1
|
Deviers J, Cailliez F, de la Lande A, Kattnig DR. Avian cryptochrome 4 binds superoxide. Comput Struct Biotechnol J 2024; 26:11-21. [PMID: 38204818 PMCID: PMC10776438 DOI: 10.1016/j.csbj.2023.12.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2023] [Revised: 12/06/2023] [Accepted: 12/12/2023] [Indexed: 01/12/2024] Open
Abstract
Flavin-binding cryptochromes are blue-light sensitive photoreceptors that have been implicated with magnetoreception in some species. The photocycle involves an intra-protein photo-reduction of the flavin cofactor, generating a magnetosensitive radical pair, and its subsequent re-oxidation. Superoxide (O2 • - ) is generated in the re-oxidation with molecular oxygen. The resulting O2 • - -containing radical pairs have also been hypothesised to underpin various magnetosensitive traits, but due to fast spin relaxation when tumbling in solution would require immobilisation. We here describe our insights in the binding of superoxide to cryptochrome 4 from C. livia based on extensive all-atom molecular dynamics studies and density-functional theory calculations. The positively charged "crypt" region that leads to the flavin binding pocket transiently binds O2 • - at 5 flexible binding sites centred on arginine residues. Typical binding times amounted to tens of nanoseconds, but exceptional binding events extended to several hundreds of nanoseconds and slowed the rotational diffusion, thereby realising rotational correlation times as large as 1 ns. The binding sites are particularly efficient in scavenging superoxide escaping from a putative generation site close to the flavin-cofactor, possibly implying a functional relevance. We discuss our findings in view of a potential magnetosensitivity of biological flavin semiquinone/superoxide radical pairs.
Collapse
Affiliation(s)
- Jean Deviers
- Living Systems Institute and Department of Physics, University of Exeter, Stocker Road, Exeter, Devon, EX4 4QD, United Kingdom
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, 91405 Orsay, France
| | - Fabien Cailliez
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, 91405 Orsay, France
| | - Aurélien de la Lande
- Institut de Chimie Physique, CNRS UMR 8000, Université Paris-Saclay, 91405 Orsay, France
| | - Daniel R. Kattnig
- Living Systems Institute and Department of Physics, University of Exeter, Stocker Road, Exeter, Devon, EX4 4QD, United Kingdom
| |
Collapse
|
2
|
Babajani A, Eftekharinasab A, Bekeschus S, Mehdian H, Vakhshiteh F, Madjd Z. Reactive oxygen species from non-thermal gas plasma (CAP): implication for targeting cancer stem cells. Cancer Cell Int 2024; 24:344. [PMID: 39438918 PMCID: PMC11515683 DOI: 10.1186/s12935-024-03523-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2024] [Accepted: 10/05/2024] [Indexed: 10/25/2024] Open
Abstract
Cancer remains a major global health challenge, with the persistence of cancer stem cells (CSCs) contributing to treatment resistance and relapse. Despite advancements in cancer therapy, targeting CSCs presents a significant hurdle. Non-thermal gas plasma, also known as CAP, represents an innovative cancer treatment. It has recently gained attention for its often found to be selective, immunogenic, and potent anti-cancer properties. CAP is composed of a collection of transient, high-energy, and physically and chemically active entities, such as reactive oxygen species (ROS). It is acknowledged that the latter are responsible for a major portion of biomedical CAP effects. The dynamic interplay of CAP-derived ROS and other components contributes to the unique and versatile properties of CAP, enabling it to interact with biological systems and elicit various therapeutic effects, including its potential in cancer treatment. While CAP has shown promise in various cancer types, its application against CSCs is relatively unexplored. This review assesses the potential of CAP as a therapeutic strategy for targeting CSCs, focusing on its ability to regulate cellular states and achieve redox homeostasis. This is done by providing an overview of CSC characteristics and demonstrating recent findings on CAP's efficacy in targeting these cells. By contributing insights into the unique attributes of CSCs and the potential of CAP, this work contributes to an advanced understanding of innovative oncology strategies.
Collapse
Affiliation(s)
- Amirhesam Babajani
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | | | - Sander Bekeschus
- ZIK Plasmatis, Leibniz Institute for Plasma Science and Technology (INP), Felix-Hausdorff-Str. 2, 17489, Greifswald, Germany
| | - Hassan Mehdian
- Plasma Medicine Group, Plasma Research Institute, Kharazmi University, Tehran, Iran
| | - Faezeh Vakhshiteh
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| | - Zahra Madjd
- Oncopathology Research Center, Iran University of Medical Sciences (IUMS), Tehran, Iran.
- Department of Molecular Medicine, Faculty of Advanced Technologies in Medicine, Iran University of Medical Sciences (IUMS), Tehran, Iran.
| |
Collapse
|
3
|
Hebchen DM, Schröder K. Redox Signaling in Endosomes Using the Example of EGF Receptors: A Graphical Review. Antioxidants (Basel) 2024; 13:1215. [PMID: 39456468 PMCID: PMC11504029 DOI: 10.3390/antiox13101215] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2024] [Revised: 10/06/2024] [Accepted: 10/08/2024] [Indexed: 10/28/2024] Open
Abstract
Early endosomes represent first-line sorting compartments or even organelles for internalized molecules. They enable the transport of molecules or ligands to other compartments of the cell, such as lysosomes, for degradation or recycle them back to the membrane by various mechanisms. Moreover, early endosomes function as signaling and scaffolding platforms to initiate or prolong distinct signaling pathways. Accordingly, early endosomes have to be recognized as either part of a degradation or recycling pathway. The physical proximity of many ligand-binding receptors with other membrane-bound proteins or complexes such as NADPH oxidases may result in an interaction of second messengers, like reactive oxygen species (ROS) and early endosomes, that promote the correct recognition of individual early endosomes. In fact, redoxosomes comprise an endosomal subsection of signaling endosomes. One example of such potential interaction is epidermal growth factor receptor (EGFR) signaling. Here we summarize recent findings on EGFR signaling as a well-studied example for receptor trafficking and trans-activation and illustrate the interplay between cellular and endosomal ROS.
Collapse
Affiliation(s)
| | - Katrin Schröder
- Institute of Physiology, Medical Faculty, Goethe University, Theodor-Stern-Kai 7, 60590 Frankfurt am Main, Germany;
| |
Collapse
|
4
|
Krishnamurthy HK, Pereira M, Rajavelu I, Jayaraman V, Krishna K, Wang T, Bei K, Rajasekaran JJ. Oxidative stress: fundamentals and advances in quantification techniques. Front Chem 2024; 12:1470458. [PMID: 39435263 PMCID: PMC11491411 DOI: 10.3389/fchem.2024.1470458] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Accepted: 09/09/2024] [Indexed: 10/23/2024] Open
Abstract
Oxidative species, generated endogenously via metabolism or from exogenous sources, play crucial roles in the body. At low levels, these species support immune functions by participating in phagocytosis. They also aid in cellular signaling and contribute to vasomodulation. However, when the levels of oxidative species exceed the body's antioxidant capacity to neutralize them, oxidative stress occurs. This stress can damage cellular macromolecules such as lipids, DNA, RNA, and proteins, driving the pathogenesis of diseases and aging through the progressive deterioration of physiological functions and cellular structures. Therefore, the body's ability to manage oxidative stress and maintain it at optimal levels is essential for overall health. Understanding the fundamentals of oxidative stress, along with its reliable quantification, can enable consistency and comparability in clinical practice across various diseases. While direct quantification of oxidant species in the body would be ideal for assessing oxidative stress, it is not feasible due to their high reactivity, short half-life, and the challenges of quantification using conventional techniques. Alternatively, quantifying lipid peroxidation, damage products of nucleic acids and proteins, as well as endogenous and exogenous antioxidants, serves as appropriate markers for indicating the degree of oxidative stress in the body. Along with the conventional oxidative stress markers, this review also discusses the role of novel markers, focusing on their biological samples and detection techniques. Effective quantification of oxidative stress may enhance the understanding of this phenomenon, aiding in the maintenance of cellular integrity, prevention of age-associated diseases, and promotion of longevity.
Collapse
Affiliation(s)
| | | | | | | | | | - Tianhao Wang
- Vibrant Sciences LLC., Santa Clara, CA, United States
| | - Kang Bei
- Vibrant Sciences LLC., Santa Clara, CA, United States
| | | |
Collapse
|
5
|
Panda B, Tripathy A, Patra S, Kullu B, Tabrez S, Jena M. Imperative connotation of SODs in cancer: Emerging targets and multifactorial role of action. IUBMB Life 2024; 76:592-613. [PMID: 38600696 DOI: 10.1002/iub.2821] [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: 01/08/2024] [Accepted: 03/14/2024] [Indexed: 04/12/2024]
Abstract
Superoxide dismutase (SOD) is a crucial enzyme responsible for the redox homeostasis inside the cell. As a part of the antioxidant defense system, it plays a pivotal role in the dismutation of the superoxide radicals (O 2 - ) generated mainly by the oxidative phosphorylation, which would otherwise bring out the redox dysregulation, leading to higher reactive oxygen species (ROS) generation and, ultimately, cell transformation, and malignancy. Several studies have shown the involvement of ROS in a wide range of human cancers. As SOD is the key enzyme in regulating ROS, any change, such as a transcriptional change, epigenetic remodeling, functional alteration, and so forth, either activates the proto-oncogenes or aberrant signaling cascades, which results in cancer. Interestingly, in some cases, SODs act as tumor promoters instead of suppressors. Furthermore, SODs have also been known to switch their role during tumor progression. In this review, we have tried to give a comprehensive account of SODs multifactorial role in various human cancers so that SODs-based therapeutic strategies could be made to thwart cancers.
Collapse
Affiliation(s)
- Biswajit Panda
- Department of Zoology, College of Basic Science and Humanities, Odisha University of Agriculture and Technology, Bhubaneswar, India
| | - Ankita Tripathy
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Srimanta Patra
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| | - Bandana Kullu
- Post Graduate Department of Botany, Utkal University, Bhubaneswar, India
| | - Shams Tabrez
- King Fahd Medical Research Center, King Abdulaziz University, Jeddah, Saudi Arabia
- Department of Medical Laboratory Sciences, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah, Saudi Arabia
| | - Mrutyunjay Jena
- Post Graduate Department of Botany, Berhampur University, Berhampur, India
| |
Collapse
|
6
|
Schanne G, Demignot S, Policar C, Delsuc N. Cellular evaluation of superoxide dismutase mimics as catalytic drugs: Challenges and opportunities. Coord Chem Rev 2024; 514:215906. [DOI: 10.1016/j.ccr.2024.215906] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2025]
|
7
|
Mizuno D, Kawahara M, Konoha-Mizuno K, Hama R, Ogawara T. The Role of Zinc in the Development of Vascular Dementia and Parkinson's Disease and the Potential of Carnosine as Their Therapeutic Agent. Biomedicines 2024; 12:1296. [PMID: 38927502 PMCID: PMC11201809 DOI: 10.3390/biomedicines12061296] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Revised: 05/10/2024] [Accepted: 06/07/2024] [Indexed: 06/28/2024] Open
Abstract
Synaptic zinc ions (Zn2+) play an important role in the development of vascular dementia (VD) and Parkinson's disease (PD). In this article, we reviewed the current comprehension of the Zn2+-induced neurotoxicity that leads to the pathogenesis of these neuronal diseases. Zn2+-induced neurotoxicity was investigated by using immortalised hypothalamic neurons (GT1-7 cells). This cell line is useful for the development of a rapid and convenient screening system for investigating Zn2+-induced neurotoxicity. GT1-7 cells were also used to search for substances that prevent Zn2+-induced neurotoxicity. Among the tested substances was a protective substance in the extract of Japanese eel (Anguilla japonica), and we determined its structure to be like carnosine (β-alanylhistidine). Carnosine may be a therapeutic drug for VD and PD. Furthermore, we reviewed the molecular mechanisms that involve the role of carnosine as an endogenous protector and its protective effect against Zn2+-induced cytotoxicity and discussed the prospects for the future therapeutic applications of this dipeptide for neurodegenerative diseases and dementia.
Collapse
Affiliation(s)
- Dai Mizuno
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Masahiro Kawahara
- Research Institute of Pharmaceutical Sciences, Faculty of Pharmacy, Musashino University, 1-1-20 Shin-machi, Nishitokyo-shi 202-8585, Tokyo, Japan;
| | - Keiko Konoha-Mizuno
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Ryoji Hama
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| | - Terumasa Ogawara
- Department of Forensic Medicine, Faculty of Medicine, Yamagata University, 2-2-2 Iida-Nishi, Yamagata-shi 990-9585, Yamagata, Japan; (K.K.-M.); (R.H.); (T.O.)
| |
Collapse
|
8
|
Pai HL, Lin DPC, Chang HH. Current updates for hyperuricemia and gout in age-related macular degeneration. FASEB J 2024; 38:e23676. [PMID: 38783765 DOI: 10.1096/fj.202400421r] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2024] [Revised: 04/13/2024] [Accepted: 05/07/2024] [Indexed: 05/25/2024]
Abstract
The escalating prevalence of metabolic syndrome poses a significant public health challenge, particularly among aging populations, with metabolic dysfunctions contributing to pro-inflammatory states. In this review, we delved into the less recognized association between hyperuricemia (HUA), a manifestation of metabolic syndrome and a primary risk factor for gout, and age-related macular degeneration (AMD), a sight-threatening ailment predominantly affecting the elderly. In recent years, inflammation, particularly its involvement in complement pathway dysregulation, has gained prominence in AMD pathophysiology. The contradictory role of uric acid (UA) in intercellular and intracellular environments was discussed, highlighting its antioxidant properties in plasma and its pro-oxidant effects intracellularly. Emerging evidence suggests a potential link between elevated serum uric acid levels and choroid neovascularization in AMD, providing insights into the role of HUA in retinal pathologies. Various pathways, including crystal-induced and non-crystal-induced mechanisms, were proposed to indicate the need for further research into the precise molecular interactions. The implication of HUA in AMD underscores its potential involvement in retinal pathologies, which entails interdisciplinary collaboration for a comprehensive understanding of its impact on retina and related clinical manifestations.
