1
|
Jourd'heuil F, Mathai C, Cat Pham LG, Gilliard K, Balnis J, Overmyer KA, Coon JJ, Jaitovich A, Boivin B, Jourd'heuil D. Cytoglobin scavenges intracellular hydrogen peroxide and regulates redox signals in the vasculature. Redox Biol 2025; 83:103633. [PMID: 40252320 PMCID: PMC12023880 DOI: 10.1016/j.redox.2025.103633] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2025] [Revised: 04/01/2025] [Accepted: 04/08/2025] [Indexed: 04/21/2025] Open
Abstract
The oxidant hydrogen peroxide (H2O2) serves as a signaling molecule that alters many aspects of cardiovascular functions and contributes to cardiovascular diseases. Recent studies suggest that cytoglobin - a member of the globin family - may promote electron transfer reactions with proposed functions in H2O2 decomposition. In the present study, we directly examined the ability of cytoglobin to decompose H2O2. Carotid arteries from cytoglobin knockout mice were more sensitive to glycolytic inhibition by H2O2 than arteries from wild type mice. In addition, the ectopic expression of cytoglobin in cultured cells limited the inhibitory effect of H2O2 on glycolysis and reversed the oxidative inactivation of the glycolytic enzyme GAPDH. Cytoglobin facilitated the reduction of the thiol-based H2O2 sensor Hyper7 after H2O2 challenge. The specific substitution of one of two cysteine residues on cytoglobin (C83) inhibited its antioxidant activity, as did the substitutions at the proximal and distal histidine residues. In vitro, direct measurements of H2O2 concentrations indicated that purified cytoglobin consumes H2O2 at rates comparable to that of peroxiredoxin 2 and that it competitively inhibits the hyperoxidation of peroxiredoxin 2. We propose that cytoglobin may serve as a regulator of intracellular redox signals initiated by H2O2.
Collapse
Affiliation(s)
- Frances Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Clinton Mathai
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Le Gia Cat Pham
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Kurrim Gilliard
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA
| | - Joseph Balnis
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Katherine A Overmyer
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA
| | - Joshua J Coon
- Department of Biomolecular Chemistry, University of Wisconsin-Madison, Madison, WI, USA; Morgridge Institute for Research, Madison, WI, USA; Department of Chemistry, University of Wisconsin-Madison, Madison, WI, USA
| | - Ariel Jaitovich
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA; Division of Pulmonary and Critical Care Medicine, Albany Medical College, Albany, NY, USA
| | - Benoit Boivin
- Department of Nanoscale Science & Engineering, University at Albany, Albany, NY, USA
| | - David Jourd'heuil
- Department of Molecular and Cellular Physiology, Albany Medical College, Albany, NY, USA.
| |
Collapse
|
2
|
Kano R, Takeda R, Sotani Y, Takagi R, Tabuchi A, Shirakawa H, Poole DC, Kano Y, Hoshino D. Cooling-induced changes in intracellular hydrogen peroxide and gene expression in mouse skeletal muscle in vivo. Am J Physiol Regul Integr Comp Physiol 2025; 328:R758-R766. [PMID: 40332827 DOI: 10.1152/ajpregu.00014.2025] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 02/13/2025] [Accepted: 04/22/2025] [Indexed: 05/08/2025]
Abstract
Changes in intracellular hydrogen peroxide concentration ([H2O2]) constitute an important signal-controlling cellular adaptations. In response to cooling, decreases in [H2O2] and changes in antioxidant-related gene expression have been observed in skeletal muscle. However, the specific temperature dependence of cooling-induced [H2O2] changes and their quantitative relationship to induced gene expression are unknown. This investigation tested the hypothesis that differences in muscle cytosolic and mitochondrial [H2O2] changes during cooling/rewarming determine the pattern of H2O2-related gene expression. H2O2-sensitive cytosolic (HyPer7) and mitochondrial (MLS-HyPer7) fluorescent proteins were expressed into tibialis anterior (TA) muscle of male C57BL/6J mice. The temperature dependence of [H2O2] was determined via in vivo imaging during a 3-min cooling protocol from 35°C to 0°C. Two cooling patterns [6 bouts of intermittent cooling (I-Cool) vs. sustained cooling (S-Cool); both to 13°C] were applied over 60 min. Three hours after cooling, the muscles were removed, and gene expression was evaluated using real-time PCR. The decrease in [H2O2] was observed in both cytosolic and mitochondrial compartments from 35°C to 13°C but was of greater magnitude in the cytosol; in contrast, further cooling from 12°C to 0°C induced a rebound increase especially in cytosolic [H2O2]. I-Cool increased the mRNA level of Nrf2 (+15%, P < 0.001). S-Cool decreased the mRNA levels of Sod2, Cat, and Ucp3 (i.e., -20, -23, and -30%, respectively, P < 0.05). In conclusion, the greatest decrease in temperature-dependent [H2O2] occurred at 13°C in the cytosolic and mitochondrial compartments of muscle fibers, and I-Cool increased Nrf2 mRNA expression, whereas S-Cool decreased several antioxidant-related genes.NEW & NOTEWORTHY This in vivo model successfully characterized the effects of cooling on cytosolic and mitochondrial [H2O2] in mouse tibialis anterior skeletal muscle. Cooling decreased [H2O2] down to ∼13°C, but the effect was reversed at still lower temperatures. Sustained cooling decreased mRNA levels of antioxidant-related genes (Sod2, Cat, and Ucp3), whereas intermittent cooling increased Nrf mRNA expression. These results help elucidate the mechanistic bases for skeletal muscle adaptation to cooling.
Collapse
Affiliation(s)
- Ryotaro Kano
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
- Research Fellowship for Young Scientists, Japan Society for the Promotion of Science, Chiyoda, Japan
| | - Reo Takeda
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
- Cellular and Molecular Biotechnology Research Institute, National Institute of Advanced Industrial Science and Technology, Tsukuba, Japan
| | - Yuta Sotani
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
| | - Ryo Takagi
- Department of Physical Therapy, Showa University School of Nursing and Rehabilitation Sciences, Kanagawa, Japan
| | - Ayaka Tabuchi
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
| | - Hideki Shirakawa
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
| | - David C Poole
- Department of Anatomy and Physiology and Kinesiology, Kansas State University, Manhattan, Kansas, United States
- Department of Kinesiology, Kansas State University, Manhattan, Kansas, United States
| | - Yutaka Kano
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
| | - Daisuke Hoshino
- Department of Engineering Science, Bioscience and Technology Program, University of Electro-Communications, Chofu, Japan
| |
Collapse
|
3
|
Miri SM, Ata BN, Çimen Ş, Barakat S, Ghaffari Zaki A, Armouch J, Vatandaşlar E, Vilain S, Öztürk G, Eroğlu E. Development of an Oxygen-Insensitive Nrf2 Reporter Reveals Redox Regulation under Physiological Normoxia. ACS Sens 2025; 10:3402-3411. [PMID: 40021628 DOI: 10.1021/acssensors.4c03167] [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] [Indexed: 03/03/2025]
Abstract
Reactive oxygen species, particularly hydrogen peroxide (H2O2), play crucial roles in cellular signaling, with Nrf2 serving as a key transcription factor in maintaining redox homeostasis. However, the precise influence of H2O2 on Nrf2 activity under physiological normoxia remains unclear due to the limitations of oxygen-sensitive imaging methods. To address this, we developed and validated an oxygen-insensitive Nrf2 reporter named pericellular oxygen-insensitive Nrf2 transcriptional performance reporter (POINTER). We employed this reporter in human cerebral microvascular endothelial cells (hCMEC/D3). Using POINTER, we investigated how varying intracellular H2O2 concentrations affect Nrf2 regulation under normoxia (5 kPa O2) compared to hyperoxia (ambient air, 21 kPa O2). We manipulated intracellular H2O2 levels through exogenous application, chemogenetic production using a modified amino acid oxidase, and pharmacological induction with Auranofin. Our findings reveal that Nrf2 transcriptional activity is significantly lower under normoxia than under hyperoxia, supporting previous literature and expectations. Using POINTER, we found that both antioxidant pathway inhibition and sustained H2O2 elevation are essential for modulating Nrf2 activity. These findings provide new insights into the regulation of Nrf2 by H2O2.
Collapse
Affiliation(s)
- Seyed Mohammad Miri
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Büşra N Ata
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Şeyma Çimen
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Department of Nutrition and Dietetics, Institution of Health Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Sarah Barakat
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Asal Ghaffari Zaki
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Joudi Armouch
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Emre Vatandaşlar
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Sven Vilain
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Department of Physiology, School of Medicine, Bolu Abant Izzet Baysal University, Bolu 14030, Turkey
| | - Emrah Eroğlu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
4
|
Chen D, Guo Z, Yao L, Sun Y, Dian Y, Zhao D, Ke Y, Zeng F, Zhang C, Deng G, Li L. Targeting oxidative stress-mediated regulated cell death as a vulnerability in cancer. Redox Biol 2025; 84:103686. [PMID: 40424719 DOI: 10.1016/j.redox.2025.103686] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2025] [Accepted: 05/17/2025] [Indexed: 05/29/2025] Open
Abstract
Reactive oxygen species (ROS), regulators of cellular behaviors ranging from signaling to cell death, have complex production and control mechanisms to maintain a dynamic redox balance under physiological conditions. Redox imbalance is frequently observed in tumor cells, where ROS within tolerable limits promote oncogenic transformation, while excessive ROS induce a range of regulated cell death (RCD). As such, targeting ROS-mediated regulated cell death as a vulnerability in cancer. However, the precise regulatory networks governing ROS-mediated cancer cell death and their therapeutic applications remain inadequately characterized. In this Review, we first provide a comprehensive overview of the mechanisms underlying ROS production and control within cells, highlighting their dynamic balance. Next, we discuss the paradoxical nature of the redox system in tumor cells, where ROS can promote tumor growth or suppress it, depending on the context. We also systematically explored the role of ROS in tumor signaling pathways and revealed the complex ROS-mediated cross-linking networks in cancer cells. Following this, we focus on the intricate regulation of ROS in RCD and its current applications in cancer therapy. We further summarize the potential of ROS-induced RCD-based therapies, particularly those mediated by drugs targeting specific redox balance mechanisms. Finally, we address the measurement of ROS and oxidative damage in research, discussing existing challenges and future prospects of targeting ROS-mediated RCD in cancer therapy. We hope this review will offer promise for the clinical application of targeting oxidative stress-mediated regulated cell death in cancer therapy.
Collapse
Affiliation(s)
- Danyao Chen
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China; Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Ziyu Guo
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Lei Yao
- Department of Liver Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yuming Sun
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, 87 Xiangya Road, Changsha, 410008, Hunan Province, China
| | - Yating Dian
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China
| | - Deze Zhao
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Yizhe Ke
- The First Affliated Hospital of Shihezi University, China
| | - Furong Zeng
- Department of Oncology, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Chunfang Zhang
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| | - Guangtong Deng
- Department of Dermatology, Xiangya Hospital, Central South University, Changsha, China; National Engineering Research Center of Personalized Diagnostic and Therapeutic Technology, China; Furong Laboratory, Changsha, Hunan, China; Hunan Key Laboratory of Skin Cancer and Psoriasis, Hunan Engineering Research Center of Skin Health and Disease, Xiangya Hospital, Central South University, Changsha, China; National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, China.
| | - Linfeng Li
- Department of Thoracic Surgery, Xiangya Hospital, Central South University, Changsha, Hunan, China.
| |
Collapse
|
5
|
Kotova DA, Ivanova AD, Kelmanson IV, Morozova KI, Khramova YV, Solotenkov MA, Stepanov EA, Moshchenko AA, Tiaglik AB, Fedotova AA, Zalygin AV, Oleinikov VA, Katrukha AG, Semyanov A, Belousov VV, Fedotov AB, Fedotov IV, Brazhe NA, Bilan DS. Redox Differences Between Neurons and Astrocytes In Vivo in Ischemic Brain Tissues of Rodents. Antioxid Redox Signal 2025. [PMID: 40387599 DOI: 10.1089/ars.2024.0876] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 05/20/2025]
Abstract
Aims: Reactive oxygen species (ROS) are considered to play a key damaging role in brain during the development of ischemic stroke. To clarify how different ROS contribute to ischemic pathogenesis, innovative approaches for real-time in vivo detection of redox parameters are necessary. Results: Using highly sensitive genetically encoded biosensor HyPer7 and a fiber-optic neurointerface technology, we demonstrated that the level of hydrogen peroxide (H2O2) slowly increases in neurons and astrocytes of the ischemic area of the rat brain after middle cerebral artery occlusion during next 40 h; notably, in astrocytes the level is somewhat higher. Raman microspectroscopy in awake mice also revealed redox differences between mitochondria of neurons and astrocytes during acute ischemia caused by photothrombosis. Astrocytes demonstrated the overloading of the electron transport chain (ETC) with electrons after 1 h of ischemia, whereas neurons do not demonstrate changes in the amount of reduced electron carries. Innovation and Conclusion: The combination of novel in vivo approaches allows to detail redox events with spatiotemporal resolution. We demonstrated redox difference between neurons and astrocytes in damaged brain areas in vivo. An elevated loading of astrocytic ETC with electrons during the acute ischemia phase provides basis for the increased generation of superoxide anion radical (O2•-) with its following conversion to other reactive species. However, we observed increased H2O2 concentrations in astrocytes and neurons at later pathogenesis stages. During this period, ETC did not demonstrate an elevated loading with electrons, and therefore, increased H2O2 generation may be a phenomenon of secondary redox events. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Aleksandra D Ivanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Kseniia I Morozova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Yulia V Khramova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Maxim A Solotenkov
- Physics Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- Life Improvement by Future Technologies Center, Moscow, Russia
| | - Evgeny A Stepanov
- Physics Department, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Aleksandr A Moshchenko
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Alisa B Tiaglik
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anna A Fedotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Anton V Zalygin
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vladimir A Oleinikov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexey G Katrukha
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Alexey Semyanov
- College of Medicine, Jiaxing University, Jiaxing, China
- Sechenov First Moscow State Medical University, Moscow, Russia
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Life Improvement by Future Technologies Center, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Andrei B Fedotov
- Physics Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- Life Improvement by Future Technologies Center, Moscow, Russia
| | - Ilya V Fedotov
- Physics Department, M.V. Lomonosov Moscow State University, Moscow, Russia
- Life Improvement by Future Technologies Center, Moscow, Russia
| | - Nadezda A Brazhe
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
6
|
Becker A, Chen X, Dresselhaus T, Gutsche N, Müller-Schüssele SJ, Sprunck S, Theißen G, de Vries S, Zachgo S. Sexual reproduction in land plants: an evolutionary perspective. PLANT REPRODUCTION 2025; 38:12. [PMID: 40355640 PMCID: PMC12069490 DOI: 10.1007/s00497-025-00522-4] [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] [Subscribe] [Scholar Register] [Received: 11/01/2024] [Accepted: 03/23/2025] [Indexed: 05/14/2025]
Abstract
KEY MESSAGE We link key aspects of land plant reproductive evolution and detail how successive molecular changes leading to novel tissues and organs require co-evolution of communication systems between tissues. The transition of water-dependent reproduction of algae to mechanisms with very limited water dependence in many land plant lineages allowed plants to colonize diverse terrestrial environments, leading to the vast variety of extant plant species. The emergence of modified cell types, novel tissues, and organs enabled this transition; their origin is associated with the co-evolution of novel or adapted molecular communication systems and gene regulatory networks. In the light of an increasing number of genome sequences in combination with the establishment of novel genetic model organisms from diverse green plant lineages, our knowledge and understanding about the origin and evolution of individual traits that arose in a concerted way increases steadily. For example, novel members of gene families in signaling pathways emerged for communication between gametes and gametophytes with additional tissues surrounding the gametes. Here, we provide a comprehensive overview on the origin and evolution of reproductive novelties such as pollen grains, immobile sperms, ovules and seeds, carpels, gamete/gametophytic communication systems, double fertilization, and the molecular mechanisms that have arisen anew or have been co-opted during evolution, including but not limited to the incorporation of phytohormones, reactive oxygen species and redox signaling as well as small RNAs in regulatory modules that contributed to the evolution of land plant sexual reproduction.
