1
|
Aryal B, Kwakye J, Ariyo OW, Ghareeb AFA, Milfort MC, Fuller AL, Khatiwada S, Rekaya R, Aggrey SE. Major Oxidative and Antioxidant Mechanisms During Heat Stress-Induced Oxidative Stress in Chickens. Antioxidants (Basel) 2025; 14:471. [PMID: 40298812 PMCID: PMC12023971 DOI: 10.3390/antiox14040471] [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: 02/18/2025] [Revised: 04/08/2025] [Accepted: 04/10/2025] [Indexed: 04/30/2025] Open
Abstract
Heat stress (HS) is one of the most important stressors in chickens, and its adverse effects are primarily caused by disturbing the redox homeostasis. An increase in electron leakage from the mitochondrial electron transport chain is the major source of free radical production under HS, which triggers other enzymatic systems to generate more radicals. As a defense mechanism, cells have enzymatic and non-enzymatic antioxidant systems that work cooperatively against free radicals. The generation of free radicals, particularly the reactive oxygen species (ROS) and reactive nitrogen species (RNS), under HS condition outweighs the cellular antioxidant capacity, resulting in oxidative damage to macromolecules, including lipids, carbohydrates, proteins, and DNA. Understanding these detrimental oxidative processes and protective defense mechanisms is important in developing mitigation strategies against HS. This review summarizes the current understanding of major oxidative and antioxidant systems and their molecular mechanisms in generating or neutralizing the ROS/RNS. Importantly, this review explores the potential mechanisms that lead to the development of oxidative stress in heat-stressed chickens, highlighting their unique behavioral and physiological responses against thermal stress. Further, we summarize the major findings associated with these oxidative and antioxidant mechanisms in chickens.
Collapse
Affiliation(s)
- Bikash Aryal
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Josephine Kwakye
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Oluwatomide W. Ariyo
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Ahmed F. A. Ghareeb
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
- Boehringer Ingelheim Animal Health (BIAH), Gainesville, GA 30501, USA
| | - Marie C. Milfort
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Alberta L. Fuller
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| | - Saroj Khatiwada
- Department of Animal Sciences, The Ohio State University, Wooster, OH 44691, USA;
| | - Romdhane Rekaya
- Department of Animal and Dairy Science, The University of Georgia, Athens, GA 30602, USA;
| | - Samuel E. Aggrey
- NutriGenomics Laboratory, Department of Poultry Science, The University of Georgia, Athens, GA 30602, USA or (B.A.); (J.K.); (O.W.A.); (A.F.A.G.); (M.C.M.); (A.L.F.)
| |
Collapse
|
2
|
Zaher A, Stephens SB. Breaking the Feedback Loop of β-Cell Failure: Insight into the Pancreatic β-Cell's ER-Mitochondria Redox Balance. Cells 2025; 14:399. [PMID: 40136648 PMCID: PMC11941261 DOI: 10.3390/cells14060399] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/01/2025] [Accepted: 03/07/2025] [Indexed: 03/27/2025] Open
Abstract
Pancreatic β-cells rely on a delicate balance between the endoplasmic reticulum (ER) and mitochondria to maintain sufficient insulin stores for the regulation of whole animal glucose homeostasis. The ER supports proinsulin maturation through oxidative protein folding, while mitochondria supply the energy and redox buffering that maintain ER proteostasis. In the development of Type 2 diabetes (T2D), the progressive decline of β-cell function is closely linked to disruptions in ER-mitochondrial communication. Mitochondrial dysfunction is a well-established driver of β-cell failure, whereas the downstream consequences for ER redox homeostasis have only recently emerged. This interdependence of ER-mitochondrial functions suggests that an imbalance is both a cause and consequence of metabolic dysfunction. In this review, we discuss the regulatory mechanisms of ER redox control and requirements for mitochondrial function. In addition, we describe how ER redox imbalances may trigger mitochondrial dysfunction in a vicious feed forward cycle that accelerates β-cell dysfunction and T2D onset.
Collapse
Affiliation(s)
- Amira Zaher
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA;
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
| | - Samuel B. Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52246, USA;
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52246, USA
- Department of Anatomy and Cell Biology, University of Iowa, Iowa City, IA 52246, USA
| |
Collapse
|
3
|
Speranza E, Sorrentino I, Boletta A, Sitia R. Exquisite sensitivity of Polycystin-1 to H 2O 2 concentration in the endoplasmic reticulum. Redox Biol 2025; 80:103486. [PMID: 39787899 PMCID: PMC11763840 DOI: 10.1016/j.redox.2024.103486] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2024] [Revised: 12/09/2024] [Accepted: 12/30/2024] [Indexed: 01/12/2025] Open
Abstract
Aquaporin11 (AQP11) is an endoplasmic reticulum (ER) resident peroxiporin. It allows H2O2 transport from the lumen to the cytosol, guaranteeing redox homeostasis and signaling in and between the two organelles. Interestingly, Aqp11-/- mice develop a fatal, early onset polycystic kidney disease (PKD) similar to Autosomal Dominant PKD, a condition frequently associated with mutations of polycystin-1 (PC-1) in human patients. Here we investigated the molecular mechanisms of AQP11-associated PKD. Using different cell models, we show that transient downregulation of AQP11 selectively prevents the biogenesis of overexpressed PC-1. Expression of catalase in the ER lumen rescues the phenotype, demonstrating a direct role of (H2O2)ER in controlling the complex maturation of PC-1. Analysis of endogenous Pc-1 revealed an additional regulatory role at the pre-translational level. Taken together, our results show that AQP11 controls the complex biogenesis of PC-1 at multiple levels governing H2O2 intra and inter-organellar fluxes, with important implications in the pathogenesis and onset of PKD.
Collapse
Affiliation(s)
- Elisa Speranza
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Ilaria Sorrentino
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy
| | - Alessandra Boletta
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy
| | - Roberto Sitia
- Division of Genetics and Cell Biology, Istituto di Ricovero e Cura a Carattere Scientifico (IRCCS) Ospedale San Raffaele, 20132, Milan, Italy; Università Vita-Salute San Raffaele, 20132, Milan, Italy.
| |
Collapse
|
4
|
An N, Zhang X, Lin H, Xu Q, Dai Q, Kong Y, Han S, Li X, Yang X, Xing Y, Shang H. The role and mechanism of TXNDC5 in cardio-oncology: Killing two birds with one stone? Curr Probl Cardiol 2025; 50:102951. [PMID: 39643150 DOI: 10.1016/j.cpcardiol.2024.102951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Cardio-oncology has emerged as a new translational and clinical field owing to the growing repertory of cancer therapy. To date, there is a lack of effective pharmacological therapy to target cardiotoxicity. Cardio-oncology, which began by investigating the negative effects of cancer medicines on cardiovascular system, has since grown to include research into the similarities between cardiovascular disease (CVD) and cancer. Thioredoxin domain-containing protein 5 (TXNDC5) belongs to the protein disulfide isomerase (PDI) family. Many diseases, including CVD and cancer, improperly express TXNDC5. This review provides a comprehensive analysis of the expression patterns of TXNDC5 in diseases. It outlines the processes via which TXNDC5 contributes to the advancement of malignant diseases such as CVD and cancer. Additionally, it summarizes prospective therapeutic approaches that can be used to target TXNDC5 for the treatment of these diseases. This will offer novel perspectives for enhancing anticancer therapy and advancing cardio-oncology research and drug development.
Collapse
Affiliation(s)
- Na An
- DongZhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xiaoyu Zhang
- Institute of Basic Research in Clinical Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Hongyuan Lin
- College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China
| | - Qianqian Xu
- Institute of Basic Theory for Traditional Chinese Medicine, China Academy of Chinese Medical Sciences, Beijing, China
| | - Qianqian Dai
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - YiFan Kong
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Songjie Han
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xiao Li
- DongZhimen Hospital, Beijing University of Chinese Medicine, Beijing, China; Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China
| | - Xinyu Yang
- Fangshan Hospital Beijing University of Chinese Medicine, Beijing, China
| | - Yanwei Xing
- Guang'anmen Hospital, China Academy of Chinese Medical Sciences, Beijing, China.
| | - Hongcai Shang
- Key Laboratory of Chinese Internal Medicine of Ministry of Education, Beijing University of Chinese Medicine, Beijing, China; Dongfang Hospital, Beijing University of Chinese Medicine, Beijing, China; College of Integrated Chinese and Western Medicine, Hunan University of Chinese Medicine, Changsha, China.
| |
Collapse
|
5
|
Varone E, Retini M, Cherubini A, Chernorudskiy A, Marrazza A, Guidarelli A, Cagnotto A, Beeg M, Gobbi M, Fumagalli S, Bolis M, Guarrera L, Barbera MC, Grasselli C, Bleve A, Generali D, Milani M, Mari M, Salmona M, Piersanti G, Bottegoni G, Broggini M, Janssen-Heininger YMW, Cho J, Cantoni O, Zito E. Small molecule-mediated inhibition of the oxidoreductase ERO1A restrains aggressive breast cancer by impairing VEGF and PD-L1 in the tumor microenvironment. Cell Death Dis 2025; 16:105. [PMID: 39962052 PMCID: PMC11833095 DOI: 10.1038/s41419-025-07426-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2024] [Revised: 01/09/2025] [Accepted: 02/03/2025] [Indexed: 02/20/2025]
Abstract
Cancer cells adapt to harsh environmental conditions by inducing the Unfolded Protein Response (UPR), of which ERO1A is a mediator. ERO1A aids protein folding by acting as a protein disulfide oxidase, and under cancer-related hypoxia conditions, it favors the folding of angiogenic VEGFA, leading tumor cells to thrive and spread. The upregulation of ERO1A in cancer cells, oppositely to the dispensability of ERO1A activity in healthy cells, renders ERO1A a perfect target for cancer therapy. Here, we report the upregulation of ERO1A in a cohort of aggressive triple-negative breast cancer (TNBC) patients in which ERO1A levels correlate with a higher risk of breast tumor recurrence and metastatic spread. For ERO1A target validation and therapy in TNBC, we designed new ERO1A inhibitors in a structure-activity campaign of the prototype EN460. Cell-based screenings showed that the presence of the Micheal acceptor in the compound is necessary to engage the cysteine 397 of ERO1A but not sufficient to set out the inhibitory effect on ERO1A. Indeed, the ERO1 inhibitor must adopt a non-coplanar rearrangement within the ERO1A binding site. I2 and I3, two new EN460 analogs with different phenyl-substituted moieties, efficiently inhibited ERO1A, blunting VEGFA secretion. Accordingly, in vitro assays to measure ERO1A engagement and inhibition confirmed that I2 and I3 bind ERO1A and restrain its activity with a IC50 in a low micromolar range. EN460, I2 and I3 triggered breast cancer cytotoxicity while specifically inhibiting ERO1A in a dose-dependent manner. I2 more efficiently impaired cancer-relevant features such as VEGFA secretion and related cell migration. I2 also acted on the tumor microenvironment and viability of xenografts and syngeneic TNBC. Thus, small molecule-mediated ERO1A pharmacological inhibition is feasible and promises to lead to effective therapy for the still incurable TNBC.
Collapse
Affiliation(s)
- Ersilia Varone
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Michele Retini
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alessandro Cherubini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alexander Chernorudskiy
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- School of Medicine, Taizhou University, Taizhou, 318000, Zhejiang, China
| | - Alice Marrazza
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Alfredo Cagnotto
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marten Beeg
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Marco Gobbi
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Marco Bolis
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
- Bioinformatics Core Unit, Institute of Oncology Research (IOR), Bellinzona, Switzerland
| | - Luca Guarrera
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | | | - Chiara Grasselli
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Augusto Bleve
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Daniele Generali
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Manuela Milani
- U.O. Patologia Mammaria e Tumori Cerebrali, Azienda Socio-Sanitaria Territoriale, Cremona, Italia
| | - Michele Mari
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mario Salmona
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Giovanni Piersanti
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Giovanni Bottegoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
- Institute of Clinical Sciences, University of Birmingham, Edgbaston, B15 2TT, Birmingham, UK
| | - Massimo Broggini
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Yvonne M W Janssen-Heininger
- Departments of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Jaehyung Cho
- Division of Hematology, Department of Medicine and Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, USA
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy.