Collapse
Affiliation(s)
- Hung-Liang Pai
- Department of Medicine, Chung Shan Medical University, Taichung City, Taiwan
| | - David Pei-Cheng Lin
- Department of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung City, Taiwan
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City, Taiwan
| | - Han-Hsin Chang
- Department of Ophthalmology, Chung Shan Medical University Hospital, Taichung City, Taiwan
- Department of Nutrition, Chung Shan Medical University, Taichung City, Taiwan
| |
Collapse
|
9
|
Shuvalova M, Dmitrieva A, Belousov V, Nosov G. The role of reactive oxygen species in the regulation of the blood-brain barrier. Tissue Barriers 2024:2361202. [PMID: 38808582 DOI: 10.1080/21688370.2024.2361202] [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: 02/28/2024] [Accepted: 05/23/2024] [Indexed: 05/30/2024] Open
Abstract
The blood-brain barrier (BBB) regulates the exchange of metabolites and cells between the blood and brain, and maintains central nervous system homeostasis. Various factors affect BBB barrier functions, including reactive oxygen species (ROS). ROS can act as stressors, damaging biological molecules, but they also serve as secondary messengers in intracellular signaling cascades during redox signaling. The impact of ROS on the BBB has been observed in multiple sclerosis, stroke, trauma, and other neurological disorders, making blocking ROS generation a promising therapeutic strategy for BBB dysfunction. However, it is important to consider ROS generation during normal BBB functioning for signaling purposes. This review summarizes data on proteins expressed by BBB cells that can be targets of redox signaling or oxidative stress. It also provides examples of signaling molecules whose impact may cause ROS generation in the BBB, as well as discusses the most common diseases associated with BBB dysfunction and excessive ROS generation, open questions that arise in the study of this problem, and possible ways to overcome them.
Collapse
Affiliation(s)
- Margarita Shuvalova
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasiia Dmitrieva
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
| | - Vsevolod Belousov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Department of metabolism and redox biology, Shemyakin and Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| | - Georgii Nosov
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Life Improvement by Future Technologies (LIFT) Center, Skolkovo, Moscow, Russia
| |
Collapse
|
10
|
Lamb FS, Choi H, Miller MR, Stark RJ. Vascular Inflammation and Smooth Muscle Contractility: The Role of Nox1-Derived Superoxide and LRRC8 Anion Channels. Hypertension 2024; 81:752-763. [PMID: 38174563 PMCID: PMC10954410 DOI: 10.1161/hypertensionaha.123.19434] [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] [Indexed: 01/05/2024]
Abstract
Vascular inflammation underlies the development of hypertension, and the mechanisms by which it increases blood pressure remain the topic of intense investigation. Proinflammatory factors including glucose, salt, vasoconstrictors, cytokines, wall stress, and growth factors enhance contractility and impair relaxation of vascular smooth muscle cells. These pathways share a dependence upon redox signaling, and excessive activation promotes oxidative stress that promotes vascular aging. Vascular smooth muscle cell phenotypic switching and migration into the intima contribute to atherosclerosis, while hypercontractility increases systemic vascular resistance and vasospasm that can trigger ischemia. Here, we review factors that drive the initiation and progression of this vasculopathy in vascular smooth muscle cells. Emphasis is placed on the contribution of reactive oxygen species generated by the Nox1 NADPH oxidase which produces extracellular superoxide (O2•-). The mechanisms of O2•- signaling remain poorly defined, but recent evidence demonstrates physical association of Nox1 with leucine-rich repeat containing 8 family volume-sensitive anion channels. These may provide a pathway for influx of O2•- to the cytoplasm, creating an oxidized cytoplasmic nanodomain where redox-based signals can affect both cytoskeletal structure and vasomotor function. Understanding the mechanistic links between inflammation, O2•- and vascular smooth muscle cell contractility may facilitate targeting of anti-inflammatory therapy in hypertension.
Collapse
Affiliation(s)
- Fred S Lamb
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Hyehun Choi
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Michael R Miller
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| | - Ryan J Stark
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, TN
| |
Collapse
|
11
|
Peng M, Jiang J, Chen S, Li K, Lin Y. Cu single-atom catalyst-based flexible hydrogen peroxide electrochemical sensor with oxygen resistance for monitoring ROS bursts. Analyst 2023; 148:5667-5672. [PMID: 37812430 DOI: 10.1039/d3an01464a] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/10/2023]
Abstract
The study of cellular responses linked to oxidative stress mechanisms is crucial in comprehending diverse physiological and pathological life processes, including mitochondrial dysfunction. Nonetheless, despite the interference of O2, the monitoring of ROS released from cells poses a challenging task. In this study, carbon-based copper single-atom catalysts (Cu SACs) were synthesized that exhibits excellent electrocatalytic performance for H2O2 reduction with an initial potential at 0.23 V and effectively avoids interference from O2. Based on this catalyst, a flexible and stretchable oxygen-tolerant sensor was constructed and applied to monitor the calcium ion-induced ROS burst in human umbilical vein endothelial cells (HUVECs) in a simulated physiological condition. This study effectively eradicates interference that may arise from the reduction of O2 and presents a dependable platform for real-time in situ monitoring of physiologically active molecules by utilizing H2O2 detection.
Collapse
Affiliation(s)
- Meihong Peng
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Jing Jiang
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Shutong Chen
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Kai Li
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| | - Yuqing Lin
- Department of Chemistry, Capital Normal University, Beijing 100048, China.
| |
Collapse
|
12
|
Wu YF, De La Toba EA, Dvoretskiy S, Jung R, Kim N, Daniels L, Romanova EV, Drnevich J, Sweedler JV, Boppart MD. Development of a cell-free strategy to recover aged skeletal muscle after disuse. J Physiol 2023; 601:5011-5031. [PMID: 35318675 PMCID: PMC9492804 DOI: 10.1113/jp282867] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2022] [Accepted: 03/14/2022] [Indexed: 12/25/2022] Open
Abstract
Extended periods of bed rest and limb immobilization are required for healing post-injury or disease, yet disuse can result in significant muscle atrophy and decreased quality of life in older adults. Physical rehabilitation is commonly prescribed to recover these deficits, yet accumulation of reactive oxygen species and sustained rates of protein degradation persist during the rehabilitation period that can significantly delay or prevent recovery. Pericytes, considered the primary mesenchymal and vascular stromal cell in skeletal muscle, secrete beneficial factors that maintain baseline muscle mass, yet minimal information exists regarding the pericyte response to disuse and recovery. In the current study, single-cell RNA sequencing and functional assays were performed to demonstrate that pericytes in mouse skeletal muscle lose the capacity to synthesize antioxidants during disuse and recovery. This information was used to guide the design of a strategy in which healthy donor pericytes were stimulated with hydrogen peroxide (H2 O2 ) to produce small extracellular vesicles (sEVs) that effectively restored myofibre size in adult and aged muscle after disuse. Proteomic assessment detected 11 differentially regulated proteins in primed sEVs that may account for recovery of muscle, including proteins associated with extracellular matrix composition and anti-inflammatory and antioxidant processes. This study demonstrates that healthy H2 O2 -primed pericyte-derived sEVs effectively improve skeletal muscle recovery after immobilization, presenting a novel acellular approach to rebuild muscle mass in older adults after a period of disuse. KEY POINTS: Previous studies suggest that prolonged oxidative stress is a barrier to skeletal muscle recovery after a period of immobilization. In this study we demonstrate that muscle-resident perivascular stromal cells (pericytes) become dysfunctional and lack the capacity to mount an antioxidant defence after disuse in mice. Hydrogen peroxide treatment of healthy pericytes in vitro simulates the release of small extracellular vesicles (sEVs) that effectively recover skeletal muscle fibre size and extracellular matrix remodelling in young adult and aged mice after disuse. Pericyte-derived sEVs present a novel acellular strategy to recover skeletal muscle after disuse.
Collapse
Affiliation(s)
- Yu-Fu Wu
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Eduardo A. De La Toba
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Svyatoslav Dvoretskiy
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Rebecca Jung
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Noah Kim
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Laureen Daniels
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Elena V. Romanova
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Jenny Drnevich
- Roy J. Carver Biotechnology Center, High Performance Biological Computing, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| | - Jonathan V. Sweedler
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Chemistry, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
| | - Marni D. Boppart
- Department of Kinesiology and Community Health, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Beckman Institute for Advanced Science and Technology, University of Illinois at Urbana-Champaign, Urbana, IL, 61801, USA
- Department of Cell and Developmental Biology, University of Illinois at Urbana-Champaign, Urbana, IL 61801, USA
| |
Collapse
|
13
|
Xie J, Wu Y, Tao Q, Liu H, Wang J, Zhang C, Zhou Y, Wei C, Chang Y, Jin Y, Ding Z. The role of lncRNA in the pathogenesis of chronic obstructive pulmonary disease. Heliyon 2023; 9:e22460. [PMID: 38034626 PMCID: PMC10687241 DOI: 10.1016/j.heliyon.2023.e22460] [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: 09/26/2023] [Revised: 11/09/2023] [Accepted: 11/13/2023] [Indexed: 12/02/2023] Open
Abstract
Chronic obstructive pulmonary disease (COPD) is characterized by progressive and irreversible airflow obstruction with abnormal lung function. Because its pathogenesis involves multiple aspects of oxidative stress, immunity and inflammation, apoptosis, airway and lung repair and destruction, the clinical approach to COPD treatment is not further updated. Therefore, it is crucial to discover a new means of COPD diagnosis and treatment. COPD etiology is associated with complex interactions between environmental and genetic determinants. Numerous genes are involved in the pathogenic process of this illness in research samples exposed to hazardous environmental conditions. Among them, Long non-coding RNAs (lncRNAs) have been reported to be involved in the molecular mechanisms of COPD development induced by different environmental exposures and genetic susceptibility encounters, and some potential lncRNA biomarkers have been identified as early diagnostic, disease course determination, and therapeutic targets for COPD. In this review, we summarize the expression profiles of the reported lncRNAs that have been reported in COPD studies related to environmental risk factors such as smoking and air pollution exposure and provided an overview of the roles of those lncRNAs in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Jing Xie
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yongkang Wu
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Qing Tao
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Hua Liu
- Anhui Institute for Food and Drug Control, Hefei, Anhui, China
| | - Jingjing Wang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chunwei Zhang
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yuanzhi Zhou
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Chengyan Wei
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Yan Chang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| | - Yong Jin
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
| | - Zhen Ding
- Inflammation and Immune-Mediated Diseases Laboratory of Anhui Province, Department of Respiratory, The Third Affiliated Hospital of Anhui Medical University (The Binhu Hospital of Hefei), School of Pharmacy, Anhui Medical University, Hefei, Anhui, China
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-Inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-Inflammatory and Immune Medicine, Hefei, Anhui, China
| |
Collapse
|
14
|
Stavely R, Ott LC, Rashidi N, Sakkal S, Nurgali K. The Oxidative Stress and Nervous Distress Connection in Gastrointestinal Disorders. Biomolecules 2023; 13:1586. [PMID: 38002268 PMCID: PMC10669114 DOI: 10.3390/biom13111586] [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: 09/08/2023] [Revised: 10/19/2023] [Accepted: 10/23/2023] [Indexed: 11/26/2023] Open
Abstract
Oxidative stress is increasingly recognized as a central player in a range of gastrointestinal (GI) disorders, as well as complications stemming from therapeutic interventions. This article presents an overview of the mechanisms of oxidative stress in GI conditions and highlights a link between oxidative insult and disruption to the enteric nervous system (ENS), which controls GI functions. The dysfunction of the ENS is characteristic of a spectrum of disorders, including neurointestinal diseases and conditions such as inflammatory bowel disease (IBD), diabetic gastroparesis, and chemotherapy-induced GI side effects. Neurons in the ENS, while essential for normal gut function, appear particularly vulnerable to oxidative damage. Mechanistically, oxidative stress in enteric neurons can result from intrinsic nitrosative injury, mitochondrial dysfunction, or inflammation-related pathways. Although antioxidant-based therapies have shown limited efficacy, recognizing the multifaceted role of oxidative stress in GI diseases offers a promising avenue for future interventions. This comprehensive review summarizes the literature to date implicating oxidative stress as a critical player in the pathophysiology of GI disorders, with a focus on its role in ENS injury and dysfunction, and highlights opportunities for the development of targeted therapeutics for these diseases.