Collapse
Affiliation(s)
- Annette Becker
- Institute of Botany, Justus Liebig University, Heinrich-Buff-Ring 38, 35392, Giessen, Germany.
| | - Xia Chen
- Institute of Plant Sciences, Cell Biology and Plant Biochemistry, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Thomas Dresselhaus
- Institute of Plant Sciences, Cell Biology and Plant Biochemistry, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Nora Gutsche
- Division of Botany, Osnabrück University, Barbarastr. 11, 49076, Osnabrück, Germany
| | | | - Stefanie Sprunck
- Institute of Plant Sciences, Cell Biology and Plant Biochemistry, Universitätsstraße 31, 93053, Regensburg, Germany
| | - Günter Theißen
- Matthias Schleiden Institute/Genetics I, Friedrich Schiller University Jena, Philosophenweg 12, 07743, Jena, Germany
| | - Sophie de Vries
- Department of Applied Bioinformatics, Institute of Microbiology and Genetics, University of Göttingen, Goldschmidtstraße 1, 37077, Göttingen, Germany
| | - Sabine Zachgo
- Division of Botany, Osnabrück University, Barbarastr. 11, 49076, Osnabrück, Germany
| |
Collapse
|
7
|
Tian Y, Kang L, Ha NT, Deng J, Liu D. Hydrogen peroxide in midbrain sleep neurons regulates sleep homeostasis. Cell Metab 2025:S1550-4131(25)00254-2. [PMID: 40378838 DOI: 10.1016/j.cmet.2025.04.016] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/15/2024] [Revised: 01/13/2025] [Accepted: 04/22/2025] [Indexed: 05/19/2025]
Abstract
Sleep could protect animals from oxidative damage, yet the dynamic interplay between the redox state and sleep homeostasis remains unclear. Here, we show that acute sleep deprivation (SD) in mice caused a general increase in brain oxidation, particularly in sleep-promoting regions. In vivo imaging of intracellular hydrogen peroxide (H2O2) real-time dynamics revealed that in nigra sleep neurons, the increase in cytosolic but not mitochondrial H2O2 reflects sleep debt and tracks spontaneous wakefulness by positively correlating with wake duration. By controllably manipulating intraneuronal H2O2, we discovered that H2O2 elevation is required for compensatory sleep and causally promotes sleep initiation, at least partly dependent on transient receptor potential melastatin 2 (TRPM2) channel. However, excessive H2O2 induced brain inflammation and sleep fragmentation. Together, our study demonstrates intraneuronal H2O2 as a crucial signaling molecule that translates brain redox imbalance into sleep drive and underscores the significance of oxidative eustress in sleep homeostasis.
Collapse
Affiliation(s)
- Yujing Tian
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China; University of Chinese Academy of Sciences, Beijing 100049, China
| | - Luwei Kang
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China
| | - Ngoc T Ha
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Juan Deng
- Department of Anesthesiology, Huashan Hospital, State Key Laboratory of Medical Neurobiology, Institute for Translational Brain Research, MOE Frontiers Center for Brain Science, Fudan University, Shanghai 200032, China
| | - Danqian Liu
- Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai 200031, China.
| |
Collapse
|
8
|
Aleixo-Silva RL, Domingos RM, Trujillo M, Gomes F, Machado LO, Oliveira CLP, Baldini R, Netto LES. Interaction between 1-Cys peroxiredoxin and ascorbate in the response to H 2O 2 exposure in Pseudomonas aeruginosa. Redox Biol 2025; 84:103658. [PMID: 40367862 DOI: 10.1016/j.redox.2025.103658] [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: 03/19/2025] [Revised: 04/28/2025] [Accepted: 05/01/2025] [Indexed: 05/16/2025] Open
Abstract
Pseudomonas aeruginosa, a leading cause of hospital-acquired infections, triggers host defenses, including oxidant release by phagocytes. Targeting bacterial antioxidants could reduce pathogen infectivity. This study investigates LsfA, a 1-Cys peroxiredoxin (Prx), member of the Prx6 subfamily, involved in P. aeruginosa virulence. LsfA efficiently reduced various peroxides (106-107 M-1s-1), while exhibiting hyperoxidation resistance (khyperoxidation ∼102 M-1s-1). Despite its substrate oxidizing promiscuity, LsfA displayed specific reduction by ascorbate (2.2 × 103 M-1s-1). Moreover, elucidating the LsfA's crystallographic structures in the reduced and sulfinic/sulfonic acid states at 2.4 and 2.0 Å resolutions unveiled possible residues related to ascorbate binding. Small-angle X-ray scattering (SAXS) and size-exclusion chromatography (SEC) confirmed LsfA as a dimer regardless of its oxidative state. Microbiological assays, including a real-time analysis employing Hyper7, a genetically encoded probe, showed that ascorbate enhanced H2O2 removal in a LsfA-dependent manner. Hence, our integrated structural, biochemical, and microbiological analyses underscored the significance of the ascorbate-LsfA pathway in P. aeruginosa response to H2O2.
Collapse
Affiliation(s)
- Rogerio L Aleixo-Silva
- From the Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Renato M Domingos
- From the Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | - Madia Trujillo
- Departamento de Bioquímica, Facultad de Medicina y Centro de Investigaciones Biomédicas, Universidad de la República, Uruguay
| | - Fernando Gomes
- From the Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil
| | | | | | - Regina Baldini
- Departamento de Bioquímica, Instituto de Química, Universidade de São Paulo, Brazil
| | - Luis E S Netto
- From the Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, Brazil.
| |
Collapse
|
9
|
Kaesler N, Kaushik S, Frisch J, Ziegler S, Grommes J, Gombert A, Roma LP, Kuppe C, Jankowski J, Floege J, de la Puente‐Secades S, Kramann R, Jankowski V. Vitamin K preserves gamma-glutamyl carboxylase activity against carbamylations in uremia: Implications for vascular calcification and adjunct therapies. Acta Physiol (Oxf) 2025; 241:e70040. [PMID: 40202064 PMCID: PMC11979876 DOI: 10.1111/apha.70040] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2024] [Revised: 02/28/2025] [Accepted: 03/20/2025] [Indexed: 04/10/2025]
Abstract
AIM Vascular calcification contributes to morbidity and mortality in aging and is accelerated in diabetes and in chronic kidney disease. Matrix Gla Protein is a potent inhibitor of vascular calcification, which is activated by the vitamin K-dependent gamma-glutamyl carboxylase (GGCX). However, through a currently unidentified mechanism, the activity of GGCX is reduced in experimental uremia, thereby contributing to the promotion of vascular calcifications. In this study, we aim to identify the cause of these functional alterations and to stimulate the enzyme activity by potential GGCX binding compounds as a new avenue of therapy. METHODS Two rodent models of experimental uremia and human carotid plaques were assessed for GGCX activity and modifications, as well as calcification. In silico compound screening via BindScope identified potential binding partners of GGCX which were further validated in functional assays for enzymatic activity changes and for in vitro calcification. Mass spectrometry was applied to monitor molecular mass changes of the GGCX. RESULTS Mass spectrometry analysis revealed post-translational modifications of the GGCX in uremic rats and mice, as well as in calcified human carotid plaques. Functional assays showed that the post-translational carbamylation of GGCX reduced the enzyme activity, which was prevented by vitamin K2. Chrysin, identified by compound screening, stimulated GGCX activity, reduced calcium deposition in VSMCs, and oxidized GGCX at lysine 517. CONCLUSION In conclusion, this study clearly demonstrates that the vitamin K-dependent enzyme GGCX plays a significant role in uremic calcification and may be modulated to help prevent pathological changes.
Collapse
Affiliation(s)
- Nadine Kaesler
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
| | - Suresh Kaushik
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
- BiosciencesCardiff UniversityCardiffUK
| | - Janina Frisch
- Institute of Biophysics, Center of Human and Molecular Biology (ZHMB), Center for Gender‐Specific Biology and Medicine (CGBM)Saarland UniversityHomburgGermany
| | - Susanne Ziegler
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
| | - Jochen Grommes
- Marienhospital AachenClinic for Vascular SurgeryAachenGermany
| | - Alexander Gombert
- Clinic for Vascular SurgeryUniversity Hospital of the RWTH AachenAachenGermany
| | - Leticia Prates Roma
- Institute of Biophysics, Center of Human and Molecular Biology (ZHMB), Center for Gender‐Specific Biology and Medicine (CGBM)Saarland UniversityHomburgGermany
| | - Christoph Kuppe
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
| | - Joachim Jankowski
- Institute of Molecular Cardiovascular Research (IMCAR)University Hospital of the RWTH AachenAachenGermany
- Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM)University of MaastrichtMaastrichtThe Netherlands
- Aachen‐Maastricht Institute for CardioRenal Disease (AMICARE)University Hospital RWTH AachenAachenGermany
| | - Jürgen Floege
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
| | - Sofia de la Puente‐Secades
- Institute of Molecular Cardiovascular Research (IMCAR)University Hospital of the RWTH AachenAachenGermany
| | - Rafael Kramann
- Medical Clinic IIUniversity Hospital of the RWTH AachenAachenGermany
| | - Vera Jankowski
- Institute of Molecular Cardiovascular Research (IMCAR)University Hospital of the RWTH AachenAachenGermany
| |
Collapse
|
10
|
Shannon N, Raymond C, Palmer C, Homa S, Bonini M, Seward D, Cunniff B. Miro1 expression alters global gene expression, ERK1/2 phosphorylation, oxidation and cell cycle progression. J Cell Sci 2025; 138:jcs263554. [PMID: 40067243 PMCID: PMC11993262 DOI: 10.1242/jcs.263554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2024] [Accepted: 02/26/2025] [Indexed: 03/19/2025] Open
Abstract
Mitochondrial positioning supports localized energy and signaling requirements. Miro1 is necessary for attachment of mitochondria to microtubule motor proteins for trafficking. When Miro1 is deleted (Miro1-/-) from mouse embryonic fibroblasts (MEFs), mitochondria become sequestered to the perinuclear space, disrupting subcellular signaling gradients. Here, we show that Miro1-/- MEFs grow slower than Miro1+/+ and Miro1-/- MEFs stably re-expressing a Myc-Miro1 plasmid. Miro1-/- MEFs have a decreased percentage of cells in G1 and increased percentage of cells in S phase. We conducted the first ever RNA sequencing experiment dependent upon Miro1 expression and found differentially expressed genes related to MAPK signaling, cell proliferation and migration. ERK1 and ERK2 (ERK1/2, also known as MAPK3 and MAPK1, respectively) phosphorylation is elevated both spatially and temporally following serum stimulation in Miro1-/- MEFs, whereas the expression levels and oxidation of the dual specificity phosphatases (DUSP1-DUSP6) is unchanged. Finally, we found the oxidation status of ERK1/2 is increased in Miro1-/- MEFs compared to that seen in Miro1+/+ and Myc-Miro1 MEFs. These results highlight transcriptional control based off Miro1 expression and demonstrate the dynamic regulation of ERK1/2 upon deletion of Miro1 which might support the observed cell cycle and proliferation defects.
Collapse
Affiliation(s)
- Nathaniel Shannon
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Cory Raymond
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Chloe Palmer
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Silver Homa
- Department of Medicine and Biochemistry, Feinberg School of Medicine Northwestern University, Chicago, IL 60611, USA
| | - Marcelo Bonini
- Department of Medicine and Biochemistry, Feinberg School of Medicine Northwestern University, Chicago, IL 60611, USA
| | - David Seward
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| |
Collapse
|
11
|
Henríquez-Olguín C, Gallero S, Reddy A, Persson KW, Schlabs FL, Voldstedlund CT, Valentinaviciute G, Meneses-Valdés R, Sigvardsen CM, Kiens B, Chouchani ET, Richter EA, Jensen TE. Revisiting insulin-stimulated hydrogen peroxide dynamics reveals a cytosolic reductive shift in skeletal muscle. Redox Biol 2025; 82:103607. [PMID: 40194326 PMCID: PMC12001130 DOI: 10.1016/j.redox.2025.103607] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2025] [Revised: 03/06/2025] [Accepted: 03/20/2025] [Indexed: 04/09/2025] Open
Abstract
The intracellular redox state is crucial for insulin responses in peripheral tissues. Despite the longstanding belief that insulin signaling increases hydrogen peroxide (H2O2) production leading to reversible oxidation of cysteine thiols, evidence is inconsistent and rarely involves human tissues. In this study, we systematically investigated insulin-dependent changes in subcellular H2O2 levels and reversible cysteine modifications across mouse and human skeletal muscle models. Utilizing advanced redox tools-including genetically encoded H2O2 sensors and non-reducing immunoblotting-we consistently observed no increase in subcellular H2O2 levels following insulin stimulation. Instead, stoichiometric cysteine proteome analyses revealed a selective pro-reductive shift in cysteine modifications affecting insulin transduction related proteins, including Cys179 on GSK3β and Cys416 on Ras and Rab Interactor 2 (RIN2). Our findings challenge the prevailing notion that insulin promotes H2O2 generation in skeletal muscle and suggest that an insulin-stimulated pro-reductive shift modulates certain aspects of insulin signal transduction.
Collapse
Affiliation(s)
- Carlos Henríquez-Olguín
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark; Exercise Science Laboratory, Faculty of Medicine, Universidad Finis Terrae, Av. Pedro de Valdivia 1509, Santiago, Chile.
| | - Samantha Gallero
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Anita Reddy
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Kaspar W Persson
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Farina L Schlabs
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Christian T Voldstedlund
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Gintare Valentinaviciute
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Roberto Meneses-Valdés
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Casper M Sigvardsen
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Bente Kiens
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Edward T Chouchani
- Department of Cell Biology, Harvard Medical School, Boston, MA, USA; Department of Cancer Biology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Erik A Richter
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark
| | - Thomas E Jensen
- August Krogh Section for Human and Molecular Physiology, Department of Nutrition, Exercise and Sports, Faculty of Science, University of Copenhagen, Denmark.
| |
Collapse
|
12
|
Zhang J, Ali MY, Chong HB, Tien PC, Woods J, Noble C, Vornbäumen T, Ordulu Z, Possemato AP, Harry S, Fonticella JM, Fellah L, Harrison D, Ge M, Khandelwal N, Huang Y, Chauvin M, Bischof AT, Hambelton GM, Gohar MF, Zhang S, Choi M, Bouberhan S, Oliva E, Mino-Kenudson M, Pavlova NN, Lawrence M, Gainor JF, Beausoleil SA, Bardeesy N, Mostoslavsky R, Pépin D, Ott CJ, Liau B, Bar-Peled L. Oxidation of retromer complex controls mitochondrial translation. Nature 2025; 641:1048-1058. [PMID: 40140582 DOI: 10.1038/s41586-025-08756-y] [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: 06/04/2024] [Accepted: 02/07/2025] [Indexed: 03/28/2025]
Abstract
Reactive oxygen species (ROS) underlie human pathologies including cancer and neurodegeneration1,2. However, the proteins that sense ROS levels and regulate their production through their cysteine residues remain ill defined. Here, using systematic base-editing and computational screens, we identify cysteines in VPS35, a member of the retromer trafficking complex3, that phenocopy inhibition of mitochondrial translation when mutated. We find that VPS35 underlies a reactive metabolite-sensing pathway that lowers mitochondrial translation to decrease ROS levels. Intracellular hydrogen peroxide oxidizes cysteine residues in VPS35, resulting in retromer dissociation from endosomal membranes and subsequent plasma membrane remodelling. We demonstrate that plasma membrane localization of the retromer substrate SLC7A1 is required to sustain mitochondrial translation. Furthermore, decreasing VPS35 levels or oxidation of its ROS-sensing cysteines confers resistance to ROS-generating chemotherapies, including cisplatin, in ovarian cancer models. Thus, we identify that intracellular ROS levels are communicated to the plasma membrane through VPS35 to regulate mitochondrial translation, connecting cytosolic ROS sensing to mitochondrial ROS production.
