1
|
Goldschmidt-Clermont PJ, Sevilla BA. Redox and actin, a fascinating story. Redox Biol 2025; 83:103630. [PMID: 40328105 DOI: 10.1016/j.redox.2025.103630] [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: 03/29/2025] [Revised: 04/06/2025] [Accepted: 04/06/2025] [Indexed: 05/08/2025] Open
Abstract
Actin is an extraordinarily complex protein whose functions are essential to cell motility, division, contraction, signaling, transport, tissular structures, DNA repair, and many more cellular activities critical to life for both animals and plants. It is one of the most abundant and conserved proteins and it exists in either a soluble, globular (monomeric, G-actin) or an insoluble, self-assembled (polymerized or filamentous actin, F-actin) conformation as a key component of the cytoskeleton. In the early 1990's little, if anything, was known about the impact of reactive oxygen species (ROS) on the biology of actin except that ROS could disrupt the actin cytoskeleton. Instructively, G-actin is susceptible to alteration by ROS, and thus, purification of G-actin is typically performed in the presence of strong antioxidants (like dithiothreitol) to limit its oxidative degradation. In contrast, F-actin is a more stable conformation and thus actin can be kept relatively intact in purified preparations as filaments at low temperature for extended periods of time. Both G- and F-actin interact with a myriad of intracellular proteins and at least with a couple of extracellular proteins, and these interactions are essential to the many actin functions. This review will show how, over the past 30 years, our understanding of the role of ROS for actin biology has evolved from noxious denaturizing agents to remarkable regulators of the actin cytoskeleton in cells and consequent cellular functions.
Collapse
|
2
|
Oo HK, Galicia-Medina CM, Nishiuchi T, Tanida R, Goto H, Nakano Y, Takeshita Y, Saito Y, Takayama H, Takamura T. Cysteine redoxome landscape in mouse brown adipose tissue under acute cold exposure. iScience 2025; 28:112051. [PMID: 40104075 PMCID: PMC11915156 DOI: 10.1016/j.isci.2025.112051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2024] [Revised: 11/26/2024] [Accepted: 02/13/2025] [Indexed: 03/20/2025] Open
Abstract
Reversible cysteine post-translational modifications serve as a "switch" for protein structure-function dynamics. While reversible cysteine oxidation in uncoupling protein 1 is known to play a role in brown fat thermogenesis, the full cysteine redoxome affected by cold exposure remains unexplored. We established a strategy for comprehensively mapping the cysteine redoxome by pinpointing oxidized and reduced cysteine residues in the brown adipose tissue of mice under room temperature and acute cold exposure. We identified over 1,000 labeled cysteine residues under room and cold temperatures. Cold exposure shifted the cysteine redox states toward oxidation. Cold-sensitive reactive cysteine residues were enriched in biological processes and molecular functions associated with thermogenesis pathways. The presence of proximal positively charged and negatively charged amino acids determined the highly reactive and non-reactive cysteine residues, respectively, under cold exposure. Our findings broaden the landscape of cold-sensitive proteome and identify potential therapeutic targets to fine-tune thermogenesis.