Collapse
Affiliation(s)
- Rhian Stavely
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Leah C. Ott
- Department of Pediatric Surgery, Massachusetts General Hospital, Harvard Medical School, Boston, MA 02114, USA
| | - Niloufar Rashidi
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Samy Sakkal
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
| | - Kulmira Nurgali
- Institute for Health and Sport, Victoria University, St Albans, VIC 3021, Australia
- Department of Medicine Western Health, The University of Melbourne, St Albans, VIC 3021, Australia
- Regenerative Medicine and Stem Cell Program, Australian Institute for Musculoskeletal Science (AIMSS), St Albans, VIC 3021, Australia
| |
Collapse
|
15
|
Zheng M, Liu Y, Zhang G, Yang Z, Xu W, Chen Q. The Applications and Mechanisms of Superoxide Dismutase in Medicine, Food, and Cosmetics. Antioxidants (Basel) 2023; 12:1675. [PMID: 37759978 PMCID: PMC10525108 DOI: 10.3390/antiox12091675] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2023] [Revised: 08/17/2023] [Accepted: 08/25/2023] [Indexed: 09/29/2023] Open
Abstract
Superoxide dismutase (SOD) is a class of enzymes that restrict the biological oxidant cluster enzyme system in the body, which can effectively respond to cellular oxidative stress, lipid metabolism, inflammation, and oxidation. Published studies have shown that SOD enzymes (SODs) could maintain a dynamic balance between the production and scavenging of biological oxidants in the body and prevent the toxic effects of free radicals, and have been shown to be effective in anti-tumor, anti-radiation, and anti-aging studies. This research summarizes the types, biological functions, and regulatory mechanisms of SODs, as well as their applications in medicine, food production, and cosmetic production. SODs have proven to be a useful tool in fighting disease, and mimetics and conjugates that report SODs have been developed successively to improve the effectiveness of SODs. There are still obstacles to solving the membrane permeability of SODs and the persistence of enzyme action, which is still a hot spot and difficulty in mining the effect of SODs and promoting their application in the future.
Collapse
Affiliation(s)
| | | | | | | | | | - Qinghua Chen
- College of Animal Science and Technology, Hunan Agricultural University, Changsha 410128, China
| |
Collapse
|
16
|
Choi S, Hatzihristidis T, Gaud G, Dutta A, Lee J, Arya A, Clubb LM, Stamos DB, Markovics A, Mikecz K, Love P. GRB2 promotes thymocyte positive selection by facilitating THEMIS-mediated inactivation of SHP1. J Exp Med 2023; 220:e20221649. [PMID: 37067793 PMCID: PMC10114920 DOI: 10.1084/jem.20221649] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2022] [Revised: 02/25/2023] [Accepted: 03/24/2023] [Indexed: 04/18/2023] Open
Abstract
The T-lineage restricted protein THEMIS has been shown to play a critical role in T cell development. THEMIS, via its distinctive CABIT domains, inhibits the catalytic activity of the tyrosine phosphatase SHP1 (PTPN6). SHP1 and THEMIS bind to the ubiquitous cytosolic adapter GRB2, and the purported formation of a tri-molecular THEMIS-GRB2-SHP1 complex facilitates inactivation of SHP1 by THEMIS. The importance of this function of GRB2 among its numerous documented activities is unclear as GRB2 binds to multiple proteins and participates in several signaling responses in thymocytes. Here, we show that similar to Themis-/- thymocytes, the primary molecular defect in GRB2-deficient thymocytes is increased catalytically active SHP1 and the developmental block in GRB2-deficient thymocytes is alleviated by deletion or inhibition of SHP1 and is exacerbated by SHP1 overexpression. Thus, the principal role of GRB2 during T cell development is to promote THEMIS-mediated inactivation of SHP1 thereby enhancing the sensitivity of TCR signaling in CD4+CD8+ thymocytes to low affinity positively selecting self-ligands.
Collapse
Affiliation(s)
- Seeyoung Choi
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Teri Hatzihristidis
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Guillaume Gaud
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Avik Dutta
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Jan Lee
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Awadhesh Arya
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Lauren M. Clubb
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Daniel B. Stamos
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| | - Adrienn Markovics
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Katalin Mikecz
- Department of Orthopedic Surgery, Rush University Medical Center, Chicago, IL, USA
| | - Paul Love
- Section on Hematopoiesis and Lymphocyte Biology, Eunice Kennedy Shriver, National Institute of Child Health and Human Development, Bethesda, MD, USA
| |
Collapse
|
17
|
Lee J, Han M, Shin Y, Lee JM, Heo G, Lee Y. How Extracellular Reactive Oxygen Species Reach Their Intracellular Targets in Plants. Mol Cells 2023; 46:329-336. [PMID: 36799103 PMCID: PMC10258463 DOI: 10.14348/molcells.2023.2158] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2022] [Revised: 12/06/2022] [Accepted: 12/20/2022] [Indexed: 02/18/2023] Open
Abstract
Reactive oxygen species (ROS) serve as secondary messengers that regulate various developmental and signal transduction processes, with ROS primarily generated by NADPH OXIDASEs (referred to as RESPIRATORY BURST OXIDASE HOMOLOGs [RBOHs] in plants). However, the types and locations of ROS produced by RBOHs are different from those expected to mediate intracellular signaling. RBOHs produce O2•- rather than H2O2 which is relatively long-lived and able to diffuse through membranes, and this production occurs outside the cell instead of in the cytoplasm, where signaling cascades occur. A widely accepted model explaining this discrepancy proposes that RBOH-produced extracellular O2•- is converted to H2O2 by superoxide dismutase and then imported by aquaporins to reach its cytoplasmic targets. However, this model does not explain how the specificity of ROS targeting is ensured while minimizing unnecessary damage during the bulk translocation of extracellular ROS (eROS). An increasing number of studies have provided clues about eROS action mechanisms, revealing various mechanisms for eROS perception in the apoplast, crosstalk between eROS and reactive nitrogen species, and the contribution of intracellular organelles to cytoplasmic ROS bursts. In this review, we summarize these recent advances, highlight the mechanisms underlying eROS action, and provide an overview of the routes by which eROS-induced changes reach the intracellular space.
Collapse
Affiliation(s)
- Jinsu Lee
- Research Institute of Basic Sciences, Seoul National University, Seoul 08826, Korea
| | - Minsoo Han
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Yesol Shin
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Jung-Min Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Geon Heo
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
| | - Yuree Lee
- School of Biological Sciences, Seoul National University, Seoul 08826, Korea
- Plant Genomics and Breeding Institute, Seoul National University, Seoul 08826, Korea
| |
Collapse
|
18
|
Fujii J, Soma Y, Matsuda Y. Biological Action of Singlet Molecular Oxygen from the Standpoint of Cell Signaling, Injury and Death. Molecules 2023; 28:molecules28104085. [PMID: 37241826 DOI: 10.3390/molecules28104085] [Citation(s) in RCA: 18] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Revised: 05/11/2023] [Accepted: 05/12/2023] [Indexed: 05/28/2023] Open
Abstract
Energy transfer to ground state triplet molecular oxygen results in the generation of singlet molecular oxygen (1O2), which has potent oxidizing ability. Irradiation of light, notably ultraviolet A, to a photosensitizing molecule results in the generation of 1O2, which is thought to play a role in causing skin damage and aging. It should also be noted that 1O2 is a dominant tumoricidal component that is generated during the photodynamic therapy (PDT). While type II photodynamic action generates not only 1O2 but also other reactive species, endoperoxides release pure 1O2 upon mild exposure to heat and, hence, are considered to be beneficial compounds for research purposes. Concerning target molecules, 1O2 preferentially reacts with unsaturated fatty acids to produce lipid peroxidation. Enzymes that contain a reactive cysteine group at the catalytic center are vulnerable to 1O2 exposure. Guanine base in nucleic acids is also susceptible to oxidative modification, and cells carrying DNA with oxidized guanine units may experience mutations. Since 1O2 is produced in various physiological reactions in addition to photodynamic reactions, overcoming technical challenges related to its detection and methods used for its generation would allow its potential functions in biological systems to be better understood.
Collapse
Affiliation(s)
- Junichi Fujii
- Department of Biochemistry and Molecular Biology, Graduate School of Medical Science, Yamagata University, Yamagata 990-9585, Japan
| | - Yuya Soma
- Graduate School of Nursing, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| | - Yumi Matsuda
- Graduate School of Nursing, Yamagata University Faculty of Medicine, Yamagata 990-9585, Japan
| |
Collapse
|
19
|
Lee SJ, Maeda S, Gao J, Nichols CG. Oxidation Driven Reversal of PIP 2-dependent Gating in GIRK2 Channels. FUNCTION 2023; 4:zqad016. [PMID: 37168492 PMCID: PMC10165546 DOI: 10.1093/function/zqad016] [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: 02/06/2023] [Revised: 03/24/2023] [Accepted: 04/03/2023] [Indexed: 05/13/2023] Open
Abstract
Physiological activity of G protein gated inward rectifier K+ (GIRK, Kir3) channel, dynamically regulated by three key ligands, phosphoinositol-4,5-bisphosphate (PIP2), Gβγ, and Na+, underlies cellular electrical response to multiple hormones and neurotransmitters in myocytes and neurons. In a reducing environment, matching that inside cells, purified GIRK2 (Kir3.2) channels demonstrate low basal activity, and expected sensitivity to the above ligands. However, under oxidizing conditions, anomalous behavior emerges, including rapid loss of PIP2 and Na+-dependent activation and a high basal activity in the absence of any agonists, that is now paradoxically inhibited by PIP2. Mutagenesis identifies two cysteine residues (C65 and C190) as being responsible for the loss of PIP2 and Na+-dependent activity and the elevated basal activity, respectively. The results explain anomalous findings from earlier studies and illustrate the potential pathophysiologic consequences of oxidation on GIRK channel function, as well as providing insight to reversed ligand-dependence of Kir and KirBac channels.
Collapse
Affiliation(s)
- Sun-Joo Lee
- Department of Cell Biology and Physiology and the Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Shoji Maeda
- Department of Pharmacology, Medical School, University of Michigan, Ann Arbor, Michigan, USA
| | - Jian Gao
- Department of Cell Biology and Physiology and the Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Colin G Nichols
- Department of Cell Biology and Physiology and the Center for Investigation of Membrane Excitability Diseases, Washington University School of Medicine, St. Louis, Missouri, USA
| |
Collapse
|
20
|
García-Rodríguez FJ, Buchrieser C, Escoll P. Legionella and mitochondria, an intriguing relationship. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2023; 374:37-81. [PMID: 36858656 DOI: 10.1016/bs.ircmb.2022.10.001] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Legionella pneumophila is the causative agent of Legionnaires' disease, a severe pneumonia. L. pneumophila injects via a type-IV-secretion-system (T4SS) more than 300 bacterial proteins into macrophages, its main host cell in humans. Certain of these bacterial effectors target organelles in the infected cell and hijack multiple processes to facilitate all steps of the intracellular life cycle of this pathogen. In this review, we discuss the interplay between L. pneumophila, an intracellular bacterium fully armed with virulence tools, and mitochondria, the extraordinary eukaryotic organelles playing prominent roles in cellular bioenergetics, cell-autonomous immunity and cell death. We present and discuss key findings concerning the multiple interactions of L. pneumophila with mitochondria during infection and the mechanisms employed by T4SS effectors that target mitochondrial functions to subvert infected cells.
Collapse
Affiliation(s)
| | - Carmen Buchrieser
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| | - Pedro Escoll
- Institut Pasteur, Université Paris Cité, Biologie des Bactéries Intracellulaires and CNRS UMR 6047, Paris, France.
| |
Collapse
|
21
|
Puente-Cobacho B, Varela-López A, Quiles JL, Vera-Ramirez L. Involvement of redox signalling in tumour cell dormancy and metastasis. Cancer Metastasis Rev 2023; 42:49-85. [PMID: 36701089 PMCID: PMC10014738 DOI: 10.1007/s10555-022-10077-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/06/2022] [Accepted: 12/27/2022] [Indexed: 01/27/2023]
Abstract
Decades of research on oncogene-driven carcinogenesis and gene-expression regulatory networks only started to unveil the complexity of tumour cellular and molecular biology. This knowledge has been successfully implemented in the clinical practice to treat primary tumours. In contrast, much less progress has been made in the development of new therapies against metastasis, which are the main cause of cancer-related deaths. More recently, the role of epigenetic and microenviromental factors has been shown to play a key role in tumour progression. Free radicals are known to communicate the intracellular and extracellular compartments, acting as second messengers and exerting a decisive modulatory effect on tumour cell signalling. Depending on the cellular and molecular context, as well as the intracellular concentration of free radicals and the activation status of the antioxidant system of the cell, the signalling equilibrium can be tilted either towards tumour cell survival and progression or cell death. In this regard, recent advances in tumour cell biology and metastasis indicate that redox signalling is at the base of many cell-intrinsic and microenvironmental mechanisms that control disseminated tumour cell fate and metastasis. In this manuscript, we will review the current knowledge about redox signalling along the different phases of the metastatic cascade, including tumour cell dormancy, making emphasis on metabolism and the establishment of supportive microenvironmental connections, from a redox perspective.