Collapse
Affiliation(s)
- Junbing Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- CAS Key Laboratory of Nutrition, Metabolism and Food Safety, Shanghai Institute of Nutrition and Health, University of Chinese Academy of Sciences, Chinese Academy of Sciences, Shanghai, China.
| | - Md Yousuf Ali
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Harrison Byron Chong
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Pei-Chieh Tien
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - James Woods
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Carolina Noble
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Tristan Vornbäumen
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Zehra Ordulu
- Brigham and Women's Hospital, Department of Pathology, Harvard Medical School, MA, USA
| | | | - Stefan Harry
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Jay Miguel Fonticella
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Lina Fellah
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Drew Harrison
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maolin Ge
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Neha Khandelwal
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Yingfei Huang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Maëva Chauvin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Anica Tamara Bischof
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | | | - Magdy Farag Gohar
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - Siwen Zhang
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
| | - MinGyu Choi
- Department of Chemistry, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Sara Bouberhan
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Division of Hematology/Oncology, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Esther Oliva
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Mari Mino-Kenudson
- Department of Pathology, Massachusetts General Hospital, Boston, MA, USA
| | - Natalya N Pavlova
- Department of Oncological Sciences, Huntsman Cancer Institute, University of Utah, Salt Lake City, UT, USA
| | - Michael Lawrence
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Justin F Gainor
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | | | - Nabeel Bardeesy
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Raul Mostoslavsky
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - David Pépin
- Pediatric Surgical Research Laboratories, Massachusetts General Hospital, Boston, MA, USA
| | - Christopher J Ott
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Medicine, Harvard Medical School, Boston, MA, USA
| | - Brian Liau
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Liron Bar-Peled
- Krantz Family Center for Cancer Research, Massachusetts General Hospital Cancer Center, Charlestown, MA, USA.
- Department of Medicine, Massachusetts General Hospital, Boston, MA, USA.
- Department of Medicine, Harvard Medical School, Boston, MA, USA.
| |
Collapse
|
13
|
Lee JD, Nguyen A, Gibbs CE, Jin ZR, Wang Y, Moghadasi A, Wait SJ, Choi H, Evitts KM, Asencio A, Bremner SB, Zuniga S, Chavan V, Pranoto IKA, Williams CA, Smith A, Moussavi-Harami F, Regnier M, Baker D, Young JE, Mack DL, Nance E, Boyle PM, Berndt A. Monitoring in real time and far-red imaging of H 2O 2 dynamics with subcellular resolution. Nat Chem Biol 2025:10.1038/s41589-025-01891-7. [PMID: 40295764 DOI: 10.1038/s41589-025-01891-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2024] [Accepted: 03/25/2025] [Indexed: 04/30/2025]
Abstract
Monitoring H2O2 dynamics in conjunction with key biological interactants is critical for elucidating the physiological outcome of cellular redox regulation. Optogenetic hydrogen peroxide sensor with HaloTag with JF635 (oROS-HT635) allows fast and sensitive chemigenetic far-red H2O2 imaging while overcoming drawbacks of existing red fluorescent H2O2 indicators, including oxygen dependency, high pH sensitivity, photoartifacts and intracellular aggregation. The compatibility of oROS-HT635 with blue-green-shifted optical tools allows versatile optogenetic dissection of redox biology. In addition, targeted expression of oROS-HT635 and multiplexed H2O2 imaging enables spatially resolved imaging of H2O2 targeting the plasma membrane and neighboring cells. Here we present multiplexed use cases of oROS-HT635 with other green fluorescence reporters by capturing acute and real-time changes in H2O2 with intracellular redox potential and Ca2+ levels in response to auranofin, an inhibitor of antioxidative enzymes, via dual-color imaging. oROS-HT635 enables detailed insights into intricate intracellular and intercellular H2O2 dynamics, along with their interactants, through spatially resolved, far-red H2O2 imaging in real time.
Collapse
Affiliation(s)
- Justin Daho Lee
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Amanda Nguyen
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Chelsea E Gibbs
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Zheyu Ruby Jin
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Yuxuan Wang
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Aida Moghadasi
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Sarah J Wait
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Hojun Choi
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
| | - Kira M Evitts
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Anthony Asencio
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Samantha B Bremner
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Shani Zuniga
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Vedant Chavan
- Department of Bioengineering, University of Washington, Seattle, WA, USA
| | - Inez K A Pranoto
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - C Andrew Williams
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - Annette Smith
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
| | - Farid Moussavi-Harami
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
- Division of Cardiology, University of Washington, Seattle, WA, USA
| | - Michael Regnier
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - David Baker
- Department of Biochemistry, University of Washington, Seattle, WA, USA
- Institute for Protein Design, University of Washington, Seattle, WA, USA
- Howard Hughes Medical Institute, University of Washington, Seattle, WA, USA
| | - Jessica E Young
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Laboratory Medicine and Pathology, University of Washington, Seattle, WA, USA
| | - David L Mack
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Department of Rehabilitation Medicine, University of Washington, Seattle, WA, USA
| | - Elizabeth Nance
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Department of Chemical Engineering, University of Washington, Seattle, WA, USA
| | - Patrick M Boyle
- Department of Bioengineering, University of Washington, Seattle, WA, USA
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA
- Center for Cardiovascular Biology, University of Washington, Seattle, WA, USA
| | - Andre Berndt
- Molecular Engineering and Sciences Institute, University of Washington, Seattle, WA, USA.
- Department of Bioengineering, University of Washington, Seattle, WA, USA.
- Institute for Stem Cell and Regenerative Medicine, University of Washington, Seattle, WA, USA.
- Center for Translational Muscle Research, University of Washington, Seattle, WA, USA.
| |
Collapse
|
14
|
Paulenda T, Echalar B, Potuckova L, Vachova V, Kleverov DA, Mehringer J, Potekhina E, Jacoby A, Sen D, Nelson C, Stegeman R, Sukhov V, Kemper D, Lichti CF, Day NJ, Zhang T, Husarcikova K, Bambouskova M, Fremont DH, Qian WJ, Djuranovic S, Pavlovic-Djuranovic S, Belousov VV, Krezel AM, Artyomov MN. Itaconate modulates immune responses via inhibition of peroxiredoxin 5. Nat Metab 2025:10.1038/s42255-025-01275-0. [PMID: 40251412 DOI: 10.1038/s42255-025-01275-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/29/2024] [Accepted: 03/12/2025] [Indexed: 04/20/2025]
Abstract
The immunoregulatory metabolite itaconate accumulates in innate immune cells upon Toll-like receptor stimulation. In response to macrophage activation by lipopolysaccharide, itaconate inhibits inflammasome activation and boosts type I interferon signalling; however, the molecular mechanism of this immunoregulation remains unclear. Here, we show that the enhancement of type I interferon secretion by itaconate depends on the inhibition of peroxiredoxin 5 and on mitochondrial reactive oxygen species. We find that itaconate non-covalently inhibits peroxiredoxin 5, leading to the modulation of mitochondrial peroxide in activating macrophages. Through genetic manipulation, we confirm that peroxiredoxin 5 modulates type I interferon secretion in macrophages. The non-electrophilic itaconate mimetic 2-methylsuccinate inhibits peroxiredoxin 5 and phenocopies immunoregulatory action of itaconate on type I interferon and inflammasome activation, providing further support for a non-covalent inhibition of peroxiredoxin 5 by itaconate. Our work provides insight into the molecular mechanism of actions and biological rationale for the predominantly immune specification of itaconate.
Collapse
Affiliation(s)
- Tomas Paulenda
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Barbora Echalar
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Lucie Potuckova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Veronika Vachova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Denis A Kleverov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Johannes Mehringer
- Bruker Biosensors, Munich, Germany
- Kurt Schwabe Institute for Sensor Technologies, Waldheim, Germany
| | - Ekaterina Potekhina
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Alex Jacoby
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Devashish Sen
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Chris Nelson
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Rick Stegeman
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Vladimir Sukhov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Danielle Kemper
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Cheryl F Lichti
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Bursky Center for Human Immunology and Immunotherapy Programs, St. Louis, MO, USA
| | - Nicholas J Day
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Tong Zhang
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Kamila Husarcikova
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
| | - Monika Bambouskova
- Department of Medicine, Washington University School of Medicine, St. Louis, MO, USA
| | - Daved H Fremont
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
- Department of Molecular Microbiology, Washington University School of Medicine, St. Louis, MO, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Sergej Djuranovic
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO, USA
| | | | - Vsevolod V Belousov
- Pirogov Russian National Research Medical University, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Andrzej M Krezel
- Department of Biochemistry and Molecular Biophysics, Washington University School of Medicine, St. Louis, MO, USA
| | - Maxim N Artyomov
- Department of Pathology and Immunology, Washington University School of Medicine, St. Louis, MO, USA.
| |
Collapse
|
15
|
Yu K, Yang S, Song H, Sun Z, Wang K, Zhu Y, Yang C, Hao R, Cao Y. High-Resolution Tracking of Aging-Related Small Molecules: Bridging Pollutant Exposure, Brain Aging Mechanisms, and Detection Innovations. BIOSENSORS 2025; 15:242. [PMID: 40277555 PMCID: PMC12024821 DOI: 10.3390/bios15040242] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/09/2025] [Revised: 03/23/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025]
Abstract
Brain aging is a complex process regulated by genetic, environmental, and metabolic factors, and increasing evidence suggests that environmental pollutants can significantly accelerate this process by interfering with oxidative stress, neuroinflammation, and mitochondrial function-related signaling pathways. Traditional studies have focused on the direct damage of pollutants on macromolecules (e.g., proteins, DNA), while the central role of senescence-associated small molecules (e.g., ROS, PGE2, lactate) in early regulatory mechanisms has been long neglected. In this study, we innovatively proposed a cascade framework of "small molecule metabolic imbalance-signaling pathway dysregulation-macromolecule collapse", which reveals that pollutants exacerbate the dynamics of brain aging through activation of NLRP3 inflammatory vesicles and inhibition of HIF-1α. Meanwhile, to address the technical bottleneck of small molecule spatiotemporal dynamics monitoring, this paper systematically reviews the cutting-edge detection tools such as electrochemical sensors, genetically encoded fluorescent probes and antioxidant quantum dots (AQDs). Among them, AQDs show unique advantages in real-time monitoring of ROS fluctuations and intervention of oxidative damage by virtue of their ultra-high specific surface area, controllable surface modification, and free radical scavenging ability. By integrating multimodal detection techniques and mechanism studies, this work provides a new perspective for analyzing pollutant-induced brain aging and lays a methodological foundation for early intervention strategies based on small molecule metabolic networks.
Collapse
Affiliation(s)
- Keying Yu
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Sirui Yang
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Hongxu Song
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Zhou Sun
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Kaichao Wang
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuqi Zhu
- School of Basic Medical Sciences, Capital Medical University, Beijing 100069, China; (S.Y.); (H.S.); (Z.S.); (Y.Z.)
| | - Chengkai Yang
- Beijing Friendship Hospital, Capital Medical University, Beijing 100069, China;
| | - Rongzhang Hao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| | - Yuanyuan Cao
- Department of Toxicology and Sanitary Chemistry, School of Public Health, Capital Medical University, Beijing 100069, China; (K.Y.); (K.W.)
- Beijing Key Laboratory of Environment and Aging, Capital Medical University, Beijing 100069, China
| |
Collapse
|
16
|
Simonyan TR, Protasova EA, Mamontova AV, Shakhov AM, Bodunova DV, Sidorenko SV, Maksimov EG, Bogdanov AM. Fluorescent protein with environmentally-sensitive fluorescence lifetime for quantitative pH measurement. Arch Biochem Biophys 2025; 766:110350. [PMID: 39971109 DOI: 10.1016/j.abb.2025.110350] [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: 09/09/2024] [Revised: 01/16/2025] [Accepted: 02/17/2025] [Indexed: 02/21/2025]
Abstract
Intracellular pH is a key factor in cell homeostasis, regulated within specific compartments, and changes in pH can result from or affect biochemical pathways. This study explores a yellow fluorescent protein EYFP-G65T as a core for a time-resolved pH-indicator. Among the tested designs-a circular permutant, a chimeric SypHer3s-like construct, and an unmodified protein-the unmodified EYFP-G65T performed best for live-cell imaging. Upon two-photon excitation, purified EYFP-G65T exhibited a 4.5-fold increase in mean fluorescence lifetime across pH 5.5-7 and a 7-fold change in its major component's lifetime from pH 6.5-8. Using this indicator, we measured pH values ranging from 6 to 8 in various organelles, and mapped pH shifts in mitochondria and the Golgi apparatus in response to stimuli.
Collapse
Affiliation(s)
- Tatiana R Simonyan
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia
| | - Elena A Protasova
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992, Moscow, Russia
| | | | - Aleksander M Shakhov
- Bio&Nanophotonics Lab, N.N. Semenov Federal Research Center for Chemical Physics, Moscow, 119991, Moscow, Russia
| | - Daria V Bodunova
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992, Moscow, Russia
| | - Svetlana V Sidorenko
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992, Moscow, Russia
| | - Eugene G Maksimov
- Faculty of Biology, M.V. Lomonosov Moscow State University, 119992, Moscow, Russia
| | - Alexey M Bogdanov
- Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Moscow, 117997, Russia; Department of Photonics, İzmir Institute of Technology, 35430, İzmir, Turkey.
| |
Collapse
|
17
|
Eaglesfield R, Fernandez-Vizarra E, Lacko E, Caldwell ST, Sloan NL, Siciarz D, Hartley RC, Tokatlidis K. Sub-organellar mitochondrial hydrogen peroxide observed using a SNAP tag targeted coumarin-based fluorescent reporter. Redox Biol 2025; 80:103502. [PMID: 39864323 PMCID: PMC11802384 DOI: 10.1016/j.redox.2025.103502] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2024] [Revised: 01/11/2025] [Accepted: 01/15/2025] [Indexed: 01/28/2025] Open
Abstract
Mitochondria are major sites of reactive oxygen species (ROS) production within cells. ROS are important signalling molecules, but excessive production can cause cellular damage and dysfunction. It is therefore crucial to accurately determine when, how and where ROS are produced within mitochondria. Previously, ROS detection involved various chemical probes and fluorescent proteins. These have limitations due to accumulation of the molecules only in the mitochondrial matrix, or the need for a new protein to be expressed for every different species. We report dynamic H2O2 flux changes within all mitochondrial sub-compartments with striking spatial resolution. We combined specific targeting of self-labeling proteins with novel H2O2-reactive probes. The approach is broad-ranging and flexible, with the same expressed proteins loadable with different dyes and sensors. It provides a framework for concomitant analysis of other chemical species, beyond ROS, whose dynamics within mitochondria are yet unknown, without needing to engineer new proteins.