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
6
|
Heaton RA, Ball ST, Staunton CA, Mouly V, Jones SW, McArdle A, Jackson MJ. Peroxiredoxin 2 mediates redox-stimulated adaptations to oxidative phosphorylation induced by contractile activity in human skeletal muscle myotubes. Free Radic Biol Med 2025; 227:395-406. [PMID: 39643135 DOI: 10.1016/j.freeradbiomed.2024.11.053] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/27/2024] [Revised: 11/25/2024] [Accepted: 11/30/2024] [Indexed: 12/09/2024]
Abstract
Skeletal muscle generates superoxide during contractions, which is converted to hydrogen peroxide (H2O2). H2O2 has been proposed to activate signalling pathways and transcription factors that regulate adaptive responses to exercise, but the concentration required to oxidize and activate key redox-sensitive signalling proteins in vitro is much higher than the typical intracellular levels seen in muscle after exercise. We hypothesized that 2-Cys-peroxiredoxins (PRDX), which rapidly oxidize in the presence of physiological concentrations of H2O2, serve as intermediary signalling molecules and play a crucial role in activating adaptive pathways following muscle contractions. This study has examined the human muscle myotube responses to contractile activity, or exposure to low extracellular concentrations (2.5-5 μM) of H2O2 and whether knock down of muscle PRDX2 alters the differential gene expression (DEG) that results from these stresses. Exposure of human skeletal muscle myotubes to a 15 min period of aerobic electrically stimulated isometric contractions or 5 μM H2O2 induced substantial changes in DEG with modification of many genes associated with adaptations of skeletal muscle to contractile activity. Common DEG in these conditions included upregulation of genes associated with increased mitochondrial oxidative phosphorylation, including COX1, COX2, COX3 and ATP6. In myotubes with PRDX2 knock down (94 % decrease in PRDX2 mRNA), the upregulation of genes associated with increased mitochondrial oxidative phosphorylation was abolished following contractile activity or exposure to H2O2. These data indicate that a common effect of contractile activity and exposure to "physiological" levels of H2O2 in human myotubes is to increase the expression of multiple genes associated with increased mitochondrial oxidative phosphorylation. Furthermore, these effects were abolished in PRDX2 knock down myotubes indicating that adaptations to upregulate multiple genes related to increased mitochondrial capacity in human muscle myotubes in response to exercise is both redox regulated and requires PRDX2 as an essential mediator of the effects of H2O2.
Collapse
Affiliation(s)
- Robert A Heaton
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France
| | - Sam Tm Ball
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France
| | - Caroline A Staunton
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France
| | - Vincent Mouly
- Sorbonne Université, Inserm, Institut de Myologie, Centre de Recherche en Myologie, Paris, France
| | - Samantha W Jones
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France
| | - Anne McArdle
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France
| | - Malcolm J Jackson
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool L7 8TX, France.
| |
Collapse
|
7
|
Fujii J, Ochi H, Yamada S. A comprehensive review of peroxiredoxin 4, a redox protein evolved in oxidative protein folding coupled with hydrogen peroxide detoxification. Free Radic Biol Med 2025; 227:336-354. [PMID: 39643136 DOI: 10.1016/j.freeradbiomed.2024.12.015] [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] [Received: 10/14/2024] [Revised: 11/29/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Peroxiredoxin (PRDX) primarily employs electrons from thioredoxin in order to reduce peroxides. PRDX4 mainly resides either in the endoplasmic reticulum (ER) lumen or in extracellular spaces. Due to the usage of alternative promoters, a first exon is transcribed from different regions of the Prdx4 gene, which results in two types of mRNAs. The first type is designated as Prdx4. It is translated with a cleavable, hydrophobic signal sequence and is expressed in most cells throughout the body. The second type is designated as Prdx4t. The peroxidase activity of PRDX4 is involved in both the reduction of hydrogen peroxides and in the oxidative folding of nascent proteins in the ER. Prdx4 appears to have evolved from an ancestral gene in Eutherians simultaneously with the evolution of sperm protamine to cysteine-rich peptides, and, therefore, the testis-specific PRDX4t is likely involved in spermatogenesis through the oxidative folding of protamine. The dysfunction of PRDX4 leads to oxidative damage and ER stress, and is related to various diseases including diabetes and cancer. In this review article we refer to the results of biological and medical research in order to unveil the functional consequences of this unique member of the PRDX family.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, 2-2-2 Iidanishi, Yamagata, 990-9585, Japan.
| | - Haruki Ochi
- Institute for Promotion of Medical Science Research, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata, 990-9585, Japan
| | - Sohsuke Yamada
- Departments of Pathology and Laboratory Medicine, Kanazawa Medical University, 1-1 Daigaku, Uchinada, Ishikawa, 920-0293, Japan
| |
Collapse
|
8
|
Lai Y, Gao FF, Ge RT, Liu R, Ma S, Liu X. Metal ions overloading and cell death. Cell Biol Toxicol 2024; 40:72. [PMID: 39162885 PMCID: PMC11335907 DOI: 10.1007/s10565-024-09910-4] [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: 04/10/2024] [Accepted: 08/06/2024] [Indexed: 08/21/2024]
Abstract
Cell death maintains cell morphology and homeostasis during development by removing damaged or obsolete cells. The concentration of metal ions whithin cells is regulated by various intracellular transporters and repositories to maintain dynamic balance. External or internal stimuli might increase the concentration of metal ions, which results in ions overloading. Abnormal accumulation of large amounts of metal ions can lead to disruption of various signaling in the cell, which in turn can produce toxic effects and lead to the occurrence of different types of cell deaths. In order to further study the occurrence and development of metal ions overloading induced cell death, this paper reviewed the regulation of Ca2+, Fe3+, Cu2+ and Zn2+ metal ions, and the internal mechanism of cell death induced by overloading. Furthermore, we found that different metal ions possess a synergistic and competitive relationship in the regulation of cell death. And the enhanced level of oxidative stress was present in all the processes of cell death due to metal ions overloading, which possibly due to the combination of factors. Therefore, this review offers a theoretical foundation for the investigation of the toxic effects of metal ions, and presents innovative insights for targeted regulation and therapeutic intervention. HIGHLIGHTS: • Metal ions overloading disrupts homeostasis, which in turn affects the regulation of cell death. • Metal ions overloading can cause cell death via reactive oxygen species (ROS). • Different metal ions have synergistic and competitive relationships for regulating cell death.
Collapse
Affiliation(s)
- Yun Lai
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Fen Fen Gao
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Ruo Ting Ge
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Rui Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China
| | - Shumei Ma
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
| | - Xiaodong Liu
- School of Public Health, Wenzhou Medical University, Wenzhou, China.
- South Zhejiang Institute of Radiation Medicine and Nuclear Technology, Wenzhou Medical University, Wenzhou, China.
- Key Laboratory of Watershed Science and Health of Zhejiang Province, Wenzhou Medical University, Wenzhou, China.
| |
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
|
Hendershot LM, Buck TM, Brodsky JL. The Essential Functions of Molecular Chaperones and Folding Enzymes in Maintaining Endoplasmic Reticulum Homeostasis. J Mol Biol 2024; 436:168418. [PMID: 38143019 PMCID: PMC12015986 DOI: 10.1016/j.jmb.2023.168418] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2023] [Revised: 12/18/2023] [Accepted: 12/19/2023] [Indexed: 12/26/2023]
Abstract
It has been estimated that up to one-third of the proteins encoded by the human genome enter the endoplasmic reticulum (ER) as extended polypeptide chains where they undergo covalent modifications, fold into their native structures, and assemble into oligomeric protein complexes. The fidelity of these processes is critical to support organellar, cellular, and organismal health, and is perhaps best underscored by the growing number of disease-causing mutations that reduce the fidelity of protein biogenesis in the ER. To meet demands encountered by the diverse protein clientele that mature in the ER, this organelle is populated with a cadre of molecular chaperones that prevent protein aggregation, facilitate protein disulfide isomerization, and lower the activation energy barrier of cis-trans prolyl isomerization. Components of the lectin (glycan-binding) chaperone system also reside within the ER and play numerous roles during protein biogenesis. In addition, the ER houses multiple homologs of select chaperones that can recognize and act upon diverse peptide signatures. Moreover, redundancy helps ensure that folding-compromised substrates are unable to overwhelm essential ER-resident chaperones and enzymes. In contrast, the ER in higher eukaryotic cells possesses a single member of the Hsp70, Hsp90, and Hsp110 chaperone families, even though several homologs of these molecules reside in the cytoplasm. In this review, we discuss specific functions of the many factors that maintain ER quality control, highlight some of their interactions, and describe the vulnerabilities that arise from the absence of multiple members of some chaperone families.
Collapse
Affiliation(s)
- Linda M Hendershot
- Department of Tumor Cell Biology, St. Jude Children's Research Hospital, Memphis, TN 38105, United States.
| | - Teresa M Buck
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, United States
| | - Jeffrey L Brodsky
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, United States
| |
Collapse
|
11
|
Liu DD, Ding W, Cheng JT, Wei Q, Lin Y, Zhu TY, Tian J, Sun K, Zhang L, Lu P, Yang F, Liu C, Tang S, Yang B. Characterize direct protein interactions with enrichable, cleavable and latent bioreactive unnatural amino acids. Nat Commun 2024; 15:5221. [PMID: 38890329 PMCID: PMC11189575 DOI: 10.1038/s41467-024-49517-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 06/10/2024] [Indexed: 06/20/2024] Open
Abstract
Latent bioreactive unnatural amino acids (Uaas) have been widely used in the development of covalent drugs and identification of protein interactors, such as proteins, DNA, RNA and carbohydrates. However, it is challenging to perform high-throughput identification of Uaa cross-linking products due to the complexities of protein samples and the data analysis processes. Enrichable Uaas can effectively reduce the complexities of protein samples and simplify data analysis, but few cross-linked peptides were identified from mammalian cell samples with these Uaas. Here we develop an enrichable and multiple amino acids reactive Uaa, eFSY, and demonstrate that eFSY is MS cleavable when eFSY-Lys and eFSY-His are the cross-linking products. An identification software, AixUaa is developed to decipher eFSY mass cleavable data. We systematically identify direct interactomes of Thioredoxin 1 (Trx1) and Selenoprotein M (SELM) with eFSY and AixUaa.
Collapse
Affiliation(s)
- Dan-Dan Liu
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Wenlong Ding
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jin-Tao Cheng
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Qiushi Wei
- School of Biological Science and Medical Engineering & School of Engineering Medicine, Beihang University, Beijing, 100191, China
| | - Yinuo Lin
- State Key Laboratory of Respiratory Disease, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Tian-Yi Zhu
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Jing Tian
- State Key Laboratory of Respiratory Disease, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China
| | - Ke Sun
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Long Zhang
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China
| | - Peilong Lu
- Key Laboratory of Structural Biology of Zhejiang Province, School of Life Sciences, Westlake University, Hangzhou, Zhejiang, 310030, China
| | - Fan Yang
- Department of Biophysics, Kidney Disease Center of the First Affiliated Hospital, Zhejiang University School of Medicine, Hangzhou, Zhejiang, 310058, China
| | - Chao Liu
- School of Biological Science and Medical Engineering & School of Engineering Medicine, Beihang University, Beijing, 100191, China.
| | - Shibing Tang
- State Key Laboratory of Respiratory Disease, Center for Chemical Biology and Drug Discovery, Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, Guangdong, 510530, China.
- China-New Zealand Joint Laboratory on Biomedicine and Health, Guangzhou, 510530, China.
| | - Bing Yang
- Life Sciences Institute, Department of Medical Oncology, The Second Affiliated Hospital of Zhejiang University School of Medicine, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
- Cancer Center, Zhejiang University, Hangzhou, Zhejiang, 310058, China.
| |
Collapse
|
12
|
Jiao M, Zhang Y, Song X, Xu B. The role and mechanism of TXNDC5 in disease progression. Front Immunol 2024; 15:1354952. [PMID: 38629066 PMCID: PMC11019510 DOI: 10.3389/fimmu.2024.1354952] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2023] [Accepted: 03/19/2024] [Indexed: 04/19/2024] Open
Abstract
Thioredoxin domain containing protein-5 (TXNDC5), also known as endothelial protein-disulfide isomerase (Endo-PDI), is confined to the endoplasmic reticulum through the structural endoplasmic reticulum retention signal (KDEL), is a member of the PDI protein family and is highly expressed in the hypoxic state. TXNDC5 can regulate the rate of disulfide bond formation, isomerization and degradation of target proteins through its function as a protein disulfide isomerase (PDI), thereby altering protein conformation, activity and improving protein stability. Several studies have shown that there is a significant correlation between TXNDC5 gene polymorphisms and genetic susceptibility to inflammatory diseases such as rheumatoid, fibrosis and tumors. In this paper, we detail the expression characteristics of TXNDC5 in a variety of diseases, summarize the mechanisms by which TXNDC5 promotes malignant disease progression, and summarize potential therapeutic strategies to target TXNDC5 for disease treatment.