Collapse
Affiliation(s)
- Beatriz Puente-Cobacho
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain
| | - Alfonso Varela-López
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - José L Quiles
- Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain
| | - Laura Vera-Ramirez
- Department of Genomic Medicine, GENYO, Centre for Genomics and Oncology, Pfizer-University of Granada and Andalusian Regional Government, PTS, Granada, Spain. .,Department of Physiology, Institute of Nutrition and Food Technology "José Mataix Verdú", Biomedical Research Center, University of Granada, Granada, Spain.
| |
Collapse
|
22
|
Ghaedamini H, Duanghathaipornsuk S, Onusko P, Binsheheween AM, Kim DS. Reduced Glutathione-Modified Electrode for the Detection of Hydroxyl Free Radicals. BIOSENSORS 2023; 13:254. [PMID: 36832020 PMCID: PMC9953857 DOI: 10.3390/bios13020254] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/05/2023] [Accepted: 02/08/2023] [Indexed: 06/18/2023]
Abstract
Hydroxyl radicals (•OH) are known as essential chemicals for cells to maintain their normal functions and defensive responses. However, a high concentration of •OH may cause oxidative stress-related diseases, such as cancer, inflammation, and cardiovascular disorders. Therefore, •OH can be used as a biomarker to detect the onset of these disorders at an early stage. Reduced glutathione (GSH), a well-known tripeptide for its antioxidant capacity against reactive oxygen species (ROS), was immobilized on a screen-printed carbon electrode (SPCE) to develop a real-time detection sensor with a high selectivity towards •OH. The signals produced by the interaction of the GSH-modified sensor and •OH were characterized using cyclic voltammetry (CV) and electrochemical impedance spectroscopy (EIS). The CV curve of the GSH-modified sensor in the Fenton reagent exhibited a pair of well-defined peaks, demonstrating the redox reaction of the electrochemical sensor and •OH. The sensor showed a linear relationship between the redox response and the concentration of •OH with a limit of detection (LOD) of 49 µM. Furthermore, using EIS studies, the proposed sensor demonstrated the capability of differentiating •OH from hydrogen peroxide (H2O2), a similar oxidizing chemical. After being immersed in the Fenton solution for 1 hr, redox peaks in the CV curve of the GSH-modified electrode disappeared, revealing that the immobilized GSH on the electrode was oxidized and turned to glutathione disulfide (GSSG). However, it was demonstrated that the oxidized GSH surface could be reversed back to the reduced state by reacting with a solution of glutathione reductase (GR) and nicotinamide adenine dinucleotide phosphate (NADPH), and possibly reused for •OH detection.
Collapse
Affiliation(s)
| | | | | | | | - Dong-Shik Kim
- Department of Chemical Engineering, University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
23
|
Andrés CMC, Pérez de la Lastra JM, Juan CA, Plou FJ, Pérez-Lebeña E. The Role of Reactive Species on Innate Immunity. Vaccines (Basel) 2022; 10:vaccines10101735. [PMID: 36298601 PMCID: PMC9609844 DOI: 10.3390/vaccines10101735] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/06/2022] [Accepted: 10/14/2022] [Indexed: 11/16/2022] Open
Abstract
This review examines the role of reactive species RS (of oxygen ROS, nitrogen RNS and halogen RHS) on innate immunity. The importance of these species in innate immunity was first recognized in phagocytes that underwent a “respiratory burst” after activation. The anion superoxide •O2− and hydrogen peroxide H2O2 are detrimental to the microbial population. NADPH oxidase NOx, as an •O2− producer is essential for microbial destruction, and patients lacking this functional oxidase are more susceptible to microbial infections. Reactive nitrogen species RNS (the most important are nitric oxide radical -•NO, peroxynitrite ONOO— and its derivatives), are also harmful to microorganisms, including bacteria, viruses, and parasites. Hypochlorous acid HOCl and hypothiocyanous acid HOSCN synthesized through the enzyme myeloperoxidase MPO, which catalyzes the reaction between H2O2 and Cl− or SCN−, are important inorganic bactericidal molecules, effective against a wide range of microbes. This review also discusses the role of antimicrobial peptides AMPs and their induction of ROS. In summary, reactive species RS are the heart of the innate immune system, and they are necessary for microbial lysis in infections that can affect mammals throughout their lives.
Collapse
Affiliation(s)
| | - José Manuel Pérez de la Lastra
- Institute of Natural Products and Agrobiology, CSIC-Spanish Research Council, Avda. Astrofísico Fco. Sánchez 3, 38206 La Laguna, Spain
- Correspondence:
| | - Celia Andrés Juan
- Cinquima Institute and Department of Organic Chemistry, Faculty of Sciences, Valladolid University, Paseo de Belén 7, 47011 Valladolid, Spain
| | - Francisco J. Plou
- Institute of Catalysis and Petrochemistry, CSIC-Spanish Research Council, 28049 Madrid, Spain
| | | |
Collapse
|
24
|
Brandt M, Dörschmann H, Khraisat S, Knopp T, Ringen J, Kalinovic S, Garlapati V, Siemer S, Molitor M, Göbel S, Stauber R, Karbach SH, Münzel T, Daiber A, Wenzel P. Telomere Shortening in Hypertensive Heart Disease Depends on Oxidative DNA Damage and Predicts Impaired Recovery of Cardiac Function in Heart Failure. Hypertension 2022; 79:2173-2184. [PMID: 35862118 DOI: 10.1161/hypertensionaha.121.18935] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Heart failure (HF) coincides with cardiomyocyte telomere shortening. Arterial hypertension is the most prominent risk factor for HF. Both HF and arterial hypertension are associated with dysregulation of the neurohormonal axis. How neurohormonal activation is linked to telomere shortening in the pathogenesis of HF is incompletely understood. METHODS Cardiomyocyte telomere length was assessed in a mouse model of hypertensive HF induced by excess neurohormonal activation (AngII [angiotensin II] infusion, high salt diet, and uninephrectomy), in AngII-stimulated cardiomyocytes and in endomyocardial biopsies from patients with HF. Superoxide production, expression of NOX2 (NADPH oxidase 2) and PRDX1 (peroxiredoxin 1) and HDAC6 (histone deacetylase 6) activity were assessed. RESULTS Telomere shortening occurred in vitro and in vivo, correlating with both left ventricular (LV) dilatation and LV systolic function impairment. Telomere shortening coincided with increased superoxide production, increased NOX2 expression, increased HDAC6 activity, loss of the telomere-specific antioxidant PRDX1, and increased oxidative DNA-damage. NOX2 knockout prevented PRDX1 depletion, DNA-damage and telomere shortening confirming this enzyme as a critical source of reactive oxygen species. Cotreatment with the NOX inhibitor apocynin ameliorated hypertensive HF and telomere shortening. Similarly, treatment with the HDAC6 inhibitor tubastatin A, which increases PRDX1 bioavailability, prevented telomere shortening in adult cardiomyocytes. To explore the clinical relevance of our findings, we examined endomyocardial biopsies from an all-comer population of patients with HF with reduced ejection fraction. Here, cardiomyocyte telomere length predicted the recovery of cardiac function. CONCLUSIONS Cardiomyocyte telomere shortening and oxidative damage in heart failure with reduced ejection fraction induced by excess neurohormonal activation depends on NOX2-derived superoxide and may help to stratify HF therapy.
Collapse
Affiliation(s)
- Moritz Brandt
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Hendrik Dörschmann
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sana'a Khraisat
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Tanja Knopp
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Julia Ringen
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sanela Kalinovic
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Venkata Garlapati
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Svenja Siemer
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Michael Molitor
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Sebastian Göbel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Roland Stauber
- Department of Otolaryngology, Head and Neck Surgery, University Medical Center Mainz' Mainz' Germany (S.S., R.S.)
| | - Susanne Helena Karbach
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Thomas Münzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Andreas Daiber
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.)
| | - Philip Wenzel
- Department of Cardiology' University Medical Center Mainz' Mainz' Germany (M.B., H.D., S.K., T.K., J.R., S.K., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Center for Thrombosis and Hemostasis' University Medical Center Mainz' Mainz' Germany (M.B., H.D., T.K., J.R., V.G., M.M., S.H.K., T.M., A.D., P.W.).,German Center for Cardiovascular Research (DZHK) - Partner site Rhine-Main (M.B., T.K., J.R., V.G., M.M., S.G., S.H.K., T.M., A.D., P.W.).,Department of Biochemistry, Cardiovascular Research Institute Maastricht School for Cardiovascular Diseases (CARIM), Maastricht University, Maastricht, the Netherlands (P.W.)
| |
Collapse
|
25
|
Morrow JR, Raymond JJ, Chowdhury MSI, Sahoo PR. Redox-Responsive MRI Probes Based on First-Row Transition-Metal Complexes. Inorg Chem 2022; 61:14487-14499. [PMID: 36067522 DOI: 10.1021/acs.inorgchem.2c02197] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The presence of multiple oxidation and spin states of first-row transition-metal complexes facilitates the development of switchable MRI probes. Redox-responsive probes capitalize on a change in the magnetic properties of the different oxidation states of the paramagnetic metal ion center upon exposure to biological oxidants and reductants. Transition-metal complexes that are useful for MRI can be categorized according to whether they accelerate water proton relaxation (T1 or T2 agents), induce paramagnetic shifts of 1H or 19F resonances (paraSHIFT agents), or are chemical exchange saturation transfer (CEST) agents. The various oxidation state couples and their properties as MRI probes are summarized with a focus on Co(II)/Co(III) or Fe(II)/Fe(III) complexes as small molecules or as liposomal agents. Solution studies of these MRI probes are reviewed with an emphasis on redox changes upon treatment with oxidants or with enzymes that are physiologically important in inflammation and disease. Finally, we outline the challenges of developing these probes further for in vivo MRI applications.
Collapse
Affiliation(s)
- Janet R Morrow
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Jaclyn J Raymond
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Md Saiful I Chowdhury
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| | - Priya Ranjan Sahoo
- Department of Chemistry, University at Buffalo, the State University of New York, Amherst, New York 14260, United States
| |
Collapse
|
26
|
Grishanova AY, Perepechaeva ML. Aryl Hydrocarbon Receptor in Oxidative Stress as a Double Agent and Its Biological and Therapeutic Significance. Int J Mol Sci 2022; 23:6719. [PMID: 35743162 PMCID: PMC9224361 DOI: 10.3390/ijms23126719] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2022] [Revised: 06/14/2022] [Accepted: 06/14/2022] [Indexed: 12/02/2022] Open
Abstract
The aryl hydrocarbon receptor (AhR) has long been implicated in the induction of a battery of genes involved in the metabolism of xenobiotics and endogenous compounds. AhR is a ligand-activated transcription factor necessary for the launch of transcriptional responses important in health and disease. In past decades, evidence has accumulated that AhR is associated with the cellular response to oxidative stress, and this property of AhR must be taken into account during investigations into a mechanism of action of xenobiotics that is able to activate AhR or that is susceptible to metabolic activation by enzymes encoded by the genes that are under the control of AhR. In this review, we examine various mechanisms by which AhR takes part in the oxidative-stress response, including antioxidant and prooxidant enzymes and cytochrome P450. We also show that AhR, as a participant in the redox balance and as a modulator of redox signals, is being increasingly studied as a target for a new class of therapeutic compounds and as an explanation for the pathogenesis of some disorders.
Collapse
Affiliation(s)
| | - Maria L. Perepechaeva
- Federal Research Center of Fundamental and Translational Medicine, Institute of Molecular Biology and Biophysics, Timakova Str. 2, 630117 Novosibirsk, Russia;
| |
Collapse
|
27
|
Meganathan V, Hamilton CE, Natarajan K, Keshava S, Boggaram V. NADPH and xanthine oxidases control induction of inflammatory mediator expression by organic dust in the lung. FASEB J 2022; 36:e22381. [PMID: 35661421 DOI: 10.1096/fj.202100732r] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2021] [Revised: 04/29/2022] [Accepted: 05/16/2022] [Indexed: 11/11/2022]
Abstract
Exposure to organic dust in animal and agricultural farms and the ensuing lung inflammation are linked to the development of respiratory diseases. We found previously that elevated production of reactive oxygen species (ROS) by aqueous poultry organic dust extract (hereafter referred to as dust extract) mediates induction of proinflammatory mediators in airway epithelial cells. In the present study, we investigated whether ROS generated by NADPH oxidases (NOX) and xanthine oxidase (XO) controls induction of inflammatory mediators by dust extract and the underlying mechanisms in bronchial epithelial cells. Using chemical inhibitors and siRNA targeted knockdown, we found that NOX1, NOX2, NOX4, and XO-derived ROS regulates induction of proinflammatory mediator levels. Like airway epithelial cells in vitro, NOX inhibitor VAS2870 reduced keratinocyte chemoattractant (KC), IL-6, and TNF-α production and 4-hydroxynonenal (4-HNE) staining induced by dust extract in mouse lungs. VAS2870 inhibition of proinflammatory mediators was associated with reduced NFκB and Stat3 activation indicating that NOX generated ROS activates NFκB and Stat3 to induce proinflammatory gene expression. Dust extract increased the membrane association of p47phox in airway epithelial cells indicating NOX2 activation but had no effect on NOX2 protein levels. In summary, our studies have shown that NOX and XO generated ROS control organic dust induction of proinflammatory mediators in airway epithelial cells via NFκB and Stat3 activation.