Collapse
Affiliation(s)
- Ross Eaglesfield
- School of Molecular Biosciences, University of Glasgow, G12 8QQ, UK; National Renewable Energy Laboratory, Golden, CO, USA
| | - Erika Fernandez-Vizarra
- School of Molecular Biosciences, University of Glasgow, G12 8QQ, UK; Department of Biochemistry and Molecular and Cellular Biology, Faculty of Health and Sport Sciences, University of Zaragoza, 22002, Spain
| | - Erik Lacko
- School of Molecular Biosciences, University of Glasgow, G12 8QQ, UK
| | | | - Nikki L Sloan
- School of Chemistry, University of Glasgow, G12 8QQ, UK
| | - Daniel Siciarz
- School of Molecular Biosciences, University of Glasgow, G12 8QQ, UK
| | | | - Kostas Tokatlidis
- School of Molecular Biosciences, University of Glasgow, G12 8QQ, UK.
| |
Collapse
|
18
|
Liu J, Yao L, Zhao F, Zhang J, Gong J, Li J, Bian Y, Yin Y, Zhao R, Wang Y, Wang W. Dual-domain superoxide dismutase: In silico prediction directed combinatorial mutation for enhanced robustness and catalytic efficiency. Int J Biol Macromol 2025; 292:139179. [PMID: 39732240 DOI: 10.1016/j.ijbiomac.2024.139179] [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: 09/30/2024] [Revised: 12/18/2024] [Accepted: 12/23/2024] [Indexed: 12/30/2024]
Abstract
The robustness and catalytic activity of superoxide dismutase (SOD) are still the main factors limiting their application in industrial fields. This study aims to further improve the properties of a natural thermophilic iron/manganese dual-domain SOD (Fe/Mn-SODA fused with N-terminal polypeptide) from Geobacillus thermodenitrificans NG80-2 (GtSOD) by modifying its each domain using in-depth in silico prediction analysis as well as protein engineering. First, computational analysis of the N-terminal domain and GtSODA domain was respectively performed by using homologous sequence alignment and virtual mutagenesis. Seven proposed mutation sites favoring increased robustness were screened out for single-point mutants (SPMs) construction. Enzymatic characterization of these SPMs identified the most favorable mutation sites E107 and S265 located in two different domains. Subsequently, the dual-domain site combinatorial mutant (DDSCM) E107L/S265K showed significant superposition effects and additional improvement in catalytic efficiency, with a Kcat/Km value of 145.45 %, 33.66 %, and 60.33 % higher than the wild type (WT), the SPMs E107L and S265K, respectively. Molecular dynamics simulations, structural and surface charge analysis revealed the possible mechanism by which combinatorial mutations improve the robustness and catalytic activity of GtSOD. Furthermore, DDSCM showed more significant resistance to ultraviolet B and various stress than WT, indicating its highly competitive industrial application prospects.
Collapse
Affiliation(s)
- Jingjing Liu
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Lei Yao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Fang Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Jingjing Zhang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Jingbo Gong
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Jiabin Li
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Ya Bian
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Yalin Yin
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Rui Zhao
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China
| | - Yijia Wang
- Laboratory of Oncologic Molecular Medicine, Tianjin Union Medical Center, Nankai University, Tianjin 300121, PR China
| | - Wei Wang
- Key Laboratory of Molecular Microbiology and Technology, Ministry of Education, TEDA Institute of Biological Sciences and Biotechnology, Nankai University, TEDA, Tianjin 300457, PR China; Tianjin Key Laboratory of Microbial Functional Genomics, Tianjin 300457, PR China.
| |
Collapse
|
19
|
Stein CS, Linzer CR, Heer CD, Witmer NH, Cochran JD, Spitz DR, Boudreau RL. Mitoregulin Promotes Cell Cycle Progression in Non-Small Cell Lung Cancer Cells. Int J Mol Sci 2025; 26:1939. [PMID: 40076565 PMCID: PMC11899852 DOI: 10.3390/ijms26051939] [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: 12/23/2024] [Revised: 02/18/2025] [Accepted: 02/20/2025] [Indexed: 03/14/2025] Open
Abstract
Mitoregulin (MTLN) is a 56-amino-acid mitochondrial microprotein known to modulate mitochondrial energetics. MTLN gene expression is elevated broadly across most cancers and has been proposed as a prognostic biomarker for non-small cell lung cancer (NSCLC). In addition, lower MTLN expression in lung adenocarcinoma (LUAD) correlates with significantly improved patient survival. In our studies, we have found that MTLN silencing in A549 NSCLC cells slowed proliferation and, in accordance with this, we observed the following: (1) increased proportion of cells in the G1 phase of cell cycle; (2) protein changes consistent with G1 arrest (e.g., reduced levels and/or reduced phosphorylation of ERK, MYC, CDK2, and RB, and elevated p27Kip1); (3) reduction in clonogenic cell survival and; (4) lower steady-state cytosolic and mitochondrial H2O2 levels as indicated by use of the roGFP2-Orp1 redox sensor. Conflicting with G1 arrest, we observed a boost in cyclin D1 abundance. We also tested MTLN silencing in combination with buthionine sulfoximine (BSO) and auranofin (AF), drugs that inhibit GSH synthesis and thioredoxin reductase, respectively, to elevate the reactive oxygen species (ROS) amount to a toxic range. Interestingly, clonogenic survival after drug treatment was greater for MTLN-silenced cultures versus the control cultures. Lower H2O2 output and reduced vulnerability to ROS damage due to G1 status may have jointly contributed to the partial BSO + AF resistance. Overall, our results provide evidence that MTLN fosters H2O2 signaling to propel G1/S transition and suggest MTLN silencing as a therapeutic strategy to limit NSCLC growth.
Collapse
Affiliation(s)
- Colleen S. Stein
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Connor R. Linzer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Collin D. Heer
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Nathan H. Witmer
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Jesse D. Cochran
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| | - Douglas R. Spitz
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, Holden Comprehensive Cancer Center, University of Iowa, Iowa City, IA 52242, USA; (C.D.H.); (D.R.S.)
| | - Ryan L. Boudreau
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA; (C.R.L.); (N.H.W.); (J.D.C.)
| |
Collapse
|
20
|
Sergeeva AD, Panova AS, Ivanova AD, Khramova YV, Morozova KI, Kotova DA, Guryleva AV, Khokhlov DD, Kelmanson IV, Vasilev AV, Kostyuk AI, Semyanov AV, Oleinikov VA, Belousov VV, Machikhin AS, Brazhe NA, Bilan DS. Where in the Tissues of Danio rerio Is More H 2O 2 Produced During Acute Hypoxia? Antioxid Redox Signal 2025; 42:292-300. [PMID: 39086238 DOI: 10.1089/ars.2024.0563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The lack of oxygen (O2) causes changes in the cell functioning. Modeling hypoxic conditions in vitro is challenging given that different cell types exhibit different sensitivities to tissue O2 levels. We present an effective in vivo platform for assessing various tissue and organ parameters in Danio rerio larvae under acute hypoxic conditions. Our system allows simultaneous positioning of multiple individuals within a chamber where O2 level in the water can be precisely and promptly regulated, all while conducting microscopy. We applied this approach in combination with a genetically encoded pH-biosensor SypHer3s and a highly H2O2-sensitive HyPer7 biosensor. Hypoxia causes H2O2 production in areas of brain, heart, and skeletal muscles, exclusively in the mitochondrial matrix; it is noteworthy that H2O2 does not penetrate into the cytosol and is neutralized in the matrix upon reoxygenation. Hypoxia causes pronounced tissue acidosis, expressed by a decrease in pH by 0.4-0.6 units everywhere. Using imaging photoplethysmography, we measured in D. rerio larvae real-time heart rate decrease under conditions of hypoxia and subsequent reoxygenation. Our observations in this experimental system lead to the hypothesis that mitochondria are the only source of H2O2 in cells of D. rerio under hypoxia. Antioxid. Redox Signal. 42, 292-300.
Collapse
Affiliation(s)
- Anastasia D Sergeeva
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Anastasiya S Panova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra D Ivanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yulia V Khramova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ksenia I Morozova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia V Guryleva
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Demid D Khokhlov
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr V Vasilev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander I Kostyuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexey V Semyanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Vladimir A Oleinikov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexander S Machikhin
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Nadezda A Brazhe
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
21
|
Radhakrishnan K, Zhang Y, Mustapha O, Weigel TK, Upchurch CM, Tian X, Herbert F, Huang W, Leitinger N, Eyo UB, Ai H, Ferris HA. 7-ketocholesterol contributes to microglia-driven increases in astrocyte reactive oxygen species in Alzheimer's disease. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.19.633810. [PMID: 39868327 PMCID: PMC11761689 DOI: 10.1101/2025.01.19.633810] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Oxidative stress is a prominent feature of Alzheimer's disease. Within this context, cholesterol undergoes oxidation, producing the pro-inflammatory product 7-ketocholesterol (7-KC). In this study, we observe elevated levels of 7-KC in the brains of the 3xTg mouse model of AD. To further understand the contribution of 7-KC on the oxidative environment, we developed a method to express a genetically encoded fluorescent hydrogen peroxide (H2O2) sensor in astrocytes, the primary source of cholesterol in the brain. With this sensor, we discovered that 7-KC increases H2O2 levels in astrocytes in vivo, but not when directly applied to astrocytes in vitro. Interestingly, when 7-KC was applied to a microglia cell line alone or mixed astrocyte and microglia cultures, it resulted in microglia activation and increased oxidative stress in astrocytes. Depletion of microglia from 3xTg mice resulted in reduced 7-KC in the brains of these mice. Taken together, these findings suggest that 7-KC, acting through microglia, contributes to increased astrocyte oxidative stress in AD. This study sheds light on the complex interplay between cholesterol oxidation, microglia activation, and astrocyte oxidative stress in the pathogenesis of AD.
Collapse
Affiliation(s)
- Kayalvizhi Radhakrishnan
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| | - Yiyu Zhang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Oluwaseun Mustapha
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Thaddeus K. Weigel
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Clint M. Upchurch
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Xiaodong Tian
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Franklin Herbert
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Wenyuan Huang
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Norbert Leitinger
- Department of Pharmacology, University of Virginia, Charlottesville, VA, USA
| | - Ukpong B. Eyo
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
| | - Huiwang Ai
- Department of Molecular Physiology and Biological Physics, University of Virginia, Charlottesville, VA, USA
- Center for Membrane and Cell Physiology, University of Virginia, Charlottesville, VA, USA
| | - Heather A. Ferris
- Center for Brain Immunology and Glia, University of Virginia, Charlottesville, VA, USA
- Department of Neuroscience, University of Virginia, Charlottesville, VA, USA
- Division of Endocrinology and Metabolism, University of Virginia, Charlottesville, VA, USA
| |
Collapse
|
22
|
Shah V, Lam HY, Leong CHM, Sakaizawa R, Shah JS, Kumar AP. Epigenetic Control of Redox Pathways in Cancer Progression. Antioxid Redox Signal 2025. [PMID: 39815993 DOI: 10.1089/ars.2023.0465] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/18/2025]
Abstract
Significance: Growing evidence indicates the importance of redox reactions homeostasis, mediated predominantly by reactive oxygen species (ROS) in influencing the development, differentiation, progression, metastasis, programmed cell death, tumor microenvironment, and therapeutic resistance of cancer. Therefore, reviewing the ROS-linked epigenetic changes in cancer is fundamental to understanding the progression and prevention of cancer. Recent Advances: We review in depth the molecular mechanisms involved in ROS-mediated epigenetic changes that lead to alteration of gene expression by altering DNA, modifying histones, and remodeling chromatin and noncoding RNA. Critical Issues: In cancerous cells, alterations of the gene-expression regulatory elements could be generated by the virtue of imbalance in tumor microenvironment. Various oxidizing agents and mitochondrial electron transport chain are the major pathways that generate ROS. ROS plays a key role in carcinogenesis by activating pro-inflammatory signaling pathways and DNA damage. This loss of ROS-mediated epigenetic regulation of the signaling pathways may promote tumorigenesis. We address all such aspects in this review. Future Directions: Developments in this growing field of epigenetics are expected to contribute to further our understanding of human health and diseases such as cancer and to test the clinical applications of redox-based therapy. Recent studies of the cancer-epigenetic landscape have revealed pervasive deregulation of the epigenetic factors in cancer. Thus, the study of interaction between ROS and epigenetic factors in cancer holds a great promise in the development of effective and targeted treatment modalities. Antioxid. Redox Signal. 00, 000-000.
Collapse
Affiliation(s)
- Vandit Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Hiu Yan Lam
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Charlene Hoi-Mun Leong
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Reo Sakaizawa
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| | - Jigna S Shah
- Department of Pharmacology, Institute of Pharmacy, Nirma University, Ahmedabad, India
| | - Alan Prem Kumar
- Department of Pharmacology, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
- NUS Center for Cancer Research, Yong Loo Lin School of Medicine, National University of Singapore, Singapore, Singapore
| |
Collapse
|
23
|
Mena D, Arusei RJ, Rahhali K, Di Lisa F, Kaludercic N. Measurement of Mitochondrial ROS Formation. Methods Mol Biol 2025; 2878:99-116. [PMID: 39546259 DOI: 10.1007/978-1-0716-4264-1_6] [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] [Indexed: 11/17/2024]
Abstract
Reactive oxygen species (ROS) play important roles in physiological and pathological processes. Mitochondria, particularly in skeletal and cardiac myocytes, are recognized as the primary site of ROS generation. Detecting oxidative modifications of intracellular or circulating molecules, such as lipids, proteins, and nucleic acids, is commonly employed to indicate ROS formation. However, this approach is indirect and provides limited insights into the spatiotemporal aspects of ROS generation. Understanding these aspects is crucial for comprehending the role of ROS in various pathophysiological conditions. To address this, fluorescent probes can be employed to measure ROS formation, offering a means to investigate ROS generation in both isolated mitochondria and intact cells. This chapter outlines three prominent examples for the use of fluorescent sensors to evaluate mitochondrial ROS formation in either isolated organelles or intact cells. The methods are explained in detail, and an analysis of the limitations of each technique is provided, underscoring potential sources of errors during the assay and the subsequent interpretation of results.
Collapse
Affiliation(s)
- Débora Mena
- Department of Biomedical Sciences, University of Padova, Padova, Italy
- Center for Neuroscience and Cell Biology (CNC), University of Coimbra, Coimbra, Portugal
- Centre for Innovative Biomedicine and Biotechnology (CIBB), University of Coimbra, Coimbra, Portugal
- Institute for Interdisciplinary Research (IIIUC), Doctoral Programme in Experimental Biology and Biomedicine (PDBEB), University of Coimbra, Coimbra, Portugal
| | | | - Karim Rahhali
- Department of Biomedical Sciences, University of Padova, Padova, Italy
| | - Fabio Di Lisa
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Neuroscience Institute, National Research Council of Italy (CNR), Padova, Italy.
| | - Nina Kaludercic
- Department of Biomedical Sciences, University of Padova, Padova, Italy.
- Fondazione Istituto di Ricerca Pediatrica Città della Speranza (IRP), Padova, Italy.
| |
Collapse
|
24
|
Rogers ZJ, Flood D, Bencherif SA, Taylor CT. Oxygen control in cell culture - Your cells may not be experiencing what you think! Free Radic Biol Med 2025; 226:279-287. [PMID: 39577817 DOI: 10.1016/j.freeradbiomed.2024.11.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Oxygen (O2)-controlled cell culture has been pivotal in studying mammalian mechanisms of O2 sensing, regulation, and utilization. We posit, however, that O2-controlled cell culture is paradoxically not controlling O2. There is overwhelming evidence that the pericellular O2 is lower than the surrounding gas phase due to cellular O2 consumption. Standard hypoxic cell culture is at high risk of inducing pericellular anoxia. We discuss the implications of poor O2 control for cellular O2 regulation mechanisms, bioenergetics, and redox signaling. We also highlight the evidence of frequent under-oxygenation in standard (i.e., normoxic) cell culture. This issue has been largely overlooked because strategies to control pericellular O2 have been lacking. Here, we propose a framework to control pericellular O2 based on our recent investigation into the nature of the gas/pericellular O2 gradient. Implementing this framework into standard practice will unlock quantitative O2 control in vitro, improving our ability to understand the role of O2 in biology.