Collapse
Affiliation(s)
- Hein Ko Oo
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Cynthia M Galicia-Medina
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Takumi Nishiuchi
- Division of Natural System, Kanazawa University Graduate School of Natural Science and Technology, Kanazawa, Japan
| | - Ryota Tanida
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Hisanori Goto
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yujiro Nakano
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yumie Takeshita
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Yoshiro Saito
- Laboratory of Molecular Biology and Metabolism, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Hiroaki Takayama
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
- Life Sciences Division, Engineering and Technology Department, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| | - Toshinari Takamura
- Department of Endocrinology and Metabolism, Kanazawa University Graduate School of Medical Sciences, Kanazawa, Japan
| |
Collapse
|
3
|
Dinda R, Garribba E, Sanna D, Crans DC, Costa Pessoa J. Hydrolysis, Ligand Exchange, and Redox Properties of Vanadium Compounds: Implications of Solution Transformation on Biological, Therapeutic, and Environmental Applications. Chem Rev 2025; 125:1468-1603. [PMID: 39818783 DOI: 10.1021/acs.chemrev.4c00475] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2025]
Abstract
Vanadium is a transition metal with important industrial, technological, biological, and biomedical applications widespread in the environment and in living beings. The different reactions that vanadium compounds (VCs) undergo in the presence of proteins, nucleic acids, lipids and metabolites under mild physiological conditions are reviewed. In the environment vanadium is present naturally or through anthropogenic sources, the latter having an environmental impact caused by the dispersion of VCs in the atmosphere and aquifers. Vanadium has a versatile chemistry with interconvertible oxidation states, variable coordination number and geometry, and ability to form polyoxidovanadates with various nuclearity and structures. If a VC is added to a water-containing environment it can undergo hydrolysis, ligand-exchange, redox, and other types of changes, determined by the conditions and speciation chemistry of vanadium. Importantly, the solution is likely to differ from the VC introduced into the system and varies with concentration. Here, vanadium redox, hydrolytic and ligand-exchange chemical reactions, the influence of pH, concentration, salt, specific solutes, biomolecules, and VCs on the speciation are described. One of our goals with this work is highlight the need for assessment of the VC speciation, so that beneficial or toxic species might be identified and mechanisms of action be elucidated.
Collapse
Affiliation(s)
- Rupam Dinda
- Department of Chemistry, National Institute of Technology, Rourkela, 769008 Odisha, India
| | - Eugenio Garribba
- Dipartimento di Medicina, Chirurgia e Farmacia, Università di Sassari, Viale San Pietro, I-07100 Sassari, Italy
| | - Daniele Sanna
- Istituto di Chimica Biomolecolare, Consiglio Nazionale delle Ricerche, Trav. La Crucca 3, I-07040 Sassari, Italy
| | - Debbie C Crans
- Department Chemistry and Cell and Molecular Biology, Colorado State University, Fort Collins, Colorado 80523, United States
| | - João Costa Pessoa
- Centro de Química Estrutural and Departamento de Engenharia Química, Institute of Molecular Sciences, Instituto Superior Técnico, Av. Rovisco Pais, 1049-001 Lisboa, Portugal
| |
Collapse
|
4
|
Quilaqueo-Millaqueo N, Brown-Brown DA, Vidal-Vidal JA, Niechi I. NOX proteins and ROS generation: role in invadopodia formation and cancer cell invasion. Biol Res 2024; 57:98. [PMID: 39696702 DOI: 10.1186/s40659-024-00577-z] [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: 07/18/2024] [Accepted: 12/04/2024] [Indexed: 12/20/2024] Open
Abstract
NADPH oxidases (NOX) are membrane-bound proteins involved in the localized generation of reactive oxygen species (ROS) at the cellular surface. In cancer, these highly reactive molecules primarily originate in mitochondria and via NOX, playing a crucial role in regulating fundamental cellular processes such as cell survival, angiogenesis, migration, invasion, and metastasis. The NOX protein family comprises seven members (NOX1-5 and DUOX1-2), each sharing a catalytic domain and an intracellular dehydrogenase site. NOX-derived ROS promote invadopodia formation, aberrant tyrosine kinase activation, and upregulation of matrix metalloproteinases (MMPs). Specifically, NOX5 modulates adhesion, motility, and proteolytic activation, while NOX1 likely contributes to invadopodia formation and adhesive capacity. NOX2 and NOX4 are implicated in regulating the invasive phenotype, expression of MMPs and EMT markers. DUOX1-2 participate in epithelial-mesenchymal transition (EMT), crucial for invasive phenotype development. Soluble molecules such as TGF-β and EGF modulate NOX protein activation, enhancing cell invasion through localized ROS production. This review focuses on elucidating the specific role of NOX proteins in regulating signaling pathways promoting cancer cell spread, particularly EMT, invadopodia formation and invasive capacity.