Collapse
Affiliation(s)
- Velmurugan Meganathan
- Department of Cellular and Molecular Biology, Health Science Center, University of Texas at Tyler, Tyler, Texas, USA
| | - Cory E Hamilton
- Department of Cellular and Molecular Biology, Health Science Center, University of Texas at Tyler, Tyler, Texas, USA
| | - Kartiga Natarajan
- Department of Cellular and Molecular Biology, Health Science Center, University of Texas at Tyler, Tyler, Texas, USA
| | - Shiva Keshava
- Department of Cellular and Molecular Biology, Health Science Center, University of Texas at Tyler, Tyler, Texas, USA
| | - Vijay Boggaram
- Department of Cellular and Molecular Biology, Health Science Center, University of Texas at Tyler, Tyler, Texas, USA
| |
Collapse
|
28
|
Bai J, Tan R, An Z, Xu Y. Quantitative estimation of intracellular oxidative stress in human tissues. Brief Bioinform 2022; 23:6599072. [PMID: 35653708 PMCID: PMC9294418 DOI: 10.1093/bib/bbac206] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 04/28/2022] [Accepted: 05/04/2022] [Indexed: 11/18/2022] Open
Abstract
Oxidative stress is known to be involved in and possibly a key driver of the development of numerous chronic diseases, including cancer. It is highly desired to have a capability to reliably estimate the level of intracellular oxidative stress as it can help to identify functional changes and disease phenotypes associated with such a stress, but the problem proves to be very challenging. We present a novel computational model for quantitatively estimating the level of oxidative stress in tissues and cells based on their transcriptomic data. The model consists of (i) three sets of marker genes found to be associated with the production of oxidizing molecules, the activated antioxidation programs and the intracellular stress attributed to oxidation, respectively; (ii) three polynomial functions defined over the expression levels of the three gene sets are developed aimed to capture the total oxidizing power, the activated antioxidation capacity and the oxidative stress level, respectively, with their detailed parameters estimated by solving an optimization problem and (iii) the optimization problem is so formulated to capture the relevant known insights such as the oxidative stress level generally goes up from normal to chronic diseases and then to cancer tissues. Systematic assessments on independent datasets indicate that the trained predictor is highly reliable and numerous insights are made based on its application results to samples in the TCGA, GTEx and GEO databases.
Collapse
Affiliation(s)
- Jun Bai
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,School of Artificial Intelligence, Jilin University, Changchun, China
| | - Renbo Tan
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,College of Computer Science and Technology, Jilin University, Changchun, China
| | - Zheng An
- Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, the University of Georgia, Athens, GA, USA
| | - Ying Xu
- Cancer Systems Biology Center, China-Japan Union Hospital of Jilin University, Jilin University, Changchun, China.,College of Computer Science and Technology, Jilin University, Changchun, China.,Computational Systems Biology Lab, Department of Biochemistry and Molecular Biology, and Institute of Bioinformatics, the University of Georgia, Athens, GA, USA
| |
Collapse
|
29
|
CeO2-Zn Nanocomposite Induced Superoxide, Autophagy and a Non-Apoptotic Mode of Cell Death in Human Umbilical-Vein-Derived Endothelial (HUVE) Cells. TOXICS 2022; 10:toxics10050250. [PMID: 35622663 PMCID: PMC9147432 DOI: 10.3390/toxics10050250] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/18/2022] [Revised: 05/12/2022] [Accepted: 05/13/2022] [Indexed: 02/01/2023]
Abstract
In this study, a nanocomposite of cerium oxide-zinc (CeO2-Zn; 26 ± 11 nm) based on the antioxidant rare-earth cerium oxide (CeO2) nanoparticles (NPs) with the modifier zinc (Zn) was synthesized by sintering method and characterized. Its bio-response was examined in human umbilical-vein-derived endothelial (HUVE) cells to get insight into the components of vascular system. While NPs of CeO2 did not significantly alter cell viability up to a concentration of 200 µg/mL for a 24 h exposure, 154 ± 6 µg/mL of nanocomposite CeO2-Zn induced 50% cytotoxicity. Mechanism of cytotoxicity occurring due to nanocomposite by its Zn content was compared by choosing NPs of ZnO, possibly the closest nanoparticulate form of Zn. ZnO NPs lead to the induction of higher reactive oxygen species (ROS) (DCF-fluorescence), steeper depletion in antioxidant glutathione (GSH) and a greater loss of mitochondrial membrane potential (MMP) as compared to that induced by CeO2-Zn nanocomposite. Nanocomposite of CeO2-Zn, on the other hand, lead to significant higher induction of superoxide radical (O2•−, DHE fluorescence), nitric oxide (NO, determined by DAR-2 imaging and Griess reagent) and autophagic vesicles (determined by Lysotracker and monodansylcadeverine probes) as compared to that caused by ZnO NP treatment. Moreover, analysis after triple staining (by annexin V-FITC, PI, and Hoechst) conducted at their respective IC50s revealed an apoptosis mode of cell death due to ZnO NPs, whereas CeO2-Zn nanocomposite induced a mechanism of cell death that was significantly different from apoptosis. Our findings on advanced biomarkers such as autophagy and mode of cell death suggested the CeO2-Zn nanocomposite might behave as independent nanostructure from its constituent ones. Since nanocomposites can behave independently of their constituent NPs/elements, by creating nanocomposites, NP versatility can be increased manifold by just manipulating existing NPs. Moreover, data in this study can furnish early mechanistic insight about the potential damage that could occur in the integrity of vascular systems.
Collapse
|
30
|
Ağaşcıoğlu E, Çolak R, Çakatay U. Redox status biomarkers in the fast-twitch extensor digitorum longus resulting from the hypoxic exercise. NAGOYA JOURNAL OF MEDICAL SCIENCE 2022; 84:433-447. [PMID: 35967949 PMCID: PMC9350571 DOI: 10.18999/nagjms.84.2.433] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 11/01/2021] [Indexed: 11/29/2022]
Abstract
The fast-twitch muscle may be affected from over-produced reactive oxygen species (ROS) during hypoxia/hypoxic exercise. The study aims to investigate redox status biomarkers in the fast-twitch extensor digitorum longus (EDL) muscle after hypoxic exercise. Male Sprague Dawley rats (eight-week-old) were randomly divided into six groups of the experimental "live high train high (LHTH), live high train low (LHTL) and live low train low (LLTL)" and their respective controls. Before the EDLs' extraction, the animals exercised for a 4-week familiarization period, then they exercised for four-weeks at different altitudes. The LHTH group had higher ratios of lipid hydroperoxides (LHPs) than the experimental groups of LHTL (p=0.004) and LLTL (p=0.002), while having no difference than its control 'LH'. Similarly, a higher percentage of advanced oxidation protein products (AOPP) was determined in the LHTH than the LHTL (p=0.041) and LLTL (p=0.048). Furthermore, oxidation of thiol fractions was the lowest in the LHTH and LH. However, redox biomarkers and thiol fractions illustrated no significant change in the LHTL and LLTL that might ensure redox homeostasis due to higher oxygen consumption. The study shows that not hypoxic exercise/exercise, but hypoxia might itself lead to a redox imbalance in the fast-twitch EDL muscle.
Collapse
Affiliation(s)
- Eda Ağaşcıoğlu
- Department of Recreation, Faculty of Sports Sciences, Lokman Hekim University, Ankara, Turkey
| | - Rıdvan Çolak
- Department of Physical Education and Sports, Ardahan University, Ardahan, Turkey
| | - Ufuk Çakatay
- Department of Medical Biochemistry, Cerrahpasa Faculty of Medicine, Istanbul University-Cerrahpasa, Istanbul, Turkey
| |
Collapse
|
31
|
Tamura H, Nishio R, Saeki N, Katahira M, Morioka H, Tamano H, Takeda A. Paraquat-induced intracellular Zn 2+ dysregulation causes dopaminergic degeneration in the substantia nigra, but not in the striatum. Neurotoxicology 2022; 90:136-144. [PMID: 35339517 DOI: 10.1016/j.neuro.2022.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2022] [Revised: 03/08/2022] [Accepted: 03/19/2022] [Indexed: 11/15/2022]
Abstract
Parkinson's disease is characterized by a selective death of nigrostriatal dopaminergic neurons, while the difference in the vulnerability to the death between the substantia nigra pars compacta (SNpc) and the striatum is poorly understood. Here we tested the difference focused on paraquat (PQ)-induced intracellular Zn2+ toxicity via extracellular glutamate accumulation. When PQ was locally injected into the SNpc and the striatum, dopaminergic degeneration was observed in the SNpc, but not in the striatum. Intracellular hydrogen peroxide (H2O2) produced by PQ was increased in both the SNpc and the striatum. In contrast, extracellular glutamate accumulation was observed only in the SNpc and rescued in the presence of N-(p-amylcinnamoyl)anthranilic acid (ACA), a blocker of the transient receptor potential melastatin 2 (TRPM2) cation channels. PQ increased intracellular Zn2+ level in the SNpc, but not in the striatum. The increase was rescued by 1-naphthyl acetyl spermine (NASPM), a selective blocker of Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptors. PQ-induced dopaminergic degeneration in the SNpc was rescued by ACA, NASPM, and GBR, a dopamine reuptake inhibitor. The present study indicates intracellular H2O2 produced by PQ, which is taken up through dopamine transporters, is retrogradely transported to presynaptic glutamatergic terminals, activates TRPM2 channels, accumulates glutamate in the extracellular compartment, and induces intracellular Zn2+ dysregulation via Ca2+- and Zn2+-permeable GluR2-lacking AMPA receptor activation, resulting in dopaminergic degeneration in the SNpc. However, H2O2 signaling is not the case in the striatum. Paraquat-induced Zn2+ dysregulation plays a key role for neurodegeneration in the SNpc, but not in the striatum.
Collapse
Affiliation(s)
- Haruna Tamura
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryusuke Nishio
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Nana Saeki
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Misa Katahira
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiroki Morioka
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Atsushi Takeda
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
32
|
Mycobacterium tuberculosis Acetyltransferase Suppresses Oxidative Stress by Inducing Peroxisome Formation in Macrophages. Int J Mol Sci 2022; 23:ijms23052584. [PMID: 35269727 PMCID: PMC8909987 DOI: 10.3390/ijms23052584] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2021] [Revised: 11/20/2021] [Accepted: 11/20/2021] [Indexed: 02/01/2023] Open
Abstract
Mycobacterium tuberculosis (Mtb) inhibits host oxidative stress responses facilitating its survival in macrophages; however, the underlying molecular mechanisms are poorly understood. Here, we identified a Mtb acetyltransferase (Rv3034c) as a novel counter actor of macrophage oxidative stress responses by inducing peroxisome formation. An inducible Rv3034c deletion mutant of Mtb failed to induce peroxisome biogenesis, expression of the peroxisomal β-oxidation pathway intermediates (ACOX1, ACAA1, MFP2) in macrophages, resulting in reduced intracellular survival compared to the parental strain. This reduced virulence phenotype was rescued by repletion of Rv3034c. Peroxisome induction depended on the interaction between Rv3034c and the macrophage mannose receptor (MR). Interaction between Rv3034c and MR induced expression of the peroxisomal biogenesis proteins PEX5p, PEX13p, PEX14p, PEX11β, PEX19p, the peroxisomal membrane lipid transporter ABCD3, and catalase. Expression of PEX14p and ABCD3 was also enhanced in lungs from Mtb aerosol-infected mice. This is the first report that peroxisome-mediated control of ROS balance is essential for innate immune responses to Mtb but can be counteracted by the mycobacterial acetyltransferase Rv3034c. Thus, peroxisomes represent interesting targets for host-directed therapeutics to tuberculosis.
Collapse
|
33
|
Guo B, Li W, Liu Y, Xu D, Liu Z, Huang C. Aberrant Expressional Profiling of Small RNA by Cold Atmospheric Plasma Treatment in Human Chronic Myeloid Leukemia Cells. Front Genet 2022; 12:809658. [PMID: 35186012 PMCID: PMC8851033 DOI: 10.3389/fgene.2021.809658] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2021] [Accepted: 12/23/2021] [Indexed: 12/15/2022] Open
Abstract
Small RNAs (sRNAs), particularly microRNAs (miRNAs), are functional molecules that modulate mRNA transcripts and have been implicated in the etiology of various types of cancer. Cold atmospheric plasma (CAP) is a physical technology widely used in the field of cancer treatment after exhibiting extensive lethality on cancer cells. However, few studies have reported the exact role of miRNAs in CAP-induced anti-cancer effects. The aim of the present study was to determine whether miRNAs are involved in CAP-induced cytotoxicity by using high-throughput sequencing. Our research demonstrated that 28 miRNAs were significantly changed (17 upregulated and 11downregulated) following 24 h of treatment with a room-temperature argon plasma jet for 90 s compared with that of the untreated group in human chronic myeloid leukemia K562 cells. GO enrichment analysis revealed that these target genes were related to cell organelles, protein binding, and single-organism processes. Furthermore, KEGG pathway analysis demonstrated that the target genes of differentially expressed miRNAs were primarily involved in the cAMP signaling pathway, AMPK signaling pathway, and phosphatidylinositol signaling system. Taken together, our study demonstrated that CAP treatment could significantly alter the small RNA expression profile of chronic myeloid leukemia cells and provide a novel theoretical insight for elucidating the molecular mechanisms in CAP biomedicine application.
