protein folding might be improving proteostasis in SAAR.
Collapse
Affiliation(s)
| | | | - Virginia L Malloy
- Orentreich Foundation for the Advancement of Science, Cold Spring, New York
| | - John T Pinto
- Department of Biochemistry and Molecular Biology, New York Medical College, Valhalla, New York
| |
Collapse
|
48
|
Delaunay-Moisan A, Ponsero A, Toledano MB. Reexamining the Function of Glutathione in Oxidative Protein Folding and Secretion. Antioxid Redox Signal 2017; 27:1178-1199. [PMID: 28791880 DOI: 10.1089/ars.2017.7148] [Citation(s) in RCA: 33] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
SIGNIFICANCE Disturbance of glutathione (GSH) metabolism is a hallmark of numerous diseases, yet GSH functions are poorly understood. One key to this question is to consider its functional compartmentation. GSH is present in the endoplasmic reticulum (ER), where it competes with substrates for oxidation by the oxidative folding machinery, composed in eukaryotes of the thiol oxidase Ero1 and proteins from the disulfide isomerase family (protein disulfide isomerase). Yet, whether GSH is required for proper ER oxidative protein folding is a highly debated question. Recent Advances: Oxidative protein folding has been thoroughly dissected over the past decades, and its actors and their mode of action elucidated. Genetically encoded GSH probes have recently provided an access to subcellular redox metabolism, including the ER. CRITICAL ISSUES Of the few often-contradictory models of the role of GSH in the ER, the most popular suggest it serves as reducing power. Yet, as a reductant, GSH also activates Ero1, which questions how GSH can nevertheless support protein reduction. Hence, whether GSH operates in the ER as a reductant, an oxidant, or just as a "blank" compound mirroring ER/periplasm redox activity is a highly debated question, which is further stimulated by the puzzling occurrence of GSH in the Escherichia coli periplasmic "secretory" compartment, aside from the Dsb thiol-reducing and oxidase pathways. FUTURE DIRECTIONS Addressing the mechanisms controlling GSH traffic in and out of the ER/periplasm and its recycling will help address GSH function in secretion. In addition, as thioredoxin reductase was recently implicated in ER oxidative protein folding, the relative contribution of each of these two reducing pathways should now be addressed. Antioxid. Redox Signal. 27, 1178-1199.
Collapse
Affiliation(s)
- Agnès Delaunay-Moisan
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| | - Alise Ponsero
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| | - Michel B Toledano
- Institute for Integrative Biology of the Cell (I2BC), LSOC, SBIGEM, CEA, CNRS, Université Paris-Sud , Université Paris-Saclay, Gif-sur-Yvette, France
| |
Collapse
|
49
|
Ma WW, Zhao L, Yuan LH, Yu HL, Wang H, Gong XY, Wei F, Xiao R. Elaidic acid induces cell apoptosis through induction of ROS accumulation and endoplasmic reticulum stress in SH‑SY5Y cells. Mol Med Rep 2017; 16:9337-9346. [PMID: 29152653 PMCID: PMC5779995 DOI: 10.3892/mmr.2017.7830] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2016] [Accepted: 06/26/2017] [Indexed: 01/09/2023] Open
Abstract
Elaidic acid, which is a major trans fatty acid, has been reported to be involved in neurotoxicity; however, the underlying molecular mechanisms underlying its neurotoxic effects remain largely unknown. Therefore, the present study aimed to investigate the potential mechanisms underlying elaidic acid‑induced neuronal damage in vitro. The SH‑SY5Y neuroblastoma cell line was used as a model in the present study. Following treatment of cells with various concentrations of elaidic acid or with vehicle for 24 h, cell viability was measured using the MTT assay. Mitochondrial membrane potential (MMP) and reactive oxygen species (ROS) release were measured using flow cytometry. Cell apoptosis was measured by Annexin V‑fluorescein isothiocyanate/propidium iodide double staining, and cellular redox status was determined using ELISA analysis. Furthermore, western blotting was used to detect the protein expression levels of factors associated with oxidative damage and components of the endoplasmic reticulum (ER) stress/unfolded protein response (UPR) signaling pathways. The results demonstrated that elaidic acid treatment inhibited cell viability, elevated cell apoptosis and resulted in a loss of MMP. In addition, elaidic acid induced marked alterations in cellular redox status. Treatment with high doses of elaidic acid treatment also enhanced the release of ROS, and upregulated lipid peroxide and malondialdehyde levels; however, it reduced superoxide dismutase and glutathione peroxidase activities. Furthermore, elaidic acid resulted in upregulation of nuclear factor erythroid 2‑related factor 2 and downregulation of heme oxygenase 1, which are two key antioxidative factors. Elaidic acid treatment also induced or inhibited the expression of numerous ER stress/UPR‑associated molecules. It induced glucose‑regulated protein 78 (GRP78) expression, whereas the expression levels of activating transcription factor 4 (ATF4) and CCAAT/enhancer‑binding protein homologous protein (CHOP) were upregulated and then downregulated following treatment with various doses of elaidic acid. These results indicated that elaidic acid inhibited SH‑SY5Y cell growth and induced apoptosis by enhancing oxidative stress and activating the ER stress/UPR signaling pathway and the GRP78/ATF4/CHOP pathway.
