1
|
Han D, Sojic N, Jiang D. Spatial Profiling of Multiple Enzymatic Activities at Single Tissue Sections via Fenton-Promoted Electrochemiluminescence. J Am Chem Soc 2025; 147:9610-9619. [PMID: 40063963 DOI: 10.1021/jacs.4c17749] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/20/2025]
Abstract
Profiling multiple enzymatic activities in tissue is crucial for understanding complex metabolic and signaling networks, yet remains a challenge with existing optical microscopies. Here, we developed a Fenton-promoted luminol electrochemiluminescence (ECL) imaging method to achieve the spatial mapping of multiple enzymatic activities within a single tissue section. This method quantitatively visualizes individual enzymatic activity by combining the enzymatic conversion of substrates with the chemical confinement of the locally produced hydrogen peroxide. To achieve high-resolution spatial imaging by limiting the diffusion (∼500 μm) of hydrogen peroxide, iron oxide nanoparticles were coated on the tissue surface to initiate the Fenton process, locally converting hydrogen peroxide into short-lived hydroxyl radicals with a nanometer-scale diffusion range. The Fenton-promoted ECL emission is confined at the enzymatic conversion sites, offering unprecedented spatial visualization of four tumor-associated oxidases within a single tissue section. Colocalization revealed a synergistic effect between lysyl oxidase and quiescin sulfhydryl oxidase on post-translational modifications of tumor extracellular matrix proteins, along with a previously undiscovered interaction with amiloride-sensitive amine oxidase, which could not be distinguished based on expressions or single enzymatic activity alone. This approach offers a novel activity-based protein profiling tool at the tissue level, providing new data for future enzynomic research and multimodal imaging.
Collapse
Affiliation(s)
- Dongni Han
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| | - Neso Sojic
- University of Bordeaux, CNRS, Bordeaux INP, ISM, UMR, 5255, F-33400 Talence, France
| | - Dechen Jiang
- State Key Laboratory of Analytical Chemistry for Life Science and School of Chemistry and Chemical Engineering, Chemistry and Biomedicine Innovation Center (ChemBIC), Nanjing University, Nanjing 210093, China
| |
Collapse
|
2
|
Liu C, Liu Z, Dong Z, Liu S, Kan H, Zhang S. Multifaceted interplays between the essential players and lipid peroxidation in ferroptosis. J Genet Genomics 2025:S1673-8527(25)00024-4. [PMID: 39862922 DOI: 10.1016/j.jgg.2025.01.009] [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: 09/07/2024] [Revised: 01/17/2025] [Accepted: 01/17/2025] [Indexed: 01/27/2025]
Abstract
Ferroptosis, a type of programmed cell death, represents a distinct paradigm in cell biology. It is characterized by the iron-dependent accumulation of reactive oxygen species, which induce lipid peroxidation (LPO), and is orchestrated by the interplay between iron, lipid peroxides, and glutathione. In this review, we emphasize the frequently overlooked role of iron in LPO beyond the classical iron-driven Fenton reaction in several crucial processes that regulate cellular iron homeostasis, including iron intake and export as well as ferritinophagy, and the emerging roles of endoplasmic reticulum-resident flavoprotein oxidoreductases, especially P450 oxidoreductases, in modulating LPO. We summarize how various types of fatty acids (FAs), including saturated, monounsaturated, and polyunsaturated FAs, differentially influence ferroptosis when incorporated into phospholipids. Furthermore, we highlight the therapeutic potential of targeting LPO to mitigate ferroptosis and discuss the regulatory mechanisms of endogenous lipophilic radical-trapping antioxidants that confer resistance to ferroptosis, shedding light on therapeutic avenues for ferroptosis-associated diseases.
Collapse
Affiliation(s)
- Conghe Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zhihao Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; School of Public Health, Shandong First Medical University and Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Zheng Dong
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Sijin Liu
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China
| | - Haidong Kan
- School of Public Health, Key Lab of Public Health Safety of the Ministry of Education and NHC Key Lab of Health Technology Assessment, Shanghai Institute of Infectious Disease and Biosecurity, Fudan University, Shanghai 200032, China
| | - Shuping Zhang
- Medical Science and Technology Innovation Center, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China; Biomedical Sciences College & Shandong Medicinal Biotechnology Centre, Shandong First Medical University & Shandong Academy of Medical Sciences, Jinan, Shandong 250117, China.
| |
Collapse
|
3
|
Fan Z, Yang C, Qu X, Zhang J, Wu H, Yang Y, Huang Y, Zeng P, Xiang Z, Yang J. Association of Oxidative Stress on Cognitive Function: A Bidirectional Mendelian Randomisation Study. Mol Neurobiol 2024; 61:10551-10560. [PMID: 38753129 DOI: 10.1007/s12035-024-04231-3] [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: 02/29/2024] [Accepted: 05/10/2024] [Indexed: 11/24/2024]
Abstract
The purpose of this study was to investigate the relationship between oxidative stress and cognitive function, encompassing cognitive performance, intelligence, memory, reaction time, speech and vision by a bidirectional Mendelian randomisation study. Independent genetic variants associated with glutathione S-transferase (GST), catalase (CAT), superoxide dismutase (SOD), glutathione peroxidase (GPX), peroxiredoxin (PRDX), sulfhydryl oxidase (SOX) and thyroid peroxidase (TPO) were explored using a genome-wide association study (GWAS). The inverse variance weighted (IVW) or Wald ratio method was employed to ascertain the relationship between antioxidant enzymes and cognitive function. The MR analyses indicated that the MR effect estimates of GST (β = 0.0352, P = 0.0047, FDR = 0.0164) and TPO (β = 0.0531, P = 0.0003, FDR = 0.0021) were significantly associated with cognitive performance elevation. Furthermore, genetically predicted GST (β = 0.0334, P = 0.0043, FDR = 0.0151) and TPO (β = 0.0496, P = 0.0031, FDR = 0.0151) were found to be associated with high intelligence. Additionally, there were also some associations of SOX (β = 0.0243, P = 0.0283, FDR = 0.066) on high cognitive performance, TPO (β = 0.1189, P = 0.0315, FDR = 0.2205) on larger maximum digits remembered correctly, and SOX (β = - 0.2435, P = 0.0395, FDR = 0.1185) on reaction time. Nevertheless, the associations between antioxidant enzymes and speech and linguistic disorders, as well as visual disturbances, were not significant. We did not find reverse causation between antioxidant enzymes and cognitive function traits. This study provides evidence of potential causal relationships between oxidative stress and cognitive function.
Collapse
Affiliation(s)
- Zhixing Fan
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
- Department of Medical Record Management, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
| | - Chaojun Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Xiaoling Qu
- Department of Medical Record Management, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
| | - Jing Zhang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Hui Wu
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Ying Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Yifan Huang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Ping Zeng
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Zujin Xiang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China
| | - Jian Yang
- Department of Cardiology, The First College of Clinical Medical Sciences, China Three Gorges University, Yichang, 443003, China.
- Institute of Cardiovascular Diseases, Three Gorges University, Yichang, 443003, China.
| |
Collapse
|
4
|
Dutt R, Thorpe C, Galileo DS. QSOX1 Modulates Glioblastoma Cell Proliferation and Migration In Vitro and Invasion In Vivo. Cancers (Basel) 2024; 16:3620. [PMID: 39518060 PMCID: PMC11545231 DOI: 10.3390/cancers16213620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2024] [Revised: 10/16/2024] [Accepted: 10/21/2024] [Indexed: 11/16/2024] Open
Abstract
Background: Quiescin Sulfhydryl Oxidase 1 (QSOX1) is an enzyme that catalyzes the oxidation of free thiols to generate disulfide bonds in a variety of proteins, including the cell surface and extracellular matrix. QSOX1 has been reported to be upregulated in a number of cancers, and the overexpression of QSOX1 has been correlated with aggressive cancers and poor patient prognosis. Glioblastoma (GBM) brain cancer has been practically impossible to treat effectively, with cells that rapidly invade normal brain tissue and escape surgery and other treatment. Thus, there is a crucial need to understand the multiple mechanisms that facilitate GBM cell invasion and to determine if QSOX1 is involved. Methods and Results: Here, we investigated the function of QSOX1 in human glioblastoma cells using two cell lines derived from T98G cells, whose proliferation, motility, and invasiveness has been shown by us to be dependent on disulfide bond-containing adhesion and receptor proteins, such as L1CAM and the FGFR. We lentivirally introduced shRNA to attenuate the QSOX1 protein expression in one cell line, and a Western blot analysis confirmed the decreased QSOX1 expression. A DNA content/cell cycle analysis using flow cytometry revealed 27% fewer knockdown cells in the S-phase of the cell cycle, indicating a reduced proliferation. A cell motility analysis utilizing our highly quantitative SuperScratch time-lapse microscopy assay revealed that knockdown cells migrated more slowly, with a 45% decrease in migration velocity. Motility was partly rescued by the co-culture of knockdown cells with control cells, indicating a paracrine effect. Surprisingly, knockdown cells exhibited increased motility when assayed using a Transwell migration assay. Our novel chick embryo orthotopic xenograft model was used to assess the in vivo invasiveness of knockdown vs. control cells, and tumors developed from both cell types. However, fewer invasive knockdown cells were observed after about a week. Conclusions: Our results indicate that an experimental reduction in QSOX1 expression in GBM cells leads to decreased cell proliferation, altered in vitro migration, and decreased in vivo invasion.
Collapse
Affiliation(s)
- Reetika Dutt
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA; (R.D.); (C.T.)
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, USA; (R.D.); (C.T.)
| | - Deni S. Galileo
- Department of Biological Sciences, University of Delaware, Newark, DE 19716, USA
| |
Collapse
|
5
|
Hebchen DM, Schröder K. Redox Signaling in Endosomes Using the Example of EGF Receptors: A Graphical Review. Antioxidants (Basel) 2024; 13:1215. [PMID: 39456468 PMCID: PMC11504029 DOI: 10.3390/antiox13101215] [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: 09/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Early endosomes represent first-line sorting compartments or even organelles for internalized molecules. They enable the transport of molecules or ligands to other compartments of the cell, such as lysosomes, for degradation or recycle them back to the membrane by various mechanisms. Moreover, early endosomes function as signaling and scaffolding platforms to initiate or prolong distinct signaling pathways. Accordingly, early endosomes have to be recognized as either part of a degradation or recycling pathway. The physical proximity of many ligand-binding receptors with other membrane-bound proteins or complexes such as NADPH oxidases may result in an interaction of second messengers, like reactive oxygen species (ROS) and early endosomes, that promote the correct recognition of individual early endosomes. In fact, redoxosomes comprise an endosomal subsection of signaling endosomes. One example of such potential interaction is epidermal growth factor receptor (EGFR) signaling. Here we summarize recent findings on EGFR signaling as a well-studied example for receptor trafficking and trans-activation and illustrate the interplay between cellular and endosomal ROS.
Collapse
Affiliation(s)
| | - Katrin Schröder
- Institute of Physiology, Medical Faculty, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| |
Collapse
|
6
|
Pierre AS, Gavriel N, Guilbard M, Ogier-Denis E, Chevet E, Delom F, Igbaria A. Modulation of Protein Disulfide Isomerase Functions by Localization: The Example of the Anterior Gradient Family. Antioxid Redox Signal 2024; 41:675-692. [PMID: 38411504 DOI: 10.1089/ars.2024.0561] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/28/2024]
Abstract
Significance: Oxidative folding within the endoplasmic reticulum (ER) introduces disulfide bonds into nascent polypeptides, ensuring proteins' stability and proper functioning. Consequently, this process is critical for maintaining proteome integrity and overall health. The productive folding of thousands of secretory proteins requires stringent quality control measures, such as the unfolded protein response (UPR) and ER-Associated Degradation (ERAD), which contribute significantly to maintaining ER homeostasis. ER-localized protein disulfide isomerases (PDIs) play an essential role in each of these processes, thereby contributing to various aspects of ER homeostasis, including maintaining redox balance, proper protein folding, and signaling from the ER to the nucleus. Recent Advances: Over the years, there have been increasing reports of the (re)localization of PDI family members and other ER-localized proteins to various compartments. A prime example is the anterior gradient (AGR) family of PDI proteins, which have been reported to relocate to the cytosol or the extracellular environment, acquiring gain of functions that intersect with various cellular signaling pathways. Critical Issues: Here, we summarize the functions of PDIs and their gain or loss of functions in non-ER locations. We will focus on the activity, localization, and function of the AGR proteins: AGR1, AGR2, and AGR3. Future Directions: Targeting PDIs in general and AGRs in particular is a promising strategy in different human diseases. Thus, there is a need for innovative strategies and tools aimed at targeting PDIs; those strategies should integrate the specific localization and newly acquired functions of these PDIs rather than solely focusing on their canonical roles.