Collapse
Affiliation(s)
- Nelson Quilaqueo-Millaqueo
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - David A Brown-Brown
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - Jetzabel A Vidal-Vidal
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile
| | - Ignacio Niechi
- Instituto de Bioquímica y Microbiología, Facultad de Ciencias, Universidad Austral de Chile, 5090000, Valdivia, Chile.
| |
Collapse
|
5
|
Boiero Sanders M, Oosterheert W, Hofnagel O, Bieling P, Raunser S. Phalloidin and DNase I-bound F-actin pointed end structures reveal principles of filament stabilization and disassembly. Nat Commun 2024; 15:7969. [PMID: 39261469 PMCID: PMC11390976 DOI: 10.1038/s41467-024-52251-3] [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: 06/08/2024] [Accepted: 08/30/2024] [Indexed: 09/13/2024] Open
Abstract
Actin filament turnover involves subunits binding to and dissociating from the filament ends, with the pointed end being the primary site of filament disassembly. Several molecules modulate filament turnover, but the underlying mechanisms remain incompletely understood. Here, we present three cryo-EM structures of the F-actin pointed end in the presence and absence of phalloidin or DNase I. The two terminal subunits at the undecorated pointed end adopt a twisted conformation. Phalloidin can still bind and bridge these subunits, inducing a conformational shift to a flattened, F-actin-like state. This explains how phalloidin prevents depolymerization at the pointed end. Interestingly, two DNase I molecules simultaneously bind to the phalloidin-stabilized pointed end. In the absence of phalloidin, DNase I binding would disrupt the terminal actin subunit packing, resulting in filament disassembly. Our findings uncover molecular principles of pointed end regulation and provide structural insights into the kinetic asymmetry between the actin filament ends.
Collapse
Affiliation(s)
- Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227, Dortmund, Germany.
| |
Collapse
|
6
|
Hegde S, Modi S, Deihl EW, Glomb OV, Yogev S, Hoerndli FJ, Koushika SP. Axonal mitochondria regulate gentle touch response through control of axonal actin dynamics. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.13.607780. [PMID: 39185223 PMCID: PMC11343141 DOI: 10.1101/2024.08.13.607780] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 08/27/2024]
Abstract
Actin in neuronal processes is both stable and dynamic. The origin & functional roles of the different pools of actin is not well understood. We find that mutants that lack mitochondria, ric-7 and mtx-2; miro-1, in neuronal processes also lack dynamic actin. Mitochondria can regulate actin dynamics upto a distance ~80 μm along the neuronal process. Absence of axonal mitochondria and dynamic actin does not markedly alter the Spectrin Membrane Periodic Skeleton (MPS) in touch receptor neurons (TRNs). Restoring mitochondria inTRNs cell autonomously restores dynamic actin in a sod-2 dependent manner. We find that dynamic actin is necessary and sufficient for the localization of gap junction proteins in the TRNs and for the C. elegans gentle touch response. We identify an in vivo mechanism by which axonal mitochondria locally facilitate actin dynamics through reactive oxygen species that we show is necessary for electrical synapses & behaviour.
Collapse
Affiliation(s)
- Sneha Hegde
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Souvik Modi
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| | - Ennis W. Deihl
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Oliver Vinzenz Glomb
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
- Current address: Institute of Clinical Anatomy and Cell Analysis, University of Tübingen, 72074 Tübingen, Germany
| | - Shaul Yogev
- Yale University, Boyer Center for Molecular Medicine, 295 Congress Ave, New Haven, CT 06510
| | - Frederic J. Hoerndli
- Colorado State University, Anatomy and Zoology W309, 1617 Campus Delivery, Fort Collins, 80523 Colorado
| | - Sandhya P. Koushika
- Tata Institute of Fundamental Research, Homi Bhabha Road, Navy Nagar, Colaba, Mumbai-400005, India
| |
Collapse
|
7
|
Yeo XY, Kwon S, Rinai KR, Lee S, Jung S, Park R. A Consolidated Understanding of the Contribution of Redox Dysregulation in the Development of Hearing Impairment. Antioxidants (Basel) 2024; 13:598. [PMID: 38790703 PMCID: PMC11118506 DOI: 10.3390/antiox13050598] [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: 02/22/2024] [Revised: 04/26/2024] [Accepted: 05/09/2024] [Indexed: 05/26/2024] Open
Abstract
The etiology of hearing impairment is multifactorial, with contributions from both genetic and environmental factors. Although genetic studies have yielded valuable insights into the development and function of the auditory system, the contribution of gene products and their interaction with alternate environmental factors for the maintenance and development of auditory function requires further elaboration. In this review, we provide an overview of the current knowledge on the role of redox dysregulation as the converging factor between genetic and environmental factor-dependent development of hearing loss, with a focus on understanding the interaction of oxidative stress with the physical components of the peripheral auditory system in auditory disfunction. The potential involvement of molecular factors linked to auditory function in driving redox imbalance is an important promoter of the development of hearing loss over time.