Collapse
Affiliation(s)
- Mingxia Jiao
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Province Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| | - Yeyong Zhang
- Department of Orthopedic Surgery, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Shandong Key Laboratory of Rheumatic Disease and Translational Medicine, Jinan, Shandong, China
| | - Xie Song
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
- Department of Hepatobiliary Surgery, Shandong Provincial Hospital affiliated to Shandong First Medical University, Jinan, Shandong, China
| | - Bing Xu
- Department of Urology, The First Affiliated Hospital of Shandong First Medical University & Shandong Province Qianfoshan Hospital, Shandong Medicine and Health Key Laboratory of Organ Transplantation and Nephrosis, Shandong Institute of Nephrology, Jinan, Shandong, China
- Shandong Provincial Key Laboratory for Rheumatic Disease and Translational Medicine, The First Affiliated Hospital of Shandong First Medical University & Shandong Provincial Qianfoshan Hospital, Jinan, Shandong, China
| |
Collapse
|
13
|
Solanki K, Bezsonov E, Orekhov A, Parihar SP, Vaja S, White FA, Obukhov AG, Baig MS. Effect of reactive oxygen, nitrogen, and sulfur species on signaling pathways in atherosclerosis. Vascul Pharmacol 2024; 154:107282. [PMID: 38325566 DOI: 10.1016/j.vph.2024.107282] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2024] [Accepted: 02/04/2024] [Indexed: 02/09/2024]
Abstract
Atherosclerosis is a chronic inflammatory disease in which fats, lipids, cholesterol, calcium, proliferating smooth muscle cells, and immune cells accumulate in the intima of the large arteries, forming atherosclerotic plaques. A complex interplay of various vascular and immune cells takes place during the initiation and progression of atherosclerosis. Multiple reports indicate that tight control of reactive oxygen species (ROS), reactive nitrogen species (RNS), and reactive sulfur species (RSS) production is critical for maintaining vascular health. Unrestricted ROS and RNS generation may lead to activation of various inflammatory signaling pathways, facilitating atherosclerosis. Given these deleterious consequences, it is important to understand how ROS and RNS affect the signaling processes involved in atherogenesis. Conversely, RSS appears to exhibit an atheroprotective potential and can alleviate the deleterious effects of ROS and RNS. Herein, we review the literature describing the effects of ROS, RNS, and RSS on vascular smooth muscle cells, endothelial cells, and macrophages and focus on how changes in their production affect the initiation and progression of atherosclerosis. This review also discusses the contribution of ROS, RNS, and RSS in mediating various post-translational modifications, such as oxidation, nitrosylation, and sulfation, of the molecules involved in inflammatory signaling.
Collapse
Affiliation(s)
- Kundan Solanki
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Evgeny Bezsonov
- Laboratory of Angiopathology, Institute of General Pathology and Pathophysiology, Moscow, Russia; Laboratory of Cellular and Molecular Pathology of Cardiovascular System, Avtsyn Research Institute of Human Morphology, Petrovsky National Research Centre of Surgery, Moscow, Russia; Department of Biology and General Genetics, Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia; The Cell Physiology and Pathology Laboratory, Turgenev State University of Orel, Orel, Russia
| | - Alexander Orekhov
- Institute for Atherosclerosis Research, Skolkovo Innovative Center, Moscow, Russia
| | - Suraj P Parihar
- Wellcome Centre for Infectious Diseases Research in Africa (CIDRI-Africa) and Institute of Infectious Diseases and Molecular Medicine (IDM), Division of Medical Microbiology, Faculty of Health Sciences, University of Cape Town, Cape Town, South Africa; Department of Biochemistry, Human Metabolomics, Faculty of Natural and Agricultural Sciences, North-West University, Potchefstroom, South Africa
| | - Shivani Vaja
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India
| | - Fletcher A White
- Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Department of Anesthesia, Indiana University School of Medicine, Indianapolis, IN 46202, USA
| | - Alexander G Obukhov
- Department of Anatomy, Cell Biology & Physiology, Indiana University School of Medicine, Indianapolis, IN 46202, USA; Stark Neurosciences Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202, USA.
| | - Mirza S Baig
- Department of Biosciences and Biomedical Engineering (BSBE), Indian Institute of Technology Indore (IITI), Simrol, Indore, India.
| |
Collapse
|
14
|
Shu P, Liang H, Zhang J, Lin Y, Chen W, Zhang D. Reactive oxygen species formation and its effect on CD4 + T cell-mediated inflammation. Front Immunol 2023; 14:1199233. [PMID: 37304262 PMCID: PMC10249013 DOI: 10.3389/fimmu.2023.1199233] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Accepted: 05/16/2023] [Indexed: 06/13/2023] Open
Abstract
Reactive oxygen species (ROS) are produced both enzymatically and non-enzymatically in vivo. Physiological concentrations of ROS act as signaling molecules that participate in various physiological and pathophysiological activities and play an important role in basic metabolic functions. Diseases related to metabolic disorders may be affected by changes in redox balance. This review details the common generation pathways of intracellular ROS and discusses the damage to physiological functions when the ROS concentration is too high to reach an oxidative stress state. We also summarize the main features and energy metabolism of CD4+ T-cell activation and differentiation and the effects of ROS produced during the oxidative metabolism of CD4+ T cells. Because the current treatment for autoimmune diseases damages other immune responses and functional cells in the body, inhibiting the activation and differentiation of autoreactive T cells by targeting oxidative metabolism or ROS production without damaging systemic immune function is a promising treatment option. Therefore, exploring the relationship between T-cell energy metabolism and ROS and the T-cell differentiation process provides theoretical support for discovering effective treatments for T cell-mediated autoimmune diseases.
Collapse
Affiliation(s)
| | | | | | | | | | - Dunfang Zhang
- Department of Biotherapy, State Key Laboratory of Biotherapy and Cancer Center, Collaborative Innovation Center of Biotherapy, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| |
Collapse
|
15
|
Thapa P, Jiang H, Ding N, Hao Y, Alshahrani A, Wei Q. The Role of Peroxiredoxins in Cancer Development. BIOLOGY 2023; 12:666. [PMID: 37237480 PMCID: PMC10215932 DOI: 10.3390/biology12050666] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/04/2023] [Revised: 04/21/2023] [Accepted: 04/26/2023] [Indexed: 05/28/2023]
Abstract
Peroxiredoxins (Prxs) are antioxidant enzymes with ubiquitous expression in human tissues. Prxs are expressed in archaea, bacteria, and eukaryota, often in multiple isoforms. Because of their abundant expression in different cellular organelles and extraordinary sensitivity to H2O2, Prxs are among the first defenses against oxidative stress. Prxs undergo reversible oxidation to disulfides, and some family members perform chaperone or phospholipase functions upon further oxidation. Prxs are upregulated in cancer cells. Research has suggested that Prxs can function as tumor promoters in various cancers. The major objective of this review is to summarize novel findings regarding the roles of Prxs in common cancer types. Prxs have been shown to influence differentiation of inflammatory cells and fibroblasts, remodeling of extracellular matrix, and regulation of stemness. Since aggressive cancer cells have higher intracellular levels of ROS that they can utilize to proliferate and metastasize compared to normal cells, it is critical that we understand the regulation and functions of primary antioxidants such as Prxs. These small but mighty proteins could prove to be key for improving cancer therapeutics and patient survival.
Collapse
Affiliation(s)
- Pratik Thapa
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Hong Jiang
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Na Ding
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Yanning Hao
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Aziza Alshahrani
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
| | - Qiou Wei
- Department of Toxicology and Cancer Biology, University of Kentucky College of Medicine, 1095 Veterans Dr, Lexington, KY 40508, USA
- Markey Cancer Center, University of Kentucky College of Medicine, 800 Rose Street, Lexington, KY 40536, USA
| |
Collapse
|
16
|
In silico analysis reveals PRDX4 as a prognostic and oncogenic marker in renal papillary cell carcinoma. Gene 2023; 859:147201. [PMID: 36646187 DOI: 10.1016/j.gene.2023.147201] [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: 11/23/2022] [Revised: 01/07/2023] [Accepted: 01/10/2023] [Indexed: 01/14/2023]
Abstract
BACKGROUND Alterations in the tumor microenvironment leads to the accumulation of reactive oxygen species (ROS). When in low levels, ROS act as a signaling molecule and contribute to tumor cell proliferation whereas its elevation results in oxidative stress and eventually cell death. It is known that antioxidant systems regulate the ROS levels and thus cell fate. Among these systems, peroxiredoxins (PRDXs) were found to be upregulated in various cancers. However their exact contribution to carcinogenesis is not yet clear. AIM Herein, the expression pattern and prognostic value of PRDXs were explored in cancer setting by using in silico analysis tools and publicly available datasets. RESULTS Pan-cancer analysis revealed that PRDXs are differentially expressed in normal and tumor tissues. Further analysis showed that higher PRDX4 levels was associated with poor prognosis and clinicopathological and histological features associated with a more aggressive renal papillary cell carcinoma (KIRP) profile. Hypoxia, ER stress and protein folding were shown to be pathways positively correlated with PRDX4 levels. Furthermore, PRDX4 was found to be strong regulator of protein homeostasis. Kaplan-Meier analysis revealed that PRDX4 is a potent prognostic marker in Type 2 KIRP and this might be due to increased ER stress and oxidative stress levels in this subtype. CONCLUSIONS The data suggest that PRDX4 can be used as a prognostic marker for KIRP patients. Its association with more aggressive tumor characteristics also underlines that it might be used for targeted therapy.
Collapse
|
17
|
Wang L, Wang CC. Oxidative protein folding fidelity and redoxtasis in the endoplasmic reticulum. Trends Biochem Sci 2023; 48:40-52. [PMID: 35871147 DOI: 10.1016/j.tibs.2022.06.011] [Citation(s) in RCA: 43] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2022] [Revised: 06/16/2022] [Accepted: 06/29/2022] [Indexed: 02/09/2023]
Abstract
In eukaryotic cells, oxidative protein folding occurs in the lumen of the endoplasmic reticulum (ER), catalyzed by ER sulfhydryl oxidase 1 (Ero1) and protein disulfide isomerase (PDI). The efficiency and fidelity of oxidative protein folding are vital for the function of secretory cells. Here, we summarize oxidative protein folding in yeast, plants, and mammals, and discuss how the conformation and activity of human Ero1-PDI machinery is regulated through various post-translational modifications (PTMs). We propose that oxidative protein folding fidelity and ER redox homeostasis are maintained by both the precise control of Ero1 oxidase activity and the division of labor between PDI family members. We also discuss how deregulated Ero1-PDI functions contribute to human diseases and can be leveraged for therapeutic interventions.
Collapse
Affiliation(s)
- Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China.
| | - Chih-Chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing 100101, China; College of Life Sciences, University of Chinese Academy of Sciences, Beijing 100049, China
| |
Collapse
|
18
|
Essential Roles of Peroxiredoxin IV in Inflammation and Cancer. Molecules 2022; 27:molecules27196513. [PMID: 36235049 PMCID: PMC9573489 DOI: 10.3390/molecules27196513] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2022] [Revised: 09/21/2022] [Accepted: 09/29/2022] [Indexed: 11/22/2022] Open
Abstract
Peroxiredoxin IV (Prx4) is a 2-Cysteine peroxidase with ubiquitous expression in human tissues. Prx4 scavenges hydrogen peroxide and participates in oxidative protein folding in the endoplasmic reticulum. In addition, Prx4 is secreted outside the cell. Prx4 is upregulated in several cancers and is a potential therapeutic target. We have summarized historical and recent advances in the structure, function and biological roles of Prx4, focusing on inflammatory diseases and cancer. Oxidative stress is known to activate pro-inflammatory pathways. Chronic inflammation is a risk factor for cancer development. Hence, redox enzymes such as Prx4 are important players in the crosstalk between inflammation and cancer. Understanding molecular mechanisms of regulation of Prx4 expression and associated signaling pathways in normal physiological and disease conditions should reveal new therapeutic strategies. Thus, although Prx4 is a promising therapeutic target for inflammatory diseases and cancer, further research needs to be conducted to bridge the gap to clinical application.