Collapse
Affiliation(s)
- Arvin S Pierre
- INSERM U1242, University of Rennes, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Noa Gavriel
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Marianne Guilbard
- ARTiSt Group, Univ. Bordeaux, INSERM U1312, Institut Bergonié, Bordeaux, France
- Thabor Therapeutics, Paris, France
| | - Eric Ogier-Denis
- INSERM U1242, University of Rennes, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Eric Chevet
- INSERM U1242, University of Rennes, Rennes, France
- Centre de Lutte Contre le Cancer Eugène Marquis, Rennes, France
| | - Frederic Delom
- ARTiSt Group, Univ. Bordeaux, INSERM U1312, Institut Bergonié, Bordeaux, France
| | - Aeid Igbaria
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| |
Collapse
|
7
|
Rafalska KT, Orzołek A, Ner-Kluza J, Wysocki P. A Comparison of White and Yellow Seminal Plasma Phosphoproteomes Obtained from Turkey ( Meleagris gallopavo) Semen. Int J Mol Sci 2024; 25:9941. [PMID: 39337428 PMCID: PMC11432639 DOI: 10.3390/ijms25189941] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2024] [Revised: 09/10/2024] [Accepted: 09/12/2024] [Indexed: 09/30/2024] Open
Abstract
Seminal plasma is rich in proteins originating from various male reproductive organs. The phosphorylation of these proteins can significantly impact sperm motility, capacitation, and acrosome reaction. Phosphoproteomics identifies, catalogues, and characterizes phosphorylated proteins. The phosphoproteomic profiling of seminal plasma offers valuable insights into the molecular mechanisms that influence semen quality and male fertility. Thus, the aim of this study was a phosphoproteomic analysis of white and yellow turkey seminal plasma. The experimental material consisted of 100 ejaculates from BIG-6 turkeys between 39 and 42 weeks of age. The collected white and yellow turkey seminal plasmas were analyzed for total protein content; the activity of selected enzymes, i.e., alkaline phosphatase (ALP), acid phosphatase (ACP), superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT); and the content of reduced glutathione (GSH) and malondialdehyde (MDA). Phosphoproteins were isolated from white and yellow seminal fluids, and the resulting protein fractions were separated by SDS-PAGE and Western blotting. Phosphorylated residues were immunodetected, and the isolated phosphoproteins were identified (nano LC-MS/MS). Yellow seminal plasmas were characterized by higher levels of total protein, GSH, and MDA, as well as higher levels of ALP, ACP, and GPx activity. There were no significant differences in the activity of SOD and CAT. A total of 113 phosphoproteins were identified in turkey seminal fluids. The functional analysis demonstrated that these phosphoproteins were mainly involved in oocyte fertilization, organization and metabolism of the actin cytoskeleton, amplification of the intracellular signal transduction pathway, general regulation of transport, vesicular transport, proteome composition of individual cellular compartments, and the organization and localization of selected cellular components and macromolecules. Increased phosphorylation of the fractions containing proteins encoded by SPARC, PPIB, TRFE, QSOX1, PRDX1, PRDX6, and FASN genes in white plasmas and the proteins encoded by CKB, ORM2, APOA1, SSC5D, RAP1B, CDC42, FTH, and TTH genes in yellow plasmas was observed based on differences in the optical density of selected bands. The obtained results indicate that the phosphorylation profiles of turkey seminal plasma proteins vary depending on the type of ejaculate.
Collapse
Affiliation(s)
- Katarzyna T Rafalska
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
| | - Aleksandra Orzołek
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
| | - Joanna Ner-Kluza
- Department of Biochemistry and Neurobiology, Faculty of Materials Science and Ceramics, AGH University, Mickiewicza 30, 30-059 Kraków, Poland
| | - Paweł Wysocki
- Department of Animal Biochemistry and Biotechnology, Faculty of Animal Bioengineering, University of Warmia and Mazury in Olsztyn, Oczapowskiego 5, 10-719 Olsztyn, Poland
| |
Collapse
|
8
|
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
|
9
|
Melo EP, El-Guendouz S, Correia C, Teodoro F, Lopes C, Martel PJ. A Conformational-Dependent Interdomain Redox Relay at the Core of Protein Disulfide Isomerase Activity. Antioxid Redox Signal 2024; 41:181-200. [PMID: 38497737 DOI: 10.1089/ars.2023.0288] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 03/19/2024]
Abstract
Aims: Protein disulfide isomerases (PDIs) are a family of chaperones resident in the endoplasmic reticulum (ER). In addition to holdase function, some members catalyze disulfide bond formation and isomerization, a crucial step for native folding and prevention of aggregation of misfolded proteins. PDIs are characterized by an arrangement of thioredoxin-like domains, with the canonical protein disulfide isomerase A1 (PDIA1) organized as four thioredoxin-like domains forming a horseshoe with two active sites, a and a', at the extremities. We aimed to clarify important aspects underlying the catalytic cycle of PDIA1 in the context of the full pathways of oxidative protein folding operating in the ER. Results: Using two fluorescent redox sensors, redox green fluorescent protein 2 (roGFP2) and HyPer (circularly permutated yellow fluorescent protein containing the regulatory domain of the H2O2-sensing protein OxyR), either unfolded or native, as client substrates, we identified the N-terminal a active site of PDIA1 as the main oxidant of thiols. From there, electrons can flow to the C-terminal a' active site, with the redox-dependent conformational flexibility of PDIA1 allowing the formation of an interdomain disulfide bond. The a' active site then acts as a crossing point to redirect electrons to ER downstream oxidases or back to client proteins to reduce scrambled disulfide bonds. Innovation and Conclusions: The two active sites of PDIA1 work cooperatively as an interdomain redox relay mechanism that explains PDIA1 oxidative activity to form native disulfides and PDIA1 reductase activity to resolve scrambled disulfides. This mechanism suggests a new rationale for shutting down oxidative protein folding under ER redox imbalance. Whether it applies to physiological substrates in cells remains to be shown.
Collapse
Affiliation(s)
- Eduardo P Melo
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | | | - Cátia Correia
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | - Fernando Teodoro
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | - Carlos Lopes
- Centro de Ciências do Mar (CCMAR), University of Algarve, Faro, Portugal
| | | |
Collapse
|
10
|
Kim AI, Oh JH, Cho JY. QSOX2 Upregulated in triple-negative breast cancer exacerbates patient prognosis by stabilizing integrin β1. Heliyon 2024; 10:e27148. [PMID: 38500982 PMCID: PMC10945127 DOI: 10.1016/j.heliyon.2024.e27148] [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: 10/31/2023] [Revised: 02/07/2024] [Accepted: 02/25/2024] [Indexed: 03/20/2024] Open
Abstract
Breast cancer (BC) remains a significant global health threat, with triple-negative breast cancer (TNBC) standing out as a particularly aggressive subtype lacking targeted therapies. Addressing this gap, we propose Quiescin Q6 sulfhydryl oxidase 2 (QSOX2) as a potential therapeutic target, a disulfide bond-forming enzyme implicated in cancer progression. Using publicly available datasets, we conducted a comprehensive analysis of QSOX2 expression in BC tumor and non-tumor tissues, assessing its specificity across different molecular subtypes. We further explored correlations between QSOX2 expression and patient outcomes, utilizing datasets like TCGA and METABRIC. In addition, we performed in vitro experiments to evaluate QSOX2 expression in BC cell lines and investigate the effects of QSOX2 knockdown on various TNBC cellular processes, including cell proliferation, apoptosis resistance, migration, and the epithelial-to-mesenchymal transition (EMT). Our results reveal significantly elevated QSOX2 expression in BC tumor tissues, particularly in TNBC, and establish an association between high QSOX2 expression and increased patient mortality, cancer progression, and recurrence across various BC subtypes. Notably, QSOX2 knockdown in TNBC cell lines reduces cell proliferation, enhances apoptosis, and suppresses migration, potentially mediated through its influence on the EMT process. Furthermore, we identify a significant link between QSOX2 and integrin β1 (ITGB1), suggesting that QSOX2 enhances ITGB1 stability, subsequently exacerbating the malignancy of TNBC. In conclusion, elevated QSOX2 expression emerges as a key factor associated with adverse patient outcomes in BC, particularly in TNBC, contributing to disease progression through various mechanisms, including the modulation of ITGB1 stability. Our findings underscore the potential of targeting QSOX2 as a therapeutic strategy for improving patient prognoses not only in TNBC but also in other BC subtypes.
Collapse
Affiliation(s)
- A-In Kim
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Ji Hoon Oh
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| | - Je-Yoel Cho
- Department of Biochemistry, Brain Korea 21 Project and Research Institute for Veterinary Science, Seoul National University College of Veterinary Medicine, Seoul, 08826, Republic of Korea
- Comparative Medicine Disease Research Center, Seoul National University, Seoul, 08826, Republic of Korea
| |
Collapse
|
11
|
Mazepa E, Furlanetto ALDDM, Brum H, Nakao LS, Martinez PA, Cadena SMSC, Rocha MEM, Cunha ES, Martinez GR. Effects of redox modulation on quiescin/sulfhydryl oxidase activity of melanoma cells. Mol Cell Biochem 2024; 479:511-524. [PMID: 37103678 DOI: 10.1007/s11010-023-04745-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2023] [Accepted: 04/14/2023] [Indexed: 04/28/2023]
Abstract
Secreted quiescin/sulfhydryl oxidase (QSOX) is overexpressed in many tumor cell lines, including melanoma, and is usually associated with a pro-invasive phenotype. Our previous work described that B16-F10 cells enter in a quiescent state as a protective mechanism against damage generated by reactive oxygen species (ROS) during melanogenesis stimulation. Our present results show that QSOX activity was two-fold higher in cells with stimulated melanogenesis when compared to control cells. Considering that glutathione (GSH) is one of the main factor responsible for controlling redox homeostasis in cells, this work also aimed to investigate the relationship between QSOX activity, GSH levels and melanogenesis stimulation in B16-F10 murine melanoma cell line. The redox homeostasis was impaired by treating cells with GSH in excess or depleting its intracellular levels through BSO treatment. Interestingly, GSH-depleted cells without stimulation of melanogenesis kept high levels of viability, suggesting a possible adaptive mechanism of survival even under low GSH levels. They also showed lower extracellular activity of QSOX, and higher QSOX intracellular immunostaining, suggesting that this enzyme was less excreted from cells and corroborating with a diminished extracellular QSOX activity. On the other hand, cells under melanogenesis stimulation showed a lower GSH/GSSG ratio (8:1) in comparison with control (non-stimulated) cells (20:1), indicating a pro-oxidative state after stimulation. This was accompanied by decreased cell viability after GSH-depletion, no alterations in QSOX extracellular activity, but higher QSOX nucleic immunostaining. We suggest that melanogenesis stimulation and redox impairment caused by GSH-depletion enhanced the oxidative stress in these cells, contributing to additional alterations of its metabolic adaptive response.
Collapse
Affiliation(s)
- Ester Mazepa
- Postgraduate Program in Sciences (Biochemistry), Department of Biochemistry and Molecular Biology, UFPR, Curitiba, PR, Brazil
| | | | - Hulyana Brum
- Postgraduate Program in Sciences (Biochemistry), Department of Biochemistry and Molecular Biology, UFPR, Curitiba, PR, Brazil
| | | | | | | | - Maria Eliane Merlin Rocha
- Postgraduate Program in Sciences (Biochemistry), Department of Biochemistry and Molecular Biology, UFPR, Curitiba, PR, Brazil
| | - Elizabeth Sousa Cunha
- Postgraduate Program in Sciences (Biochemistry), Department of Biochemistry and Molecular Biology, UFPR, Curitiba, PR, Brazil
| | - Glaucia Regina Martinez
- Postgraduate Program in Sciences (Biochemistry), Department of Biochemistry and Molecular Biology, UFPR, Curitiba, PR, Brazil.
| |
Collapse
|
12
|
Koelbel C, Ruiz Y, Wan Z, Wang S, Ho T, Lake D. Development of tandem antigen capture ELISAs measuring QSOX1 isoforms in plasma and serum. Free Radic Biol Med 2024; 210:212-220. [PMID: 38036070 PMCID: PMC10843750 DOI: 10.1016/j.freeradbiomed.2023.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 10/12/2023] [Accepted: 11/20/2023] [Indexed: 12/02/2023]
Abstract
QSOX1 is a sulfhydryl oxidase that has been identified as a potential biomarker in multiple cancer types as well as acute decompensated heart failure. Three anti-QSOX1 monoclonal antibodies (mAbs) were generated: 2F1, 3A10, and 56-3. MAbs 2F1 and 3A10 were generated against the short isoform of recombinant QSOX1 (rQSOX1-S), and mAb 56-3 was generated against a peptide (NEQEQPLGQWHLS) from the long isoform of QSOX1 (QSOX1-L). Using these mAbs, tandem antigen capture ELISAs were developed to quantify both short and long isoforms of QSOX1 (Total QSOX1 ELISA) and QSOX1-L (QSOX1-L ELISA) in serum and plasma samples. The Total QSOX1 ELISA pairs mAbs 2F1 and 3A10 and has a limit of detection of 109.5 pM, while the QSOX1-L ELISA pairs mAbs 2F1 and 56-3 and has a limit of detection of 10 pM. The levels of total QSOX1 and QSOX1-L were measured in a cohort of paired sera and plasma from 61 donors ≥40 years old and 15 donors <40 years old. No difference in QSOX1 levels was detected between QSOX1-L and QSOX1-S in serum, but the mean concentration of QSOX1-L was found to be 3.21 nM in serum and 5.63 nM in plasma (**p = 0.006). Our tandem ELISAs demonstrate the wide range of concentrations of QSOX1-L and QSOX1-S among individual serum and plasma samples. Since the epitope of mAb 2F1 was mapped to the first CxxC motif at residues C70 and C73 and mAb 56-3 was generated against NEQEQPLGQWHLS in QSOX1-L, our findings support previous research which suggested that QSOX1-L is secreted from cells despite a putative transmembrane domain. The ELISAs reported here may be a useful tool for investigating QSOX1 isoforms as potential biomarkers in cancer and/or heart failure.