Collapse
Affiliation(s)
- Xin Yi Yeo
- Department of Psychological Medicine, Yong Loo Lin School of Medicine, National University of Singapore, Singapore 119228, Singapore;
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Soohyun Kwon
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
- Department of BioNanotechnology, Gachon University, Seongnam 13120, Republic of Korea
| | - Kimberley R. Rinai
- Department of Life Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Sungsu Lee
- Department of Otolaryngology-Head and Neck Surgery, Chonnam National University Hospital and Medical School, Gwangju 61469, Republic of Korea;
| | - Sangyong Jung
- Department of Medical Science, College of Medicine, CHA University, Seongnam 13488, Republic of Korea;
| | - Raekil Park
- Department of Biomedical Science and Engineering, Gwangju Institute of Science & Technology (GIST), Gwangju 61005, Republic of Korea
| |
Collapse
|
8
|
Oosterheert W, Boiero Sanders M, Funk J, Prumbaum D, Raunser S, Bieling P. Molecular mechanism of actin filament elongation by formins. Science 2024; 384:eadn9560. [PMID: 38603491 DOI: 10.1126/science.adn9560] [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: 01/09/2024] [Accepted: 03/05/2024] [Indexed: 04/13/2024]
Abstract
Formins control the assembly of actin filaments (F-actin) that drive cell morphogenesis and motility in eukaryotes. However, their molecular interaction with F-actin and their mechanism of action remain unclear. In this work, we present high-resolution cryo-electron microscopy structures of F-actin barbed ends bound by three distinct formins, revealing a common asymmetric formin conformation imposed by the filament. Formation of new intersubunit contacts during actin polymerization sterically displaces formin and triggers its translocation. This "undock-and-lock" mechanism explains how actin-filament growth is coordinated with formin movement. Filament elongation speeds are controlled by the positioning and stability of actin-formin interfaces, which distinguish fast and slow formins. Furthermore, we provide a structure of the actin-formin-profilin ring complex, which resolves how profilin is rapidly released from the barbed end during filament elongation.
Collapse
Affiliation(s)
- Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Johanna Funk
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Daniel Prumbaum
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, 44227 Dortmund, Germany
| |
Collapse
|
9
|
Li Y, Zhang J, Li F. Gastrodin improves osteoblast function and adhesion to titanium surface in a high glucose environment. Biochem Biophys Rep 2024; 37:101623. [PMID: 38225991 PMCID: PMC10788200 DOI: 10.1016/j.bbrep.2023.101623] [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: 08/29/2023] [Revised: 11/27/2023] [Accepted: 12/18/2023] [Indexed: 01/17/2024] Open
Abstract
Objective To investigate the effects of gastrodin on the biological behavior of osteoblasts and osseointegration on the surface of the titanium plate in a high glucose environment, and to explore the possible regulatory mechanisms involved. Methods A high glucose-induced oxidative damage model of MC3T3-E1 cells was established in vitro to observe the effects of gastrodin on cellular oxidative stress, cell viability, osteogenic differentiation, mineralization, migration, and adhesion ability on the titanium surface. Results High glucose environment can cause oxidative stress damage to MC3T3-E1 cells, leading to a decrease in cell viability, osteogenesis, migration, adhesion and other functions. Gastrodin can upregulate the expression of antioxidant enzymes (Nrf2 and HO-1) and osteogenic differentiation related proteins (RUNX2 and BMP2) in MC3T3-E1 cells in high glucose environment, thereby inhibiting the excessive production of intracellular reactive oxygen species (ROS), reversing the decrease in cell viability, and improving the osteogenic differentiation and mineralization ability of osteoblasts. And gastrodin alleviated the decline in cell migration ability, improved the morphology of the cytoskeleton and increased the adhesion ability of osteoblasts on the surface of titanium plates in high glucose environment. However, gastrodin itself did not affect the cell viability, osteogenic differentiation and mineralization ability of osteoblasts in normal environment. Conclusions Gastrodin may protect MC3T3-E1 cells osteogenesis and osseointegration on the surface of the titanium plate in vitro by upregulating antioxidant enzymes expression, and attenuating high glucose-induced oxidative stress. Therefore, gastrodin may be a potential drug to address the problem of poor implant osseointegration in patients with diabetes.