Collapse
|
19
|
Varone E, Chernorudskiy A, Cherubini A, Cattaneo A, Bachi A, Fumagalli S, Erol G, Gobbi M, Lenardo MJ, Borgese N, Zito E. ERO1 alpha deficiency impairs angiogenesis by increasing N-glycosylation of a proangiogenic VEGFA. Redox Biol 2022; 56:102455. [PMID: 36063727 PMCID: PMC9463388 DOI: 10.1016/j.redox.2022.102455] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2022] [Revised: 08/16/2022] [Accepted: 08/22/2022] [Indexed: 11/23/2022] Open
Abstract
N-glycosylation and disulfide bond formation are two essential steps in protein folding that occur in the endoplasmic reticulum (ER) and reciprocally influence each other. Here, to analyze crosstalk between N-glycosylation and oxidation, we investigated how the protein disulfide oxidase ERO1-alpha affects glycosylation of the angiogenic VEGF121, a key regulator of vascular homeostasis. ERO1 deficiency, while retarding disulfide bond formation in VEGF121, increased utilization of its single N-glycosylation sequon, which lies close to an intra-polypeptide disulfide bridge, and concomitantly slowed its secretion. Unbiased mass-spectrometric analysis revealed interactions between VEGF121 and N-glycosylation pathway proteins in ERO1-knockout (KO), but not wild-type cells. Notably, MAGT1, a thioredoxin-containing component of the post-translational oligosaccharyltransferase complex, was a major hit exclusive to ERO1-deficient cells. Thus, both a reduced rate of formation of disulfide bridges, and the increased trapping potential of MAGT1 may increase N-glycosylation of VEGF121. Extending our investigation to tissues, we observed altered lectin staining of ERO1 KO breast tumor xenografts, implicating ERO1 as a physiologic regulator of protein N-glycosylation. Our study, highlighting the effect of ERO1 loss on N-glycosylation of proteins, is particularly relevant not only to angiogenesis but also to other cancer patho-mechanisms in light of recent findings suggesting a close causal link between alterations in protein glycosylation and cancer development.
Collapse
|
20
|
Rohli KE, Boyer CK, Bearrows SC, Moyer MR, Elison WS, Bauchle CJ, Blom SE, Zhang J, Wang Y, Stephens SB. ER Redox Homeostasis Regulates Proinsulin Trafficking and Insulin Granule Formation in the Pancreatic Islet β-Cell. FUNCTION 2022; 3:zqac051. [PMID: 36325514 PMCID: PMC9614934 DOI: 10.1093/function/zqac051] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2022] [Revised: 09/11/2022] [Accepted: 09/21/2022] [Indexed: 01/07/2023] Open
Abstract
Defects in the pancreatic β-cell's secretion system are well-described in type 2 diabetes (T2D) and include impaired proinsulin processing and a deficit in mature insulin-containing secretory granules; however, the cellular mechanisms underlying these defects remain poorly understood. To address this, we used an in situ fluorescent pulse-chase strategy to study proinsulin trafficking. We show that insulin granule formation and the appearance of nascent granules at the plasma membrane are decreased in rodent and cell culture models of prediabetes and hyperglycemia. Moreover, we link the defect in insulin granule formation to an early trafficking delay in endoplasmic reticulum (ER) export of proinsulin, which is independent of overt ER stress. Using a ratiometric redox sensor, we show that the ER becomes hyperoxidized in β-cells from a dietary model of rodent prediabetes and that addition of reducing equivalents restores ER export of proinsulin and insulin granule formation and partially restores β-cell function. Together, these data identify a critical role for the regulation of ER redox homeostasis in proinsulin trafficking and suggest that alterations in ER redox poise directly contribute to the decline in insulin granule production in T2D. This model highlights a critical link between alterations in ER redox and ER function with defects in proinsulin trafficking in T2D. Hyperoxidation of the ER lumen, shown as hydrogen peroxide, impairs proinsulin folding and disulfide bond formation that prevents efficient exit of proinsulin from the ER to the Golgi. This trafficking defect limits available proinsulin for the formation of insulin secretory granules during the development of T2D.
Collapse
Affiliation(s)
- Kristen E Rohli
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Cierra K Boyer
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Shelby C Bearrows
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Marshall R Moyer
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Weston S Elison
- Department of Nutrition, Dietetics, and Food Science, Brigham Young University, Provo, UT 84602, USA
| | - Casey J Bauchle
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Sandra E Blom
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| | - Jianchao Zhang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48103, USA
| | - Yanzhuang Wang
- Department of Molecular, Cellular, and Developmental Biology, University of Michigan, Ann Arbor, MI 48103, USA
- Department of Neurology, School of Medicine, University of Michigan, Ann Arbor, MI 48103, USA
| | - Samuel B Stephens
- Fraternal Order of Eagles Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
- Interdisciplinary Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
- Department of Internal Medicine, Division of Endocrinology and Metabolism, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
21
|
Ugalde JM, Aller I, Kudrjasova L, Schmidt RR, Schlößer M, Homagk M, Fuchs P, Lichtenauer S, Schwarzländer M, Müller-Schüssele SJ, Meyer AJ. Endoplasmic reticulum oxidoreductin provides resilience against reductive stress and hypoxic conditions by mediating luminal redox dynamics. THE PLANT CELL 2022; 34:4007-4027. [PMID: 35818121 PMCID: PMC9516139 DOI: 10.1093/plcell/koac202] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Accepted: 07/05/2022] [Indexed: 05/28/2023]
Abstract
Oxidative protein folding in the endoplasmic reticulum (ER) depends on the coordinated action of protein disulfide isomerases and ER oxidoreductins (EROs). Strict dependence of ERO activity on molecular oxygen as the final electron acceptor implies that oxidative protein folding and other ER processes are severely compromised under hypoxia. Here, we isolated viable Arabidopsis thaliana ero1 ero2 double mutants that are highly sensitive to reductive stress and hypoxia. To elucidate the specific redox dynamics in the ER in vivo, we expressed the glutathione redox potential (EGSH) sensor Grx1-roGFP2iL-HDEL with a midpoint potential of -240 mV in the ER of Arabidopsis plants. We found EGSH values of -241 mV in wild-type plants, which is less oxidizing than previously estimated. In the ero1 ero2 mutants, luminal EGSH was reduced further to -253 mV. Recovery to reductive ER stress induced by dithiothreitol was delayed in ero1 ero2. The characteristic signature of EGSH dynamics in the ER lumen triggered by hypoxia was affected in ero1 ero2 reflecting a disrupted balance of reductive and oxidizing inputs, including nascent polypeptides and glutathione entry. The ER redox dynamics can now be dissected in vivo, revealing a central role of EROs as major redox integrators to promote luminal redox homeostasis.
Collapse
Affiliation(s)
| | - Isabel Aller
- INRES-Chemical Signalling, University of Bonn, D-53113 Bonn, Germany
| | - Lika Kudrjasova
- INRES-Chemical Signalling, University of Bonn, D-53113 Bonn, Germany
| | - Romy R Schmidt
- Plant Biotechnology, Bielefeld University, D-33615 Bielefeld, Germany
| | - Michelle Schlößer
- INRES-Chemical Signalling, University of Bonn, D-53113 Bonn, Germany
| | - Maria Homagk
- INRES-Chemical Signalling, University of Bonn, D-53113 Bonn, Germany
| | | | - Sophie Lichtenauer
- Institute for Biology and Biotechnology of Plants, University of Münster, D-48143 Münster, Germany
| | - Markus Schwarzländer
- Institute for Biology and Biotechnology of Plants, University of Münster, D-48143 Münster, Germany
| | - Stefanie J Müller-Schüssele
- INRES-Chemical Signalling, University of Bonn, D-53113 Bonn, Germany
- Molecular Botany, Department of Biology, TU Kaiserslautern, D-67663, Kaiserslautern, Germany
| | | |
Collapse
|
22
|
West JD. Experimental Approaches for Investigating Disulfide-Based Redox Relays in Cells. Chem Res Toxicol 2022; 35:1676-1689. [PMID: 35771680 DOI: 10.1021/acs.chemrestox.2c00123] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Reversible oxidation of cysteine residues within proteins occurs naturally during normal cellular homeostasis and can increase during oxidative stress. Cysteine oxidation often leads to the formation of disulfide bonds, which can impact protein folding, stability, and function. Work in both prokaryotic and eukaryotic models over the past five decades has revealed several multiprotein systems that use thiol-dependent oxidoreductases to mediate disulfide bond reduction, formation, and/or rearrangement. Here, I provide an overview of how these systems operate to carry out disulfide exchange reactions in different cellular compartments, with a focus on their roles in maintaining redox homeostasis, transducing redox signals, and facilitating protein folding. Additionally, I review thiol-independent and thiol-dependent approaches for interrogating what proteins partner together in such disulfide-based redox relays. While the thiol-independent approaches rely either on predictive measures or standard procedures for monitoring protein-protein interactions, the thiol-dependent approaches include direct disulfide trapping methods as well as thiol-dependent chemical cross-linking. These strategies may prove useful in the systematic characterization of known and newly discovered disulfide relay mechanisms and redox switches involved in oxidant defense, protein folding, and cell signaling.
Collapse
Affiliation(s)
- James D West
- Biochemistry & Molecular Biology Program, Departments of Biology and Chemistry, The College of Wooster, Wooster, Ohio 44691, United States
| |
Collapse
|
23
|
Isomoto A, Shoguchi E, Hisata K, Inoue J, Sun Y, Inaba K, Satoh N, Ogawa T, Shibata H. Active Expression of Genes for Protein Modification Enzymes in Habu Venom Glands. Toxins (Basel) 2022; 14:toxins14050300. [PMID: 35622547 PMCID: PMC9146206 DOI: 10.3390/toxins14050300] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 04/09/2022] [Accepted: 04/16/2022] [Indexed: 01/27/2023] Open
Abstract
Genes encoding snake venom toxins have been studied extensively. However, genes involved in the modification and functioning of venom proteins are little known. Protobothrops is a genus of pit vipers, which are venomous and inhabit the Nansei (Southwest) islands of Japan, Taiwan China, Vietnam, Thailand, Myanmar, Nepal, Bhutan, and India. Our previous study decoded the genome of Protobothrops flavoviridis, a species endemic to the Nansei Islands, Japan, and revealed unique evolutionary processes of some venom genes. In this study, we analyzed genes that are highly expressed in venom glands to survey genes for candidate enzymes or chaperone proteins involved in toxin folding and modification. We found that, in addition to genes that encode venom proteins and ribosomal proteins, genes that encode protein disulfide isomerase (PDI) family members (orthologs of human P4HB and PDIA3), Selenoprotein M (SELENOM), and Calreticulin (CALR) are highly expressed in venom glands. Since these enzymes or chaperones are involved in protein modification and potentially possess protein folding functions, we propose that P4HB, SELENOM, CALR, and PDIA3 encode candidate enzymes or chaperones to confer toxic functions upon the venom transcriptome.
Collapse
Affiliation(s)
- Akiko Isomoto
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan;
- Department of Biological Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: (A.I.); (T.O.); (H.S.)
| | - Eiichi Shoguchi
- Marine Genomics Unit, Okinawa Institute of Science, Technology Graduate University, Onna 904-0495, Japan; (E.S.); (K.H.); (J.I.); (N.S.)
| | - Kanako Hisata
- Marine Genomics Unit, Okinawa Institute of Science, Technology Graduate University, Onna 904-0495, Japan; (E.S.); (K.H.); (J.I.); (N.S.)
| | - Jun Inoue
- Marine Genomics Unit, Okinawa Institute of Science, Technology Graduate University, Onna 904-0495, Japan; (E.S.); (K.H.); (J.I.); (N.S.)
| | - Yinrui Sun
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan;
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Material, Tohoku University, Sendai 980-8577, Japan;
- Department of Biomolecular Science, Graduate School of Life Sciences, Tohoku University, Sendai 980-8577, Japan
| | - Noriyuki Satoh
- Marine Genomics Unit, Okinawa Institute of Science, Technology Graduate University, Onna 904-0495, Japan; (E.S.); (K.H.); (J.I.); (N.S.)
| | - Tomohisa Ogawa
- Department of Biological Chemistry, Graduate School of Agricultural Science, Tohoku University, Sendai 980-8572, Japan
- Correspondence: (A.I.); (T.O.); (H.S.)
| | - Hiroki Shibata
- Division of Genomics, Medical Institute of Bioregulation, Kyushu University, Fukuoka 812-8582, Japan;
- Correspondence: (A.I.); (T.O.); (H.S.)
| |
Collapse
|
24
|
Qiao X, Zhang Y, Ye A, Zhang Y, Xie T, Lv Z, Shi C, Wu D, Chu B, Wu X, Zhang W, Wang P, Liu GH, Wang CC, Wang L, Chen C. ER reductive stress caused by Ero1α S-nitrosation accelerates senescence. Free Radic Biol Med 2022; 180:165-178. [PMID: 35033630 DOI: 10.1016/j.freeradbiomed.2022.01.006] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/17/2021] [Revised: 12/24/2021] [Accepted: 01/10/2022] [Indexed: 02/07/2023]
Abstract
Oxidative stress in aging has attracted much attention; however, the role of reductive stress in aging remains largely unknown. Here, we report that the endoplasmic reticulum (ER) undergoes reductive stress during replicative senescence, as shown by specific glutathione and H2O2 fluorescent probes. We constructed an ER-specific reductive stress cell model by ER-specific catalase overexpression and observed accelerated senescent phenotypes accompanied by disrupted proteostasis and a compromised ER unfolded protein response (UPR). Mechanistically, S-nitrosation of the pivotal ER sulfhydryl oxidase Ero1α led to decreased activity, therefore resulting in reductive stress in the ER. Inhibition of inducible nitric oxide synthase decreased the level of Ero1α S-nitrosation and decreased cellular senescence. Moreover, the expression of constitutively active Ero1α restored an oxidizing state in the ER and successfully rescued the senescent phenotypes. Our results uncover a new mechanism of senescence promoted by ER reductive stress and provide proof-of-concept that maintaining the oxidizing power of the ER and organelle-specific precision redox regulation could be valuable future geroprotective strategies.