Collapse
Affiliation(s)
- Calvin Koelbel
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Yvette Ruiz
- School of Life Sciences, Arizona State University, Tempe, AZ, USA
| | - Zijian Wan
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, USA
| | - Shaopeng Wang
- Biodesign Center for Bioelectronics and Biosensors, Arizona State University, Tempe, AZ, USA; School of Biological and Health Systems Engineering, Arizona State University, Tempe, AZ, USA
| | - Thai Ho
- Divison of Hematology and Medical Oncology, Hollings Cancer Center, Medical University of South Carolina College of Medicine, Charleston, SC, USA
| | - Douglas Lake
- School of Life Sciences, Arizona State University, Tempe, AZ, USA.
| |
Collapse
|
13
|
Ong G, Logue SE. Unfolding the Interactions between Endoplasmic Reticulum Stress and Oxidative Stress. Antioxidants (Basel) 2023; 12:antiox12050981. [PMID: 37237847 DOI: 10.3390/antiox12050981] [Citation(s) in RCA: 50] [Impact Index Per Article: 25.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2023] [Revised: 04/16/2023] [Accepted: 04/19/2023] [Indexed: 05/28/2023] Open
Abstract
Oxidative stress is caused by an imbalance in cellular redox state due to the accumulation of reactive oxygen species (ROS). While homeostatic levels of ROS are important for cell physiology and signaling, excess ROS can induce a variety of negative effects ranging from damage to biological macromolecules to cell death. Additionally, oxidative stress can disrupt the function of redox-sensitive organelles including the mitochondria and endoplasmic reticulum (ER). In the case of the ER, the accumulation of misfolded proteins can arise due to oxidative stress, leading to the onset of ER stress. To combat ER stress, cells initiate a highly conserved stress response called the unfolded protein response (UPR). While UPR signaling, within the context of resolving ER stress, is well characterised, how UPR mediators respond to and influence oxidative stress is less defined. In this review, we evaluate the interplay between oxidative stress, ER stress and UPR signaling networks. Specifically, we assess how UPR signaling mediators can influence antioxidant responses.
Collapse
Affiliation(s)
- Gideon Ong
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
| | - Susan E Logue
- Department of Human Anatomy and Cell Science, Rady Faculty of Health Sciences, University of Manitoba, Winnipeg, MB R3E 0J9, Canada
- CancerCare Manitoba Research Institute, Winnipeg, MB R3E 0V9, Canada
- The Children's Hospital Research Institute of Manitoba (CHRIM), Winnipeg, MB R3E 3P4, Canada
| |
Collapse
|
14
|
Resende R, Fernandes T, Pereira AC, Marques AP, Pereira CF. Endoplasmic Reticulum-Mitochondria Contacts Modulate Reactive Oxygen Species-Mediated Signaling and Oxidative Stress in Brain Disorders: The Key Role of Sigma-1 Receptor. Antioxid Redox Signal 2022; 37:758-780. [PMID: 35369731 DOI: 10.1089/ars.2020.8231] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
Abstract
Significance: Mitochondria-Associated Membranes (MAMs) are highly dynamic endoplasmic reticulum (ER)-mitochondria contact sites that, due to the transfer of lipids and Ca2+ between these organelles, modulate several physiologic processes, such as ER stress response, mitochondrial bioenergetics and fission/fusion events, autophagy, and inflammation. In addition, these contacts are implicated in the modulation of the cellular redox status since several MAMs-resident proteins are involved in the generation of reactive oxygen species (ROS), which can act as both signaling mediators and deleterious molecules, depending on their intracellular levels. Recent Advances: In the past few years, structural and functional alterations of MAMs have been associated with the pathophysiology of several neurodegenerative diseases that are closely associated with the impairment of several MAMs-associated events, including perturbation of the redox state on the accumulation of high ROS levels. Critical Issues: Inter-organelle contacts must be tightly regulated to preserve cellular functioning by maintaining Ca2+ and protein homeostasis, lipid metabolism, mitochondrial dynamics and energy production, as well as ROS signaling. Simultaneously, these contacts should avoid mitochondrial Ca2+ overload, which might lead to energetic deficits and deleterious ROS accumulation, culminating in oxidative stress-induced activation of apoptotic cell death pathways, which are common features of many neurodegenerative diseases. Future Directions: Given that Sig-1R is an ER resident chaperone that is highly enriched at the MAMs and that controls ER to mitochondria Ca2+ flux, as well as oxidative and ER stress responses, its potential as a therapeutic target for neurodegenerative diseases such as Amyotrophic Lateral Sclerosis, Alzheimer, Parkinson, and Huntington diseases should be further explored. Antioxid. Redox Signal. 37, 758-780.
Collapse
Affiliation(s)
- Rosa Resende
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Tânia Fernandes
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Catarina Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Patrícia Marques
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Cláudia Fragão Pereira
- Center for Neuroscience and Cell Biology, Centre for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| |
Collapse
|
15
|
Critical roles of protein disulfide isomerases in balancing proteostasis in the nervous system. J Biol Chem 2022; 298:102087. [PMID: 35654139 PMCID: PMC9253707 DOI: 10.1016/j.jbc.2022.102087] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2021] [Revised: 05/05/2022] [Accepted: 05/08/2022] [Indexed: 02/08/2023] Open
Abstract
Protein disulfide isomerases (PDIs) constitute a family of oxidoreductases promoting redox protein folding and quality control in the endoplasmic reticulum. PDIs catalyze disulfide bond formation, isomerization, and reduction, operating in concert with molecular chaperones to fold secretory cargoes in addition to directing misfolded proteins to be refolded or degraded. Importantly, PDIs are emerging as key components of the proteostasis network, integrating protein folding status with central surveillance mechanisms to balance proteome stability according to cellular needs. Recent advances in the field driven by the generation of new mouse models, human genetic studies, and omics methodologies, in addition to interventions using small molecules and gene therapy, have revealed the significance of PDIs to the physiology of the nervous system. PDIs are also implicated in diverse pathologies, ranging from neurodevelopmental conditions to neurodegenerative diseases and traumatic injuries. Here, we review the principles of redox protein folding in the ER with a focus on current evidence linking genetic mutations and biochemical alterations to PDIs in the etiology of neurological conditions.
Collapse
|
16
|
Gansemer ER, Rutkowski DT. Pathways Linking Nicotinamide Adenine Dinucleotide Phosphate Production to Endoplasmic Reticulum Protein Oxidation and Stress. Front Mol Biosci 2022; 9:858142. [PMID: 35601828 PMCID: PMC9114485 DOI: 10.3389/fmolb.2022.858142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum (ER) lumen is highly oxidizing compared to other subcellular compartments, and maintaining the appropriate levels of oxidizing and reducing equivalents is essential to ER function. Both protein oxidation itself and other essential ER processes, such as the degradation of misfolded proteins and the sequestration of cellular calcium, are tuned to the ER redox state. Simultaneously, nutrients are oxidized in the cytosol and mitochondria to power ATP generation, reductive biosynthesis, and defense against reactive oxygen species. These parallel needs for protein oxidation in the ER and nutrient oxidation in the cytosol and mitochondria raise the possibility that the two processes compete for electron acceptors, even though they occur in separate cellular compartments. A key molecule central to both processes is NADPH, which is produced by reduction of NADP+ during nutrient catabolism and which in turn drives the reduction of components such as glutathione and thioredoxin that influence the redox potential in the ER lumen. For this reason, NADPH might serve as a mediator linking metabolic activity to ER homeostasis and stress, and represent a novel form of mitochondria-to-ER communication. In this review, we discuss oxidative protein folding in the ER, NADPH generation by the major pathways that mediate it, and ER-localized systems that can link the two processes to connect ER function to metabolic activity.
Collapse
Affiliation(s)
- Erica R. Gansemer
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - D. Thomas Rutkowski
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
17
|
Wang TE, Yeh LY, Kuo-Kuang Lee R, Lu CH, Yang TH, Kuo YW, Joshi R, Tsai PS, Li SH. Secretory mouse quiescin sulfhydryl oxidase 1 aggregates defected human and mouse spermatozoa in vitro and in vivo. iScience 2021; 24:103167. [PMID: 34667943 PMCID: PMC8506963 DOI: 10.1016/j.isci.2021.103167] [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: 03/25/2021] [Revised: 07/08/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
A flavin-dependent enzyme quiescin Q6 sulfhydryl oxidase 1 (QSOX1) catalyzes the oxidation of thiol groups into disulfide bonds. QSOX1 is prominently expressed in the seminal plasma. However, its role in male reproduction is elusive. Here, we purified the secreted form of QSOX1, i.e., QSOX1c, from mouse seminal vesicle secretions and revealed for the first time its function involved in sperm physiology. Exogenous addition of QSOX1c time-dependently promoted the in vitro aggregation of thiol-rich, oxidative stressed, and apoptotic mouse and human sperm cells. Also, in vivo aggregated sperm cells collected from mouse uterine and human ejaculates also showed high levels of QSOX1c, intracellular reactive oxygen species, annexin V, and free thiols. In summary, our studies demonstrated that QSOX1c could agglutinate spermatozoa susceptible to free radical attack and apoptosis. This characteristic may provide an opportunity to separate defective sperm cells and improve sperm quality before artificial insemination in humans and animals. QSOX1c is expressed in the seminal vesicle and presented in the seminal plasma QSOX1c agglutinates thiol-rich, oxidatively stressed, and apoptotic sperm QSOX1c aggregates impaired sperm presented in the mouse uterine and human ejaculates QSOX1c-treated semen may improve the sperm quality for artificial insemination
Collapse
Affiliation(s)
- Tse-En Wang
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Cellular and Molecular Physiology, Yale School of Medicine, New Haven, USA
| | - Ling-Yu Yeh
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Robert Kuo-Kuang Lee
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Chung-Hao Lu
- Department of Obstetrics and Gynecology, MacKay Memorial Hospital, Taipei, Taiwan
| | - Tsung-Hsien Yang
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan
| | - Yu-Wen Kuo
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Department of Clinical Sciences, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Radhika Joshi
- Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan
| | - Pei-Shiue Tsai
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, Taipei, Taiwan.,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, MacKay Memorial Hospital, Tamsui, Taiwan.,MacKay Junior College of Medicine, Nursing, and Management, Taipei, Taiwan
| |
Collapse
|
18
|
Nazempour N, Taleqani MH, Taheri N, Haji Ali Asgary Najafabadi AH, Shokrollahi A, Zamani A, Fattahi Dolatabadi N, Peymani M, Mahdevar M. The role of cell surface proteins gene expression in diagnosis, prognosis, and drug resistance of colorectal cancer: In silico analysis and validation. Exp Mol Pathol 2021; 123:104688. [PMID: 34592197 DOI: 10.1016/j.yexmp.2021.104688] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2021] [Revised: 09/21/2021] [Accepted: 09/22/2021] [Indexed: 12/13/2022]
Abstract
Cell surface proteins (CSPs) are an important type of protein in different essential cell functions. This study aimed to distinguish overexpressed CSPs in colorectal cancer to investigate their biomarker, prognosis, and drug resistance potential. Raw data of three datasets including 1187 samples was downloaded then normalization and differential expression were performed. By the combination of the cancer genome atlas (TCGA) clinical data, survival analysis was carried out. Information of all CSPs was collected from cell surface protein atlas. The role of each candidate gene expression was investigated in drug resistance by CCEL and GDSC data from PharmacoGX. CRC samples including 30 tumor samples and adjacent normal were used to confirm data by RT-qPCR. Outcomes showed that 66 CSPs overexpressed in three datasets, and 146 CSPs expression associated with poor prognosis features in TCGA data that TIMP1 and QSOX2 can associate with poor patient survival independently. High-risk patients illustrated more fatality than low-risk patients based on the risk score calculated by the expression level of these genes. Receiver operating characteristic curve analysis showed that 39 CSPs as perfect biomarkers for diagnosis in CRC. Furthermore, QSOX2 and TIMP1 expression levels increased in tumor samples compared to adjacent normal samples. The Drug resistance analysis demonstrated ADAM12 and COL1A2 up-regulation among 66 overexpressed CSPs caused resistance to Venetoclax and Cyclophosphamide with a high estimate, respectively. Many CSPs are deregulated in CRC, and can be valuable candidates as biomarkers for diagnosis, prognosis, and drug resistance.