Collapse
Affiliation(s)
- Yi Li
- Shanxi Medical University School of Stomatology, Taiyuan 030000, China
| | - Jingyi Zhang
- Shanxi Medical University School of Stomatology, Taiyuan 030000, China
| | - Fenglan Li
- Department of Prosthodontics, Shanxi Provincial People's Hospital, Shanxi Medical University, Taiyuan 030000, China
| |
Collapse
|
10
|
Chan E, Dirk BS, Honda T, Stathopulos PB, Dikeakos JD, Di Guglielmo GM. Acetylenic tricyclic bis-(cyano enone) interacts with Cys 374 of actin, a residue necessary for stress fiber formation and cell migration. BIOCHIMICA ET BIOPHYSICA ACTA. MOLECULAR CELL RESEARCH 2024; 1871:119629. [PMID: 37981034 DOI: 10.1016/j.bbamcr.2023.119629] [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: 08/22/2023] [Revised: 10/31/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023]
Abstract
The migratory and invasive potential of tumour cells relies on the actin cytoskeleton. We previously demonstrated that the tricyclic compound, TBE-31, inhibits actin polymerization and here we further examine the precise interaction between TBE-31 and actin. We demonstrate that iodoacetamide, a cysteine (Cys) alkylating agent, interferes with the ability of TBE-31 to interact with actin. In addition, in silico analysis identified Cys 217, Cys 272, Cys 285 and Cys 374 as potential binding sites for TBE-31. Using mass spectrometry analysis, we determined that TBE-31 associates with actin with a stoichiometric ratio of 1:1. We mutated the identified cysteines of actin to alanine and performed a pull-down analysis with a biotin labeled TBE-31 and demonstrated that by mutating Cys 374 to alanine the association between TBE-31 and actin was significantly reduced, suggesting that TBE-31 binds to Cys 374. A characterization of the NIH3T3 cells overexpressing eGFP-actin-C374A showed reduced stress fiber formation, suggesting Cys 374 is necessary for efficient incorporation into filamentous actin. Furthermore, migration of eGFP-Actin-WT expressing cells were observed to be inhibited by TBE-31, however fewer eGFP-Actin-C374A expressing cells were observed to migrate compared to the cells expressing eGFP-Actin-WT in the presence or absence of TBE-31. Taken together, our results suggest that TBE-31 binds to Cys 374 of actin to inhibit actin stress fiber formation and may potentially be a mechanism through which TBE-31 inhibits cell migration.
Collapse
Affiliation(s)
- Eddie Chan
- Western University, Department of Physiology and Pharmacology, London N6A5C1, Canada
| | - Brennan S Dirk
- Western University, Department of Microbiology and Immunology, London N6A5C1, Canada
| | - Tadashi Honda
- Stony Brook University, Department of Chemistry, Institute of Chemical Biology & Drug Discovery, Stony Brook 11790-3400, USA
| | - Peter B Stathopulos
- Western University, Department of Physiology and Pharmacology, London N6A5C1, Canada
| | - Jimmy D Dikeakos
- Western University, Department of Microbiology and Immunology, London N6A5C1, Canada
| | - Gianni M Di Guglielmo
- Western University, Department of Physiology and Pharmacology, London N6A5C1, Canada.