Collapse
Affiliation(s)
- Xinhua Qiao
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yingmin Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Aojun Ye
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yini Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Ting Xie
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Zhenyu Lv
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Chang Shi
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Dongli Wu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; School of Basic Medical Sciences of Southwest Medical University, Luzhou, 646000, China
| | - Boyu Chu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Xun Wu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Weiqi Zhang
- CAS Key Laboratory of Genomic and Precision Medicine, Beijing Institute of Genomics, Chinese Academy of Sciences, Beijing, 100101, China
| | - Ping Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Guang-Hui Liu
- State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101, China
| | - Chih-Chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Chang Chen
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, 100101, China; University of Chinese Academy of Sciences, Beijing, 100049, China; Beijing Institute for Brain Disorders, Capital Medical University, Beijing, 100069, China.
| |
Collapse
|
25
|
Liang X, Zhu D, Li Y, Liao L, Yang C, Liu L, Huang R, Wang W, Zhu Z, He L, Wang Y. Molecular characterization and functional analysis of peroxiredoxin 4 in grass carp (Ctenopharyngodon idella). DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2021; 125:104213. [PMID: 34324900 DOI: 10.1016/j.dci.2021.104213] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 07/22/2021] [Accepted: 07/23/2021] [Indexed: 06/13/2023]
Abstract
Peroxiredoxins (Prxs) are a group of evolutionarily conserved selenium-independent thiol-specific antioxidant proteins. In this study, the peroxiredoxin-4 (CiPrx4) gene from grass carp was identified and characterized. The full-length of CiPrx4 is 1339 bp, encoding 260 amino acids that contain two peroxiredoxin signature motifs and two GVL motifs. CiPrx4 belongs to the typical 2-Cys subfamily and shows the highest homology with Prx4 from Cyprinus carpio (95.4%). CiPrx4 mRNA was constitutively expressed in all tested tissues and was upregulated by grass carp reovirus and pathogen-associated molecular pattern (PAMP) stimulation. CiPrx4 was localized in the cytoplasm and co-localized with the endoplasmic reticulum. The purified CiPrx4 protein protected DNA from degradation in a dose-dependent manner. Moreover, the overexpression of CiPrx4 in Escherichia coli and fish cells showed apparent antioxidant and antiviral activities. Collectively, the results of the present study provide new insights for further understanding the functions of Prx4 in teleost fish.
Collapse
Affiliation(s)
- Xinyu Liang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Denghui Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China
| | - Yongming Li
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Lanjie Liao
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Cheng Yang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Liyue Liu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; China Zebrafish Resource Center, National Aquatic Biological Resource Center, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Rong Huang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Weiyan Wang
- The Yellow Lake Fishery Co. Ltd of Anhui Province, Anqing, 246524, China
| | - Zuoyan Zhu
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China
| | - Libo He
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; University of Chinese Academy of Sciences, Beijing, 100049, China.
| | - Yaping Wang
- State Key Laboratory of Freshwater Ecology and Biotechnology, Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, 430072, China; Innovative Academy of Seed Design, Chinese Academy of Sciences, Beijing, 100101, China.
| |
Collapse
|
26
|
Abstract
Import and oxidative folding of proteins in the mitochondrial intermembrane space differ among eukaryotic lineages. While opisthokonts such as yeast rely on the receptor and oxidoreductase Mia40 in combination with the Mia40:cytochrome c oxidoreductase Erv, kinetoplastid parasites and other Excavata/Discoba lack Mia40 but have a functional Erv homologue. Whether excavate Erv homologues rely on a Mia40 replacement or directly interact with imported protein substrates remains controversial. Here, we used the CRISPR-Cas9 system to generate a set of tagged and untagged homozygous mutants of LTERV from the kinetoplastid model parasite Leishmania tarentolae. Modifications of the shuttle cysteine motif of LtErv were lethal, whereas replacement of clamp residue Cys17 or removal of the kinetoplastida-specific second (KISS) domain had no impact on parasite viability under standard growth conditions. However, removal of the KISS domain rendered parasites sensitive to heat stress and led to the accumulation of homodimeric and mixed LtErv disulfides. We therefore determined and compared the redox interactomes of tagged wild-type LtErv and LtErvΔKISS using stable isotope labeling by amino acids in cell culture (SILAC) and quantitative mass spectrometry. While the Mia40-replacement candidate Mic20 and all but one typical substrate with twin Cx3/9C-motifs were absent in both redox interactomes, we identified a small set of alternative potential interaction partners with putative redox-active cysteine residues. In summary, our study reveals parasite-specific intracellular structure-function relationships and redox interactomes of LtErv with implications for current hypotheses on mitochondrial protein import in nonopisthokonts. IMPORTANCE The discovery of the redox proteins Mia40/CHCHD4 and Erv1/ALR, as well as the elucidation of their relevance for oxidative protein folding in the mitochondrial intermembrane space of yeast and mammals, founded a new research topic in redox biology and mitochondrial protein import. The lack of Mia40/CHCHD4 in protist lineages raises fundamental and controversial questions regarding the conservation and evolution of this essential pathway. Do protist Erv homologues act alone, or do they use the candidate Mic20 or another protein as a Mia40 replacement? Furthermore, we previously showed that Erv homologues in L. tarentolae and the human pathogen L. infantum are not only essential but also differ structurally and mechanistically from yeast and human Erv1/ALR. Here, we analyzed the relevance of such structural differences in vivo and determined the first redox interactomes of a nonopisthokont Erv homologue. Our data challenge recent hypotheses on mitochondrial protein import in nonopisthokonts.
Collapse
|
27
|
Buchmann GK, Schürmann C, Spaeth M, Abplanalp W, Tombor L, John D, Warwick T, Rezende F, Weigert A, Shah AM, Hansmann ML, Weissmann N, Dimmeler S, Schröder K, Brandes RP. The hydrogen-peroxide producing NADPH oxidase 4 does not limit neointima development after vascular injury in mice. Redox Biol 2021; 45:102050. [PMID: 34218201 PMCID: PMC8256285 DOI: 10.1016/j.redox.2021.102050] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2021] [Revised: 06/15/2021] [Accepted: 06/15/2021] [Indexed: 11/26/2022] Open
Abstract
Objective The NADPH oxidase Nox4 is an important source of H2O2. Nox4-derived H2O2 limits vascular inflammation and promotes smooth muscle differentiation. On this basis, the role of Nox4 for restenosis development was determined in the mouse carotid artery injury model. Methods and results Genetic deletion of Nox4 by a tamoxifen-activated Cre-Lox-system did not impact on neointima formation in the carotid artery wire injury model. To understand this unexpected finding, time-resolved single-cell RNA-sequencing (scRNAseq) from injured carotid arteries of control mice and massive-analysis-of-cDNA-ends (MACE)-RNAseq from the neointima harvested by laser capture microdissection of control and Nox4 knockout mice was performed. This revealed that resting smooth muscle cells (SMCs) and fibroblasts exhibit high Nox4 expression, but that the proliferating de-differentiated SMCs, which give rise to the neointima, have low Nox4 expression. In line with this, the first weeks after injury, gene expression was unchanged between the carotid artery neointimas of control and Nox4 knockout mice. Conclusion Upon vascular injury, Nox4 expression is transiently lost in the cells which comprise the neointima. NADPH oxidase 4 therefore does not interfere with restenosis development after wire-induced vascular injury.
Collapse
Affiliation(s)
- Giulia K Buchmann
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Christoph Schürmann
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Manuela Spaeth
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Wesley Abplanalp
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Lukas Tombor
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - David John
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Timothy Warwick
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Flávia Rezende
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Andreas Weigert
- Institute of Biochemistry I, Faculty of Medicine, Goethe-University Frankfurt, Frankfurt, Germany
| | - Ajay M Shah
- School of Cardiovascular Medicine & Sciences, King's College London, British Heart Foundation Centre, London, UK
| | | | - Norbert Weissmann
- Excellence Cluster Cardio-Pulmonary Institute (CPI), Universities of Giessen and Marburg Lung Center (UGMLC), Member of the German Center for Lung Research (DZL), Gießen, Germany
| | - Stefanie Dimmeler
- German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany; Institute of Cardiovascular Regeneration, Centre for Molecular Medicine, Goethe University Frankfurt, Germany
| | - Katrin Schröder
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany
| | - Ralf P Brandes
- Institute for Cardiovascular Physiology, Goethe-University, Frankfurt Am Main, Germany; German Center for Cardiovascular Research (DZHK), Partner Site Rhein Main, Frankfurt Am Main, Germany.
| |
Collapse
|
28
|
Nakada EM, Sun R, Fujii U, Martin JG. The Impact of Endoplasmic Reticulum-Associated Protein Modifications, Folding and Degradation on Lung Structure and Function. Front Physiol 2021; 12:665622. [PMID: 34122136 PMCID: PMC8188853 DOI: 10.3389/fphys.2021.665622] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 04/23/2021] [Indexed: 12/15/2022] Open
Abstract
The accumulation of unfolded/misfolded proteins in the endoplasmic reticulum (ER) causes ER stress and induces the unfolded protein response (UPR) and other mechanisms to restore ER homeostasis, including translational shutdown, increased targeting of mRNAs for degradation by the IRE1-dependent decay pathway, selective translation of proteins that contribute to the protein folding capacity of the ER, and activation of the ER-associated degradation machinery. When ER stress is excessive or prolonged and these mechanisms fail to restore proteostasis, the UPR triggers the cell to undergo apoptosis. This review also examines the overlooked role of post-translational modifications and their roles in protein processing and effects on ER stress and the UPR. Finally, these effects are examined in the context of lung structure, function, and disease.
Collapse
Affiliation(s)
- Emily M. Nakada
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Rui Sun
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - Utako Fujii
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| | - James G. Martin
- Meakins-Christie Laboratories, Research Institute of the McGill University Health Centre (RI-MUHC), McGill University, Montreal, QC, Canada
- McGill University, Montreal, QC, Canada
| |
Collapse
|
29
|
Elko EA, Manuel AM, White S, Zito E, van der Vliet A, Anathy V, Janssen-Heininger YMW. Oxidation of peroxiredoxin-4 induces oligomerization and promotes interaction with proteins governing protein folding and endoplasmic reticulum stress. J Biol Chem 2021; 296:100665. [PMID: 33895140 PMCID: PMC8141880 DOI: 10.1016/j.jbc.2021.100665] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2021] [Revised: 04/07/2021] [Accepted: 04/12/2021] [Indexed: 02/08/2023] Open
Abstract
Peroxiredoxins (PRDXs) catalyze the reduction of hydrogen peroxide (H2O2). PRDX4 is the only peroxiredoxin located within the endoplasmic reticulum (ER) and is the most highly expressed H2O2 scavenger in the ER. PRDX4 has emerged as an important player in numerous diseases, such as fibrosis and metabolic syndromes, and its overoxidation is a potential indicator of ER redox stress. It is unclear how overoxidation of PRDX4 governs its oligomerization state and interacting partners. Herein, we addressed these questions via nonreducing Western blots, mass spectrometry, and site-directed mutagenesis. We report that the oxidation of PRDX4 in lung epithelial cells treated with tertbutyl hydroperoxide caused a shift of PRDX4 from monomer/dimer to high molecular weight (HMW) species, which contain PRDX4 modified with sulfonic acid residues (PRDX4-SO3), as well as of a complement of ER-associated proteins, including protein disulfide isomerases important in protein folding, thioredoxin domain-containing protein 5, and heat shock protein A5, a key regulator of the ER stress response. Mutation of any of the four cysteines in PRDX4 altered the HMW species in response to tertbutyl hydroperoxide as well as the secretion of PRDX4. We also demonstrate that the expression of ER oxidoreductase 1 alpha, which generates H2O2 in the ER, increased PRDX4 HMW formation and secretion. These results suggest a link between SO3 modification in the formation of HMW PRDX4 complexes in cells, whereas the association of key regulators of ER homeostasis with HMW oxidized PRDX4 point to a putative role of PRDX4 in regulating ER stress responses.