Collapse
Affiliation(s)
- Nasrin Nazempour
- Department of Chemistry, Shahreza Branch, Islamic Azad University, Shahreza, Isfahan, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | - Mohammad Hossein Taleqani
- Department of Biology, Faculty of Science, University of Yazd, Yazd, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | - Navid Taheri
- Department of Microbiology, Zanjan Branch, Islamic Azad University, Zanjan, Iran; Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | | | - Alireza Shokrollahi
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| | - Atefeh Zamani
- Gene Raz Bu Ali, Genetic and Biotechnology Academy, Isfahan, Iran
| | | | - Maryam Peymani
- Department of Biology, Faculty of Basic Sciences, Shahrekord Branch, Islamic Azad University, Sharekord, Iran.
| | - Mohammad Mahdevar
- Department of Cell and Molecular Biology and Microbiology, Faculty of Biological Science and Technology, University of Isfahan, Isfahan, Iran
| |
Collapse
|
19
|
Redox and Inflammatory Signaling, the Unfolded Protein Response, and the Pathogenesis of Pulmonary Hypertension. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1304:333-373. [PMID: 34019276 DOI: 10.1007/978-3-030-68748-9_17] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Protein folding overload and oxidative stress disrupt endoplasmic reticulum (ER) homeostasis, generating reactive oxygen species (ROS) and activating the unfolded protein response (UPR). The altered ER redox state induces further ROS production through UPR signaling that balances the cell fates of survival and apoptosis, contributing to pulmonary microvascular inflammation and dysfunction and driving the development of pulmonary hypertension (PH). UPR-induced ROS production through ER calcium release along with NADPH oxidase activity results in endothelial injury and smooth muscle cell (SMC) proliferation. ROS and calcium signaling also promote endothelial nitric oxide (NO) synthase (eNOS) uncoupling, decreasing NO production and increasing vascular resistance through persistent vasoconstriction and SMC proliferation. C/EBP-homologous protein further inhibits eNOS, interfering with endothelial function. UPR-induced NF-κB activity regulates inflammatory processes in lung tissue and contributes to pulmonary vascular remodeling. Conversely, UPR-activated nuclear factor erythroid 2-related factor 2-mediated antioxidant signaling through heme oxygenase 1 attenuates inflammatory cytokine levels and protects against vascular SMC proliferation. A mutation in the bone morphogenic protein type 2 receptor (BMPR2) gene causes misfolded BMPR2 protein accumulation in the ER, implicating the UPR in familial pulmonary arterial hypertension pathogenesis. Altogether, there is substantial evidence that redox and inflammatory signaling associated with UPR activation is critical in PH pathogenesis.
Collapse
|
20
|
The aftermath of the interplay between the endoplasmic reticulum stress response and redox signaling. Exp Mol Med 2021; 53:151-167. [PMID: 33558590 PMCID: PMC8080639 DOI: 10.1038/s12276-021-00560-8] [Citation(s) in RCA: 161] [Impact Index Per Article: 40.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Accepted: 12/14/2020] [Indexed: 02/07/2023] Open
Abstract
The endoplasmic reticulum (ER) is an essential organelle of eukaryotic cells. Its main functions include protein synthesis, proper protein folding, protein modification, and the transportation of synthesized proteins. Any perturbations in ER function, such as increased demand for protein folding or the accumulation of unfolded or misfolded proteins in the ER lumen, lead to a stress response called the unfolded protein response (UPR). The primary aim of the UPR is to restore cellular homeostasis; however, it triggers apoptotic signaling during prolonged stress. The core mechanisms of the ER stress response, the failure to respond to cellular stress, and the final fate of the cell are not yet clear. Here, we discuss cellular fate during ER stress, cross talk between the ER and mitochondria and its significance, and conditions that can trigger ER stress response failure. We also describe how the redox environment affects the ER stress response, and vice versa, and the aftermath of the ER stress response, integrating a discussion on redox imbalance-induced ER stress response failure progressing to cell death and dynamic pathophysiological changes. The endoplasmic reticulum (ER), a cellular organelle responsible for protein folding, is sensitive to chemical imbalances that can induce stress, leading to cell death and disease. Researchers in South Korea, led by Han-Jung Chae from Jeonbuk National University in Jeonju and Hyung-Ryong Kim from Dankook University in Cheonan, review how the ER counters changes in its environment that spur protein folding defects by activating a series of signaling pathways, known collectively as the unfolded protein response. Redox imbalance, may fail adaptive ER stress response that can damage the ER and surrounding mitochondria by modifying cysteine residues. The interaction between the two stress systems, ER stress and oxidative stress, has profound negative impacts on normal physiology. Targeting one or both of these stress mechanisms may therefore be an effective means of treating disease.
Collapse
|
21
|
Autophagy and Redox Homeostasis in Parkinson's: A Crucial Balancing Act. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2020; 2020:8865611. [PMID: 33224433 PMCID: PMC7671810 DOI: 10.1155/2020/8865611] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 09/23/2020] [Accepted: 10/14/2020] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) and reactive nitrogen species (RNS) are generated primarily from endogenous biochemical reactions in mitochondria, endoplasmic reticulum (ER), and peroxisomes. Typically, ROS/RNS correlate with oxidative damage and cell death; however, free radicals are also crucial for normal cellular functions, including supporting neuronal homeostasis. ROS/RNS levels influence and are influenced by antioxidant systems, including the catabolic autophagy pathways. Autophagy is an intracellular lysosomal degradation process by which invasive, damaged, or redundant cytoplasmic components, including microorganisms and defunct organelles, are removed to maintain cellular homeostasis. This process is particularly important in neurons that are required to cope with prolonged and sustained operational stress. Consequently, autophagy is a primary line of protection against neurodegenerative diseases. Parkinson's is caused by the loss of midbrain dopaminergic neurons (mDANs), resulting in progressive disruption of the nigrostriatal pathway, leading to motor, behavioural, and cognitive impairments. Mitochondrial dysfunction, with associated increases in oxidative stress, and declining proteostasis control, are key contributors during mDAN demise in Parkinson's. In this review, we analyse the crosstalk between autophagy and redoxtasis, including the molecular mechanisms involved and the detrimental effect of an imbalance in the pathogenesis of Parkinson's.
Collapse
|
22
|
Tanaka LY, Oliveira PVS, Laurindo FRM. Peri/Epicellular Thiol Oxidoreductases as Mediators of Extracellular Redox Signaling. Antioxid Redox Signal 2020; 33:280-307. [PMID: 31910038 DOI: 10.1089/ars.2019.8012] [Citation(s) in RCA: 26] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Significance: Supracellular redox networks regulating cell-extracellular matrix (ECM) and organ system architecture merge with structural and functional (catalytic or allosteric) properties of disulfide bonds. This review addresses emerging evidence that exported thiol oxidoreductases (TORs), such as thioredoxin, protein disulfide isomerases (PDIs), quiescin sulfhydryl oxidases (QSOX)1, and peroxiredoxins, composing a peri/epicellular (pec)TOR pool, mediate relevant signaling. pecTOR functions depend mainly on kinetic and spatial regulation of thiol-disulfide exchange reactions governed by redox potentials, which are modulated by exported intracellular low-molecular-weight thiols, together conferring signal specificity. Recent Advances: pecTOR redox-modulates several targets including integrins, ECM proteins, surface molecules, and plasma components, although clear-cut documentation of direct effects is lacking in many cases. TOR catalytic pathways, displaying common patterns, culminate in substrate thiol reduction, oxidation, or isomerization. Peroxiredoxins act as redox/peroxide sensors, contrary to PDIs, which are likely substrate-targeted redox modulators. Emerging evidence suggests important pecTOR roles in patho(physio)logical processes, including blood coagulation, vascular remodeling, mechanosensing, endothelial function, immune responses, and inflammation. Critical Issues: Effects of pecPDIs supporting thrombosis/platelet activation have been well documented and reached the clinical arena. Roles of pecPDIA1 in vascular remodeling/mechanosensing are also emerging. Extracellular thioredoxin and pecPDIs redox-regulate immunoinflammation. Routes of TOR externalization remain elusive and appear to involve Golgi-independent routes. pecTORs are particularly accessible drug targets. Future Directions: Further understanding mechanisms of thiol redox reactions and developing assays for assessing pecTOR redox activities remain important research avenues. Also, addressing pecTORs as disease markers and achieving more efficient/specific drugs for pecTOR modulation are major perspectives for diagnostic/therapeutic improvements.
Collapse
Affiliation(s)
- Leonardo Y Tanaka
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Percillia V S Oliveira
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| | - Francisco R M Laurindo
- Vascular Biology Laboratory, LIM-64 (Translational Cardiovascular Biology), Instituto do Coracao (InCor), Hospital das Clinicas HCFMUSP, Faculdade de Medicina, Universidade de Sao Paulo, Sao Paulo, Brazil
| |
Collapse
|
23
|
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
|
24
|
Riboflavin Deficiency-Implications for General Human Health and Inborn Errors of Metabolism. Int J Mol Sci 2020; 21:ijms21113847. [PMID: 32481712 PMCID: PMC7312377 DOI: 10.3390/ijms21113847] [Citation(s) in RCA: 93] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Revised: 05/20/2020] [Accepted: 05/26/2020] [Indexed: 01/13/2023] Open
Abstract
As an essential vitamin, the role of riboflavin in human diet and health is increasingly being highlighted. Insufficient dietary intake of riboflavin is often reported in nutritional surveys and population studies, even in non-developing countries with abundant sources of riboflavin-rich dietary products. A latent subclinical riboflavin deficiency can result in a significant clinical phenotype when combined with inborn genetic disturbances or environmental and physiological factors like infections, exercise, diet, aging and pregnancy. Riboflavin, and more importantly its derivatives, flavin mononucleotide (FMN) and flavin adenine dinucleotide (FAD), play a crucial role in essential cellular processes including mitochondrial energy metabolism, stress responses, vitamin and cofactor biogenesis, where they function as cofactors to ensure the catalytic activity and folding/stability of flavoenzymes. Numerous inborn errors of flavin metabolism and flavoenzyme function have been described, and supplementation with riboflavin has in many cases been shown to be lifesaving or to mitigate symptoms. This review discusses the environmental, physiological and genetic factors that affect cellular riboflavin status. We describe the crucial role of riboflavin for general human health, and the clear benefits of riboflavin treatment in patients with inborn errors of metabolism.
Collapse
|
25
|
França KC, Martinez PA, Prado ML, Lo SM, Borges BE, Zanata SM, San Martin A, Nakao LS. Quiescin/sulfhydryl oxidase 1b (QSOX1b) induces migration and proliferation of vascular smooth muscle cells by distinct redox pathways. Arch Biochem Biophys 2020; 679:108220. [PMID: 31812669 DOI: 10.1016/j.abb.2019.108220] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2019] [Revised: 12/04/2019] [Accepted: 12/04/2019] [Indexed: 01/24/2023]
Abstract
Quiescent and contractile VSMC can switch to proliferative and migratory phenotype in response to growth factors and cytokines, an effect underscored by Nox family NADPH oxidases, particularly Nox1. We previously showed that quiescin/sulfhydryl oxidase 1 (QSOX1) has a role in neointima formation in balloon-injured rat carotid. Here, we investigated the intracellular redox mechanisms underlying these effects in primary VSMC. Our results show that exogenous incubation with wild type QSOX1b (wt QSOX), or with secreted QSOX1, but not with the inactive C452S QSOX 1b (C452S QSOX) or secreted inactive C455S QSOX1, induces VSMC migration and chemotaxis. PEG-catalase (PEG-CAT) prevented, while PEG-superoxide dismutase (PEG-SOD) increased migration induced by wt QSOX. Moreover, wt QSOX-induced migration was abrogated in NOX1-null VSMC. In contrast, both wt QSOX and C452S QSOX, and both secreted QSOX1 and C455S QSOX1, induce cell proliferation. Such effect was unaltered by PEG-CAT, while being inhibited by PEG-SOD. However, QSOX1-induced proliferation was not significantly affected in NOX1-null VSMC, compared with WT VSMC. These results indicate that hydrogen peroxide and superoxide mediate, respectively, migration and proliferation. However, Nox1 was required only for QSOX1-induced migration. In parallel, QSOX1-induced proliferation was independent of its redox activity, although mediated by intracellular superoxide.
Collapse
Affiliation(s)
- Karime C França
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Pierina A Martinez
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Maiara L Prado
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Sze M Lo
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Beatriz E Borges
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | - Silvio M Zanata
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil
| | | | - Lia S Nakao
- Department of Basic Pathology, Universidade Federal Do Paraná, Curitiba, PR, 81531-980, Brazil.
| |
Collapse
|
26
|
Bayse CA, Pollard DB. Conformation dynamics of cyclic disulfides and selenosulfides in CXXC(U) (X = Gly, Ala) tetrapeptide redox motifs. J Pept Sci 2019; 25:e3160. [PMID: 30873692 DOI: 10.1002/psc.3160] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2018] [Revised: 01/26/2019] [Accepted: 02/05/2019] [Indexed: 02/01/2023]
Abstract
Thioredoxin fold proteins often contain a Cys-(Xxx)n -Cys(Sec) or CXn C(U) motif, where the active cysteine (C) or selenocysteine (U) is bridged by X residues, which vary with protein function. The effect of the X residues on the conformation space of the oxidized disulfide and selenosulfide forms of the CXXC(U) motif has been investigated using molecular dynamics (MD) and density functional theory. Multi-microsecond-length MD simulations of the CGGC, CGAC, and CAGC cyclic peptides show that CGGC rings readily exchange between several conformations over the course of the simulation, but steric interactions with the methyl group of Ala limit the conformation space available to the cyclic peptide, especially for CGAC. The potential for the motif to be reduced, as measured by the energy of the lowest unoccupied molecular orbitals, is dependent upon the ring conformation. These results suggest that control of available conformations by the bridging residues and the protein tertiary structure may be important for defining the function of the CXXC motif. Theoretical 77 Se chemical shifts of the selenosulfide moiety are dependent upon the conformation and/or intramolecular Se···O interactions with the backbone carbonyl group of the C-terminal U residue.