| |
Collapse
|
11
|
Kast DJ, Jansen S. Purification of modified mammalian actin isoforms for in vitro reconstitution assays. Eur J Cell Biol 2023; 102:151363. [PMID: 37778219 PMCID: PMC10872616 DOI: 10.1016/j.ejcb.2023.151363] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 08/19/2023] [Accepted: 09/26/2023] [Indexed: 10/03/2023] Open
Abstract
In vitro reconstitution assays using purified actin have greatly improved our understanding of cytoskeletal dynamics and their regulation by actin-binding proteins. However, early purification methods consisted of harsh conditions to obtain pure actin and often did not include correct maturation and obligate modification of the isolated actin monomers. Novel insights into the folding requirements and N-terminal processing of actin as well as a better understanding of the interaction of actin with monomer sequestering proteins such as DNaseI, profilin and gelsolin, led to the development of more gentle approaches to obtain pure recombinant actin isoforms with known obligate modifications. This review summarizes the approaches that can be employed to isolate natively folded endogenous and recombinant actin from tissues and cells. We further emphasize the use and limitations of each method and describe how these methods can be implemented to study actin PTMs, disease-related actin mutations and novel actin-like proteins.
Collapse
Affiliation(s)
- David J Kast
- Department of Cell Biology and Physiology, Washington University in St. Louis, Saint Louis, MO, 63110, United States.
| | - Silvia Jansen
- Department of Cell Biology and Physiology, Washington University in St. Louis, Saint Louis, MO, 63110, United States.
| |
Collapse
|
12
|
Oosterheert W, Blanc FEC, Roy A, Belyy A, Sanders MB, Hofnagel O, Hummer G, Bieling P, Raunser S. Molecular mechanisms of inorganic-phosphate release from the core and barbed end of actin filaments. Nat Struct Mol Biol 2023; 30:1774-1785. [PMID: 37749275 PMCID: PMC10643162 DOI: 10.1038/s41594-023-01101-9] [Citation(s) in RCA: 17] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2023] [Accepted: 08/18/2023] [Indexed: 09/27/2023]
Abstract
The release of inorganic phosphate (Pi) from actin filaments constitutes a key step in their regulated turnover, which is fundamental to many cellular functions. The mechanisms underlying Pi release from the core and barbed end of actin filaments remain unclear. Here, using human and bovine actin isoforms, we combine cryo-EM with molecular-dynamics simulations and in vitro reconstitution to demonstrate how actin releases Pi through a 'molecular backdoor'. While constantly open at the barbed end, the backdoor is predominantly closed in filament-core subunits and opens only transiently through concerted amino acid rearrangements. This explains why Pi escapes rapidly from the filament end but slowly from internal subunits. In a nemaline-myopathy-associated actin variant, the backdoor is predominantly open in filament-core subunits, resulting in accelerated Pi release and filaments with drastically shortened ADP-Pi caps. Our results provide the molecular basis for Pi release from actin and exemplify how a disease-linked mutation distorts the nucleotide-state distribution and atomic structure of the filament.
Collapse
Affiliation(s)
- Wout Oosterheert
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Florian E C Blanc
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany
| | - Ankit Roy
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Micaela Boiero Sanders
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Oliver Hofnagel
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany
| | - Gerhard Hummer
- Department of Theoretical Biophysics, Max Planck Institute of Biophysics, Frankfurt am Main, Germany.
- Institute for Biophysics, Goethe University, Frankfurt am Main, Germany.
| | - Peter Bieling
- Department of Systemic Cell Biology, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Dortmund, Germany.