Collapse
Affiliation(s)
- Bo Guo
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Wen Li
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Yijie Liu
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
| | - Dehui Xu
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi’an Jiaotong University, Xi’an, China
| | - Zhijie Liu
- State Key Laboratory of Electrical Insulation and Power Equipment, Centre for Plasma Biomedicine, Xi’an Jiaotong University, Xi’an, China
| | - Chen Huang
- Department of Cell Biology and Genetics/Key Laboratory of Environment and Genes Related to Diseases, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Institute of Genetics and Developmental Biology, Translational Medicine Institute, School of Basic Medical Sciences, Xi’an Jiaotong University Health Science Center, Xi’an, China
- Key Laboratory of Shaanxi Province for Craniofacial Precision Medicine Research, Xi’an, China
- *Correspondence: Chen Huang,
| |
Collapse
|
34
|
Santos Wagner AL, Araniti F, Ishii-Iwamoto EL, Abenavoli MR. Resveratrol exerts beneficial effects on the growth and metabolism of Lactuca sativa L. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2022; 171:26-37. [PMID: 34971953 DOI: 10.1016/j.plaphy.2021.12.023] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2021] [Revised: 12/21/2021] [Accepted: 12/22/2021] [Indexed: 06/14/2023]
Abstract
In order to assist sustainable agriculture, new strategies and methods are being used based on the utilization of new natural molecules. These natural compounds can be used as potential natural crop protectors and growth promoters, and the elucidation of their modes/mechanisms of action can represent a big step towards cleaner agriculture free of agrochemicals. In the present paper, the mechanisms underlying the effects of exogenous resveratrol (R), a natural phytoalexin found in plants, on Lactuca sativa metabolism were investigated through physiological and metabolomic approaches. The results highlighted that R stimulates the growth of lettuce. A reduction of the O2⋅- production in R-treated seedlings and an increase in the photosynthesis efficiency was observed, indicated by a higher Fv/Fm. The metabolomic analysis of lettuce seedlings treated with R identified 116 metabolites related to galactose, amino acids, sugar and nucleotide sugar, and ascorbate and aldarate metabolisms. Increased content of some polyamines and several metabolites was also observed, which may have contributed to scavenging free radicals and activating antioxidant enzymes, thus reducing oxidative damage and improving PSII protection in R-treated seedlings.
Collapse
Affiliation(s)
- Ana Luiza Santos Wagner
- Laboratory of Biological Oxidations, Department of Biochemistry, State University of Maringa, 87020900, Maringa, Brazil
| | - Fabrizio Araniti
- Department of Agricultural and Environmental Sciences (DISAA), University of Milan, Via Celoria, 2, 20133, Milan, Italy
| | - Emy Luiza Ishii-Iwamoto
- Laboratory of Biological Oxidations, Department of Biochemistry, State University of Maringa, 87020900, Maringa, Brazil.
| | - Maria Rosa Abenavoli
- Department of Agriculture, University of Reggio di Calabria, 89124, Reggio Calabria, Italy.
| |
Collapse
|
35
|
Nishio R, Morioka H, Takeuchi A, Saeki N, Furuhata R, Katahira M, Chinenn T, Tamura H, Tamano H, Takeda A. Intracellular hydrogen peroxide produced by 6-hydroxydopamine is a trigger for nigral dopaminergic degeneration of rats via rapid influx of extracellular Zn 2. Neurotoxicology 2021; 89:1-8. [PMID: 34958835 DOI: 10.1016/j.neuro.2021.12.006] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 12/08/2021] [Accepted: 12/22/2021] [Indexed: 01/06/2023]
Abstract
To elucidate the mechanism and significance of 6-hydroxydopamine (6-OHDA)-induced Zn2+ toxicity, which is involved in neurodegeneration in the substantia nigra pars compacta (SNpc) of rats, we postulated that intracellular hydrogen peroxide (H2O2) produced by 6-OHDA is a trigger for intracellular Zn2+ dysregulation in the SNpc. Intracellular H2O2 level elevated by 6-OHDA in the SNpc was completely inhibited by co-injection of GBR 13069 dihydrochloride (GBR), a dopamine reuptake inhibitor, suggesting that 6-OHDA taken up through dopamine transporters produces H2O2 in the intercellular compartment of dopaminergic neurons. When the SNpc was perfused with H2O2, glutamate accumulated in the extracellular compartment and the accumulation was inhibited in the presence of N-(p-amylcinnamoyl)anthranilic acid (ACA), a blocker of the transient receptor potential melastatin 2 (TRPM2) channels. In addition to 6-OHDA, H2O2 also induced intracellular Zn2+ dysregulation via AMPA receptor activation followed by nigral dopaminergic degeneration. Furthermore, 6-OHDA-induced nigral dopaminergic degeneration was completely inhibited by co-injection of either HYDROP, an intracellular H2O2 scavenger or GBR into the SNpc. The present study indicates that H2O2 is produced by 6-OHDA taken up through dopamine transporters in the SNpc, is retrogradely transported to presynaptic glutamatergic terminals, activates TRPM2 channels, accumulates glutamate in the extracellular compartment, and induces intracellular Zn2+ dysregulation via AMPA receptor activation, resulting in nigral dopaminergic degeneration prior to movement disorder. It is likely that intracellular H2O2, but not extracellular H2O2, is a key trigger for nigral dopaminergic degeneration via intracellular Zn2+ dysregulation.
Collapse
Affiliation(s)
- Ryusuke Nishio
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Hiroki Morioka
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Azusa Takeuchi
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Nana Saeki
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Ryo Furuhata
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Misa Katahira
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Takato Chinenn
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamura
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Haruna Tamano
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan
| | - Atsushi Takeda
- School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka 422-8526, Japan.
| |
Collapse
|
36
|
Cao Q, Wang T, Xiao M, Bai L. Increased endogenous reactive oxygen species normalizes proliferation defects of Bmi1 heterozygous knockout neural stem cells. Neuroreport 2021; 32:1388-1394. [PMID: 34718251 DOI: 10.1097/wnr.0000000000001740] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
OBJECTIVES The Bmi1gene, one of transcriptional suppressor genes in multi-comb family, maintains proliferation of neural stem cells (NSCs) and redox homeostasis. However, heterozygous deletion of the Bmi1 gene (Bmi1+/-) does not reduce the proliferative ability of NSCs. The aim of the present study was to reveal the underlying mechanism of this phenotype. METHODS NSCs derived from the cortex of newborn Bmi1+/- and wild-type (WT) mice were treated with different concentrations of hydrogen peroxide (H2O2) and antioxidant N-acetyl-L-cysteine (NAC) for 24 h followed by analyses of NSC proliferation and oxidative stress-related indexes. RESULTS The levels of reactive oxygen species (ROS) of Bmi1+/--NSCs were slightly higher than that of WT-NSCs at baseline. H2O2 increased ROS and NAC reduced ROS in a concentration-dependent pattern, but the change was significantly greater in Bmi1+/--NSCs than WT-NSCs. The proliferation and self-renewal ability of Bmi1+/--NSCs and WT-NSCs were comparable in a basic state. After 1 μM H2O2 treatment, Brdu incorporation ratio, cell viability, total antioxidant capacity (T-AOC) and total superoxide dismutase activity were increased slightly in WT-NSCs, but decreased in Bmi1+/--NSCs. H2O2 at 10 μM decreased proliferation and self-renewal ability of both genotype NSCs, with greater effect in Bmi1+/-. After treatment with 1 mM NAC, the number and diameter of neurospheres, Brdu incorporation rate, cell viability, T-AOC and total superoxide dismutase activity of Bmi1+/--NSCs were lower than those of WT-NSCs. CONCLUSION These results suggest that Bmi1+/--NSCs exhibit normal proliferation and self-renewal due to a slight increase in ROS, but are more vulnerable to changes in redox status.
Collapse
Affiliation(s)
- Qiuchen Cao
- Jiangsu Key Laboratory of Neurodegeneration, Department of Pharmacology Nanjing Medical University, Nanjing, Jiangsu, People's Republic of China
| | | | | | | |
Collapse
|
37
|
Akhtar MJ, Ahamed M, Alhadlaq H. Anti-Inflammatory CeO 2 Nanoparticles Prevented Cytotoxicity Due to Exogenous Nitric Oxide Donors via Induction Rather Than Inhibition of Superoxide/Nitric Oxide in HUVE Cells. Molecules 2021; 26:5416. [PMID: 34500851 PMCID: PMC8434366 DOI: 10.3390/molecules26175416] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2021] [Revised: 09/01/2021] [Accepted: 09/04/2021] [Indexed: 11/16/2022] Open
Abstract
The mechanism behind the cytoprotective potential of cerium oxide nanoparticles (CeO2 NPs) against cytotoxic nitric oxide (NO) donors and H2O2 is still not clear. Synthesized and characterized CeO2 NPs significantly ameliorated the lipopolysaccharide (LPS)-induced cytokines IL-1β and TNF-α. The main goal of this study was to determine the capacities of NPs regarding signaling effects that could have occurred due to reactive oxygen species (ROS) and/or NO, since NP-induced ROS/NO did not lead to toxicity in HUVE cells. Concentrations that induced 50% cell death (i.e., IC50s) of two NO donors (DETA-NO; 1250 ± 110 µM and sodium nitroprusside (SNP); 950 ± 89 µM) along with the IC50 of H2O2 (120 ± 7 µM) were utilized to evaluate cytoprotective potential and its underlying mechanism. We determined total ROS (as a collective marker of hydrogen peroxide, superoxide radical (O2•-), hydroxyl radical, etc.) by DCFH-DA and used a O2•- specific probe DHE to decipher prominent ROS. The findings revealed that signaling effects mediated mainly by O2•- and/or NO are responsible for the amelioration of toxicity by CeO2 NPs at 100 µg/mL. The unaltered effect on mitochondrial membrane potential (MMP) due to NP exposure and, again, CeO2 NPs-mediated recovery in the loss of MMP due to exogenous NO donors and H2O2 suggested that NP-mediated O2•- production might be extra-mitochondrial. Data on activated glutathione reductase (GR) and unaffected glutathione peroxidase (GPx) activities partially explain the mechanism behind the NP-induced gain in GSH and persistent cytoplasmic ROS. The promoted antioxidant capacity due to non-cytotoxic ROS and/or NO production, rather than inhibition, by CeO2 NP treatment may allow cells to develop the capacity to tolerate exogenously induced toxicity.
Collapse
Affiliation(s)
- Mohd Javed Akhtar
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Maqusood Ahamed
- King Abdullah Institute for Nanotechnology, King Saud University, Riyadh 11451, Saudi Arabia;
| | - Hisham Alhadlaq
- Department of Physics and Astronomy, College of Sciences, King Saud University, Riyadh 11451, Saudi Arabia;
| |
Collapse
|
38
|
Quan Y, Du Y, Tong Y, Gu S, Jiang JX. Connexin Gap Junctions and Hemichannels in Modulating Lens Redox Homeostasis and Oxidative Stress in Cataractogenesis. Antioxidants (Basel) 2021; 10:1374. [PMID: 34573006 PMCID: PMC8464761 DOI: 10.3390/antiox10091374] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 08/24/2021] [Indexed: 11/18/2022] Open
Abstract
The lens is continuously exposed to oxidative stress insults, such as ultraviolet radiation and other oxidative factors, during the aging process. The lens possesses powerful oxidative stress defense systems to maintain its redox homeostasis, one of which employs connexin channels. Connexins are a family of proteins that form: (1) Hemichannels that mediate the communication between the intracellular and extracellular environments, and (2) gap junction channels that mediate cell-cell communication between adjacent cells. The avascular lens transports nutrition and metabolites through an extensive network of connexin channels, which allows the passage of small molecules, including antioxidants and oxidized wastes. Oxidative stress-induced post-translational modifications of connexins, in turn, regulates gap junction and hemichannel permeability. Recent evidence suggests that dysfunction of connexins gap junction channels and hemichannels may induce cataract formation through impaired redox homeostasis. Here, we review the recent advances in the knowledge of connexin channels in lens redox homeostasis and their response to cataract-related oxidative stress by discussing two major aspects: (1) The role of lens connexins and channels in oxidative stress and cataractogenesis, and (2) the impact and underlying mechanism of oxidative stress in regulating connexin channels.
Collapse
Affiliation(s)
| | | | | | | | - Jean X. Jiang
- Department of Biochemistry and Structural Biology, University of Texas Health Science Center, San Antonio, TX 78229, USA; (Y.Q.); (Y.D.); (Y.T.); (S.G.)
| |
Collapse
|
39
|
Rac-dependent feedforward autoactivation of NOX2 leads to oxidative burst. J Biol Chem 2021; 297:100982. [PMID: 34293347 PMCID: PMC8353492 DOI: 10.1016/j.jbc.2021.100982] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2021] [Revised: 07/09/2021] [Accepted: 07/17/2021] [Indexed: 12/03/2022] Open
Abstract
NADPH oxidase 2 (NOX2) produces the superoxide anion radical (O2−), which has functions in both cell signaling and immune defense. NOX2 is a multimeric-protein complex consisting of several protein subunits including the GTPase Rac. NOX2 uniquely facilitates an oxidative burst, which is described by initially slow O2− production, which increases over time. The NOX2 oxidative burst is considered critical to immune defense because it enables expedited O2− production in response to infections. However, the mechanism of the initiation and progression of this oxidative burst and its implications for regulation of NOX2 have not been clarified. In this study, we show that the NOX2 oxidative burst is a result of autoactivation of NOX2 coupled with the redox function of Rac. NOX2 autoactivation begins when active Rac triggers NOX2 activation and the subsequent production of O2−, which in turn activates redox-sensitive Rac. This activated Rac further activates NOX2, amplifying the feedforward cycle and resulting in a NOX2-mediated oxidative burst. Using mutagenesis-based kinetic and cell analyses, we show that enzymatic activation of Rac is exclusively responsible for production of the active Rac trigger that initiates NOX2 autoactivation, whereas redox-mediated Rac activation is the main driving force of NOX2 autoactivation and contributes to generation of ∼98% of the active NOX2 in cells. The results of this study provide insight into the regulation of NOX2 function, which could be used to develop therapeutics to control immune responses associated with dysregulated NOX2 oxidative bursts.