Collapse
Affiliation(s)
- Zachary J Rogers
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Darragh Flood
- Conway Institute of Biomolecular and Biomedical Research and School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Sidi A Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Cormac T Taylor
- Conway Institute of Biomolecular and Biomedical Research and School of Medicine, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
25
|
Zimmermann J, Lang L, Calabrese G, Laporte H, Amponsah PS, Michalk C, Sukmann T, Oestreicher J, Tursch A, Peker E, Owusu TNE, Weith M, Roma LP, Deponte M, Riemer J, Morgan B. Tsa1 is the dominant peroxide scavenger and a source of H 2O 2-dependent GSSG production in yeast. Free Radic Biol Med 2025; 226:408-420. [PMID: 39515595 DOI: 10.1016/j.freeradbiomed.2024.11.004] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2024] [Revised: 10/30/2024] [Accepted: 11/02/2024] [Indexed: 11/16/2024]
Abstract
Hydrogen peroxide (H2O2) is an important biological molecule, functioning both as a second messenger in cell signaling and, especially at higher concentrations, as a cause of cell damage. Cells harbor multiple enzymes that have peroxide reducing activity in vitro. However, the contribution of each of these enzymes towards peroxide scavenging in vivo is less clear. Therefore, to directly investigate in vivo peroxide scavenging, we used the genetically encoded peroxide probes, roGFP2-Tsa2ΔCR and HyPer7, to systematically screen the peroxide scavenging capacity of baker's yeast thiol and heme peroxidase mutants. We show that the 2-Cys peroxiredoxin Tsa1 alone is responsible for almost all exogenous H2O2 and tert-butyl hydroperoxide scavenging. Furthermore, Tsa1 can become an important source of H2O2-dependent cytosolic glutathione disulfide production. The two catalases and cytochrome c peroxidase only produce observable scavenging defects at higher H2O2 concentrations when these three heme peroxidases are removed in combination. We also analyzed the reduction of Tsa1 in vitro, revealing that the enzyme is efficiently reduced by thioredoxin-1 with a rate constant of 2.8 × 106 M-1s-1 but not by glutaredoxin-2. Tsa1 reduction by reduced glutathione occurs nonenzymatically with a rate constant of 2.9 M-1s-1. Hence, the observed Tsa1-dependent glutathione disulfide production in yeast probably requires the oxidation of thioredoxins. Our findings clarify the importance of the various thiol and heme peroxidases for peroxide removal and suggest that most thiol peroxidases have alternative or specialized functions in specific subcellular compartments.
Collapse
Affiliation(s)
- Jannik Zimmermann
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Lukas Lang
- Faculty of Chemistry, Comparative Biochemistry, RPTU Kaiserslautern, D-67663, Kaiserslautern, Germany
| | - Gaetano Calabrese
- Institute for Biochemistry, Redox Biochemistry, University of Cologne, Zuelpicher Str. 47a/R. 3.49, 50674, Cologne, Germany
| | - Hugo Laporte
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Prince S Amponsah
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany; Cellular Biochemistry, RPTU Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Christoph Michalk
- Cellular Biochemistry, RPTU Kaiserslautern, 67663, Kaiserslautern, Germany
| | - Tobias Sukmann
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Julian Oestreicher
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany
| | - Anja Tursch
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Esra Peker
- Institute for Biochemistry, Redox Biochemistry, University of Cologne, Zuelpicher Str. 47a/R. 3.49, 50674, Cologne, Germany
| | - Theresa N E Owusu
- Division of Redox Regulation, DKFZ-ZMBH Alliance, German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120, Heidelberg, Germany
| | - Matthias Weith
- Institute for Biochemistry, Redox Biochemistry, University of Cologne, Zuelpicher Str. 47a/R. 3.49, 50674, Cologne, Germany
| | - Leticia Prates Roma
- Institute of Biophysics, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66424, Homburg, Germany
| | - Marcel Deponte
- Faculty of Chemistry, Comparative Biochemistry, RPTU Kaiserslautern, D-67663, Kaiserslautern, Germany.
| | - Jan Riemer
- Institute for Biochemistry, Redox Biochemistry, University of Cologne, Zuelpicher Str. 47a/R. 3.49, 50674, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-Associated Diseases (CECAD), University of Cologne, 50931, Cologne, Germany.
| | - Bruce Morgan
- Institute of Biochemistry, Centre for Human and Molecular Biology (ZHMB), Saarland University, 66123, Saarbrücken, Germany.
| |
Collapse
|
26
|
Johal AS, Al-Shekaili HH, Abedrabbo M, Kehinde AZ, Towriss M, Koe JC, Hewton KG, Thomson SB, Ciernia AV, Leavitt B, Parker SJ. Restricting lysine normalizes toxic catabolites associated with ALDH7A1 deficiency in cells and mice. Cell Rep 2024; 43:115069. [PMID: 39661514 DOI: 10.1016/j.celrep.2024.115069] [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: 04/05/2024] [Revised: 10/04/2024] [Accepted: 11/22/2024] [Indexed: 12/13/2024] Open
Abstract
Lysine metabolism converges at α-aminoadipic semialdehyde dehydrogenase (ALDH7A1). Rare loss-of-function mutations in ALDH7A1 cause a toxic accumulation of lysine catabolites, including piperideine-6-carboxylate (P6C), that are thought to cause fatal seizures in children unless strictly managed with dietary lysine reduction. In this study, we perform metabolomics and expression analysis of tissues from Aldh7a1-deficient mice, which reveal tissue-specific differences in lysine metabolism and other metabolic pathways. We also develop a fluorescent biosensor to characterize lysine transporter activity and identify competitive substrates that reduce the accumulation of lysine catabolites in ALDH7A1-deficient HEK293 cells. Lastly, we show that intravenous administration of lysine α-oxidase from Trichoderma viride reduces lysine and P6C levels by >80% in mice. Our results improve our understanding of lysine metabolism and make inroads toward improving therapeutic strategies for lysine catabolic disorders.
Collapse
Affiliation(s)
- Amritpal S Johal
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Hilal H Al-Shekaili
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada
| | - Muna Abedrabbo
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Abisola Z Kehinde
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Morgan Towriss
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Jessica C Koe
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Keeley G Hewton
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Sarah B Thomson
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Annie V Ciernia
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Blair Leavitt
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada; Department of Medical Genetics, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC V6T 1Z4, Canada
| | - Seth J Parker
- Department of Biochemistry & Molecular Biology, University of British Columbia, Vancouver, BC V6T 1Z4, Canada; Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC V5Z 4H4, Canada; British Columbia Children's Hospital Research Institute, Vancouver, BC V5Z 4H4, Canada.
| |
Collapse
|
27
|
Pérez-Chávez I, Koberstein JN, Pueyo JM, Gilglioni EH, Vertommen D, Baeyens N, Ezeriņa D, Gurzov EN, Messens J. Tracking fructose 1,6-bisphosphate dynamics in liver cancer cells using a fluorescent biosensor. iScience 2024; 27:111336. [PMID: 39640569 PMCID: PMC11617404 DOI: 10.1016/j.isci.2024.111336] [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: 06/10/2024] [Revised: 09/10/2024] [Accepted: 11/04/2024] [Indexed: 12/07/2024] Open
Abstract
HYlight is a genetically encoded fluorescent biosensor that ratiometrically monitors fructose 1,6-bisphosphate (FBP), a key glycolytic metabolite. Given the role of glucose in liver cancer metabolism, we expressed HYlight in human liver cancer cells and primary mouse hepatocytes. Through in vitro, in silico, and in cellulo experiments, we showed HYlight's ability to monitor FBP changes linked to glycolysis, not gluconeogenesis. HYlight's affinity for FBP was ∼1 μM and stable within physiological pH range. HYlight demonstrated weak binding to dihydroxyacetone phosphate, and its ratiometric response was influenced by both ionic strength and phosphate. Therefore, simulating cytosolic conditions in vitro was necessary to establish a reliable correlation between HYlight's cellular responses and FBP concentrations. FBP concentrations were found to be in the lower micromolar range, far lower than previous millimolar estimates. Altogether, this biosensor approach offers real-time monitoring of FBP concentrations at single-cell resolution, making it an invaluable tool for the understanding of cancer metabolism.
Collapse
Affiliation(s)
- Israel Pérez-Chávez
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050 Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | | | - Julia Malo Pueyo
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050 Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Eduardo H. Gilglioni
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Didier Vertommen
- de Duve Institute, MASSPROT Platform, UCLouvain, 1200 Brussels, Belgium
| | - Nicolas Baeyens
- Laboratoire de Physiologie et de Pharmacologie (LAPP), Université Libre de Bruxelles (ULB), Brussels, Belgium
| | - Daria Ezeriņa
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050 Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| | - Esteban N. Gurzov
- Signal Transduction and Metabolism Laboratory, Université Libre de Bruxelles (ULB), Brussels, Belgium
- WELBIO Department, WEL Research Institute, Avenue Pasteur 6, Wavre B-1300, Belgium
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut Voor Biotechnologie, B-1050 Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, B-1050 Brussels, Belgium
| |
Collapse
|
28
|
Scherschel M, Niemeier JO, Jacobs LJHC, Hoffmann MDA, Diederich A, Bell C, Höhne P, Raetz S, Kroll JB, Steinbeck J, Lichtenauer S, Multhoff J, Zimmermann J, Sadhanasatish T, Rothemann RA, Grashoff C, Messens J, Ampofo E, Laschke MW, Riemer J, Roma LP, Schwarzländer M, Morgan B. A family of NADPH/NADP + biosensors reveals in vivo dynamics of central redox metabolism across eukaryotes. Nat Commun 2024; 15:10704. [PMID: 39702652 PMCID: PMC11659435 DOI: 10.1038/s41467-024-55302-x] [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: 03/11/2024] [Accepted: 12/08/2024] [Indexed: 12/21/2024] Open
Abstract
The NADPH/NADP+ redox couple is central to metabolism and redox signalling. NADP redox state is differentially regulated by distinct enzymatic machineries at the subcellular compartment level. Nonetheless, a detailed understanding of subcellular NADP redox dynamics is limited by the availability of appropriate tools. Here, we introduce NAPstars, a family of genetically encoded, fluorescent protein-based NADP redox state biosensors. NAPstars offer real-time, specific measurements, across a broad-range of NADP redox states, with subcellular resolution. NAPstar measurements in yeast, plants, and mammalian cell models, reveal a conserved robustness of cytosolic NADP redox homoeostasis. NAPstars uncover cell cycle-linked NADP redox oscillations in yeast and illumination- and hypoxia-dependent NADP redox changes in plant leaves. By applying NAPstars in combination with selective impairment of the glutathione and thioredoxin antioxidative pathways under acute oxidative challenge, we find an unexpected and conserved role for the glutathione system as the primary mediator of antioxidative electron flux.
Collapse
Affiliation(s)
- Marie Scherschel
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - Jan-Ole Niemeier
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Lianne J H C Jacobs
- Redox Metabolism, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Markus D A Hoffmann
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Anika Diederich
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - Christopher Bell
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Pascal Höhne
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Sonja Raetz
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Johanna B Kroll
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Janina Steinbeck
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Sophie Lichtenauer
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Jan Multhoff
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany
| | - Jannik Zimmermann
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany
| | - Tanmay Sadhanasatish
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, Münster, Germany
| | - R Alexander Rothemann
- Redox Metabolism, Institute for Biochemistry, University of Cologne, Cologne, Germany
| | - Carsten Grashoff
- Institute of Integrative Cell Biology and Physiology, University of Münster, Schlossplatz 5, Münster, Germany
| | - Joris Messens
- VIB-VUB Center for Structural Biology, Vlaams Instituut voor Biotechnologie, Brussels, Belgium
- Brussels Center for Redox Biology, Vrije Universiteit Brussel, Brussels, Belgium
- Structural Biology Brussels, Vrije Universiteit Brussel, Brussels, Belgium
| | - Emmanuel Ampofo
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Matthias W Laschke
- Institute for Clinical & Experimental Surgery, Saarland University, Homburg, Germany
| | - Jan Riemer
- Redox Metabolism, Institute for Biochemistry, University of Cologne, Cologne, Germany
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Cologne, Cologne, Germany
| | - Leticia Prates Roma
- Department of Biophysics, Center for Human and Molecular Biology (ZHMB), Saarland University, Homburg, Germany
| | - Markus Schwarzländer
- Institute of Plant Biology and Biotechnology, University of Münster, Schlossplatz 8, Münster, Germany.
| | - Bruce Morgan
- Institute of Biochemistry, Center for Human and Molecular Biology (ZHMB), Saarland University, Saarbrücken, Germany.
| |
Collapse
|
29
|
Oh YK, Yadavalli HC, Ryu MY, Shah P, Oh TR, Choi SW, Cho SK, Kim YJ, Kim JH, Yang SW. Application of fluorescence i-motif DNA silver nanocluster sensor to visualize endogenous reactive oxygen species in plant cells. PLANT CELL REPORTS 2024; 44:6. [PMID: 39676128 DOI: 10.1007/s00299-024-03398-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/01/2024] [Accepted: 12/05/2024] [Indexed: 12/17/2024]
Abstract
KEY MESSAGE A novel fluorescent i-motif DNA silver nanoclusters system has been developed for visualization of reactive oxygen species in plants, enabling the detection of intracellular signaling in plant cells. Reactive oxygen species (ROS) are crucial in plant growth, defense, and stress responses, making them vital for improving crop resilience. Various ROS sensing methods for plants have been developed to detect ROS in vitro and in vivo. However, each method comes its own advantages and disadvantages, leading to an increasing demand for a simple and effective sensory system for ROS detection in plants. Here, we introduce novel DNA silver nanoclusters (DNA/AgNCs) sensors for visualizing ROS in plants. Two sensors, C20/AgNCs and FAM-C20/AgNCs-Cy5, detect intracellular ROS signaling in response to stimuli, such as abscisic acid, salicylic acid, ethylene, and bacterial peptide elicitor flg22. Notably, FAM-C20/AgNCs-Cy5 exceeds the sensing capabilities of HyPer7, a widely recognized ROS sensor. Taken together, we suggest that fluorescent i-motif DNA/AgNCs system is an effective tool for visualizing ROS signals in plant cells. This advancement is important to advancing our understanding of ROS-mediated processes in plant biology.
Collapse
Affiliation(s)
- Young Kyoung Oh
- Department of Systems Biology, Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Hari Chandana Yadavalli
- Department of Systems Biology, Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea
| | - Moon Young Ryu
- Xenohelix Research Institute, BT Centre 305, 56 Songdogwahakro, Yeonsugu, Incheon, 21984, Korea
| | - Pratik Shah
- Department of Science and Environment, Roskilde University, 4000, Roskilde, Denmark
| | - Tae Rin Oh
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Suk Won Choi
- Department of Biochemistry and Biophysics, Texas A&M University, College Station, TX, USA
| | - Seok Keun Cho
- Xenohelix Research Institute, BT Centre 305, 56 Songdogwahakro, Yeonsugu, Incheon, 21984, Korea
| | - Yun Ju Kim
- Department of Systems Biology, Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| | - Jong Hum Kim
- Department of Life Science, Pohang University of Science and Technology (POSTECH), Pohang, 37673, Korea.
- Institute for Convergence Research and Education in Advanced Technology, Yonsei University, Seoul, 03722, Korea.
| | - Seong Wook Yang
- Department of Systems Biology, Institute of Life Science and Biotechnology, Yonsei University, Seoul, 03722, Korea.
| |
Collapse
|
30
|
Young M, Booth DM, Smith D, Tigano M, Hajnόczky G, Joseph SK. Transcriptional regulation in the absence of inositol trisphosphate receptor calcium signaling. Front Cell Dev Biol 2024; 12:1473210. [PMID: 39712573 PMCID: PMC11659226 DOI: 10.3389/fcell.2024.1473210] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2024] [Accepted: 11/13/2024] [Indexed: 12/24/2024] Open
Abstract
The activation of IP3 receptor (IP3R) Ca2+ channels generates agonist-mediated Ca2+ signals that are critical for the regulation of a wide range of biological processes. It is therefore surprising that CRISPR induced loss of all three IP3R isoforms (TKO) in HEK293 and HeLa cell lines yields cells that can survive, grow and divide, albeit more slowly than wild-type cells. In an effort to understand the adaptive mechanisms involved, we have examined the activity of key Ca2+ dependent transcription factors (NFAT, CREB and AP-1) and signaling pathways using luciferase-reporter assays, phosphoprotein immunoblots and whole genome transcriptomic studies. In addition, the diacylglycerol arm of the signaling pathway was investigated with protein kinase C (PKC) inhibitors and siRNA knockdown. The data showed that agonist-mediated NFAT activation was lost but CREB activation was maintained in IP3R TKO cells. Under base-line conditions transcriptome analysis indicated the differential expression of 828 and 311 genes in IP3R TKO HEK293 or HeLa cells, respectively, with only 18 genes being in common. Three main adaptations in TKO cells were identified in this study: 1) increased basal activity of NFAT, CREB and AP-1; 2) an increased reliance on Ca2+- insensitive PKC isoforms; and 3) increased production of reactive oxygen species and upregulation of antioxidant defense enzymes. We suggest that whereas wild-type cells rely on a Ca2+ and DAG signal to respond to stimuli, the TKO cells utilize the adaptations to allow key signaling pathways (e.g., PKC, Ras/MAPK, CREB) to transition to the activated state using a DAG signal alone.