Collapse
Affiliation(s)
- Craig A Bayse
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA, 23529
| | - Deanna B Pollard
- Department of Chemistry and Biochemistry, Old Dominion University, Norfolk, VA, 23529
| |
Collapse
|
27
|
Meyer AJ, Riemer J, Rouhier N. Oxidative protein folding: state-of-the-art and current avenues of research in plants. THE NEW PHYTOLOGIST 2019; 221:1230-1246. [PMID: 30230547 DOI: 10.1111/nph.15436] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/21/2018] [Accepted: 08/01/2018] [Indexed: 06/08/2023]
Abstract
Contents Summary 1230 I. Introduction 1230 II. Formation and isomerization of disulfides in the ER and the Golgi apparatus 1231 III. The disulfide relay in the mitochondrial intermembrane space: why are plants different? 1236 IV. Disulfide bond formation on luminal proteins in thylakoids 1240 V. Conclusion 1242 Acknowledgements 1242 References 1242 SUMMARY: Disulfide bonds are post-translational modifications crucial for the structure and function of thousands of proteins. Their formation and isomerization, referred to as oxidative folding, require specific protein machineries found in oxidizing subcellular compartments, namely the endoplasmic reticulum and the associated endomembrane system, the intermembrane space of mitochondria and the thylakoid lumen of chloroplasts. At least one protein component is required for transferring electrons from substrate proteins to an acceptor that is usually molecular oxygen. For oxidation reactions, incoming reduced substrates are oxidized by thiol-oxidoreductase proteins (or domains in case of chimeric proteins), which are usually themselves oxidized by a single thiol oxidase, the enzyme generating disulfide bonds de novo. By contrast, the description of the molecular actors and pathways involved in proofreading and isomerization of misfolded proteins, which require a tightly controlled redox balance, lags behind. Herein we provide a general overview of the knowledge acquired on the systems responsible for oxidative protein folding in photosynthetic organisms, highlighting their particularities compared to other eukaryotes. Current research challenges are discussed including the importance and specificity of these oxidation systems in the context of the existence of reducing systems in the same compartments.
Collapse
Affiliation(s)
- Andreas J Meyer
- INRES-Chemical Signalling, University of Bonn, 53113, Bonn, Germany
| | - Jan Riemer
- Institute of Biochemistry, University of Cologne, 50674, Cologne, Germany
| | | |
Collapse
|
28
|
Javitt G, Grossman‐Haham I, Alon A, Resnick E, Mutsafi Y, Ilani T, Fass D. cis-Proline mutants of quiescin sulfhydryl oxidase 1 with altered redox properties undermine extracellular matrix integrity and cell adhesion in fibroblast cultures. Protein Sci 2019; 28:228-238. [PMID: 30367560 PMCID: PMC6295897 DOI: 10.1002/pro.3537] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2018] [Revised: 10/22/2018] [Accepted: 10/22/2018] [Indexed: 11/13/2022]
Abstract
The thioredoxin superfamily has expanded and diverged extensively throughout evolution such that distant members no longer show appreciable sequence homology. Nevertheless, redox-active thioredoxin-fold proteins functioning in diverse physiological contexts often share canonical amino acids near the active-site (di-)cysteine motif. Quiescin sulfhydryl oxidase 1 (QSOX1), a catalyst of disulfide bond formation secreted by fibroblasts, is a multi-domain thioredoxin superfamily enzyme with certain similarities to the protein disulfide isomerase (PDI) enzymes. Among other potential functions, QSOX1 supports extracellular matrix assembly in fibroblast cultures. We introduced mutations at a cis-proline in QSOX1 that is conserved across the thioredoxin superfamily and was previously observed to modulate redox interactions of the bacterial enzyme DsbA. The resulting QSOX1 variants showed a striking detrimental effect when added exogenously to fibroblasts: they severely disrupted the extracellular matrix and cell adhesion, even in the presence of naturally secreted, wild-type QSOX1. The specificity of this phenomenon for particular QSOX1 mutants inspired an investigation of the effects of mutation on catalytic and redox properties. For a series of QSOX1 mutants, the detrimental effect correlated with the redox potential of the first redox-active site, and an X-ray crystal structure of one of the mutants revealed the reorganization of the cis-proline loop caused by the mutations. Due to the conservation of the mutated residues across the PDI family and beyond, insights obtained in this study may be broadly applicable to a variety of physiologically important redox-active enzymes. IMPACT STATEMENT: We show that mutation of a conserved cis-proline amino acid, analogous to a mutation used to trap substrates of a bacterial disulfide catalyst, has a dramatic effect on the physiological function of the mammalian disulfide catalyst QSOX1. As the active-site region of QSOX1 is shared with the large family of protein disulfide isomerases in humans, the effects of such mutations on redox properties, enzymatic activity, and biological targeting may be relevant across the family.
Collapse
Affiliation(s)
- Gabriel Javitt
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Iris Grossman‐Haham
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Assaf Alon
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Efrat Resnick
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Yael Mutsafi
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Tal Ilani
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| | - Deborah Fass
- Department of Structural BiologyWeizmann Institute of ScienceRehovot7610001Israel
| |
Collapse
|
29
|
Scheuermann MJ, Forbes CR, Zondlo NJ. Redox-Responsive Protein Design: Design of a Small Protein Motif Dependent on Glutathionylation. Biochemistry 2018; 57:6956-6963. [PMID: 30511831 DOI: 10.1021/acs.biochem.8b00973] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
Cysteine S-glutathionylation is a protein post-translational modification that promotes cellular responses to changes in oxidative conditions. The design of protein motifs that directly depend on defined changes to protein side chains provides new methods for probing diverse protein post-translational modifications. A canonical, 12-residue EF-hand motif was redesigned to be responsive to cysteine glutathionylation. The key design principle was the replacement of the metal-binding Glu12 carboxylate of an EF-hand with a motif capable of metal binding via a free carboxylate in the glutathione-conjugated peptide. In the optimized peptide (DKDADGWCG), metal binding and terbium luminescence were dependent on glutathionylation, with weaker metal binding in the presence of reduced cysteine but increased metal affinity and a 3.5-fold increase in terbium luminescence at 544 nm when cysteine was glutathionylated. Nuclear magnetic resonance spectroscopy indicated that the structure at all residues of the glutathionylated peptide changed in the presence of metal, with chemical shift changes consistent with the adoption of an EF-hand-like structure in the metal-bound glutathionylated peptide. This small protein motif consists of canonical amino acids and is thus genetically encodable, for its potential use as a localized tag to probe protein glutathionylation.
Collapse
Affiliation(s)
- Michael J Scheuermann
- Department of Chemistry and Biochemistry , University of Delaware , Newark , Delaware 19716 , United States
| | - Christina R Forbes
- Department of Chemistry and Biochemistry , University of Delaware , Newark , Delaware 19716 , United States
| | - Neal J Zondlo
- Department of Chemistry and Biochemistry , University of Delaware , Newark , Delaware 19716 , United States
| |
Collapse
|
30
|
Bromage DI, Santos CX, Shah AM. Developing potential biomarkers through bedside-to-bench translation. J Mol Cell Cardiol 2018; 133:209-210. [PMID: 30472252 DOI: 10.1016/j.yjmcc.2018.07.254] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2018] [Accepted: 07/25/2018] [Indexed: 10/27/2022]
Affiliation(s)
- D I Bromage
- King's College London British Heart Foundation Centre of Excellence, London, UK
| | - C X Santos
- King's College London British Heart Foundation Centre of Excellence, London, UK
| | - A M Shah
- King's College London British Heart Foundation Centre of Excellence, London, UK.
| |
Collapse
|
31
|
Zhang Z, Zhang L, Zhou L, Lei Y, Zhang Y, Huang C. Redox signaling and unfolded protein response coordinate cell fate decisions under ER stress. Redox Biol 2018; 25:101047. [PMID: 30470534 PMCID: PMC6859529 DOI: 10.1016/j.redox.2018.11.005] [Citation(s) in RCA: 254] [Impact Index Per Article: 36.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2018] [Revised: 11/05/2018] [Accepted: 11/07/2018] [Indexed: 02/05/2023] Open
Abstract
Endoplasmic reticulum (ER) is a dynamic organelle orchestrating the folding and post-translational maturation of almost all membrane proteins and most secreted proteins. These proteins synthesized in the ER, need to form disulfide bridge to acquire specific three-dimensional structures for function. The formation of disulfide bridge is mediated via protein disulfide isomerase (PDI) family and other oxidoreductases, which contribute to reactive oxygen species (ROS) generation and consumption in the ER. Therefore, redox regulation of ER is delicate and sensitive to perturbation. Deregulation in ER homeostasis, usually called ER stress, can provoke unfolded protein response (UPR) pathways with an aim to initially restore homeostasis by activating genes involved in protein folding and antioxidative machinery. Over time, however, activated UPR involves a variety of cellular signaling pathways which determine the state and fate of cell in large part (like autophagy, apoptosis, ferroptosis, inflammation, senescence, stemness, and cell cycle, etc.). This review will describe the regulation of UPR from the redox perspective in controlling the cell survival or death, emphasizing the redox modifications of UPR sensors/transducers in the ER.
Collapse
Affiliation(s)
- Zhe Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Lu Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Li Zhou
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China
| | - Yunlong Lei
- Department of Biochemistry and Molecular Biology, and Molecular Medicine and Cancer Research Center, Chongqing Medical University, Chongqing 400016, PR China
| | - Yuanyuan Zhang
- Department of Pharmacology, West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, Chengdu 610041, PR China.
| | - Canhua Huang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, West China Hospital, and West China School of Basic Medical Sciences & Forensic Medicine, Sichuan University, and Collaborative Innovation Center for Biotherapy, Chengdu 610041, PR China.
| |
Collapse
|
32
|
Horn N, Møller LB, Nurchi VM, Aaseth J. Chelating principles in Menkes and Wilson diseases: Choosing the right compounds in the right combinations at the right time. J Inorg Biochem 2018; 190:98-112. [PMID: 30384011 DOI: 10.1016/j.jinorgbio.2018.10.009] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2018] [Revised: 10/06/2018] [Accepted: 10/19/2018] [Indexed: 01/08/2023]
Abstract
Dysregulation of copper homeostasis in humans is primarily found in two genetic diseases of copper transport, Menkes and Wilson diseases, which show symptoms of copper deficiency or overload, respectively. However, both diseases are copper storage disorders despite completely opposite clinical pictures. Clinically, Menkes disease is characterized by copper deficiency secondary to poor loading of copper-requiring enzymes although sufficient body copper. Copper accumulates in non-hepatic tissues, but is deficient in blood, liver, and brain. In contrast, Wilson disease is characterized by symptoms of copper toxicity secondary to accumulation of copper in several organs most notably brain and liver, and a saturated blood copper pool. It is a challenge to correct copper dyshomeostasis in either disease though copper depletion in Menkes disease is most challenging. Both diseases are caused by defective copper export from distinct cells, and we seek to give new angles and guidelines to improve treatment of these two complementary diseases. Therapy of Menkes disease with copper-histidine, thiocarbamate, nitrilotriacetate or lipoic acid is discussed. In Wilson disease combination of a hydrophilic chelator e.g. trientine or dimercaptosuccinate with a brain shuttle e.g. thiomolybdate or lipoate, is discussed. New chelating principles for copper removal or delivery are outlined.