Collapse
Affiliation(s)
- Wei-Wei Ma
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Lei Zhao
- Department of Molecular Physiology and Biophysics, Holden Comprehensive Cancer Center, University of Iowa Carver College of Medicine, Iowa City, IA 52242, USA
| | - Lin-Hong Yuan
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Huan-Ling Yu
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Hui Wang
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Xin-Yuan Gong
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Feng Wei
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| | - Rong Xiao
- Department of Nutrition and Food Hygiene, School of Public Health, Beijing Key Laboratory of Environmental Toxicology, Capital Medical University, Beijing 100069, P.R. China
| |
Collapse
|
50
|
Langsjoen RM, Auguste AJ, Rossi SL, Roundy CM, Penate HN, Kastis M, Schnizlein MK, Le KC, Haller SL, Chen R, Watowich SJ, Weaver SC. Host oxidative folding pathways offer novel anti-chikungunya virus drug targets with broad spectrum potential. Antiviral Res 2017; 143:246-251. [PMID: 28461071 DOI: 10.1016/j.antiviral.2017.04.014] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 04/05/2017] [Indexed: 11/15/2022]
Abstract
Alphaviruses require conserved cysteine residues for proper folding and assembly of the E1 and E2 envelope glycoproteins, and likely depend on host protein disulfide isomerase-family enzymes (PDI) to aid in facilitating disulfide bond formation and isomerization in these proteins. Here, we show that in human HEK293 cells, commercially available inhibitors of PDI or modulators thereof (thioredoxin reductase, TRX-R; endoplasmic reticulum oxidoreductin-1, ERO-1) inhibit the replication of CHIKV chikungunya virus (CHIKV) in vitro in a dose-dependent manner. Further, the TRX-R inhibitor auranofin inhibited Venezuelan equine encephalitis virus and the flavivirus Zika virus replication in vitro, while PDI inhibitor 16F16 reduced replication but demonstrated notable toxicity. 16F16 significantly altered the viral genome: plaque-forming unit (PFU) ratio of CHIKV in vitro without affecting relative intracellular viral RNA quantities and inhibited CHIKV E1-induced cell-cell fusion, suggesting that PDI inhibitors alter progeny virion infectivity through altered envelope function. Auranofin also increased the extracellular genome:PFU ratio but decreased the amount of intracellular CHIKV RNA, suggesting an alternative mechanism of action. Finally, auranofin reduced footpad swelling and viremia in the C57BL/6 murine model of CHIKV infection. Our results suggest that targeting oxidative folding pathways represents a potential new anti-alphavirus therapeutic strategy.
Collapse
Affiliation(s)
- Rose M Langsjoen
- Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Albert J Auguste
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Shannan L Rossi
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Christopher M Roundy
- Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA
| | - Heidy N Penate
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA
| | - Maria Kastis
- Center in Environmental Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | | | - Kevin C Le
- Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Sherry L Haller
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Rubing Chen
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA
| | - Stanley J Watowich
- Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Center in Environmental Toxicology, University of Texas Medical Branch, Galveston, TX, USA
| | - Scott C Weaver
- Institute for Translational Science, University of Texas Medical Branch, Galveston, TX, USA; Institute for Human Infections and Immunity, University of Texas Medical Branch, Galveston, TX, USA; Department of Pathology, University of Texas Medical Branch, Galveston, TX, USA; Department of Microbiology and Immunology, University of Texas Medical Branch, Galveston, TX, USA.
| |
Collapse
|