Collapse
Affiliation(s)
- Michael Young
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David M. Booth
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - David Smith
- Center for Single Cell Biology, Children’s Hospital of Philadelphia, Philadelphia, PA, United States
| | - Marco Tigano
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Gyӧrgy Hajnόczky
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| | - Suresh K. Joseph
- MitoCare Center, Department of Pathology and Genomic Medicine, Thomas Jefferson University, Philadelphia, PA, United States
| |
Collapse
|
31
|
Jia Q, Young D, Zhang Q, Sieburth D. Endogenous hydrogen peroxide positively regulates secretion of a gut-derived peptide in neuroendocrine potentiation of the oxidative stress response in Caenorhabditis elegans. eLife 2024; 13:RP97503. [PMID: 39636673 PMCID: PMC11620748 DOI: 10.7554/elife.97503] [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: 12/07/2024] Open
Abstract
The gut-brain axis mediates bidirectional signaling between the intestine and the nervous system and is critical for organism-wide homeostasis. Here, we report the identification of a peptidergic endocrine circuit in which bidirectional signaling between neurons and the intestine potentiates the activation of the antioxidant response in Caenorhabditis elegans in the intestine. We identify an FMRF-amide-like peptide, FLP-2, whose release from the intestine is necessary and sufficient to activate the intestinal oxidative stress response by promoting the release of the antioxidant FLP-1 neuropeptide from neurons. FLP-2 secretion from the intestine is positively regulated by endogenous hydrogen peroxide (H2O2) produced in the mitochondrial matrix by sod-3/superoxide dismutase, and is negatively regulated by prdx-2/peroxiredoxin, which depletes H2O2 in both the mitochondria and cytosol. H2O2 promotes FLP-2 secretion through the DAG and calcium-dependent protein kinase C family member pkc-2 and by the SNAP25 family member aex-4 in the intestine. Together, our data demonstrate a role for intestinal H2O2 in promoting inter-tissue antioxidant signaling through regulated neuropeptide-like protein exocytosis in a gut-brain axis to activate the oxidative stress response.
Collapse
Affiliation(s)
- Qi Jia
- Development, Stem Cells and Regenerative Medicine PhD program, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
| | - Drew Young
- Neuroscience Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Qixin Zhang
- Neuromedicine Graduate Program, University of Southern CaliforniaLos AngelesUnited States
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
| | - Derek Sieburth
- Zilkha Neurogenetic Institute, University of Southern CaliforniaLos AngelesUnited States
- Department of Physiology and Neuroscience, Keck School of Medicine, University of Southern CaliforniaLos AngelesUnited States
| |
Collapse
|
32
|
Barakat S, Çimen Ş, Miri SM, Vatandaşlar E, Yelkenci HE, San Martín A, Beker MÇ, Kök K, Öztürk G, Eroglu E. Bioenergetic shift and proteomic signature induced by lentiviral-transduction of GFP-based biosensors. Redox Biol 2024; 78:103416. [PMID: 39509993 PMCID: PMC11574814 DOI: 10.1016/j.redox.2024.103416] [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: 09/17/2024] [Revised: 10/12/2024] [Accepted: 11/01/2024] [Indexed: 11/15/2024] Open
Abstract
Fluorescent proteins (FPs) stand as pivotal tools extensively employed across diverse biological research endeavors in various model systems. However, long-standing concerns surround their use due to the numerous side effects associated with their expression. Recent investigations have brought to light the significance of hydrogen peroxide (H2O2) that is associated with the maturation process of green fluorescent protein (GFP) fluorophores. The structural and functional impairments associated with GFP expression are possibly linked to this amount of H2O2. In this study, we assess the impact of the GFP-based HyPer7 biosensor on cellular homeostasis and proteome changes, aiming to identify potential risks related to oxidative stress responses that potentially risks the application of such tools. Cells expressing genome-integrated HyPer7 demonstrated altered mitochondrial membrane potential (MMP), which was alleviated by the addition of antioxidants or culturing cells at physiological normoxia (5 kPa O2). Additionally, HyPer7-expressing cells also exhibited significant impairment in mitochondrial oxidative respiration, suggesting broader mitochondrial dysfunction. Through untargeted proteomics analysis, we identified 26 proteins exhibiting differential expression in HyPer7-expressing cells compared to respective control cells. Functional annotation analysis showed that the list of the delineated proteins is associated with cellular responses to stress and the regulation of antioxidant mechanisms. Our findings underscore the significance of caution and validation in ensuring a thorough comprehension of cellular responses when using fluorescent protein-based tools, thereby enhancing the reliability of the results.
Collapse
Affiliation(s)
- Sarah Barakat
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Şeyma Çimen
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Department of Nutrition and Dietetics, Institution of Health Sciences, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Seyed Mohammad Miri
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, 34956, Turkey
| | - Emre Vatandaşlar
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Hayriye Ecem Yelkenci
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Alejandro San Martín
- Centro de Estudios Científicos (CECs), 5110466, Valdivia, Chile; Facultad de Ciencias para el Cuidado de la Salud, Universidad San Sebastián, 5110773, Valdivia, Chile
| | - Mustafa Çağlar Beker
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Department of Physiology, School of Medicine, Istanbul Medipol University, Istanbul, 34810, Turkey
| | - Kıvanç Kök
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Department of Biostatistics and Medical Informatics, International School of Medicine, Istanbul Medipol University, Istanbul, 34810, Turkey.
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Department of Physiology, School of Medicine, Bolu Abant İzzet Baysal University, Bolu, 14030, Turkey.
| | - Emrah Eroglu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul, 34810, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul, 34956, Turkey.
| |
Collapse
|
33
|
Gomes F, Turano H, Haddad LA, Netto LES. Human mitochondrial peroxiredoxin Prdx3 is dually localized in the intermembrane space and matrix subcompartments. Redox Biol 2024; 78:103436. [PMID: 39591905 PMCID: PMC11626719 DOI: 10.1016/j.redox.2024.103436] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Revised: 11/14/2024] [Accepted: 11/17/2024] [Indexed: 11/28/2024] Open
Abstract
Peroxiredoxin 3 (Prdx3) is the major sink for H2O2 and other hydroperoxides within mitochondria, yet the mechanisms guiding the import of its cytosolic precursor into mitochondrial sub-compartments remain elusive. Prdx3 is synthesized in the cytosol as a precursor with an N-terminal cleavable presequence, which is frequently proposed to target the protein exclusively to the mitochondrial matrix. Here, we present a comprehensive analysis of the human Prdx3 biogenesis, using highly purified mitochondria from HEK293T cells. Subfractionation and probing for specific mitochondrial markers confirmed Prdx3 localization in the matrix, while unexpectedly revealed its presence in the mitochondrial intermembrane space (IMS). Both matrix and IMS isoforms were found to be soluble proteins, as demonstrated by alkaline carbonate extraction. By combining in silico analysis, in organello import assays and heterologous expression in yeast, we found that Prdx3 undergoes sequential proteolytic processing steps by mitochondrial processing peptidase (MPP) and mitochondrial intermediate peptidase (MIP) during its import into the matrix. Additionally, heterologous expression of Prdx3 in yeast revealed that its sorting to the IMS is dependent on the inner membrane peptidase (IMP) complex. Collectively, these findings uncover a complex submitochondrial distribution of Prdx3, supporting its multifaceted role in mitochondrial H2O2 metabolism.
Collapse
Affiliation(s)
- Fernando Gomes
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, 05508-090, Brazil.
| | - Helena Turano
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, 05508-090, Brazil
| | - Luciana A Haddad
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, 05508-090, Brazil
| | - Luis E S Netto
- Departamento de Genética e Biologia Evolutiva, Instituto de Biociências, Universidade de São Paulo, São Paulo, 05508-090, Brazil.
| |
Collapse
|
34
|
Nedo AO, Liang H, Sriram J, Razzak MA, Lee JY, Kambhamettu C, Dinesh-Kumar SP, Caplan JL. CHUP1 restricts chloroplast movement and effector-triggered immunity in epidermal cells. THE NEW PHYTOLOGIST 2024; 244:1864-1881. [PMID: 39415611 PMCID: PMC11583462 DOI: 10.1111/nph.20147] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2024] [Accepted: 08/29/2024] [Indexed: 10/19/2024]
Abstract
Chloroplast Unusual Positioning 1 (CHUP1) plays an important role in the chloroplast avoidance and accumulation responses in mesophyll cells. In epidermal cells, prior research showed silencing CHUP1-induced chloroplast stromules and amplified effector-triggered immunity (ETI); however, the underlying mechanisms remain largely unknown. CHUP1 has a dual function in anchoring chloroplasts and recruiting chloroplast-associated actin (cp-actin) filaments for blue light-induced movement. To determine which function is critical for ETI, we developed an approach to quantify chloroplast anchoring and movement in epidermal cells. Our data show that silencing NbCHUP1 in Nicotiana benthamiana plants increased epidermal chloroplast de-anchoring and basal movement but did not fully disrupt blue light-induced chloroplast movement. Silencing NbCHUP1 auto-activated epidermal chloroplast defense (ECD) responses including stromule formation, perinuclear chloroplast clustering, the epidermal chloroplast response (ECR), and the chloroplast reactive oxygen species (ROS), hydrogen peroxide (H2O2). These findings show chloroplast anchoring restricts a multifaceted ECD response. Our results also show that the accumulated chloroplastic H2O2 in NbCHUP1-silenced plants was not required for the increased basal epidermal chloroplast movement but was essential for increased stromules and enhanced ETI. This finding indicates that chloroplast de-anchoring and H2O2 play separate but essential roles during ETI.
Collapse
Affiliation(s)
- Alexander O Nedo
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19713, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Huining Liang
- Department of Computer & Information Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Jaya Sriram
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19713, USA
| | - Md Abdur Razzak
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19713, USA
| | - Jung-Youn Lee
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19713, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Chandra Kambhamettu
- Department of Computer & Information Sciences, University of Delaware, Newark, DE, 19716, USA
| | - Savithramma P Dinesh-Kumar
- Department of Plant Biology and The Genome Center, College of Biological Sciences, University of California, Davis, CA, 95616, USA
| | - Jeffrey L Caplan
- Department of Plant and Soil Sciences, University of Delaware, Newark, DE, 19716, USA
- Delaware Biotechnology Institute, University of Delaware, Newark, DE, 19713, USA
- Department of Biological Sciences, University of Delaware, Newark, DE, 19716, USA
| |
Collapse
|
35
|
Gest AM, Sahan AZ, Zhong Y, Lin W, Mehta S, Zhang J. Molecular Spies in Action: Genetically Encoded Fluorescent Biosensors Light up Cellular Signals. Chem Rev 2024; 124:12573-12660. [PMID: 39535501 PMCID: PMC11613326 DOI: 10.1021/acs.chemrev.4c00293] [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: 04/17/2024] [Revised: 09/07/2024] [Accepted: 09/20/2024] [Indexed: 11/16/2024]
Abstract
Cellular function is controlled through intricate networks of signals, which lead to the myriad pathways governing cell fate. Fluorescent biosensors have enabled the study of these signaling pathways in living systems across temporal and spatial scales. Over the years there has been an explosion in the number of fluorescent biosensors, as they have become available for numerous targets, utilized across spectral space, and suited for various imaging techniques. To guide users through this extensive biosensor landscape, we discuss critical aspects of fluorescent proteins for consideration in biosensor development, smart tagging strategies, and the historical and recent biosensors of various types, grouped by target, and with a focus on the design and recent applications of these sensors in living systems.
Collapse
Affiliation(s)
- Anneliese
M. M. Gest
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Ayse Z. Sahan
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Biomedical
Sciences Graduate Program, University of
California, San Diego, La Jolla, California 92093, United States
| | - Yanghao Zhong
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Wei Lin
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Sohum Mehta
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
| | - Jin Zhang
- Department
of Pharmacology, University of California,
San Diego, La Jolla, California 92093, United States
- Shu
Chien-Gene Lay Department of Bioengineering, University of California, San Diego, La Jolla, California 92093, United States
- Department
of Chemistry and Biochemistry, University
of California, San Diego, La Jolla, California 92093, United States
| |
Collapse
|
36
|
Jackson MJ. Exercise-induced adaptations to homeostasis of reactive oxygen species in skeletal muscle. Free Radic Biol Med 2024; 225:494-500. [PMID: 39427746 DOI: 10.1016/j.freeradbiomed.2024.10.270] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 10/03/2024] [Accepted: 10/07/2024] [Indexed: 10/22/2024]
Abstract
Reactive oxygen species are generated by multiple mechanisms during contractile activity in exercising skeletal muscle and are recognised to play a role in signaling adaptations to the contractions. The sources of the superoxide and hydrogen peroxide generated are now relatively well understood but how the resulting low concentrations of hydrogen peroxide induce activation of multiple signaling pathways remains obscure. Several theories are presented together with accumulating evidence that 2-Cys peroxiredoxins may play a role of "effector" proteins in mediating the signaling actions of hydrogen peroxide. Identification of the mechanisms underlying these pathways offers the potential in the longer term for development of novel interventions to maintain exercise responses in the elderly with the potential to maintain muscle mass and function and consequent quality of life.
Collapse
Affiliation(s)
- Malcolm J Jackson
- MRC-Versus Arthritis Centre for Integrated Research Into Musculoskeletal Ageing (CIMA), Department of Musculoskeletal and Ageing Science, Institute of Life Course and Medical Sciences, University of Liverpool, Liverpool, L7 8TX, UK.
| |
Collapse
|
37
|
Ma Y, Hu W, Hu J, Ruan M, Hu J, Yang M, Zhang Y, Xie H, Hu C. Bifunctional nanoprobe for simultaneous detection of intracellular reactive oxygen species and temperature in single cells. MICROSYSTEMS & NANOENGINEERING 2024; 10:171. [PMID: 39562541 PMCID: PMC11577004 DOI: 10.1038/s41378-024-00814-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2024] [Revised: 09/08/2024] [Accepted: 09/20/2024] [Indexed: 11/21/2024]
Abstract
Living cells can rapidly adjust their metabolic activities in response to external stimuli, leading to fluctuations in intracellular temperature and reactive oxygen species (ROS) levels. Monitoring these parameters is essential for understanding cellular metabolism, particularly during dynamic biological processes. In this study, we present a bifunctional nanoprobe capable of simultaneous measurement of ROS levels and temperature within single cells. The nanoprobe features two individually addressable nanoelectrodes, with platinum (Pt) and nickel (Ni) coatings on both sides. At the tip, these two metal layers form a nano-thermocouple, enabling precise intracellular temperature measurements, while the Pt layer facilitates selective ROS detection. This dual functionality allows for real-time monitoring of cellular responses during synergistic chemo-photothermal therapy of cancer cells and zebrafish embryos subjected to mitochondrial toxic stress. Our results demonstrate that the nanoprobe effectively measures increases in temperature and ROS levels in HeLa cells undergoing chemo-photothermal therapy, as well as in chemically stimulated zebrafish embryos. By providing detailed analysis of submicrometer-scale temperature and ROS variations within living cells, this nanoprobe offers valuable insights into cellular processes and holds promise for early disease detection and drug development.