Collapse
|
40
|
Alterations in the Plasma and Red Blood Cell Properties in Patients with Varicose Vein: A Pilot Study. Cardiol Res Pract 2021; 2021:5569961. [PMID: 34306747 PMCID: PMC8263278 DOI: 10.1155/2021/5569961] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/11/2021] [Revised: 06/10/2021] [Accepted: 06/22/2021] [Indexed: 01/24/2023] Open
Abstract
The varicose vein results from the inefficient functioning of the valves in the lower limb veins, making the blood flow slow down and leading to blood stasis and hypoxia. This type of vein dysfunction might be a result of the development of oxidative stress. We compared oxidative stress markers in the plasma and erythrocytes obtained from peripheral veins and varicose veins in the same patients (glutathione, nonenzymatic antioxidant capacity (NEAC), catalase (CAT) and acetylcholinesterase (AChE) activity, thiols, thiobarbituric acid-reactive substance (TBARS), and protein carbonyls). We found a decrease in NEAC in the plasma obtained from the varicose veins compared to the peripheral veins. We detected a decrease in thiols in the plasma, hemolysate, and plasma membranes and increase in protein carbonyl compounds and TBARS levels in the varicose veins. These changes were accompanied by a decrease in CAT and AChE activity. For the first time, our results show changes in the plasma, erythrocyte membrane, and hemolysate protein properties in varicose vein blood in contrast to the plasma and erythrocytes in peripheral vein blood from the same patients. The increased oxidative stress accompanying varicose vein disease might result from the local inefficiency of the antioxidant defense system.
Collapse
|
41
|
Montiel V, Bella R, Michel LYM, Esfahani H, De Mulder D, Robinson EL, Deglasse JP, Tiburcy M, Chow PH, Jonas JC, Gilon P, Steinhorn B, Michel T, Beauloye C, Bertrand L, Farah C, Dei Zotti F, Debaix H, Bouzin C, Brusa D, Horman S, Vanoverschelde JL, Bergmann O, Gilis D, Rooman M, Ghigo A, Geninatti-Crich S, Yool A, Zimmermann WH, Roderick HL, Devuyst O, Balligand JL. Inhibition of aquaporin-1 prevents myocardial remodeling by blocking the transmembrane transport of hydrogen peroxide. Sci Transl Med 2021; 12:12/564/eaay2176. [PMID: 33028705 DOI: 10.1126/scitranslmed.aay2176] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2019] [Revised: 12/24/2019] [Accepted: 08/31/2020] [Indexed: 12/31/2022]
Abstract
Pathological remodeling of the myocardium has long been known to involve oxidant signaling, but strategies using systemic antioxidants have generally failed to prevent it. We sought to identify key regulators of oxidant-mediated cardiac hypertrophy amenable to targeted pharmacological therapy. Specific isoforms of the aquaporin water channels have been implicated in oxidant sensing, but their role in heart muscle is unknown. RNA sequencing from human cardiac myocytes revealed that the archetypal AQP1 is a major isoform. AQP1 expression correlates with the severity of hypertrophic remodeling in patients with aortic stenosis. The AQP1 channel was detected at the plasma membrane of human and mouse cardiac myocytes from hypertrophic hearts, where it colocalized with NADPH oxidase-2 and caveolin-3. We show that hydrogen peroxide (H2O2), produced extracellularly, is necessary for the hypertrophic response of isolated cardiac myocytes and that AQP1 facilitates the transmembrane transport of H2O2 through its water pore, resulting in activation of oxidant-sensitive kinases in cardiac myocytes. Structural analysis of the amino acid residues lining the water pore of AQP1 supports its permeation by H2O2 Deletion of Aqp1 or selective blockade of the AQP1 intrasubunit pore inhibited H2O2 transport in mouse and human cells and rescued the myocyte hypertrophy in human induced pluripotent stem cell-derived engineered heart muscle. Treatment of mice with a clinically approved AQP1 inhibitor, Bacopaside, attenuated cardiac hypertrophy. We conclude that cardiac hypertrophy is mediated by the transmembrane transport of H2O2 by the water channel AQP1 and that inhibitors of AQP1 represent new possibilities for treating hypertrophic cardiomyopathies.
Collapse
Affiliation(s)
- Virginie Montiel
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Ramona Bella
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Lauriane Y M Michel
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Hrag Esfahani
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Delphine De Mulder
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Emma L Robinson
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KULeuven, 3000 Leuven, Belgium
| | - Jean-Philippe Deglasse
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Malte Tiburcy
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany
| | - Pak Hin Chow
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Jean-Christophe Jonas
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Patrick Gilon
- Institute of Experimental and Clinical Research (IREC), Endocrinology, Diabetes and Nutrition (EDIN), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Benjamin Steinhorn
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 2115, USA
| | - Thomas Michel
- Cardiovascular Division, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, MA 2115, USA
| | - Christophe Beauloye
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Luc Bertrand
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Charlotte Farah
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Flavia Dei Zotti
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Huguette Debaix
- Institute of Experimental and Clinical Research (IREC), Nephrology (NEFR), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.,Institute of Physiology, University of Zürich, CH 8057 Zürich, Switzerland
| | - Caroline Bouzin
- 2IP-IREC Imaging Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Davide Brusa
- Flow Cytometry Platform, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Sandrine Horman
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Jean-Louis Vanoverschelde
- Institute of Experimental and Clinical Research (IREC), Pole of Cardiovascular Research (CARD), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium
| | - Olaf Bergmann
- Center for Regenerative Therapies Dresden, Technische Universität Dresden, 01062 Dresden, Germany.,Department of Cell and Molecular Biology, Karolinska Institutet, 171 77 Stockholm, Sweden
| | - Dimitri Gilis
- Computational Biology and Bioinformatics (3BIO-BioInfo), Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Marianne Rooman
- Computational Biology and Bioinformatics (3BIO-BioInfo), Université Libre de Bruxelles (ULB), 1000 Brussels, Belgium
| | - Alessandra Ghigo
- Molecular Biotechnology Center, Università di Torino, 10124 Torino, Italy
| | | | - Andrea Yool
- Adelaide Medical School, University of Adelaide, Adelaide, SA 5000, Australia
| | - Wolfram H Zimmermann
- Institute of Pharmacology and Toxicology, University Medical Center Göttingen, 37075 Göttingen, Germany.,DZHK (German Center for Cardiovascular Research), Partner Site Göttingen, 37075 Göttingen, Germany.,Cluster of Excellence "Multiscale Bioimaging: from Molecular Machines to Networks of Excitable Cells" (MBExC), University of Göttingen, 37075 Göttingen, Germany
| | - H Llewelyn Roderick
- Laboratory of Experimental Cardiology, Department of Cardiovascular Sciences, KULeuven, 3000 Leuven, Belgium
| | - Olivier Devuyst
- Institute of Experimental and Clinical Research (IREC), Nephrology (NEFR), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.,Institute of Physiology, University of Zürich, CH 8057 Zürich, Switzerland
| | - Jean-Luc Balligand
- Institute of Experimental and Clinical Research (IREC), Pharmacology and Therapeutics (FATH), Cliniques Universitaires St Luc and Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium.
| |
Collapse
|
42
|
Macrophage-produced peroxynitrite induces antibiotic tolerance and supersedes intrinsic mechanisms of persister formation. Infect Immun 2021; 89:e0028621. [PMID: 34097475 DOI: 10.1128/iai.00286-21] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Staphylococcus aureus is a leading human pathogen that frequently causes chronic and relapsing infections. Antibiotic tolerant persister cells contribute to frequent antibiotic failure in patients. Macrophages represent an important niche during S. aureus bacteremia and recent work has identified a role for oxidative burst in the formation of antibiotic tolerant S. aureus. We find that host-derived peroxynitrite, the reaction product of superoxide and nitric oxide, is the main mediator of antibiotic tolerance in macrophages. Using a collection of S. aureus clinical isolates, we find that, despite significant variation in persister formation in pure culture, all strains were similarly enriched for antibiotic tolerance following internalization by activated macrophages. Our findings suggest that host interaction strongly induces antibiotic tolerance and may negate bacterial mechanisms of persister formation, established in pure culture. These findings emphasize the importance of studying antibiotic tolerance in the context of bacterial interaction with the host suggest that modulation of the host response may represent a viable therapeutic strategy to sensitize S. aureus to antibiotics.
Collapse
|
43
|
Muller JDAI, Lencina JDS, Souza MIL, Mortari MR, Toffoli-Kadri MC. Macrophage activation in vitro by Parachartergus fraternus venom. Toxicon 2021; 198:48-53. [PMID: 33940047 DOI: 10.1016/j.toxicon.2021.04.028] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Revised: 04/25/2021] [Accepted: 04/28/2021] [Indexed: 10/21/2022]
Abstract
Parachartergus fraternus wasp induces inflammation with a predominance of mononuclear cells, that can acquire macrophage functions at the sting site, amplifying the response. These cells can be activated by venomous animals and are involved in destruction of injurious agents and release of inflammatory mediators. The objective of this work was to evaluate the activity of P. fraternus venom (Pfv) on isolated murine macrophage function. The cells were obtained from peritoneal cavity of Swiss male mice and incubated with Pfv (2.5, 5 and 10 μg/mL). Cytotoxicity was determined using MTT assay. Adhesion and detachment were evaluated using violet crystal dye. Spreading was evaluated based on morphological parameters. Phagocytosis was performed with opsonized zymosan. Production of hydrogen peroxide (H2O2) and nitric oxide (NO) were quantified using the phenol red and Griess assays, respectively. Pfv at concentrations evaluated was not cytotoxic in MTT assay and did not cause macrophage detachment in cell culture plates. However, it increased adhesion of macrophage, spreading and phagocytosis of opsonized zymosan, as well as induced production of H2O2 and NO. Therefore, Pfv induces macrophage activation in vitro and the response of these cells can be correlated with the previously reported inflammatory process triggered by this wasp.
Collapse
Affiliation(s)
- Jéssica de Araujo Isaias Muller
- Laboratory of Pharmacology and Inflammation, FACFAN/Federal University of Mato Grosso do Sul, Brazil; PhD Student of the Multicenter Program of Post-Graduation in Biochemistry and Molecular Biology, INBIO/Federal University of Mato Grosso do Sul, Brazil
| | - Joyce Dos Santos Lencina
- Laboratory of Pharmacology and Inflammation, FACFAN/Federal University of Mato Grosso do Sul, Brazil; Master of Science Student of the Program of Post-Graduation in Pharmacy, FACFAN/ Federal University of Mato Grosso do Sul, Brazil
| | - Maria Inês Lenz Souza
- Laboratory of Biophysiopharmacology, INBIO/ Federal University of Mato Grosso do Sul, Brazil, CB/University of Brasilia, Brazil
| | | | | |
Collapse
|
44
|
Ghosh S, Liu H, Yazdankhah M, Stepicheva N, Shang P, Vaidya T, Hose S, Gupta U, Calderon MJ, Hu MW, Nair AP, Weiss J, Fitting CS, Bhutto IA, Gadde SGK, Naik NK, Jaydev C, Lutty GA, Handa JT, Jayagopal A, Qian J, Sahel JA, Rajasundaram D, Sergeev Y, Zigler JS, Sethu S, Watkins S, Ghosh A, Sinha D. βA1-crystallin regulates glucose metabolism and mitochondrial function in mouse retinal astrocytes by modulating PTP1B activity. Commun Biol 2021; 4:248. [PMID: 33627831 PMCID: PMC7904954 DOI: 10.1038/s42003-021-01763-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2020] [Accepted: 01/28/2021] [Indexed: 02/08/2023] Open
Abstract
βA3/A1-crystallin, a lens protein that is also expressed in astrocytes, is produced as βA3 and βA1-crystallin isoforms by leaky ribosomal scanning. In a previous human proteome high-throughput array, we found that βA3/A1-crystallin interacts with protein tyrosine phosphatase 1B (PTP1B), a key regulator of glucose metabolism. This prompted us to explore possible roles of βA3/A1-crystallin in metabolism of retinal astrocytes. We found that βA1-crystallin acts as an uncompetitive inhibitor of PTP1B, but βA3-crystallin does not. Loss of βA1-crystallin in astrocytes triggers metabolic abnormalities and inflammation. In CRISPR/cas9 gene-edited βA1-knockdown (KD) mice, but not in βA3-knockout (KO) mice, the streptozotocin (STZ)-induced diabetic retinopathy (DR)-like phenotype is exacerbated. Here, we have identified βA1-crystallin as a regulator of PTP1B; loss of this regulation may be a new mechanism by which astrocytes contribute to DR. Interestingly, proliferative diabetic retinopathy (PDR) patients showed reduced βA1-crystallin and higher levels of PTP1B in the vitreous humor.