Collapse
Affiliation(s)
- Evan A Elko
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Allison M Manuel
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Sheryl White
- Department of Neurological Sciences, University of Vermont, Burlington, Vermont, USA
| | - Ester Zito
- Department of Biochemistry and Molecular Pharmacology, Istituto di Ricerche Farmacologiche Mario Negri IRCCS, Milan, Italy
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Burlington, Vermont, USA
| | | |
Collapse
|
30
|
Bolduc J, Koruza K, Luo T, Malo Pueyo J, Vo TN, Ezeriņa D, Messens J. Peroxiredoxins wear many hats: Factors that fashion their peroxide sensing personalities. Redox Biol 2021; 42:101959. [PMID: 33895094 PMCID: PMC8113037 DOI: 10.1016/j.redox.2021.101959] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2020] [Revised: 03/07/2021] [Accepted: 03/25/2021] [Indexed: 02/07/2023] Open
Abstract
Peroxiredoxins (Prdxs) sense and assess peroxide levels, and signal through protein interactions. Understanding the role of the multiple structural and post-translational modification (PTM) layers that tunes the peroxiredoxin specificities is still a challenge. In this review, we give a tabulated overview on what is known about human and bacterial peroxiredoxins with a focus on structure, PTMs, and protein-protein interactions. Armed with numerous cellular and atomic level experimental techniques, we look at the future and ask ourselves what is still needed to give us a clearer view on the cellular operating power of Prdxs in both stress and non-stress conditions.
Collapse
Affiliation(s)
- Jesalyn Bolduc
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Katarina Koruza
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Ting Luo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Julia Malo Pueyo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Trung Nghia Vo
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, B-1050, Brussels, Belgium; Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050, Brussels, Belgium; Structural Biology Brussels, Vrije Universiteit Brussel, B-1050, Brussels, Belgium.
| |
Collapse
|
31
|
Pozzer D, Invernizzi RW, Blaauw B, Cantoni O, Zito E. Ascorbic Acid Route to the Endoplasmic Reticulum: Function and Role in Disease. Antioxid Redox Signal 2021; 34:845-855. [PMID: 31867990 DOI: 10.1089/ars.2019.7912] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Significance: Humans cannot synthesize ascorbic acid (AscH2) (vitamin C), so deficiencies in dietary AscH2 cause the life-threatening disease of scurvy and many other diseases. After oral ingestion, plasma AscH2 concentrations are strictly controlled by transporters, which are required for entry into the cell and into intracellular organelles. Recent Advances: Besides its general antioxidant function, AscH2 is a cofactor for endoplasmic reticulum (ER)-localized collagen hydroxylases. Its important role in ER homeostasis is also highlighted by the fact that AscH2 deficiency in auxotrophic species triggers ER stress. Critical Issues: Characterizations of the molecular basis of diseases suggest that intracellular AscH2 deficiency is due not only to limited dietary access but also to its limited intracellular transport and net loss under conditions of intracellular hyperoxidation in the ER. This essay will offer an overview of the different transporters of vitamin C regulating its intracellular concentration, its function inside the ER, and the phenotypes of the diseases that can be triggered by increased depletion of this vitamin in the ER. Future Directions: When considering the benefits of increasing dietary AscH2, it is important to consider pharmacokinetic differences in the bioavailability between orally and intravenously administered AscH2: the latter bypasses intestinal absorption and is, therefore, the only route that can lead to the high plasma concentrations that may provide some health effects, and it is this route that needs to be chosen in clinical trials for those diseases associated with a deficiency of AscH2. Antioxid. Redox Signal. 34, 845-855.
Collapse
Affiliation(s)
- Diego Pozzer
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| | | | - Bert Blaauw
- Department of Biomedical Sciences, University of Padua, Padua, Italy
- Venetian Institute of Molecular Medicine, Padua, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Ester Zito
- Istituto di Ricerche Farmacologiche Mario Negri-IRCCS, Milan, Italy
| |
Collapse
|
32
|
Hirayama C, Machida K, Noi K, Murakawa T, Okumura M, Ogura T, Imataka H, Inaba K. Distinct roles and actions of protein disulfide isomerase family enzymes in catalysis of nascent-chain disulfide bond formation. iScience 2021; 24:102296. [PMID: 33855279 PMCID: PMC8024706 DOI: 10.1016/j.isci.2021.102296] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2020] [Revised: 01/13/2021] [Accepted: 03/05/2021] [Indexed: 12/04/2022] Open
Abstract
The mammalian endoplasmic reticulum (ER) harbors more than 20 members of the protein disulfide isomerase (PDI) family that act to maintain proteostasis. Herein, we developed an in vitro system for directly monitoring PDI- or ERp46-catalyzed disulfide bond formation in ribosome-associated nascent chains of human serum albumin. The results indicated that ERp46 more efficiently introduced disulfide bonds into nascent chains with a short segment exposed outside the ribosome exit site than PDI. Single-molecule analysis by high-speed atomic force microscopy further revealed that PDI binds nascent chains persistently, forming a stable face-to-face homodimer, whereas ERp46 binds for a shorter time in monomeric form, indicating their different mechanisms for substrate recognition and disulfide bond introduction. Thus, ERp46 serves as a more potent disulfide introducer especially during the early stages of translation, whereas PDI can catalyze disulfide formation when longer nascent chains emerge out from ribosome. We developed an in vitro system for monitoring nascent-chain disulfide formation High-speed AFM visualized PDI and ERp46 molecules acting on nascent chains PDI persistently holds nascent chains via dimerization for disulfide introduction ERp46 rapidly introduces disulfide bonds to nascent chains via short-time binding
Collapse
Affiliation(s)
- Chihiro Hirayama
- Institute of Multidisciplinary Research for Advanced Materials, Katahira 2-1-1, Aoba-ku, Tohoku University, Sendai, Miyagi 980-8577, Japan
| | - Kodai Machida
- Graduate School of Engineering, University of Hyogo, Himeji, Hyogo 671-2280, Japan
| | - Kentaro Noi
- Institute for NanoScience Design, Osaka University, Toyonaka, Osaka 560-8531, Japan
| | - Tadayoshi Murakawa
- Graduate School of Life Science and Technology, Tokyo Institute of Technology, Yokohama, Kanagawa, 226-8503, Japan
| | - Masaki Okumura
- Institute of Multidisciplinary Research for Advanced Materials, Katahira 2-1-1, Aoba-ku, Tohoku University, Sendai, Miyagi 980-8577, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Sendai, Miyagi 980-8578, Japan
| | - Teru Ogura
- Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Kumamoto 860-0811, Japan.,Faculty of Life Sciences, Kumamoto University, Kumamoto, Kumamoto 862-0973, Japan
| | - Hiroaki Imataka
- Graduate School of Engineering, University of Hyogo, Himeji, Hyogo 671-2280, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Katahira 2-1-1, Aoba-ku, Tohoku University, Sendai, Miyagi 980-8577, Japan
| |
Collapse
|
33
|
Hydrogen peroxide reactivity and specificity in thiol-based cell signalling. Biochem Soc Trans 2021; 48:745-754. [PMID: 32412042 DOI: 10.1042/bst20190049] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2020] [Revised: 04/14/2020] [Accepted: 04/16/2020] [Indexed: 12/15/2022]
Abstract
Reversible oxidation of thiol proteins is an important cell signalling mechanism. In many cases, this involves generation or exposure of the cells to H2O2, and oxidation of proteins that are not particularly H2O2-reactive. There is a conundrum as to how these proteins are oxidised when other highly reactive proteins such as peroxiredoxins are present. This article discusses potential mechanisms, focussing on recent evidence for oxidation being localised within the cell, redox relays involving peroxiredoxins operating in some signalling pathways, and mechanisms for facilitated or directed oxidation of specific targets. These findings help define conditions that enable redox signalling but there is still much to learn regarding mechanisms.
Collapse
|
34
|
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: 177] [Impact Index Per Article: 44.3] [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
|
35
|
Yang J, Wang Z, Liu X, Lu P. Modulation of vascular integrity and neuroinflammation by peroxiredoxin 4 following cerebral ischemia-reperfusion injury. Microvasc Res 2021; 135:104144. [PMID: 33515567 DOI: 10.1016/j.mvr.2021.104144] [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: 08/17/2020] [Revised: 01/16/2021] [Accepted: 01/25/2021] [Indexed: 01/10/2023]
Abstract
Ischemic stroke is a leading cause of morbidity and mortality worldwide, with oxidative stress playing a key role in the injury mechanism of thrombolytic therapy. There is increasing evidence that oxidative stress damages endothelial cells (ECs), degrades tight junction proteins (TJs), and contributes to increased blood-brain barrier (BBB) permeability. It has been demonstrated that the breakdown of BBB could increase the risk of intracerebral hemorrhagic transformation in ischemic stroke. And an episode of cerebral ischemia/reperfusion (I/R) also initiates oxidative stress-mediated inflammatory processes in ECs, which further promotes BBB disruption and the progression of brain injury. Previous studies have revealed that antioxidants could inhibit ROS generation and attenuate BBB disruption after cerebral I/R. Peroxiredoxin 4 (Prx4) is a member of the antioxidant enzymes family (Prx1-6) and has been characterized to be an efficient H2O2 scavenger. It should be noted that Prx4 may be directly involved in the protection of ECs from the effects of ROS and function in ECs as a membrane-associated peroxidase. This paper reviewed the implication of Prx4 on vascular integrity and neuroinflammation following a cerebral I/R injury.
Collapse
Affiliation(s)
- Jiping Yang
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China.
| | - Zairan Wang
- Department of Neurosurgery, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Xiuying Liu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| | - Pengchao Lu
- Department of Medical Imaging, The Second Hospital of Hebei Medical University, Shijiazhuang 050000, Hebei Province, China
| |
Collapse
|
36
|
Konno T, Melo EP, Chambers JE, Avezov E. Intracellular Sources of ROS/H 2O 2 in Health and Neurodegeneration: Spotlight on Endoplasmic Reticulum. Cells 2021; 10:233. [PMID: 33504070 PMCID: PMC7912550 DOI: 10.3390/cells10020233] [Citation(s) in RCA: 67] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2020] [Revised: 01/15/2021] [Accepted: 01/18/2021] [Indexed: 02/08/2023] Open
Abstract
Reactive oxygen species (ROS) are produced continuously throughout the cell as products of various redox reactions. Yet these products function as important signal messengers, acting through oxidation of specific target factors. Whilst excess ROS production has the potential to induce oxidative stress, physiological roles of ROS are supported by a spatiotemporal equilibrium between ROS producers and scavengers such as antioxidative enzymes. In the endoplasmic reticulum (ER), hydrogen peroxide (H2O2), a non-radical ROS, is produced through the process of oxidative folding. Utilisation and dysregulation of H2O2, in particular that generated in the ER, affects not only cellular homeostasis but also the longevity of organisms. ROS dysregulation has been implicated in various pathologies including dementia and other neurodegenerative diseases, sanctioning a field of research that strives to better understand cell-intrinsic ROS production. Here we review the organelle-specific ROS-generating and consuming pathways, providing evidence that the ER is a major contributing source of potentially pathologic ROS.
Collapse
Affiliation(s)
- Tasuku Konno
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| | - Eduardo Pinho Melo
- CCMAR—Centro de Ciências do Mar, Campus de Gambelas, Universidade do Algarve, 8005-139 Faro, Portugal;
| | - Joseph E. Chambers
- Cambridge Institute for Medical Research, University of Cambridge, Cambridge CB2 0XY, UK;
| | - Edward Avezov
- Department of Clinical Neurosciences, UK Dementia Research Institute, University of Cambridge, Cambridge CB2 0AH, UK
| |
Collapse
|
37
|
Stretton C, Pugh JN, McDonagh B, McArdle A, Close GL, Jackson MJ. 2-Cys peroxiredoxin oxidation in response to hydrogen peroxide and contractile activity in skeletal muscle: A novel insight into exercise-induced redox signalling? Free Radic Biol Med 2020; 160:199-207. [PMID: 32784030 PMCID: PMC7718083 DOI: 10.1016/j.freeradbiomed.2020.06.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2020] [Revised: 06/05/2020] [Accepted: 06/06/2020] [Indexed: 12/15/2022]
Abstract
Skeletal muscle generates superoxide during contractions which is rapidly converted to H2O2. This molecule has been proposed to activate signalling pathways and transcription factors that regulate key adaptive responses to exercise but the concentration of H2O2 required to oxidise and activate key signalling proteins in vitro is much higher than the intracellular concentration in muscle fibers following exercise. We hypothesised that Peroxiredoxins (Prx), which reacts with H2O2 at the low intracellular concentrations found in muscle, would be rapidly oxidised in contracting muscle and hence potentially transmit oxidising equivalents to downstream signalling proteins as a method for their oxidation and activation. The aim of this study was to characterise the effects of muscle contractile activity on the oxidation of Prx1, 2 and 3 and determine if these were affected by aging. Prx1, 2 and 3 were all rapidly and reversibly oxidised following treatment with low micromolar concentrations of H2O2 in C2C12 myotubes and also in isolated mature flexor digitalis brevis fibers from adult mice following a protocol of repeated isometric contractions. Significant oxidation of Prx2 was seen within 1 min (i.e. after 12 contractions), whereas significant oxidation was seen after 2 min for Prx1 and 3. In muscle fibers from old mice, Prx2 oxidation was significantly attenuated following contractile activity. Thus we show for the first time that Prx are rapidly and reversibly oxidised in response to contractile activity in skeletal muscle and hypothesise that these proteins act as effectors of muscle redox signalling pathways which are key to adaptations to exercise that are attenuated during aging.