Collapse
Affiliation(s)
- Yanmei Ma
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Weikang Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Jian Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Muyang Ruan
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Jie Hu
- Department of Electrical and Electronic Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Ming Yang
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Yi Zhang
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Hanhan Xie
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China
| | - Chengzhi Hu
- Shenzhen Key Laboratory of Biomimetic Robotics and Intelligent Systems, Department of Mechanical and Energy Engineering, Southern University of Science and Technology, Shenzhen, 518000, China.
| |
Collapse
|
38
|
Chang X, Chen X, Zhang X, Chen N, Tang W, Zhang Z, Zheng S, Huang J, Ji Y, Zhao Y, Yang Y, Li X. A bright red fluorescent genetically encoded sensor for lactate imaging. Biochem Biophys Res Commun 2024; 734:150449. [PMID: 39096623 DOI: 10.1016/j.bbrc.2024.150449] [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: 07/22/2024] [Accepted: 07/23/2024] [Indexed: 08/05/2024]
Abstract
Lactate plays a crucial role in energy metabolism and greatly impacts protein activities, exerting diverse physiological and pathological effects. Therefore, convenient lactate assays for tracking spatiotemporal dynamics in living cells are desirable. In this paper, we engineered and optimized a red fluorescent protein sensor for l-lactate named FiLa-Red. This indicator exhibited a maximal fluorescence change of 730 % and an apparent dissociation constant (Kd) of approximately 460 μM. By utilizing FiLa-Red and other sensors, we monitored energy metabolism in a multiplex manner by simultaneously tracking lactate and NAD+/NADH abundance in the cytoplasm, nucleus, and mitochondria. The FiLa-Red sensor is expected to be a useful tool for performing metabolic analysis in vitro, in living cells and in vivo.
Collapse
Affiliation(s)
- Xuanming Chang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Xiaoqian Chen
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Xiuze Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Nian Chen
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Weitao Tang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Zhuo Zhang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Sulin Zheng
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Jiong Huang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yihan Ji
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China
| | - Yuzheng Zhao
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Yi Yang
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China
| | - Xie Li
- Optogenetics & Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai, 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing, 100730, China.
| |
Collapse
|
39
|
Hunt M, Torres M, Bachar-Wikstrom E, Wikstrom JD. Cellular and molecular roles of reactive oxygen species in wound healing. Commun Biol 2024; 7:1534. [PMID: 39562800 DOI: 10.1038/s42003-024-07219-w] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 11/05/2024] [Indexed: 11/21/2024] Open
Abstract
Wound healing is a highly coordinated spatiotemporal sequence of events involving several cell types and tissues. The process of wound healing requires strict regulation, and its disruption can lead to the formation of chronic wounds, which can have a significant impact on an individual's health as well as on worldwide healthcare expenditure. One essential aspect within the cellular and molecular regulation of wound healing pathogenesis is that of reactive oxygen species (ROS) and oxidative stress. Wounding significantly elevates levels of ROS, and an array of various reactive species are involved in modulating the wound healing process, such as through antimicrobial activities and signal transduction. However, as in many pathologies, ROS play an antagonistic pleiotropic role in wound healing, and can be a pathogenic factor in the formation of chronic wounds. Whilst advances in targeting ROS and oxidative stress have led to the development of novel pre-clinical therapeutic methods, due to the complex nature of ROS in wound healing, gaps in knowledge remain concerning the specific cellular and molecular functions of ROS in wound healing. In this review, we highlight current knowledge of these functions, and discuss the potential future direction of new studies, and how these pathways may be targeted in future pre-clinical studies.
Collapse
Affiliation(s)
- Matthew Hunt
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Monica Torres
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden
| | - Etty Bachar-Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden
| | - Jakob D Wikstrom
- Dermatology and Venereology Division, Department of Medicine (Solna), Karolinska Institutet, Stockholm, Sweden.
- Dermato-Venereology Clinic, Karolinska University Hospital, Stockholm, Sweden.
| |
Collapse
|
40
|
Chang J, Li Z, Yuan H, Wang X, Xu J, Yang P, Qin L. Protective role of aconitate decarboxylase 1 in neuroinflammation-induced dysfunctions of the paraventricular thalamus and sleepiness. Commun Biol 2024; 7:1484. [PMID: 39523388 PMCID: PMC11551151 DOI: 10.1038/s42003-024-07215-0] [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: 03/26/2024] [Accepted: 11/05/2024] [Indexed: 11/16/2024] Open
Abstract
Sleepiness is commonly associated with neuroinflammation; however, the underlying neuroregulatory mechanisms remain unclear. Previous research suggests that the paraventricular thalamus (PVT) plays a crucial role in regulating sleep-wake dynamics; thus, neurological abnormalities in the PVT may contribute to neuroinflammation-induced sleepiness. To test this hypothesis, we performed electroencephalography recordings in mice treated with lipopolysaccharide (LPS) and found that the mice exhibited temporary sleepiness lasting for 7 days. Using the Fos-TRAP method, fiber photometry recordings, and immunofluorescence staining, we detected temporary PVT neuron hypoactivation and microglia activation from day 1 to day 7 post-LPS treatment. Combining the results of bulk and single-cell RNA sequencing, we found upregulation of aconitate decarboxylase 1 (Acod1) in PVT microglia post-LPS treatment. To investigate the role of Acod1, we manipulated Acod1 gene expression in PVT microglia via stereotactic injection of short hairpin RNA adenovirus. Knockdown of Acod1 exacerbated inflammation, neuronal hypoactivation, and sleepiness. Itaconate is a metabolite synthesized by the enzyme encoded by Acod1. Finally, we confirmed that exogenous administration of an itaconate derivative, 4-octyl itaconate, could inhibit microglia activation, alleviate neuronal dysfunction, and relieve sleepiness. Our findings highlight PVT's role in inflammation-induced sleepiness and suggest Acod1 as a potential therapeutic target for neuroinflammation.
Collapse
Affiliation(s)
- Jianjun Chang
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Zijie Li
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Hui Yuan
- Laboratory of Hearing Research, School of Life Sciences, China Medical University, Shenyang, China
| | - Xuejiao Wang
- Department of Physiology, School of Life Sciences, China Medical University, Shenyang, China
| | - Jingyi Xu
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, China
| | - Pingting Yang
- Department of Rheumatology and Immunology, The First Hospital of China Medical University, Shenyang, China.
| | - Ling Qin
- Laboratory of Hearing Research, School of Life Sciences, China Medical University, Shenyang, China.
| |
Collapse
|
41
|
Shannon N, Raymond C, Palmer C, Seward D, Cunniff B. Miro1 expression alters global gene expression, ERK1/2 phosphorylation, oxidation, and cell cycle progression. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.11.06.622334. [PMID: 39574731 PMCID: PMC11581026 DOI: 10.1101/2024.11.06.622334] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/02/2024]
Abstract
Subcellular mitochondrial positioning in cells is necessary for localized energy and signaling requirements. Mitochondria are strategically trafficked throughout the cytoplasm via the actin cytoskeleton, microtubule motor proteins, and adaptor proteins. Miro1, an outer mitochondrial membrane adaptor protein, is necessary for attachment of mitochondria to microtubule motor proteins for trafficking. Previous work showed when Miro1 is deleted (Miro1-/-) from mouse embryonic fibroblasts (MEFs), the mitochondria become sequestered to the perinuclear space, disrupting subcellular energy and reactive oxygen species gradients. Here, we show that Miro1-/- MEFs grow slower compared to Miro1+/+ and Miro1-/- MEFs stably re-expressing the Myc-Miro1 plasmid. Miro1-/- MEFs have a have a cell cycle defect with decreased percentage of cells in G1 and increased cells in the S phase of the cell cycle. We conducted the first ever RNA sequencing experiment dependent upon Miro1 expression and found differential expression in cell proliferation and migration genes upon deletion of Miro1, including the MAP Kinase signaling pathway. We find that ERK1/2 phosphorylation is elevated both spatially (cytoplasm and nucleus) and temporally following serum stimulation in Miro1-/- MEFs. We investigated the expression levels and oxidation of the Dual Specificity Phosphatases (DUSP1-6), ERK1/2 target phosphatases. We found no differences in DUSP1-6 expression and oxidation under asynchronous and synchronized cells. Lastly, we evaluated the oxidation status of ERK1/2 and found an increase in ERK1/2 oxidation in the Miro1-/- MEFs compared to Miro1+/+ and Myc-Miro1. These data highlight transcriptional control based off Miro1 expression and demonstrate the highly dynamic regulation of ERK1/2 upon deletion of Miro1 that may support the observed cell cycle and proliferation defects.
Collapse
Affiliation(s)
- Nathaniel Shannon
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Cory Raymond
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Chloe Palmer
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - David Seward
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| | - Brian Cunniff
- Department of Pathology and Laboratory Medicine, University of Vermont Cancer Center, Larner College of Medicine, Burlington, VT 05405, USA
| |
Collapse
|
42
|
Choya-Foces C, Navarro E, Ríos CDL, López MG, Egea J, Hernansanz-Agustín P, Martínez-Ruiz A. The mitochondrial Na +/Ca 2+ exchanger NCLX is implied in the activation of hypoxia-inducible factors. Redox Biol 2024; 77:103364. [PMID: 39341036 PMCID: PMC11470253 DOI: 10.1016/j.redox.2024.103364] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2024] [Revised: 09/18/2024] [Accepted: 09/19/2024] [Indexed: 09/30/2024] Open
Abstract
Eukaryotic cells and organisms depend on oxygen for basic living functions, and they display a panoply of adaptations to situations in which oxygen availability is diminished (hypoxia). A number of these responses in animals are mediated by changes in gene expression programs directed by hypoxia-inducible factors (HIFs), whose main mechanism of stabilization and functional activation in response to decreased cytosolic oxygen concentration was elucidated two decades ago. Human acute responses to hypoxia have been known for decades, although their precise molecular mechanism for oxygen sensing is not fully understood. It is already known that a redox component, linked with reactive oxygen species (ROS) production of mitochondrial origin, is implied in these responses. We have recently described a mechanism by which the mitochondrial sodium/calcium exchanger, NCLX, participates in mitochondrial electron transport chain regulation and ROS production in response to acute hypoxia. Here we show that NCLX is also implied in the response to hypoxia mediated by the HIFs. By using a NCLX inhibitor and interference RNA we show that NCLX activity is necessary for HIF-α subunits stabilization in hypoxia and for HIF-1-dependent transcriptional activity. We also show that hypoxic mitochondrial ROS production is not required for HIF-1α stabilization under all circumstances, suggesting that the basal cytosolic redox state or other mechanism(s) could be operating in the NCLX-mediated response to hypoxia that operates through HIF-α stabilization. This finding provides a link between acute and medium-term responses to hypoxia, reinforcing a central role of mitochondrial cell signalling in the response to hypoxia.
Collapse
Affiliation(s)
- Carmen Choya-Foces
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Elisa Navarro
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Instituto Universitario de Investigación en Neuroquímica, Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Universidad Complutense, Madrid, Spain; Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Spain; Instituto Ramón y Cajal de Investigación Sanitaria (IRYCIS), Madrid, Spain
| | - Cristóbal de Los Ríos
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Grupo de Investigación de Alto Rendimiento en Fisiopatología y Farmacología del Sistema Digestivo (NeuGut), Departamento de Ciencias Básicas de la Salud, Universidad Rey Juan Carlos, Alcorcón (Madrid), Spain
| | - Manuela G López
- Instituto Teófilo Hernando, Departamento de Farmacología y Terapéutica, Facultad de Medicina, Universidad Autónoma de Madrid (UAM), Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Javier Egea
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain
| | - Pablo Hernansanz-Agustín
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain; Centro Nacional de Investigaciones Cardiovasculares Carlos III (CNIC), Madrid, Spain; Departamento de Neurobiología Molecular, Celular y del Desarrollo, Instituto Cajal, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| | - Antonio Martínez-Ruiz
- Unidad de Investigación, Hospital Universitario Santa Cristina, Instituto de Investigación Sanitaria Princesa (IIS-IP), Madrid, Spain.
| |
Collapse
|
43
|
Tsuboyama S, Okumura T, Watanabe K, Koga K, Shiratani M, Kuchitsu K. Real-time live imaging of cytosolic hydrogen peroxide and Ca 2+ of Marchantia polymorpha gemmalings reveal immediate initial responses of plant cells triggered by nonthermal plasma irradiation. PLANT PHYSIOLOGY AND BIOCHEMISTRY : PPB 2024; 216:109172. [PMID: 39395224 DOI: 10.1016/j.plaphy.2024.109172] [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: 05/06/2024] [Revised: 09/03/2024] [Accepted: 09/29/2024] [Indexed: 10/14/2024]
Abstract
Cold atmospheric pressure plasma generators capable of generating plasma under normal pressure and temperature conditions have recently been developed, and their biological applications have been extensively studied. Plasma irradiation has been reported to affect plant germination and growth; however, the molecular mechanism underlying these effects and initial cellular responses to plasma irradiation remains poorly understood. To unravel the molecular and cellular mechanisms underlying the effects of plasma irradiation on plants, we have been establishing novel experimental systems using a model liverwort Marchantia polymorpha. We here focused on the initial responses of plant cells to plasma irradiation. To investigate immediate cellular responses following plasma irradiation, we developed a new plasma device that allows irradiation under a microscope. Through integration with live fluorescence imaging, we established an experimental setup to track, the dynamics of intracellular concentration of H2O2 and Ca2+ as representative initial cellular responses. We revealed that plasma irradiation induced a rapid and transient increase in intracellular concentration of H2O2 and Ca2+ in Marchantia gemmalings. Pharmacological analyses suggested that the long-lived reactive species, H2O2, generated by the plasma generator was directly delivered into the plant cells. Competitive inhibitors of Ca2+ channels abolished the Ca2+ rise, suggesting that plasma irradiation immediately activate plasma membrane Ca2+ channel(s) to induce Ca2+ influx. Importantly, this study marks the inaugural demonstration of real-time monitoring of cytosolic H2O2 and Ca2+ dynamics in plants, triggered by plasma irradiation.
Collapse
Affiliation(s)
- Shoko Tsuboyama
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Takamasa Okumura
- Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan
| | - Kenshiro Watanabe
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba, 278-8510, Japan
| | - Kazunori Koga
- Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan.
| | - Masaharu Shiratani
- Faculty of Information Science and Electrical Engineering, Kyushu University, Fukuoka, Japan; Center of Plasma Nano-interface Engineering, Kyushu University, Fukuoka, Japan
| | - Kazuyuki Kuchitsu
- Department of Applied Biological Science, Tokyo University of Science, Noda, Chiba, 278-8510, Japan.
| |
Collapse
|
44
|
Zhang Y, Li Y, Ren T, Duan JA, Xiao P. Promising tools into oxidative stress: A review of non-rodent model organisms. Redox Biol 2024; 77:103402. [PMID: 39437623 PMCID: PMC11532775 DOI: 10.1016/j.redox.2024.103402] [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: 07/28/2024] [Revised: 10/07/2024] [Accepted: 10/16/2024] [Indexed: 10/25/2024] Open
Abstract
Oxidative stress is a crucial concept in redox biology, and significant progress has been made in recent years. Excessive levels of reactive oxygen species (ROS) can lead to oxidative damage, heightening vulnerability to various diseases. By contrast, ROS maintained within a moderate range plays a role in regulating normal physiological metabolism. Choosing suitable animal models in a complex research context is critical for enhancing research efficacy. While rodents are frequently utilized in medical experiments, they pose challenges such as high costs and ethical considerations. Alternatively, non-rodent model organisms like zebrafish, Drosophila, and C. elegans offer promising avenues into oxidative stress research. These organisms boast advantages such as their small size, high reproduction rate, availability for live imaging, and ease of gene manipulation. This review highlights advancements in the detection of oxidative stress using non-rodent models. The oxidative homeostasis regulatory pathway, Kelch-like ECH-associated protein 1-Nuclear factor erythroid 2-related factor 2 (Keap1-Nrf2), is systematically reviewed alongside multiple regulation of Nrf2-centered pathways in different organisms. Ultimately, this review conducts a comprehensive comparative analysis of different model organisms and further explores the combination of novel techniques with non-rodents. This review aims to summarize state-of-the-art findings in oxidative stress research using non-rodents and to delineate future directions.