Collapse
Affiliation(s)
| | - Lisbeth Birk Møller
- Kennedy Center, Department of Clinical Genetics, Copenhagen University Hospital, Rigshospitalet, Gl. Landevej 7, 2600 Glostrup, Denmark
| | | | - Jan Aaseth
- Innlandet Hospital, Norway; Inland Norway University of Applied Sciences, Elverum, Norway.
| |
Collapse
|
33
|
Fujii J, Homma T, Kobayashi S, Seo HG. Mutual interaction between oxidative stress and endoplasmic reticulum stress in the pathogenesis of diseases specifically focusing on non-alcoholic fatty liver disease. World J Biol Chem 2018; 9:1-15. [PMID: 30364769 PMCID: PMC6198288 DOI: 10.4331/wjbc.v9.i1.1] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/27/2018] [Revised: 09/19/2018] [Accepted: 10/11/2018] [Indexed: 02/05/2023] Open
Abstract
Reactive oxygen species (ROS) are produced during normal physiologic processes with the consumption of oxygen. While ROS play signaling roles, when they are produced in excess beyond normal antioxidative capacity this can cause pathogenic damage to cells. The majority of such oxidation occurs in polyunsaturated fatty acids and sulfhydryl group in proteins, resulting in lipid peroxidation and protein misfolding, respectively. The accumulation of misfolded proteins in the endoplasmic reticulum (ER) is enhanced under conditions of oxidative stress and results in ER stress, which, together, leads to the malfunction of cellular homeostasis. Multiple types of defensive machinery are activated in unfolded protein response under ER stress to resolve this unfavorable situation. ER stress triggers the malfunction of protein secretion and is associated with a variety of pathogenic conditions including defective insulin secretion from pancreatic β-cells and accelerated lipid droplet formation in hepatocytes. Herein we use nonalcoholic fatty liver disease (NAFLD) as an illustration of such pathological liver conditions that result from ER stress in association with oxidative stress. Protecting the ER by eliminating excessive ROS via the administration of antioxidants or by enhancing lipid-metabolizing capacity via the activation of peroxisome proliferator-activated receptors represent promising therapeutics for NAFLD.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Takujiro Homma
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Sho Kobayashi
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Han Geuk Seo
- Sanghuh College of Life Sciences, Konkuk University, Seoul 143-701, South Korea
| |
Collapse
|
34
|
Sung HJ, Ahn JM, Yoon YH, Na SS, Choi YJ, Kim YI, Lee SY, Lee EB, Cho S, Cho JY. Quiescin Sulfhydryl Oxidase 1 (QSOX1) Secreted by Lung Cancer Cells Promotes Cancer Metastasis. Int J Mol Sci 2018; 19:ijms19103213. [PMID: 30336636 PMCID: PMC6214099 DOI: 10.3390/ijms19103213] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2018] [Revised: 10/12/2018] [Accepted: 10/15/2018] [Indexed: 01/16/2023] Open
Abstract
As lung cancer shows the highest mortality in cancer-related death, serum biomarkers are demanded for lung cancer diagnosis and its treatment. To discover lung cancer protein biomarkers, secreted proteins from primary cultured lung cancer and adjacent normal tissues from patients were subjected to LC/MS⁻MS proteomic analysis. Quiescin sulfhydryl oxidase (QSOX1) was selected as a biomarker candidate from the enriched proteins in the secretion of lung cancer cells. QSOX1 levels were higher in 82% (51 of 62 tissues) of lung cancer tissues compared to adjacent normal tissues. Importantly, QSOX1 serum levels were significantly higher in cancer patients (p < 0.05, Area Under curve (AUC) = 0.89) when measured by multiple reaction monitoring (MRM). Higher levels of QSOX1 were also uniquely detected in lung cancer tissues, among several other solid cancers, by immunohistochemistry. QSOX1-knock-downed Lewis lung cancer (LLC) cells were less viable from oxidative stress and reduced migration and invasion. In addition, LLC mouse models with QSOX1 knock-down also proved that QSOX1 functions in promoting cancer metastasis. In conclusion, QSOX1 might be a lung cancer tissue-derived biomarker and be involved in the promotion of lung cancers, and thus can be a therapeutic target for lung cancers.
Collapse
Affiliation(s)
- Hye-Jin Sung
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Jung-Mo Ahn
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Yeon-Hee Yoon
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Sang-Su Na
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Young-Jin Choi
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Yong-In Kim
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| | - Soo-Youn Lee
- Departments of Laboratory Medicine & Genetics and Medicine, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul 06351, Korea.
| | - Eung-Bae Lee
- Department of Thoracic and Cardiovascular Surgery, Kyungpook National University Medical Center, Daegu 41944, Korea.
| | - Sukki Cho
- Department of Thoracic and Cardiovascular Surgery, Seoul National University Bundang Hospital, Seoungnam-si, Gyeonggi-do 13620, Korea.
| | - Je-Yoel Cho
- Department of Biochemistry, BK21 PLUS Program for Creative Veterinary Science Research and Research Institute for Veterinary Science, College of Veterinary Medicine, Seoul National University, Seoul 08826, Korea.
| |
Collapse
|
35
|
Abstract
The concept of cell signaling in the context of nonenzyme-assisted protein modifications by reactive electrophilic and oxidative species, broadly known as redox signaling, is a uniquely complex topic that has been approached from numerous different and multidisciplinary angles. Our Review reflects on five aspects critical for understanding how nature harnesses these noncanonical post-translational modifications to coordinate distinct cellular activities: (1) specific players and their generation, (2) physicochemical properties, (3) mechanisms of action, (4) methods of interrogation, and (5) functional roles in health and disease. Emphasis is primarily placed on the latest progress in the field, but several aspects of classical work likely forgotten/lost are also recollected. For researchers with interests in getting into the field, our Review is anticipated to function as a primer. For the expert, we aim to stimulate thought and discussion about fundamentals of redox signaling mechanisms and nuances of specificity/selectivity and timing in this sophisticated yet fascinating arena at the crossroads of chemistry and biology.
Collapse
Affiliation(s)
- Saba Parvez
- Department of Pharmacology and Toxicology, College of
Pharmacy, University of Utah, Salt Lake City, Utah, 84112, USA
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Marcus J. C. Long
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Jesse R. Poganik
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
| | - Yimon Aye
- Ecole Polytechnique Fédérale de Lausanne,
Institute of Chemical Sciences and Engineering, 1015, Lausanne, Switzerland
- Department of Chemistry and Chemical Biology, Cornell
University, Ithaca, New York, 14853, USA
- Department of Biochemistry, Weill Cornell Medicine, New
York, New York, 10065, USA
| |
Collapse
|
36
|
Yu T, Laird JR, Prescher JA, Thorpe C. Gaussia princeps luciferase: a bioluminescent substrate for oxidative protein folding. Protein Sci 2018; 27:1509-1517. [PMID: 29696739 DOI: 10.1002/pro.3433] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2018] [Revised: 04/20/2018] [Accepted: 04/23/2018] [Indexed: 11/07/2022]
Abstract
Gaussia princeps luciferase (GLuc) generates an intense burst of blue light when exposed to coelenterazine in the absence of ATP. Here we show that this 5-disulfide containing enzyme can be used as a facile and convenient substrate for studies of oxidative protein folding. Reduced GLuc (rGLuc), with 10 free cysteine residues, is completely inactive as a luciferase but >60% bioluminescence activity, compared to controls, can be recovered using a range of oxidizing regimens in the absence of the exogenous shuffling activity of protein disulfide isomerase (PDI). The sulfhydryl oxidase QSOX1 can be assayed using rGLuc in a simple bioluminescence plate reader format. Similarly, low concentrations of rGLuc can be oxidized by millimolar levels of dehydroascorbate, hydrogen peroxide or much lower concentrations of sodium tetrathionate. The oxidative refolding of rGLuc in the presence of a range of glutathione redox buffers is only marginally accelerated by micromolar levels of PDI. This modest rate enhancement probably results from a relatively simple disulfide connectivity in native GLuc; reflecting two homologous domains each carrying two disulfide bonds with a single interdomain disulfide. When GLuc is reoxidized under denaturing conditions the resulting scrambled protein (sGLuc) can be used in a sensitive bioluminescence assay for reduced PDI in the absence of added exogenous thiols. Finally, the general facility by which rGLuc can recover bioluminescent activity in vitro provides a sensitive method for the assessment of inhibitors of oxidative protein folding.
Collapse
Affiliation(s)
- Tiantian Yu
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, 19716
| | - Joanna R Laird
- Department of Chemistry, University of California at Irvine, Irvine, California, 92697
| | - Jennifer A Prescher
- Department of Chemistry, University of California at Irvine, Irvine, California, 92697
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware, Newark, Delaware, 19716
| |
Collapse
|
37
|
Caillard A, Sadoune M, Cescau A, Meddour M, Gandon M, Polidano E, Delcayre C, Da Silva K, Manivet P, Gomez AM, Cohen-Solal A, Vodovar N, Li Z, Mebazaa A, Samuel JL. QSOX1, a novel actor of cardiac protection upon acute stress in mice. J Mol Cell Cardiol 2018; 119:75-86. [DOI: 10.1016/j.yjmcc.2018.04.014] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 04/07/2018] [Accepted: 04/27/2018] [Indexed: 12/31/2022]
|
38
|
Liu C, He X, Wu X, Wang Z, Zuo W, Hu G. Clinicopathological and prognostic significance of GPx2 protein expression in nasopharyngeal carcinoma. Cancer Biomark 2018; 19:335-340. [PMID: 28453466 DOI: 10.3233/cbm-160542] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
Abstract
OBJECTIVE This study was designed to evaluate the relation between GPx2 (glutathione peroxidase 2) expressions and clinicopathological features as well as prognosis of patients with nasopharyngeal carcinoma (NPC). METHODS A total of 89 cases of NPC were investigated to examine the immunohistochemical expression of GPx2. Fourteen pairs of NPC and the control samples were analyzed respectively by qRT-PCR and Western blot. The correlations of GPx2 expressions with the clinicopathologic features and the prognosis of NPC patients were also analyzed. RESULTS The expression of GPx2 in NPC tissues was elevated immunohistochemically when compared with normal nasopharyngeal tissues (P< 0.05). The mRNA expression of GPx2 in carcinoma tissues was highly elevated compared with the control tissues (P< 0.05). GPx2 protein in carcinoma tissues was also over expressed than in control tissues (P< 0.05). Also GPx2 expression was significantly higher in the late clinical stage (P= 0.02). While there was no significant association between GPx2 expression and patient age, sex, T-stage, N-stage and the metastasis. CONCLUSIONS GPx2 may play an important role in the development of nasopharyngeal carcinoma. Furthermore, GPx2 may serve as a prognostic biomarker for NPC patient.
Collapse
|
39
|
Characterization of sulfhydryl oxidase from Aspergillus tubingensis. BMC BIOCHEMISTRY 2017; 18:15. [PMID: 29216817 PMCID: PMC5721475 DOI: 10.1186/s12858-017-0090-4] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/15/2017] [Accepted: 11/20/2017] [Indexed: 11/26/2022]
Abstract
Background Despite of the presence of sulfhydryl oxidases (SOXs) in the secretomes of industrially relevant organisms and their many potential applications, only few of these enzymes have been biochemically characterized. In addition, basic functions of most of the SOX enzymes reported so far are not fully understood. In particular, the physiological role of secreted fungal SOXs is unclear. Results The recently identified SOX from Aspergillus tubingensis (AtSOX) was produced, purified and characterized in the present work. AtSOX had a pH optimum of 6.5, and showed a good pH stability retaining more than 80% of the initial activity in a pH range 4-8.5 within 20 h. More than 70% of the initial activity was retained after incubation at 50 °C for 20 h. AtSOX contains a non-covalently bound flavin cofactor. The enzyme oxidised a sulfhydryl group of glutathione to form a disulfide bond, as verified by nuclear magnetic resonance spectroscopy. AtSOX preferred glutathione as a substrate over cysteine and dithiothreitol. The activity of the enzyme was totally inhibited by 10 mM zinc sulphate. Peptide- and protein-bound sulfhydryl groups in bikunin, gliotoxin, holomycin, insulin B chain, and ribonuclease A, were not oxidised by the enzyme. Based on the analysis of 33 fungal genomes, SOX enzyme encoding genes were found close to nonribosomal peptide synthetases (NRPS) but not with polyketide synthases (PKS). In the phylogenetic tree, constructed from 25 SOX and thioredoxin reductase sequences from IPR000103 InterPro family, AtSOX was evolutionary closely related to other Aspergillus SOXs. Oxidoreductases involved in the maturation of nonribosomal peptides of fungal and bacterial origin, namely GliT, HlmI and DepH, were also evolutionary closely related to AtSOX whereas fungal thioreductases were more distant. Conclusions AtSOX (55 kDa) is a fungal secreted flavin-dependent enzyme with good stability to both pH and temperature. A Michaelis-Menten behaviour was observed with reduced glutathione as a substrate. Based on the location of SOX enzyme encoding genes close to NRPSs, SOXs could be involved in the secondary metabolism and act as an accessory enzyme in the production of nonribosomal peptides. Electronic supplementary material The online version of this article (10.1186/s12858-017-0090-4) contains supplementary material, which is available to authorized users.
Collapse
|
40
|
Abstract
SIGNIFICANCE Glutathione metabolism is comparable to a jigsaw puzzle with too many pieces. It is supposed to comprise (i) the reduction of disulfides, hydroperoxides, sulfenic acids, and nitrosothiols, (ii) the detoxification of aldehydes, xenobiotics, and heavy metals, and (iii) the synthesis of eicosanoids, steroids, and iron-sulfur clusters. In addition, glutathione affects oxidative protein folding and redox signaling. Here, I try to provide an overview on the relevance of glutathione-dependent pathways with an emphasis on quantitative data. Recent Advances: Intracellular redox measurements reveal that the cytosol, the nucleus, and mitochondria contain very little glutathione disulfide and that oxidative challenges are rapidly counterbalanced. Genetic approaches suggest that iron metabolism is the centerpiece of the glutathione puzzle in yeast. Furthermore, recent biochemical studies provide novel insights on glutathione transport processes and uncoupling mechanisms. CRITICAL ISSUES Which parts of the glutathione puzzle are most relevant? Does this explain the high intracellular concentrations of reduced glutathione? How can iron-sulfur cluster biogenesis, oxidative protein folding, or redox signaling occur at high glutathione concentrations? Answers to these questions not only seem to depend on the organism, cell type, and subcellular compartment but also on different ideologies among researchers. FUTURE DIRECTIONS A rational approach to compare the relevance of glutathione-dependent pathways is to combine genetic and quantitative kinetic data. However, there are still many missing pieces and too little is known about the compartment-specific repertoire and concentration of numerous metabolites, substrates, enzymes, and transporters as well as rate constants and enzyme kinetic patterns. Gathering this information might require the development of novel tools but is crucial to address potential kinetic competitions and to decipher uncoupling mechanisms to solve the glutathione puzzle. Antioxid. Redox Signal. 27, 1130-1161.