Collapse
Affiliation(s)
- Clare Stretton
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool and MRC- Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), UK
| | - Jamie N Pugh
- School of Sport and Exercise Sciences, Tom Reilly Building, Byrom Street, Liverpool John Moores University, Liverpool, UK
| | - Brian McDonagh
- Discipline of Physiology, School of Medicine, National University of Ireland Galway, Ireland
| | - Anne McArdle
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool and MRC- Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), UK
| | - Graeme L Close
- School of Sport and Exercise Sciences, Tom Reilly Building, Byrom Street, Liverpool John Moores University, Liverpool, UK
| | - Malcolm J Jackson
- Department of Musculoskeletal Biology, Institute of Ageing and Chronic Disease, University of Liverpool and MRC- Arthritis Research UK Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), UK.
| |
Collapse
|
38
|
Okumura M, Noi K, Inaba K. Visualization of structural dynamics of protein disulfide isomerase enzymes in catalysis of oxidative folding and reductive unfolding. Curr Opin Struct Biol 2020; 66:49-57. [PMID: 33176263 DOI: 10.1016/j.sbi.2020.10.004] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/18/2020] [Accepted: 10/08/2020] [Indexed: 02/06/2023]
Abstract
Time-resolved single-molecule observations by high-speed atomic force microscopy (HS-AFM), have greatly advanced our understanding of how proteins operate to fulfill their unique functions. Using this device, we succeeded in visualizing two members of the protein disulfide isomerase family (PDIs) that act to catalyze oxidative folding and reductive unfolding in the endoplasmic reticulum (ER). ERdj5, an ER-resident disulfide reductase that promotes ER-associated degradation, reduces nonnative disulfide bonds of misfolded proteins utilizing the dynamics of its N-terminal and C-terminal clusters. With unfolded substrates, canonical PDI assembles to form a face-to-face dimer with a central hydrophobic cavity and multiple redox-active sites to accelerate oxidative folding inside the cavity. Altogether, PDIs exert highly dynamic mechanisms to ensure the protein quality control in the ER.
Collapse
Affiliation(s)
- Masaki Okumura
- Frontier Research Institute for Interdisciplinary Sciences, Aramaki aza Aoba 6-3, Aoba-ku, Sendai 980-8578, Japan
| | - Kentaro Noi
- Institute of Nanoscience Design, Osaka University, Machikaneyamatyou 1-3, Toyonaka 560-8531, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira 2-1-1, Aoba-ku, Sendai 980-8577, Japan.
| |
Collapse
|
39
|
Wang L, Yu J, Wang CC. Protein disulfide isomerase is regulated in multiple ways: Consequences for conformation, activities, and pathophysiological functions. Bioessays 2020; 43:e2000147. [PMID: 33155310 DOI: 10.1002/bies.202000147] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2020] [Revised: 10/13/2020] [Accepted: 10/15/2020] [Indexed: 12/13/2022]
Abstract
Protein disulfide isomerase (PDI) is one of the most abundant and critical protein folding catalysts in the endoplasmic reticulum of eukaryotic cells. PDI consists of four thioredoxin domains and interacts with a wide range of substrate and partner proteins due to its intrinsic conformational flexibility. PDI plays multifunctional roles in a variety of pathophysiological events, both as an oxidoreductase and a molecular chaperone. Recent studies have revealed that the conformation and activity of PDI can be regulated in multiple ways, including posttranslational modification and substrate/ligand binding. Here, we summarize recent advances in understanding the function and regulation of PDI in different pathological and physiological events. We propose that the multifunctional roles of PDI are regulated by multiple mechanisms. Furthermore, we discuss future directions for the study of PDI, emphasizing how different regulatory modes are linked to the conformational changes and biological functions of PDI in the context of diverse pathophysiologies.
Collapse
Affiliation(s)
- Lei Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Jiaojiao Yu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| | - Chih-Chen Wang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in Biomacromolecules, Institute of Biophysics, Chinese Academy of Sciences, Beijing, China.,College of Life Sciences, University of Chinese Academy of Sciences, Beijing, China
| |
Collapse
|
40
|
Pathways for Sensing and Responding to Hydrogen Peroxide at the Endoplasmic Reticulum. Cells 2020; 9:cells9102314. [PMID: 33080949 PMCID: PMC7603117 DOI: 10.3390/cells9102314] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2020] [Revised: 10/14/2020] [Accepted: 10/15/2020] [Indexed: 02/06/2023] Open
Abstract
The endoplasmic reticulum (ER) has emerged as a source of hydrogen peroxide (H2O2) and a hub for peroxide-based signaling events. Here we outline cellular sources of ER-localized peroxide, including sources within and near the ER. Focusing on three ER-localized proteins-the molecular chaperone BiP, the transmembrane stress-sensor IRE1, and the calcium pump SERCA2-we discuss how post-translational modification of protein cysteines by H2O2 can alter ER activities. We review how changed activities for these three proteins upon oxidation can modulate signaling events, and also how cysteine oxidation can serve to limit the cellular damage that is most often associated with elevated peroxide levels.
Collapse
|
41
|
Lénon M, Ke N, Szady C, Sakhtah H, Ren G, Manta B, Causey B, Berkmen M. Improved production of Humira antibody in the genetically engineered Escherichia coli SHuffle, by co-expression of human PDI-GPx7 fusions. Appl Microbiol Biotechnol 2020; 104:9693-9706. [PMID: 32997203 PMCID: PMC7595990 DOI: 10.1007/s00253-020-10920-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2020] [Revised: 09/11/2020] [Accepted: 09/18/2020] [Indexed: 12/21/2022]
Abstract
Abstract Microbial production of antibodies offers the promise of cheap, fast, and efficient production of antibodies at an industrial scale. Limiting this capacity in prokaryotes is the absence of the post-translational machinery, present in dedicated antibody producing eukaryotic cell lines, such as B cells. There has been few and limited success in producing full-length, correctly folded, and assembled IgG in the cytoplasm of prokaryotic cell lines. One such success was achieved by utilizing the genetically engineered Escherichia coli strain SHuffle with an oxidative cytoplasm. Due to the genetic disruption of reductive pathways, SHuffle cells are under constant oxidative stress, including increased levels of hydrogen peroxide (H2O2). The oxidizing capacity of H2O2 was linked to improved disulfide bond formation, by expressing a fusion of two endoplasmic reticulum-resident proteins, the thiol peroxidase GPx7 and the protein disulfide isomerase, PDI. In concert, these proteins mediate disulfide transfer from H2O2 to target proteins via PDI-Gpx7 fusions. The potential of this new strain was tested with Humira, a blockbuster antibody usually produced in eukaryotic cells. Expression results demonstrate that the new engineered SHuffle strain (SHuffle2) could produce Humira IgG four-fold better than the parental strain, both in shake-flask and in high-density fermentation. These preliminary studies guide the field in genetically engineering eukaryotic redox pathways in prokaryotes for the production of complex macromolecules. Key points • A eukaryotic redox pathway was engineered into the E. coli strain SHuffle in order to improve the yield of the blockbuster antibody Humira. • The best peroxidase-PDI fusion was selected using bioinformatics and in vivo studies. • Improved yields of Humira were demonstrated at shake-flask and high-density fermenters. Electronic supplementary material The online version of this article (10.1007/s00253-020-10920-5) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Marine Lénon
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Department of Microbiology, Stress Adaptation and Metabolism in Enterobacteria Unit, UMR CNRS 2001, Institut Pasteur, 25-28 Rue du Dr Roux, 75015, Paris, France
| | - Na Ke
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Cecily Szady
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Hassan Sakhtah
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Boston Institute of Biotechnology, LLC., Upstream Process Development, 225 Turnpike Road, Southborough, MA, 01772, USA
| | - Guoping Ren
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Bruno Manta
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
- Facultad de Medicina, Departamento de Bioquímica and Centro de Investigaciones Biomédicas, Universidad de la República, CP 11800, Montevideo, Uruguay
| | - Bryce Causey
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA
| | - Mehmet Berkmen
- New England Biolabs, 240 County Road, Ipswich, MA, 01938, USA.
| |
Collapse
|
42
|
Patel C, Saad H, Shenkman M, Lederkremer GZ. Oxidoreductases in Glycoprotein Glycosylation, Folding, and ERAD. Cells 2020; 9:cells9092138. [PMID: 32971745 PMCID: PMC7563561 DOI: 10.3390/cells9092138] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2020] [Revised: 09/17/2020] [Accepted: 09/18/2020] [Indexed: 12/17/2022] Open
Abstract
N-linked glycosylation and sugar chain processing, as well as disulfide bond formation, are among the most common post-translational protein modifications taking place in the endoplasmic reticulum (ER). They are essential modifications that are required for membrane and secretory proteins to achieve their correct folding and native structure. Several oxidoreductases responsible for disulfide bond formation, isomerization, and reduction have been shown to form stable, functional complexes with enzymes and chaperones that are involved in the initial addition of an N-glycan and in folding and quality control of the glycoproteins. Some of these oxidoreductases are selenoproteins. Recent studies also implicate glycan machinery–oxidoreductase complexes in the recognition and processing of misfolded glycoproteins and their reduction and targeting to ER-associated degradation. This review focuses on the intriguing cooperation between the glycoprotein-specific cell machineries and ER oxidoreductases, and highlights open questions regarding the functions of many members of this large family.
Collapse
Affiliation(s)
- Chaitanya Patel
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Haddas Saad
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Marina Shenkman
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
| | - Gerardo Z. Lederkremer
- The Shmunis School of Biomedicine and Cancer Research, Cell Biology Division, George Wise Faculty of Life Sciences, Tel Aviv University, Tel Aviv 69978, Israel; (C.P.); (H.S.); (M.S.)
- Sagol School of Neuroscience, Tel Aviv University, Tel Aviv 69978, Israel
- Correspondence:
| |
Collapse
|
43
|
Mechanisms of Disulfide Bond Formation in Nascent Polypeptides Entering the Secretory Pathway. Cells 2020; 9:cells9091994. [PMID: 32872499 PMCID: PMC7565403 DOI: 10.3390/cells9091994] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2020] [Revised: 08/26/2020] [Accepted: 08/28/2020] [Indexed: 12/16/2022] Open
Abstract
Disulfide bonds are an abundant feature of proteins across all domains of life that are important for structure, stability, and function. In eukaryotic cells, a major site of disulfide bond formation is the endoplasmic reticulum (ER). How cysteines correctly pair during polypeptide folding to form the native disulfide bond pattern is a complex problem that is not fully understood. In this paper, the evidence for different folding mechanisms involved in ER-localised disulfide bond formation is reviewed with emphasis on events that occur during ER entry. Disulfide formation in nascent polypeptides is discussed with focus on (i) its mechanistic relationship with conformational folding, (ii) evidence for its occurrence at the co-translational stage during ER entry, and (iii) the role of protein disulfide isomerase (PDI) family members. This review highlights the complex array of cellular processes that influence disulfide bond formation and identifies key questions that need to be addressed to further understand this fundamental process.