Collapse
Affiliation(s)
- Yuhao Zhang
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Yun Li
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Tianyi Ren
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China
| | - Jin-Ao Duan
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| | - Ping Xiao
- Jiangsu Collaborative Innovation Center of Chinese Medicinal Resources Industrialization, National and Local Collaborative Engineering Center of Chinese Medicinal Resources Industrialization and Formulae Innovative Medicine, and Jiangsu Key Laboratory for High Technology Research of TCM Formulae, Nanjing University of Chinese Medicine, Nanjing, 210023, China.
| |
Collapse
|
45
|
Zhang Z, Chen C, Li X, Zheng J, Zhao Y. Regulation of leukemogenesis via redox metabolism. Trends Cell Biol 2024; 34:928-941. [PMID: 39492031 DOI: 10.1016/j.tcb.2023.10.001] [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: 07/12/2023] [Revised: 09/27/2023] [Accepted: 10/04/2023] [Indexed: 11/05/2024]
Abstract
Redox metabolism plays a central role in the cellular metabolism network, involves catabolic and anabolic reactions of diverse biomass, and determines the redox state of cells. It can be quantitatively and conveniently measured in living cells and organisms with genetically encoded fluorescent sensors, providing novel insights that cannot be readily acquired via conventional metabolic assays. Here, we review the recent progress on the regulation of leukemogenesis via redox metabolism, especially redox biosensor-based findings. In general, low reactive oxygen species levels and high reductive capacity promote leukemogenesis and chemotherapy resistance in leukemia cells, and acute leukemia cells rewire metabolism of glucose, fatty acids, and some amino acids, together with oxidative phosphorylation, to fuel energy production, support biomass-related synthesis, and survive oxidative stress. In summary, redox metabolism is a potential target for the development of novel therapies for leukemia or beneficial dietary regimens for patients with refractory and relapsed leukemia.
Collapse
Affiliation(s)
- Zhuo Zhang
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Chiqi Chen
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | - Xie Li
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China
| | - Junke Zheng
- Hongqiao International Institute of Medicine, Shanghai Tongren Hospital, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Faculty of Basic Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China; Shanghai Key Laboratory of Reproductive Medicine, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China.
| | - Yuzheng Zhao
- Optogenetics and Synthetic Biology Interdisciplinary Research Center, Shanghai Frontiers Science Center of Optogenetic Techniques for Cell Metabolism, State Key Laboratory of Bioreactor Engineering, School of Pharmacy, East China University of Science and Technology, Shanghai 200237, China; Research Unit of New Techniques for Live-cell Metabolic Imaging, Chinese Academy of Medical Sciences, Beijing 100730, China.
| |
Collapse
|
46
|
Ali S, Tyagi A, Park S, Bae H. Understanding the mechanobiology of phytoacoustics through molecular Lens: Mechanisms and future perspectives. J Adv Res 2024; 65:47-72. [PMID: 38101748 PMCID: PMC11518948 DOI: 10.1016/j.jare.2023.12.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2023] [Revised: 12/11/2023] [Accepted: 12/12/2023] [Indexed: 12/17/2023] Open
Abstract
BACKGROUND How plants emit, perceive, and respond to sound vibrations (SVs) is a long-standing question in the field of plant sensory biology. In recent years, there have been numerous studies on how SVs affect plant morphological, physiological, and biochemical traits related to growth and adaptive responses. For instance, under drought SVs navigate plant roots towards water, activate their defence responses against stressors, and increase nectar sugar in response to pollinator SVs. Also, plants emit SVs during stresses which are informative in terms of ecological and adaptive perspective. However, the molecular mechanisms underlying the SV perception and emission in plants remain largely unknown. Therefore, deciphering the complexity of plant-SV interactions and identifying bonafide receptors and signaling players will be game changers overcoming the roadblocks in phytoacoustics. AIM OF REVIEW The aim of this review is to provide an overview of recent developments in phytoacoustics. We primarily focuss on SV signal perception and transduction with current challenges and future perspectives. KEY SCIENTIFIC CONCEPTS OF REVIEW Timeline breakthroughs in phytoacoustics have constantly shaped our understanding and belief that plants may emit and respond to SVs like other species. However, unlike other plant mechanostimuli, little is known about SV perception and signal transduction. Here, we provide an update on phytoacoustics and its ecological importance. Next, we discuss the role of cell wall receptor-like kinases, mechanosensitive channels, intracellular organelle signaling, and other key players involved in plant-SV receptive pathways that connect them. We also highlight the role of calcium (Ca2+), reactive oxygen species (ROS), hormones, and other emerging signaling molecules in SV signal transduction. Further, we discuss the importance of molecular, biophysical, computational, and live cell imaging tools for decoding the molecular complexity of acoustic signaling in plants. Finally, we summarised the role of SV priming in plants and discuss how SVs could modulate plant defense and growth trade-offs during other stresses.
Collapse
Affiliation(s)
- Sajad Ali
- Department of Biotechnology, Yeungnam University, Gyeongsan Gyeongbuk 38541, Republic of Korea
| | - Anshika Tyagi
- Department of Biotechnology, Yeungnam University, Gyeongsan Gyeongbuk 38541, Republic of Korea
| | - Suvin Park
- Department of Biotechnology, Yeungnam University, Gyeongsan Gyeongbuk 38541, Republic of Korea
| | - Hanhong Bae
- Department of Biotechnology, Yeungnam University, Gyeongsan Gyeongbuk 38541, Republic of Korea.
| |
Collapse
|
47
|
Harjung A, Fracassi A, Devaraj NK. Encoding extracellular modification of artificial cell membranes using engineered self-translocating proteins. Nat Commun 2024; 15:9363. [PMID: 39477950 PMCID: PMC11526174 DOI: 10.1038/s41467-024-53783-4] [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: 12/01/2023] [Accepted: 10/23/2024] [Indexed: 11/02/2024] Open
Abstract
The development of artificial cells has led to fundamental insights into the functional processes of living cells while simultaneously paving the way for transformative applications in biotechnology and medicine. A common method of generating artificial cells is to encapsulate protein expression systems within lipid vesicles. However, to communicate with the external environment, protein translocation across lipid membranes must take place. In living cells, protein transport across membranes is achieved with the aid of complex translocase systems which are difficult to reconstitute into artificial cells. Thus, there is need for simple mechanisms by which proteins can be encoded and expressed inside synthetic compartments yet still be externally displayed. Here we present a genetically encodable membrane functionalization system based on mutants of pore-forming proteins. We modify the membrane translocating loop of α-hemolysin to translocate functional peptides up to 52 amino acids across lipid membranes. Full membrane translocation occurs in the absence of any translocase machinery and the translocated peptides are recognized by specific peptide-binding ligands on the opposing membrane side. Engineered hemolysins can be used for genetically programming artificial cells to display interacting peptide pairs, enabling their assembly into artificial tissue-like structures.
Collapse
Affiliation(s)
- Alexander Harjung
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA
| | - Alessandro Fracassi
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA
| | - Neal K Devaraj
- Department of Chemistry and Biochemistry, University of California, San Diego, CA, USA.
| |
Collapse
|
48
|
Park JY, Park SM, Lee TS, Lee SJ, Kim JY, Oh SJ, Yoon HJ, Kim BS, Moon BS. Innovations in Nuclear Medicine Imaging for Reactive Oxygen Species: Applications and Radiopharmaceuticals. Antioxidants (Basel) 2024; 13:1254. [PMID: 39456507 PMCID: PMC11504556 DOI: 10.3390/antiox13101254] [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: 09/27/2024] [Revised: 10/16/2024] [Accepted: 10/16/2024] [Indexed: 10/28/2024] Open
Abstract
Reactive oxygen species (ROS) are generated during normal cellular energy production and play a critical role in maintaining cellular function. However, excessive ROS can damage cells and tissues, contributing to the development of diseases such as cardiovascular, inflammatory, and neurodegenerative disorders. This review explores the potential of nuclear medicine imaging techniques for detecting ROS and evaluates various radiopharmaceuticals used in these applications. Radiopharmaceuticals, which are drugs labeled with radionuclides, can bind to specific biomarkers, facilitating their identification in vivo using nuclear medicine equipment, i.e., positron emission tomography and single photon emission computed tomography, for diagnostic purposes. This review includes a comprehensive search of PubMed, covering radiopharmaceuticals such as analogs of fluorescent probes and antioxidant vitamin C, and biomarkers targeting mitochondrial complex I or cystine/glutamate transporter.
Collapse
Affiliation(s)
- Joo Yeon Park
- Department of Nuclear Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (J.Y.P.); (S.M.P.)
| | - Sun Mi Park
- Department of Nuclear Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (J.Y.P.); (S.M.P.)
| | - Tae Sup Lee
- Division of RI Applications, Korea Institute Radiological and Medical Sciences, Seoul 01812, Republic of Korea;
| | - Sang Ju Lee
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (S.J.L.); (S.J.O.)
| | - Ji-Young Kim
- Department of Nuclear Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul 07985, Republic of Korea; (J.-Y.K.); (H.-J.Y.)
| | - Seung Jun Oh
- Department of Nuclear Medicine, Asan Medical Center, University of Ulsan College of Medicine, Seoul 05505, Republic of Korea; (S.J.L.); (S.J.O.)
| | - Hai-Jeon Yoon
- Department of Nuclear Medicine, Ewha Womans University Mokdong Hospital, Ewha Womans University College of Medicine, Seoul 07985, Republic of Korea; (J.-Y.K.); (H.-J.Y.)
| | - Bom Sahn Kim
- Department of Nuclear Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (J.Y.P.); (S.M.P.)
| | - Byung Seok Moon
- Department of Nuclear Medicine, Ewha Womans University Seoul Hospital, Ewha Womans University College of Medicine, Seoul 07804, Republic of Korea; (J.Y.P.); (S.M.P.)
| |
Collapse
|
49
|
Chen S, Sun Y, Qin Y, Yang L, Hao Z, Xu Z, Björklund M, Liu W, Hong Z. Dynamic interaction of REEP5-MFN1/2 enables mitochondrial hitchhiking on tubular ER. J Cell Biol 2024; 223:e202304031. [PMID: 39133213 PMCID: PMC11318672 DOI: 10.1083/jcb.202304031] [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: 04/08/2023] [Revised: 02/15/2024] [Accepted: 06/12/2024] [Indexed: 08/13/2024] Open
Abstract
Mitochondrial functions can be regulated by membrane contact sites with the endoplasmic reticulum (ER). These mitochondria-ER contact sites (MERCs) are functionally heterogeneous and maintained by various tethers. Here, we found that REEP5, an ER tubule-shaping protein, interacts with Mitofusins 1/2 to mediate mitochondrial distribution throughout the cytosol by a new transport mechanism, mitochondrial "hitchhiking" with tubular ER on microtubules. REEP5 depletion led to reduced tethering and increased perinuclear localization of mitochondria. Conversely, increasing REEP5 expression facilitated mitochondrial distribution throughout the cytoplasm. Rapamycin-induced irreversible REEP5-MFN1/2 interaction led to mitochondrial hyperfusion, implying that the dynamic release of mitochondria from tethering is necessary for normal mitochondrial distribution and dynamics. Functionally, disruption of MFN2-REEP5 interaction dynamics by forced dimerization or silencing REEP5 modulated the production of mitochondrial reactive oxygen species (ROS). Overall, our results indicate that dynamic REEP5-MFN1/2 interaction mediates cytosolic distribution and connectivity of the mitochondrial network by "hitchhiking" and this process regulates mitochondrial ROS, which is vital for multiple physiological functions.
Collapse
Affiliation(s)
- Shue Chen
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- Nuclear Organization and Gene Expression Section, Laboratory of Biochemistry and Genetics, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, MD, USA
| | - Yang Sun
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Yuling Qin
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Lan Yang
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Zhenhua Hao
- National Center for Children's Health, Beijing Children's Hospital, Capital Medical University, Beijing, China
| | - Zhihao Xu
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
| | - Mikael Björklund
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| | - Wei Liu
- Metabolic Medicine Center, International Institutes of Medicine, the Fourth Affiliated Hospital, School of Medicine, Zhejiang University, Yiwu, China
| | - Zhi Hong
- Department of Neurology, the Second Affiliated Hospital of Zhejiang University, School of Medicine, Zhejiang University, Hangzhou, China
- Centre for Cellular Biology and Signaling, Zhejiang University-University of Edinburgh Institute, Haining, China
- University of Edinburgh Medical School, Biomedical Sciences, College of Medicine & Veterinary Medicine, University of Edinburgh, Edinburgh, UK
| |
Collapse
|
50
|
Smirnov IV, Usatova VS, Berestovoy MA, Fedotov AB, Lanin AA, Belousov VV, Sukhorukov GB. Long-term tracing of individual human neural cells using multiphoton microscopy and photoconvertible polymer capsules. J R Soc Interface 2024; 21:20240497. [PMID: 39471872 PMCID: PMC11521627 DOI: 10.1098/rsif.2024.0497] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/28/2024] [Accepted: 09/18/2024] [Indexed: 11/01/2024] Open
Abstract
The study of human neural cells, their behaviour and migration are important areas of research in the biomedical field, particularly for potential therapeutic applications. The safety of using neural cells in therapy is still a concern due to a lack of information on long-term changes that may occur. While current methods of cell tracing explore gene manipulations, we elaborate approaches to cell marking with no genetic interference. In this study, we present a novel method for labelling and tracking neural cells using cell-impregnatable photoconvertible polyelectrolyte microcapsules. These capsules demonstrated low cytotoxicity with no effect on the differentiation ability of the neural cells, maintained a high level of fluorescent signal and ability for tracing individual neural cells for over 7 days. The capsules modified with rhodamine- and fluorescein-based dyes were demonstrated to undergo photoconversion by both one- and two-photon lasers while being internalized by neural cells. The finding gives the possibility to select individual capsules inside multicellular structures like spheroids and tissues and alternate their fluorescent appearance. Thus, we can track individual cell paths in complex systems. This new method offers a promising alternative for studying neural cells' long-term behaviour and migration in complex systems such as three-dimensional cellular populations.
Collapse
Affiliation(s)
- Ivan V. Smirnov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow121205, Russia
| | - Veronika S. Usatova
- Federal Center for Brain and Neurotechnologies, Federal Medical-Biological Agency, Moscow117997, Russia
| | - Mikhail A. Berestovoy
- Federal Center for Brain and Neurotechnologies, Federal Medical-Biological Agency, Moscow117997, Russia
| | - Andrei B. Fedotov
- Physics Department, Lomonosov Moscow State University, Moscow119992, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow143025, Russia
| | - Aleksandr A. Lanin
- Physics Department, Lomonosov Moscow State University, Moscow119992, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow143025, Russia
| | - Vsevolod V. Belousov
- Federal Center for Brain and Neurotechnologies, Federal Medical-Biological Agency, Moscow117997, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow143025, Russia
- Center for Precision Genome Editing and Genetic Technologies for Biomedicine, Pirogov Russian National Research Medical University, Moscow119334, Russia
- Department of Metabolism and Redox Biology, Shemyakin-Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow117997, Russia
| | - Gleb B. Sukhorukov
- Vladimir Zelman Center for Neurobiology and Brain Rehabilitation, Skolkovo Institute of Science and Technology, Moscow121205, Russia
- Life Improvement by Future Technologies (LIFT) Center, Moscow143025, Russia
- School of Engineering and Materials Science, Queen Mary University of London, LondonE1 4NS, UK
| |
Collapse
|