Collapse
Affiliation(s)
- Marcel Deponte
- Department of Parasitology, Ruprecht-Karls University , Heidelberg, Germany
| |
Collapse
|
41
|
Zhan YA, Abskharon R, Li Y, Yuan J, Zeng L, Dang J, Martinez MC, Wang Z, Mikol J, Lehmann S, Bu S, Steyaert J, Cui L, Petersen RB, Kong Q, Wang GX, Wohlkonig A, Zou WQ. Quiescin-sulfhydryl oxidase inhibits prion formation in vitro. Aging (Albany NY) 2017; 8:3419-3429. [PMID: 27959866 PMCID: PMC5270677 DOI: 10.18632/aging.101132] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2016] [Accepted: 11/24/2016] [Indexed: 02/03/2023]
Abstract
Prions are infectious proteins that cause a group of fatal transmissible diseases in animals and humans. The scrapie isoform (PrPSc) of the cellular prion protein (PrPC) is the only known component of the prion. Several lines of evidence have suggested that the formation and molecular features of PrPSc are associated with an abnormal unfolding/refolding process. Quiescin-sulfhydryl oxidase (QSOX) plays a role in protein folding by introducing disulfides into unfolded reduced proteins. Here we report that QSOX inhibits human prion propagation in protein misfolding cyclic amplification reactions and murine prion propagation in scrapie-infected neuroblastoma cells. Moreover, QSOX preferentially binds PrPSc from prion-infected human or animal brains, but not PrPC from uninfected brains. Surface plasmon resonance of the recombinant mouse PrP (moPrP) demonstrates that the affinity of QSOX for monomer is significantly lower than that for octamer (312 nM vs 1.7 nM). QSOX exhibits much lower affinity for N-terminally truncated moPrP (PrP89-230) than for the full-length moPrP (PrP23-231) (312 nM vs 2 nM), suggesting that the N-terminal region of PrP is critical for the interaction of PrP with QSOX. Our study indicates that QSOX may play a role in prion formation, which may open new therapeutic avenues for treating prion diseases.
Collapse
Affiliation(s)
- Yi-An Zhan
- First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, The People's Republic of China.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Romany Abskharon
- VIB Center for Structural Biology, VIB, 1050 Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium.,National Institute of Oceanography and Fisheries (NIFO), 11516 Cairo, Egypt.,CNS, Van Andel Research Institute, Grand Rapids, MI 49503, USA
| | - Yu Li
- First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, The People's Republic of China.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Jue Yuan
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Liang Zeng
- First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, The People's Republic of China.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Johnny Dang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Manuel Camacho Martinez
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Zerui Wang
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, The People's Republic of China
| | - Jacqueline Mikol
- Hôpital Lariboisière, Service d'Anatomie et Cytologie Pathologiques, Paris, France
| | - Sylvain Lehmann
- IRMB -Hôpital ST ELOI, CHU de Montpellier, Montpellier, France
| | - Shizhong Bu
- Diabetes Research Center, Ningbo University, The People's Republic of China
| | - Jan Steyaert
- VIB Center for Structural Biology, VIB, 1050 Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium
| | - Li Cui
- Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, The People's Republic of China
| | - Robert B Petersen
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neuroscience, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Qingzhong Kong
- Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA
| | - Gong-Xiang Wang
- First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, The People's Republic of China
| | - Alexandre Wohlkonig
- VIB Center for Structural Biology, VIB, 1050 Brussels, Belgium.,Structural Biology Brussels, Vrije Universiteit Brussel (VUB), 1050 Brussels, Belgium
| | - Wen-Quan Zou
- First Affiliated Hospital, Nanchang University, Nanchang, Jiangxi Province, The People's Republic of China.,Department of Pathology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neurology, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,National Prion Disease Pathology Surveillance Center, Case Western Reserve University School of Medicine, Cleveland, Ohio 44106, USA.,Department of Neurology, The First Hospital of Jilin University, Changchun, Jilin Province, The People's Republic of China.,State Key Laboratory for Infectious Disease Prevention and Control, National Institute for Viral Disease Control and Prevention, Chinese Center for Disease Control and Prevention, Beijing, The People's Republic of China
| |
Collapse
|
42
|
Flowers SA, Kalamajski S, Ali L, Björkman LI, Raj JR, Aspberg A, Karlsson NG, Jin C. Cartilage oligomeric matrix protein forms protein complexes with synovial lubricin via non-covalent and covalent interactions. Osteoarthritis Cartilage 2017; 25:1496-1504. [PMID: 28373131 DOI: 10.1016/j.joca.2017.03.016] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2016] [Revised: 03/17/2017] [Accepted: 03/23/2017] [Indexed: 02/02/2023]
Abstract
OBJECTIVE Understanding the cartilage surface structure, lost in arthritic disease, is essential for developing strategies to effectively restore it. Given that adherence of the lubricating protein, lubricin, to the cartilage surface is critical for boundary lubrication, an interaction with cartilage oligomeric matrix protein (COMP) was investigated. COMP, an abundant cartilage protein, is known to be important for matrix formation. DESIGN Synovial fluid (SF) from arthritic patients was used to detect possible COMP-lubricin complexes by immunological methods. Recombinant (RC) COMP and lubricin fragments were expressed to characterize this bonding and mass spectrometry employed to specifically identify the cysteines involved in inter-protein disulfide bonds. RESULTS COMP-lubricin complexes were identified in the SF of arthritic patients by Western blot, co-immunoprecipitation and sandwich ELISA. RC fragment solid-phase binding assays showed that the C-terminal (amino acids (AA) 518-757) of COMP bound non-covalently to the N-terminal of lubricin (AA 105-202). Mass spectrometry determined that although cysteines throughout COMP were involved in binding with lubricin, the cysteines in lubricin were primarily focused to an N-terminal region (AA 64-86). The close proximity of the non-covalent and disulfide binding domains on lubricin suggest a two-step mechanism to strongly bind lubricin to COMP. CONCLUSION These data demonstrate that lubricin forms a complex network with COMP involving both non-covalent and covalent bonds. This complex between lubricin and the cartilage protein COMP can be identified in the SF of patients with arthritis conditions including osteoarthritis (OA) and rheumatoid arthritis (RA).
Collapse
Affiliation(s)
- S A Flowers
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - S Kalamajski
- Department of Clinical Sciences Lund, Division of Rheumatology and Molecular Skeletal Biology, Lund University, Lund, Sweden.
| | - L Ali
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - L I Björkman
- Department of Rheumatology and Inflammation Research, Institute of Medicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - J R Raj
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - A Aspberg
- Department of Clinical Sciences Lund, Division of Rheumatology and Molecular Skeletal Biology, Lund University, Lund, Sweden.
| | - N G Karlsson
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| | - C Jin
- Department of Medical Biochemistry and Cell Biology, Institute of Biomedicine, Sahlgrenska Academy, University of Gothenburg, Gothenburg, Sweden.
| |
Collapse
|
43
|
Hourihan JM, Moronetti Mazzeo LE, Fernández-Cárdenas LP, Blackwell TK. Cysteine Sulfenylation Directs IRE-1 to Activate the SKN-1/Nrf2 Antioxidant Response. Mol Cell 2017; 63:553-566. [PMID: 27540856 DOI: 10.1016/j.molcel.2016.07.019] [Citation(s) in RCA: 137] [Impact Index Per Article: 17.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2016] [Revised: 07/01/2016] [Accepted: 07/20/2016] [Indexed: 12/13/2022]
Abstract
Emerging evidence suggests that many proteins may be regulated through cysteine modification, but the extent and functions of this signaling remain largely unclear. The endoplasmic reticulum (ER) transmembrane protein IRE-1 maintains ER homeostasis by initiating the unfolded protein response (UPR(ER)). Here we show in C. elegans and human cells that IRE-1 has a distinct redox-regulated function in cytoplasmic homeostasis. Reactive oxygen species (ROS) that are generated at the ER or by mitochondria sulfenylate a cysteine within the IRE-1 kinase activation loop. This inhibits the IRE-1-mediated UPR(ER) and initiates the p38/SKN-1(Nrf2) antioxidant response, thereby increasing stress resistance and lifespan. Many AGC-family kinases (AKT, p70S6K, PKC, ROCK1) seem to be regulated similarly. The data reveal that IRE-1 has an ancient function as a cytoplasmic sentinel that activates p38 and SKN-1(Nrf2) and indicate that cysteine modifications induced by ROS signals can direct proteins to adopt unexpected functions and may coordinate many cellular processes.
Collapse
Affiliation(s)
- John M Hourihan
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - Lorenza E Moronetti Mazzeo
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - L Paulette Fernández-Cárdenas
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA
| | - T Keith Blackwell
- Research Division, Joslin Diabetes Center, Boston, MA 02215, USA; Department of Genetics and Harvard Stem Cell Institute, Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|
44
|
Abstract
Cysteine thiols are among the most reactive functional groups in proteins, and their pairing in disulfide linkages is a common post-translational modification in proteins entering the secretory pathway. This modest amino acid alteration, the mere removal of a pair of hydrogen atoms from juxtaposed cysteine residues, contrasts with the substantial changes that characterize most other post-translational reactions. However, the wide variety of proteins that contain disulfides, the profound impact of cross-linking on the behavior of the protein polymer, the numerous and diverse players in intracellular pathways for disulfide formation, and the distinct biological settings in which disulfide bond formation can take place belie the simplicity of the process. Here we lay the groundwork for appreciating the mechanisms and consequences of disulfide bond formation in vivo by reviewing chemical principles underlying cysteine pairing and oxidation. We then show how enzymes tune redox-active cofactors and recruit oxidants to improve the specificity and efficiency of disulfide formation. Finally, we discuss disulfide bond formation in a cellular context and identify important principles that contribute to productive thiol oxidation in complex, crowded, dynamic environments.
Collapse
Affiliation(s)
- Deborah Fass
- Department of Structural Biology, Weizmann Institute of Science , Rehovot 7610001, Israel
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware , Newark, Delaware 19716, United States
| |
Collapse
|
45
|
Kuo YW, Joshi R, Wang TE, Chang HW, Li SH, Hsiao CN, Tsai PSJ. Identification, characterization and purification of porcine Quiescin Q6-Sulfydryl Oxidase 2 protein. BMC Vet Res 2017; 13:205. [PMID: 28662655 PMCID: PMC5492681 DOI: 10.1186/s12917-017-1125-1] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2017] [Accepted: 06/20/2017] [Indexed: 01/05/2023] Open
Abstract
Background Post-spermiogenesis membrane surface modifications rely on molecules present in the reproductive tracts. Two isoforms (isoform 1 and 2) from Quiescin Q6-Sulfydryl Oxidase protein family have been identified in the male reproductive tract of rodent species. However, unlike isoform 1, scarce information is available for isoform 2, likely due to its lower expression level and lack of proper purification methods to obtain sufficient protein quantity for further assays. Results This study demonstrated the presence of short and long forms of Quiescin Q6-Sulfydryl Oxidase 2 in boar, likely representing the secretory (short form) and transmembrane (long form) forms of Quiescin Q6-Sulfydryl Oxidase 2. Immunohistochemistry studies revealed the presence of Quiescin Q6-Sulfydryl Oxidase 2 in a broad range of porcine tissues; the pronounced vesicle-contained Quiescin Q6-Sulfydryl Oxidase 2 at the apical region of epididymis and seminal vesicles epithelium suggested its involvement in sperm physiology and its participation in semen formation. The majority of porcine Quiescin Q6-Sulfydryl Oxidase 2 could be purified via either antibody affinity column or be salted out using 10%–40% ammonium sulfate. Higher amount of low molecular weight Quiescin Q6-Sulfydryl Oxidase 2 observed in the seminal vesicle likely represents the secretory form of Quiescin Q6-Sulfydryl Oxidase 2 and reflects an exuberant secretory activity in this organ. Conclusions We demonstrated for the first time, the presence of Quiescin Q6-Sulfydryl Oxidase 2 in porcine species; moreover, two forms of Quiescin Q6-Sulfydryl Oxidase 2 were identified and exhibited distinct molecular weights and properties during protein purification processes. This study also provided feasible Quiescin Q6-Sulfydryl Oxidase 2 purification methods from slaughterhouse materials that could potentially allow obtaining sufficient amount of Quiescin Q6-Sulfydryl Oxidase 2 for future functional investigations.