Collapse
Affiliation(s)
- Sayan Ghosh
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Haitao Liu
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Meysam Yazdankhah
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Nadezda Stepicheva
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Peng Shang
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Tanuja Vaidya
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Stacey Hose
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Urvi Gupta
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Michael Joseph Calderon
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Ming-Wen Hu
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Joseph Weiss
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Christopher S Fitting
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Imran A Bhutto
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Naveen Kumar Naik
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Chaitra Jaydev
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Gerard A Lutty
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - James T Handa
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | | | - Jiang Qian
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - José-Alain Sahel
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Institut de la Vision, INSERM, CNRS, Sorbonne Université, Paris, France
| | - Dhivyaa Rajasundaram
- Department of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Yuri Sergeev
- National Eye Institute, National Institutes of Health, Bethesda, MD, USA
| | - J Samuel Zigler
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Swaminathan Sethu
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Simon Watkins
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Arkasubhra Ghosh
- GROW Research Laboratory, Narayana Nethralaya Foundation, Bengaluru, India
| | - Debasish Sinha
- Department of Ophthalmology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Department of Cell Biology and Center for Biologic Imaging, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
- Wilmer Eye Institute, The Johns Hopkins University School of Medicine, Baltimore, MD, USA.
| |
Collapse
|
45
|
Bilcke G, Van den Berge K, De Decker S, Bonneure E, Poulsen N, Bulankova P, Osuna-Cruz CM, Dickenson J, Sabbe K, Pohnert G, Vandepoele K, Mangelinckx S, Clement L, De Veylder L, Vyverman W. Mating type specific transcriptomic response to sex inducing pheromone in the pennate diatom Seminavis robusta. THE ISME JOURNAL 2021; 15:562-576. [PMID: 33028976 PMCID: PMC8027222 DOI: 10.1038/s41396-020-00797-7] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 09/10/2020] [Accepted: 09/24/2020] [Indexed: 12/14/2022]
Abstract
Sexual reproduction is a fundamental phase in the life cycle of most diatoms. Despite its role as a source of genetic variation, it is rarely reported in natural circumstances and its molecular foundations remain largely unknown. Here, we integrate independent transcriptomic datasets to prioritize genes responding to sex inducing pheromones (SIPs) in the pennate diatom Seminavis robusta. We observe marked gene expression changes associated with SIP treatment in both mating types, including an inhibition of S phase progression, chloroplast division, mitosis, and cell wall formation. Meanwhile, meiotic genes are upregulated in response to SIP, including a sexually induced diatom specific cyclin. Our data further suggest an important role for reactive oxygen species, energy metabolism, and cGMP signaling during the early stages of sexual reproduction. In addition, we identify several genes with a mating type specific response to SIP, and link their expression pattern with physiological specialization, such as the production of the attraction pheromone diproline in mating type - (MT-) and mate-searching behavior in mating type + (MT+). Combined, our results provide a model for early sexual reproduction in pennate diatoms and significantly expand the suite of target genes to detect sexual reproduction events in natural diatom populations.
Collapse
Affiliation(s)
- Gust Bilcke
- Protistology and Aquatic Ecology, Department of Biology, Ghent University, 9000, Ghent, Belgium
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- VIB Center for Plant Systems Biology, 9052, Ghent, Belgium
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000, Ghent, Belgium
| | - Koen Van den Berge
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000, Ghent, Belgium
- Department of Statistics, University of California, Berkeley, Berkeley, CA, USA
| | - Sam De Decker
- Protistology and Aquatic Ecology, Department of Biology, Ghent University, 9000, Ghent, Belgium
| | - Eli Bonneure
- SynBioC, Department of Green Chemistry and Technology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Nicole Poulsen
- B CUBE Center for Molecular Bioengineering, Technical University of Dresden, Tatzberg 41, 01307, Dresden, Germany
| | - Petra Bulankova
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- VIB Center for Plant Systems Biology, 9052, Ghent, Belgium
| | - Cristina Maria Osuna-Cruz
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- VIB Center for Plant Systems Biology, 9052, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
| | - Jack Dickenson
- Marine Biological Association, The Laboratory, Citadel Hill, Plymouth, PL1 2PB, UK
- School of Ocean and Earth Science, University of Southampton, Southampton, UK
| | - Koen Sabbe
- Protistology and Aquatic Ecology, Department of Biology, Ghent University, 9000, Ghent, Belgium
| | - Georg Pohnert
- Bioorganic Analytics, Institute for Inorganic and Analytical Chemistry, Friedrich Schiller University Jena, Lessingstr. 8, 07743, Jena, Germany
| | - Klaas Vandepoele
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- VIB Center for Plant Systems Biology, 9052, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
| | - Sven Mangelinckx
- SynBioC, Department of Green Chemistry and Technology, Ghent University, Coupure Links 653, 9000, Ghent, Belgium
| | - Lieven Clement
- Department of Applied Mathematics, Computer Science and Statistics, Ghent University, 9000, Ghent, Belgium
- Bioinformatics Institute Ghent, Ghent University, Technologiepark 71, 9052, Ghent, Belgium
| | - Lieven De Veylder
- Department of Plant Biotechnology and Bioinformatics, Ghent University, 9052, Ghent, Belgium
- VIB Center for Plant Systems Biology, 9052, Ghent, Belgium
| | - Wim Vyverman
- Protistology and Aquatic Ecology, Department of Biology, Ghent University, 9000, Ghent, Belgium.
| |
Collapse
|
46
|
Duanghathaipornsuk S, Farrell EJ, Alba-Rubio AC, Zelenay P, Kim DS. Detection Technologies for Reactive Oxygen Species: Fluorescence and Electrochemical Methods and Their Applications. BIOSENSORS 2021; 11:30. [PMID: 33498809 PMCID: PMC7911324 DOI: 10.3390/bios11020030] [Citation(s) in RCA: 44] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/28/2020] [Revised: 01/12/2021] [Accepted: 01/19/2021] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) have been found in plants, mammals, and natural environmental processes. The presence of ROS in mammals has been linked to the development of severe diseases, such as diabetes, cancer, tumors, and several neurodegenerative conditions. The most common ROS involved in human health are superoxide (O2•-), hydrogen peroxide (H2O2), and hydroxyl radicals (•OH). Organic and inorganic molecules have been integrated with various methods to detect and monitor ROS for understanding the effect of their presence and concentration on diseases caused by oxidative stress. Among several techniques, fluorescence and electrochemical methods have been recently developed and employed for the detection of ROS. This literature review intends to critically discuss the development of these techniques to date, as well as their application for in vitro and in vivo ROS detection regarding free-radical-related diseases. Moreover, important insights into and further steps for using fluorescence and electrochemical methods in the detection of ROS are presented.
Collapse
Affiliation(s)
| | - Eveline J Farrell
- Department of Chemical Engineering, The University of Toledo, Toledo, OH 43606, USA
| | - Ana C Alba-Rubio
- Department of Chemical Engineering, The University of Toledo, Toledo, OH 43606, USA
| | - Piotr Zelenay
- Materials Physics and Applications Division, Los Alamos National Laboratory, Los Alamos, NM 87545, USA
| | - Dong-Shik Kim
- Department of Chemical Engineering, The University of Toledo, Toledo, OH 43606, USA
| |
Collapse
|
47
|
Nwasike C, Purr E, Yoo E, Nagi JS, Doiron AL. Activatable Nanoparticles: Recent Advances in Redox-Sensitive Magnetic Resonance Contrast Agent Candidates Capable of Detecting Inflammation. Pharmaceuticals (Basel) 2021; 14:69. [PMID: 33467028 PMCID: PMC7829999 DOI: 10.3390/ph14010069] [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: 12/31/2020] [Revised: 01/14/2021] [Accepted: 01/14/2021] [Indexed: 11/16/2022] Open
Abstract
The emergence of activatable magnetic resonance (MR) contrast agents has prompted significant interest in the detection of functional markers of diseases, resulting in the creation of a plethora of nanoprobes capable of detecting these biomarkers. These markers are commonly dysregulated in several chronic diseases, specifically select cancers and inflammatory diseases. Recently, the development of redox-sensitive nanoparticle-based contrast agents has gained momentum given advances in medicine linking several inflammatory diseases to redox imbalance. Researchers have pinpointed redox dysregulation as an opportunity to use activatable MR contrast agents to detect and stage several diseases as well as monitor the treatment of inflammatory diseases or conditions. These new classes of agents represent an advancement in the field of MR imaging as they elicit a response to stimuli, creating contrast while providing evidence of biomarker changes and commensurate disease state. Most redox-sensitive nanoparticle-based contrast agents are sensitive to reductive glutathione or oxidative reactive oxygen species. In this review, we will explore recent investigations into redox-activatable, nanoparticle-based MR contrast agent candidates.
Collapse
Affiliation(s)
- Chukwuazam Nwasike
- Department of Biomedical Engineering, Binghamton University (SUNY), Binghamton, NY 13902, USA; (C.N.); (E.P.)
| | - Erin Purr
- Department of Biomedical Engineering, Binghamton University (SUNY), Binghamton, NY 13902, USA; (C.N.); (E.P.)
| | - Eunsoo Yoo
- Department of Otolaryngology-Head & Neck Surgery, University of Pennsylvania, Philadelphia, PA 19104, USA;
| | - Jaspreet Singh Nagi
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA;
| | - Amber L. Doiron
- Department of Electrical and Biomedical Engineering, University of Vermont, Burlington, VT 05405, USA;
| |
Collapse
|
48
|
Edge R, Truscott TG. COVID-19 and the ethnicity link - is there a photochemical link? Photochem Photobiol Sci 2021; 20:183-188. [PMID: 33721239 PMCID: PMC7791943 DOI: 10.1007/s43630-020-00004-8] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2020] [Accepted: 12/10/2020] [Indexed: 11/23/2022]
Abstract
A hypothesis is proposed to explain the increased detrimental effect of COVID-19 for Black, Asian and Minority Ethnic (BAME) men and women compared to Caucasian individuals. This is based on the differing photochemistry of phaeomelanin in fair skin and eumelanin in dark/black skin. It is suggested that a range of reactive oxygen species, including, singlet oxygen and the superoxide radical anion, derived via direct photolysis of phaeomelanin, may escape the melanocyte and cause subsequent damage to the SARS-CoV-2 virus. It is further suggested that (large) carbon and sulphur peroxy radicals, from oxygen addition to radicals formed by carbon–sulphur bond cleavage, may assist via damage to the cell membranes. It is also speculated that light absorption by phaeomelanin and the subsequent C-S bond cleavage, leads to release of pre-absorbed reactive oxygen species, such as singlet oxygen and free radicals, which may also contribute to an enhanced protective effect for fair-skinned people.
Collapse
Affiliation(s)
- Ruth Edge
- Dalton Cumbrian Facility, Westlakes Science Park, The University of Manchester, Cumbria, CA24 3HA, UK
| | - T George Truscott
- School of Chemical and Physical Sciences, Keele University, Staffordshire, ST5 5BG, UK.
| |
Collapse
|
49
|
Singh N. Antioxidant metal oxide nanozymes: role in cellular redox homeostasis and therapeutics. PURE APPL CHEM 2020. [DOI: 10.1515/pac-2020-0802] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Abstract
Nanomaterials with enzyme-like activity, generally referred to as ‘nanozymes’, find myriad potential in various biomedical fields. More importantly, the nanoparticles that can functionally mimic the activity of cellular antioxidant enzymes attract tremendous interest owing to their possible therapeutic candidature in oxidative stress-mediated disorders. Oxidative stress culminating due to excess reactive oxygen species (ROS) level and dysregulated cellular antioxidant machinery is implicated in the development and progression of various pathophysiological disorders such as cancer, diabetes, cardiovascular and neurodegenerative diseases. Moreover, the optimum essentiality of ROS due to its pivotal role in cell signaling evokes the requirement of novel artificial antioxidant enzymes that can circumvent the detrimental effects of enhanced ROS levels without perturbing the basal redox status of cells. In recent years, the fast emanating artificial enzymes, i.e. nanozymes with antioxidant enzyme-like activity, has made tremendous progress with their broad applications in therapeutics, diagnostic medicine, bio-sensing, and immunoassay. Among various antioxidant nanoparticles reported till-date, the metal oxide nanozymes have emerged as the most efficient and successful candidates in mimicking the activity of first-line defense antioxidant enzymes, i.e. superoxide dismutase, catalase, and glutathione peroxidase. This review intends to exclusively highlight the development of representative metal oxide-based antioxidant nanozymes capable of maintaining the cellular redox homeostasis and their potential therapeutic significance.
Collapse
Affiliation(s)
- Namrata Singh
- Department of Inorganic and Physical Chemistry , Indian Institute of Science , Bangalore 560012 , India
| |
Collapse
|
50
|
The Universal Soldier: Enzymatic and Non-Enzymatic Antioxidant Functions of Serum Albumin. Antioxidants (Basel) 2020; 9:antiox9100966. [PMID: 33050223 PMCID: PMC7601824 DOI: 10.3390/antiox9100966] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 10/06/2020] [Accepted: 10/07/2020] [Indexed: 12/14/2022] Open
Abstract
As a carrier of many biologically active compounds, blood is exposed to oxidants to a greater extent than the intracellular environment. Serum albumin plays a key role in antioxidant defence under both normal and oxidative stress conditions. This review evaluates data published in the literature and from our own research on the mechanisms of the enzymatic and non-enzymatic activities of albumin that determine its participation in redox modulation of plasma and intercellular fluid. For the first time, the results of numerous clinical, biochemical, spectroscopic and computational experiments devoted to the study of allosteric modulation of the functional properties of the protein associated with its participation in antioxidant defence are analysed. It has been concluded that it is fundamentally possible to regulate the antioxidant properties of albumin with various ligands, and the binding and/or enzymatic features of the protein by changing its redox status. The perspectives for using the antioxidant properties of albumin in practice are discussed.
Collapse
|