Collapse
|
44
|
Tran DT, Pottekat A, Mir SA, Loguercio S, Jang I, Campos AR, Scully KM, Lahmy R, Liu M, Arvan P, Balch WE, Kaufman RJ, Itkin-Ansari P. Unbiased Profiling of the Human Proinsulin Biosynthetic Interaction Network Reveals a Role for Peroxiredoxin 4 in Proinsulin Folding. Diabetes 2020; 69:1723-1734. [PMID: 32457219 PMCID: PMC7372081 DOI: 10.2337/db20-0245] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/16/2020] [Accepted: 05/15/2020] [Indexed: 12/12/2022]
Abstract
The β-cell protein synthetic machinery is dedicated to the production of mature insulin, which requires the proper folding and trafficking of its precursor, proinsulin. The complete network of proteins that mediate proinsulin folding and advancement through the secretory pathway, however, remains poorly defined. Here we used affinity purification and mass spectrometry to identify, for the first time, the proinsulin biosynthetic interaction network in human islets. Stringent analysis established a central node of proinsulin interactions with endoplasmic reticulum (ER) folding factors, including chaperones and oxidoreductases, that is remarkably conserved in both sexes and across three ethnicities. The ER-localized peroxiredoxin PRDX4 was identified as a prominent proinsulin-interacting protein. In β-cells, gene silencing of PRDX4 rendered proinsulin susceptible to misfolding, particularly in response to oxidative stress, while exogenous PRDX4 improved proinsulin folding. Moreover, proinsulin misfolding induced by oxidative stress or high glucose was accompanied by sulfonylation of PRDX4, a modification known to inactivate peroxiredoxins. Notably, islets from patients with type 2 diabetes (T2D) exhibited significantly higher levels of sulfonylated PRDX4 than islets from healthy individuals. In conclusion, we have generated the first reference map of the human proinsulin interactome to identify critical factors controlling insulin biosynthesis, β-cell function, and T2D.
Collapse
Affiliation(s)
- Duc T Tran
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Anita Pottekat
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Saiful A Mir
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | | | - Insook Jang
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | | | - Kathleen M Scully
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Reyhaneh Lahmy
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Ming Liu
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI
- Department of Endocrinology and Metabolism, Tianjin Medical University, Tianjin, China
| | - Peter Arvan
- Division of Metabolism, Endocrinology, and Diabetes, University of Michigan Medical School, Ann Arbor, MI
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, CA
- Integrative Structural and Computational Biology, Scripps Research, La Jolla, CA
| | - Randal J Kaufman
- Degenerative Diseases Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
| | - Pamela Itkin-Ansari
- Development, Aging and Regeneration Program, Sanford Burnham Prebys Medical Discovery Institute, La Jolla, CA
- Department of Pediatrics, University of California, San Diego, La Jolla, CA
| |
Collapse
|
45
|
Kanemura S, Sofia EF, Hirai N, Okumura M, Kadokura H, Inaba K. Characterization of the endoplasmic reticulum-resident peroxidases GPx7 and GPx8 shows the higher oxidative activity of GPx7 and its linkage to oxidative protein folding. J Biol Chem 2020; 295:12772-12785. [PMID: 32719007 DOI: 10.1074/jbc.ra120.013607] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2020] [Revised: 07/17/2020] [Indexed: 12/13/2022] Open
Abstract
Oxidative protein folding occurs primarily in the mammalian endoplasmic reticulum, enabled by a diverse network comprising more than 20 members of the protein disulfide isomerase (PDI) family and more than five PDI oxidases. Although the canonical disulfide bond formation pathway involving Ero1α and PDI has been well-studied so far, the physiological roles of the newly identified PDI oxidases, glutathione peroxidase-7 (GPx7) and -8 (GPx8), are only poorly understood. We here demonstrated that human GPx7 has much higher reactivity with H2O2 and hence greater PDI oxidation activity than human GPx8. The high reactivity of GPx7 is due to the presence of a catalytic tetrad at the redox-active site, which stabilizes the sulfenylated species generated upon the reaction with H2O2 Although it was previously postulated that GPx7 catalysis involved a highly reactive peroxidatic cysteine that can be sulfenylated by H2O2, we revealed that a resolving cysteine instead regulates the PDI oxidation activity of GPx7. We also determined that GPx7 formed complexes preferentially with PDI and P5 in H2O2-treated cells. Altogether, these results suggest that human GPx7 functions as an H2O2-dependent PDI oxidase in cells, whereas PDI oxidation may not be the central physiological role of human GPx8.
Collapse
Affiliation(s)
- Shingo Kanemura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan.,School of Science and Technology, Kwansei Gakuin University, Gakuen, Sanda, Hyogo, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki, Aza, Aoba-ku, Sendai, Miyagi, Japan
| | - Elza Firdiani Sofia
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Naoya Hirai
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Masaki Okumura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan.,Frontier Research Institute for Interdisciplinary Sciences, Tohoku University, Aramaki, Aza, Aoba-ku, Sendai, Miyagi, Japan
| | - Hiroshi Kadokura
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| | - Kenji Inaba
- Institute of Multidisciplinary Research for Advanced Materials, Tohoku University, Katahira, Aoba-ku, Sendai, Miyagi, Japan
| |
Collapse
|
46
|
Álvarez-Izquierdo M, Guillermo Esteban J, Muñoz-Antoli C, Toledo R. Ileal proteomic changes associated with IL-25-mediated resistance against intestinal trematode infections. Parasit Vectors 2020; 13:336. [PMID: 32616023 PMCID: PMC7331265 DOI: 10.1186/s13071-020-04206-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2020] [Accepted: 06/24/2020] [Indexed: 01/07/2023] Open
Abstract
Background Echinostoma caproni (Trematoda: Echinostomatidae) is an intestinal trematode, which has been extensively used to investigate the factors that determine the rejection of intestinal helminths. In this sense, several studies have shown that IL-25 is critical for the development of resistance against E. caproni in mice. In fact, treatment of mice with recombinant IL-25 generates resistance against primary E. caproni infection. However, the mechanisms by which IL-25 induces resistance remain unknown. Methods To study the mechanisms responsible for resistance elicited by IL-25, we analyzed the ileal proteomic changes induced by IL-25 in mice and their potential role in resistance. To this purpose, we compared the protein expression profiles in the ileum of four experimental groups of mice: naïve controls; E. caproni-infected mice; rIL-25-treated mice; and rIL-25-treated mice exposed to E. caproni metacercariae. Results Quantitative comparison by 2D-DIGE showed significant changes in a total of 41 spots. Of these, 40 validated protein spots were identified by mass spectrometry corresponding to 24 proteins. Conclusions Our results indicate that resistance to infection is associated with the maintenance of the intestinal epithelial homeostasis and the regulation of proliferation and cell death. These results provide new insights into the proteins involved in the regulation of tissue homeostasis after intestinal infection and its transcendence in resistance.![]()
Collapse
Affiliation(s)
- María Álvarez-Izquierdo
- Área de Parasitología, Departamento de Farmacia y Tecnología Farmacéutica y Parasitología, Facultad de Farmacia, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - J Guillermo Esteban
- Área de Parasitología, Departamento de Farmacia y Tecnología Farmacéutica y Parasitología, Facultad de Farmacia, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - Carla Muñoz-Antoli
- Área de Parasitología, Departamento de Farmacia y Tecnología Farmacéutica y Parasitología, Facultad de Farmacia, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain
| | - Rafael Toledo
- Área de Parasitología, Departamento de Farmacia y Tecnología Farmacéutica y Parasitología, Facultad de Farmacia, Universitat de València, Avda. Vicent Andrés Estellés s/n, 46100, Burjassot, Valencia, Spain.
| |
Collapse
|
47
|
Cao X, Lilla S, Cao Z, Pringle MA, Oka OBV, Robinson PJ, Szmaja T, van Lith M, Zanivan S, Bulleid NJ. The mammalian cytosolic thioredoxin reductase pathway acts via a membrane protein to reduce ER-localised proteins. J Cell Sci 2020; 133:jcs241976. [PMID: 32184267 PMCID: PMC7197872 DOI: 10.1242/jcs.241976] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2019] [Accepted: 03/10/2020] [Indexed: 01/21/2023] Open
Abstract
Folding of proteins entering the mammalian secretory pathway requires the insertion of the correct disulfides. Disulfide formation involves both an oxidative pathway for their insertion and a reductive pathway to remove incorrectly formed disulfides. Reduction of these disulfides is crucial for correct folding and degradation of misfolded proteins. Previously, we showed that the reductive pathway is driven by NADPH generated in the cytosol. Here, by reconstituting the pathway using purified proteins and ER microsomal membranes, we demonstrate that the thioredoxin reductase system provides the minimal cytosolic components required for reducing proteins within the ER lumen. In particular, saturation of the pathway and its protease sensitivity demonstrates the requirement for a membrane protein to shuttle electrons from the cytosol to the ER. These results provide compelling evidence for the crucial role of the cytosol in regulating ER redox homeostasis, ensuring correct protein folding and facilitating the degradation of misfolded ER proteins.
Collapse
Affiliation(s)
- Xiaofei Cao
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Sergio Lilla
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
| | - Zhenbo Cao
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Marie Anne Pringle
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Ojore B V Oka
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Philip J Robinson
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Tomasz Szmaja
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Marcel van Lith
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| | - Sara Zanivan
- Cancer Research UK Beatson Institute, Glasgow, G61 1BD, UK
- Institute of Cancer Sciences, University of Glasgow, Glasgow, G61 1QH, UK
| | - Neil J Bulleid
- Institute of Molecular, Cell and Systems Biology, College of Medical Veterinary and Life Sciences, Davidson Building, University of Glasgow, Glasgow, G12 8QQ, UK
| |
Collapse
|
48
|
The Role of Hydrogen Peroxide and Peroxiredoxins throughout the Cell Cycle. Antioxidants (Basel) 2020; 9:antiox9040280. [PMID: 32224940 PMCID: PMC7222192 DOI: 10.3390/antiox9040280] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2020] [Revised: 03/23/2020] [Accepted: 03/24/2020] [Indexed: 01/22/2023] Open
Abstract
Hydrogen peroxide (H2O2) is an oxidizing agent that induces cellular damage at inappropriate concentrations and gives rise to an arrest during cell cycle progression, causing cell death. Recent evidence indicates that H2O2 also acts as a promoter for cell cycle progression by oxidizing specific thiol proteins. The intracellular concentration of H2O2 is regulated tightly, enabling its use as a cellular signaling molecule while minimizing its potential to cause cellular damage. Peroxiredoxins (Prxs) have peroxidase activity toward H2O2, organic hydroperoxides, and peroxynitrite for protecting cells from oxidative stress. They are suggested to work as signaling mediators, allowing the local accumulation of H2O2 by inactivating their peroxidase activity uniquely compared with other antioxidant proteins such as catalase and glutathione peroxidase. Given that Prxs are highly sensitive to oxidation by H2O2, they act as sensors and transducers of H2O2 signaling via transferring their oxidation state to effector proteins. The concentrations of intracellular H2O2 increase as the cell cycle progresses from G1 to mitosis. Here, we summarize the roles of Prxs with regard to the regulation of cell cycle-dependent kinase activity and anaphase-promoting complex/cyclosome in terms of changes in H2O2 levels. Protection of the cell from unwanted progression of the cell cycle is suggested to be a role of Prx. We discuss the possible roles of Prxs to control H2O2 levels.
Collapse
|
49
|
Janssen-Heininger Y, Reynaert NL, van der Vliet A, Anathy V. Endoplasmic reticulum stress and glutathione therapeutics in chronic lung diseases. Redox Biol 2020; 33:101516. [PMID: 32249209 PMCID: PMC7251249 DOI: 10.1016/j.redox.2020.101516] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2019] [Revised: 03/20/2020] [Accepted: 03/20/2020] [Indexed: 02/07/2023] Open
Affiliation(s)
- Yvonne Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine and School of Nutrition and Translational Research in Metabolism (NUTRIM), Maastricht University Medical Center, Maastricht, the Netherlands
| | - Albert van der Vliet
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| | - Vikas Anathy
- Department of Pathology and Laboratory Medicine, University of Vermont, Larner College of Medicine, Burlington, VT, 05405, USA
| |
Collapse
|
50
|
Redox Signaling from Mitochondria: Signal Propagation and Its Targets. Biomolecules 2020; 10:biom10010093. [PMID: 31935965 PMCID: PMC7023504 DOI: 10.3390/biom10010093] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/02/2020] [Accepted: 01/02/2020] [Indexed: 02/07/2023] Open
Abstract
Progress in mass spectroscopy of posttranslational oxidative modifications has enabled researchers to experimentally verify the concept of redox signaling. We focus here on redox signaling originating from mitochondria under physiological situations, discussing mechanisms of transient redox burst in mitochondria, as well as the possible ways to transfer such redox signals to specific extramitochondrial targets. A role of peroxiredoxins is described which enables redox relay to other targets. Examples of mitochondrial redox signaling are discussed: initiation of hypoxia-inducible factor (HIF) responses; retrograde redox signaling to PGC1α during exercise in skeletal muscle; redox signaling in innate immune cells; redox stimulation of insulin secretion, and other physiological situations.
Collapse
|