Collapse
Affiliation(s)
- Yu-Wen Kuo
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan
| | - Radhika Joshi
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan
| | - Tse-En Wang
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.,Graduate Institute of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan
| | - Hui-Wen Chang
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.,Graduate Institute of Molecular and Comparative Pathobiology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan
| | - Sheng-Hsiang Li
- Department of Medical Research, Mackay Memorial Hospital, No. 92, Section 2, Zhongshan N. Rd, 251 Tamshui, Taipei, Taiwan
| | - Chun-Ni Hsiao
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.,Graduate Institute of Molecular and Comparative Pathobiology, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.,Shui-Po International Certification Boar Semen Station, No. 71-115, 732, Tainan, Taiwan
| | - Pei-Shiue Jason Tsai
- Department of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan. .,Graduate Institute of Veterinary Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan. .,Research Center for Developmental Biology and Regenerative Medicine, National Taiwan University, No. 1, Sec. 4, Roosevelt Rd, 10617, Taipei, Taiwan.
| |
Collapse
|
46
|
Selles B, Zannini F, Couturier J, Jacquot JP, Rouhier N. Atypical protein disulfide isomerases (PDI): Comparison of the molecular and catalytic properties of poplar PDI-A and PDI-M with PDI-L1A. PLoS One 2017; 12:e0174753. [PMID: 28362814 PMCID: PMC5375154 DOI: 10.1371/journal.pone.0174753] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2017] [Accepted: 03/14/2017] [Indexed: 11/18/2022] Open
Abstract
Protein disulfide isomerases are overwhelmingly multi-modular redox catalysts able to perform the formation, reduction or isomerisation of disulfide bonds. We present here the biochemical characterization of three different poplar PDI isoforms. PDI-A is characterized by a single catalytic Trx module, the so-called a domain, whereas PDI-L1a and PDI-M display an a-b-b’-a’ and a°-a-b organisation respectively. Their activities have been tested in vitro using purified recombinant proteins and a series of model substrates as insulin, NADPH thioredoxin reductase, NADP malate dehydrogenase (NADP-MDH), peroxiredoxins or RNase A. We demonstrated that PDI-A exhibited none of the usually reported activities, although the cysteines of the WCKHC active site signature are able to form a disulfide with a redox midpoint potential of -170 mV at pH 7.0. The fact that it is able to bind a [Fe2S2] cluster upon Escherichia coli expression and anaerobic purification might indicate that it does not have a function in dithiol-disulfide exchange reactions. The two other proteins were able to catalyze oxidation or reduction reactions, PDI-L1a being more efficient in most cases, except that it was unable to activate the non-physiological substrate NADP-MDH, in contrast to PDI-M. To further evaluate the contribution of the catalytic domains of PDI-M, the dicysteinic motifs have been independently mutated in each a domain. The results indicated that the two a domains seem interconnected and that the a° module preferentially catalyzed oxidation reactions whereas the a module catalyzed reduction reactions, in line with the respective redox potentials of -170 mV and -190 mV at pH 7.0. Overall, these in vitro results illustrate that the number and position of a and b domains influence the redox properties and substrate recognition (both electron donors and acceptors) of PDI which contributes to understand why this protein family expanded along evolution.
Collapse
Affiliation(s)
- Benjamin Selles
- UMR 1136 Interactions Arbres/Microorganismes, Université de Lorraine/ INRA, Faculté des Sciences et Technologies, Vandoeuvre-lès-Nancy, France
| | - Flavien Zannini
- UMR 1136 Interactions Arbres/Microorganismes, Université de Lorraine/ INRA, Faculté des Sciences et Technologies, Vandoeuvre-lès-Nancy, France
| | - Jérémy Couturier
- UMR 1136 Interactions Arbres/Microorganismes, Université de Lorraine/ INRA, Faculté des Sciences et Technologies, Vandoeuvre-lès-Nancy, France
| | - Jean-Pierre Jacquot
- UMR 1136 Interactions Arbres/Microorganismes, Université de Lorraine/ INRA, Faculté des Sciences et Technologies, Vandoeuvre-lès-Nancy, France
| | - Nicolas Rouhier
- UMR 1136 Interactions Arbres/Microorganismes, Université de Lorraine/ INRA, Faculté des Sciences et Technologies, Vandoeuvre-lès-Nancy, France
- * E-mail:
| |
Collapse
|
47
|
Patwardhan RS, Sharma D, Checker R, Thoh M, Sandur SK. Spatio-temporal changes in glutathione and thioredoxin redox couples during ionizing radiation-induced oxidative stress regulate tumor radio-resistance. Free Radic Res 2016; 49:1218-32. [PMID: 26021764 DOI: 10.3109/10715762.2015.1056180] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Ionizing radiation (IR)-induced oxidative stress in tumor cells is effectively managed by constitutive and inducible antioxidant defense systems. This study was initiated to understand the relative contribution of different redox regulatory systems in determining the tumor radio-resistance. In this study, human T-cell lymphoma (Jurkat) cells were exposed to IR (4 Gy) and monitored for the spatio-temporal changes in cellular redox regulatory parameters. We monitored the changes in the levels of reactive oxygen species (ROS) (total, mitochondrial, primary, and secondary), thiols (total, surface, and intracellular), GSH/GSSG ratio, antioxidant enzyme activity viz. thioredoxin (Trx), Trx reductase (TrxR), glutathione peroxidase, and glutathione reductase with respect to time. We have also measured protein glutathionylation. We observed that tumor cells mount a biphasic response after IR exposure which can be divided into early (0-6 h) and late (16-48 h) responses in terms of changes in cellular redox parameters. During early response, constitutively active GSH and Trx systems respond to restore cellular redox balance to pre-exposure levels and help in activation of redox-sensitive transcription factor Nrf-2. During late response, increase in the levels of antioxidants GSH and Trx rescue cells against IR-mediated damage. We observed that disruption of either glutathione or thioredoxin metabolism led to partial impairment of ability of cells to survive against IR-induced damage. But simultaneous disruption of both the pathways significantly increased radio sensitivity of Jurkat cells. This highlighted the importance of these two antioxidant pathways in regulating redox homeostasis under conditions of IR-induced oxidative stress.
Collapse
Affiliation(s)
- R S Patwardhan
- a Radiation Biology and Health Sciences Division, Modular Laboratories, Bhabha Atomic Research Centre , Trombay, Mumbai , India
| | | | | | | | | |
Collapse
|
48
|
West KP, Cole RN, Shrestha S, Schulze KJ, Lee SE, Betz J, Nonyane BAS, Wu LSF, Yager JD, Groopman JD, Christian P. A Plasma α-Tocopherome Can Be Identified from Proteins Associated with Vitamin E Status in School-Aged Children of Nepal. J Nutr 2015; 145:2646-56. [PMID: 26446483 PMCID: PMC6619677 DOI: 10.3945/jn.115.210682] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2015] [Accepted: 09/14/2015] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND The term vitamin E describes a family of 8 vitamers, 1 of which is α-tocopherol, that is essential for human health. Vitamin E status remains largely unknown in low-income countries because of the complexity and cost of measurement. Quantitative proteomics may offer an approach for identifying plasma proteins for assessing vitamin E status in these populations. OBJECTIVE To improve options for vitamin E status assessment, we sought to detect and quantify a set of plasma proteins associated with α- and γ-tocopherol concentrations in a cohort of 500 rural Nepalese children aged 6-8 y and, based on nutrient-protein associations, to predict the prevalence of vitamin E deficiency (α-tocopherol <12 μmol/L). METHODS Study children were born to mothers enrolled in an earlier antenatal micronutrient trial in Sarlahi District, Nepal. Plasma α- and γ-tocopherol concentrations were measured by high-performance liquid chromatography. Plasma aliquots were depleted of 6 high-abundance proteins, digested with trypsin, labeled with isobaric mass tags, and assessed for relative protein abundance by tandem mass spectrometry. Linear mixed-effects models were used to evaluate the association between α-tocopherol status and relative protein abundance and to predict deficiency. RESULTS We quantified 982 plasma proteins in >10% of all child samples, of which 119 correlated with α-tocopherol (false discovery rate, q < 0.10). Proteins were primarily involved in lipid transport, coagulation, repair, innate host defenses, neural function, and homeostasis. Six proteins [apolipoprotein (apo)C-III; apoB; pyruvate kinase, muscle; forkhead box 04; unc5 homolog C; and regulator of G-protein signaling 8] explained 71% of the variability in plasma α-tocopherol, predicting an in-sample population prevalence of vitamin E deficiency of 51.4% (95% CI: 46.4%, 56.3%) compared with a measured prevalence of 54.8%. Plasma γ-tocopherol was associated with 12 proteins (q < 0.10), 2 of which (apoC-III and Misato 1) explained 20% of its variability. CONCLUSIONS In this undernourished population of children in South Asia, quantitative proteomics identified a large plasma α-tocopherome from which 6 proteins predicted the prevalence of vitamin E deficiency. The findings illustrate that protein biomarkers, once absolutely quantified, can potentially predict micronutrient deficiencies in populations. The maternal micronutrient supplementation trial from which data were derived as a follow-up activity was registered with clinicaltrials.gov as NCT00115271.
Collapse
Affiliation(s)
| | - Robert N Cole
- Department of Biological Chemistry, Johns Hopkins School of Medicine Mass
Spectrometry and Proteomics Facility, Baltimore, MD
| | - Sudeep Shrestha
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| | - Kerry J Schulze
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| | - Sun Eun Lee
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| | - Joshua Betz
- Departments of Biostatistics, Johns Hopkins University Bloomberg School of
Public Health, Baltimore, MD
| | - Bareng AS Nonyane
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| | - Lee S-F Wu
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| | - James D Yager
- Departments of Environmental Health Sciences, Center for Human Nutrition,
Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - John D Groopman
- Departments of Environmental Health Sciences, Center for Human Nutrition,
Johns Hopkins University Bloomberg School of Public Health, Baltimore, MD
| | - Parul Christian
- Departments of International Health, Johns Hopkins University Bloomberg
School of Public Health, Baltimore, MD
| |
Collapse
|
49
|
Hudson DA, Thorpe C. Mia40 is a facile oxidant of unfolded reduced proteins but shows minimal isomerase activity. Arch Biochem Biophys 2015; 579:1-7. [PMID: 26014136 PMCID: PMC4500674 DOI: 10.1016/j.abb.2015.05.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2015] [Revised: 05/18/2015] [Accepted: 05/19/2015] [Indexed: 01/22/2023]
Abstract
Mia40 participates in oxidative protein folding within the mitochondrial intermembrane space (IMS) by mediating the transfer of reducing equivalents from client proteins to FAD-linked oxidoreductases of the Erv1 family (lfALR in mammals). Here we investigate the specificity of the human Mia40/lfALR system towards non-cognate unfolded protein substrates to assess whether the efficient introduction of disulfides requires a particular amino acid sequence context or the presence of an IMS targeting signal. Reduced pancreatic ribonuclease A (rRNase), avian lysozyme, and riboflavin binding protein are all competent substrates of the Mia40/lfALR system, although they lack those sequence features previously thought to direct disulfide bond formation in cognate IMS substrates. The oxidation of rRNase by Mia40 does not limit overall turnover of unfolded substrate by the Mia40/lfALR system. Mia40 is an ineffective protein disulfide isomerase when its ability to restore enzymatic activity from scrambled RNase is compared to that of protein disulfide isomerase. Mia40's ability to bind amphipathic peptides is evident by avid binding to the isolated B-chain during the insulin reductase assay. In aggregate these data suggest that the Mia40/lfALR system has a broad sequence specificity and that potential substrates may be protected from adventitious oxidation by kinetic sequestration within the mitochondrial IMS.
Collapse
Affiliation(s)
- Devin A Hudson
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, United States
| | - Colin Thorpe
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE 19716, United States.
| |
Collapse
|
50
|
Abstract
Erv1 (essential for respiration and viability 1) is a FAD-dependent sulphydryl oxidase with a tryptophan-rich catalytic domain. We show that Trp95 and Trp183 are important for stabilizing the folding, FAD-binding, and function of Erv1, whilst other four tryptophan residues are not functionally important. Erv1 is an FAD-dependent thiol oxidase of the ERV (essential for respiration and viability)/ALR (augmenter of liver regeneration) sub-family and an essential component of the mitochondrial import and assembly pathway. Erv1 contains six tryptophan residues, which are all located in the highly conserved C-terminal FAD-binding domain. Though important structural roles were predicted for the invariable Trp95, no experimental study has been reported. In the present study, we investigated the structural and functional roles of individual tryptophan residues of Erv1. Six single tryptophan-to-phenylalanine yeast mutant strains were generated and their effects on cell viability were tested at various temperatures. Then, the mutants were purified from Escherichia coli. Their effects on folding, FAD-binding and Erv1 activity were characterized. Our results showed that Erv1W95F has the strongest effect on the stability and function of Erv1 and followed by Erv1W183F. Erv1W95F results in a decrease in the Tm of Erv1 by 23°C, a significant loss of the oxidase activity and thus causing cell growth defects at both 30°C and 37°C. Erv1W183F induces changes in the oligomerization state of Erv1, along with a pronounced effect on the stability of Erv1 and its function at 37°C, whereas the other mutants had no clear effect on the function of Erv1 including the highly conserved Trp157 mutant. Finally, computational analysis indicates that Trp95 plays a key role in stabilizing the isoalloxazine ring to interact with Cys133. Taken together, the present study provided important insights into the molecular mechanism of how thiol oxidases use FAD in catalysing disulfide bond formation.
Collapse
|