| |
Collapse
|
13
|
Lopez AJ, Andreadaki M, Vahokoski J, Deligianni E, Calder LJ, Camerini S, Freitag A, Bergmann U, Rosenthal PB, Sidén-Kiamos I, Kursula I. Structure and function of Plasmodium actin II in the parasite mosquito stages. PLoS Pathog 2023; 19:e1011174. [PMID: 36877739 PMCID: PMC10019781 DOI: 10.1371/journal.ppat.1011174] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2022] [Revised: 03/16/2023] [Accepted: 02/03/2023] [Indexed: 03/07/2023] Open
Abstract
Actins are filament-forming, highly-conserved proteins in eukaryotes. They are involved in essential processes in the cytoplasm and also have nuclear functions. Malaria parasites (Plasmodium spp.) have two actin isoforms that differ from each other and from canonical actins in structure and filament-forming properties. Actin I has an essential role in motility and is fairly well characterized. The structure and function of actin II are not as well understood, but mutational analyses have revealed two essential functions in male gametogenesis and in the oocyst. Here, we present expression analysis, high-resolution filament structures, and biochemical characterization of Plasmodium actin II. We confirm expression in male gametocytes and zygotes and show that actin II is associated with the nucleus in both stages in filament-like structures. Unlike actin I, actin II readily forms long filaments in vitro, and near-atomic structures in the presence or absence of jasplakinolide reveal very similar structures. Small but significant differences compared to other actins in the openness and twist, the active site, the D-loop, and the plug region contribute to filament stability. The function of actin II was investigated through mutational analysis, suggesting that long and stable filaments are necessary for male gametogenesis, while a second function in the oocyst stage also requires fine-tuned regulation by methylation of histidine 73. Actin II polymerizes via the classical nucleation-elongation mechanism and has a critical concentration of ~0.1 μM at the steady-state, like actin I and canonical actins. Similarly to actin I, dimers are a stable form of actin II at equilibrium.
Collapse
Affiliation(s)
- Andrea J. Lopez
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Maria Andreadaki
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Juha Vahokoski
- Department of Biomedicine, University of Bergen, Bergen, Norway
| | - Elena Deligianni
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Lesley J. Calder
- Structural Biology of Cells and Viruses Laboratory, Francis Crick Institute, London, United Kingdom
| | | | - Anika Freitag
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
| | - Ulrich Bergmann
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
| | - Peter B. Rosenthal
- Structural Biology of Cells and Viruses Laboratory, Francis Crick Institute, London, United Kingdom
| | - Inga Sidén-Kiamos
- Institute of Molecular Biology and Biotechnology, Foundation for Research and Technology-Hellas, Heraklion, Greece
- * E-mail: (ISK); (IK)
| | - Inari Kursula
- Department of Biomedicine, University of Bergen, Bergen, Norway
- Faculty of Biochemistry and Molecular Medicine, University of Oulu, Oulu, Finland
- * E-mail: (ISK); (IK)
| |
Collapse
|
14
|
Onda Y, Okino T. Thiol-disulfide oxidoreductase PDI1;1 regulates actin structures in Oryza sativa root cells. FEBS Lett 2022; 596:3015-3023. [PMID: 35781879 DOI: 10.1002/1873-3468.14445] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2022] [Revised: 06/20/2022] [Accepted: 06/21/2022] [Indexed: 12/14/2022]
Abstract
The polarized and dynamic actin cytoskeleton is essential for root cell growth. Here, we report the key role of thiol-disulfide oxidoreductase PDI1;1 in actin structures. Microscopic analyses revealed that after Oryza sativa roots were exposed to H2 O2 , both actin and PDI1;1 were depolarized and arranged in a meshwork. In H2 O2 -exposed cells, actin formed intermolecularly disulfide-bonded high-molecular-weight structures, which were thiol-trapped by PDI1;1. Recombinant PDI1;1 exhibited the ability to recognize actin in an in vitro binding assay. During recovery from H2 O2 exposure, the amount of disulfide-bonded high-molecular-weight structures of actin decreased over time, but deficiency of PDI1;1 inhibited the decrease. These results suggest a PDI1;1-dependent pathway that reduces disulfide bonds in high-molecular-weight structures of actin, thus promoting their degradation.
Collapse
Affiliation(s)
- Yayoi Onda
- Graduate School of Agriculture, Ehime University, Matsuyama, Japan
| | - Tomoya Okino
- Faculty of Agriculture, Ehime University, Matsuyama, Japan
| |
Collapse
|
15
|
Li Y, Li F. Mechanism and Prospect of Gastrodin in Osteoporosis, Bone Regeneration, and Osseointegration. Pharmaceuticals (Basel) 2022; 15:1432. [PMID: 36422561 PMCID: PMC9698149 DOI: 10.3390/ph15111432] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2022] [Revised: 11/08/2022] [Accepted: 11/16/2022] [Indexed: 11/14/2023] Open
Abstract
Gastrodin, a traditional Chinese medicine ingredient, is widely used to treat vascular and neurological diseases. However, recently, an increasing number of studies have shown that gastrodin has anti-osteoporosis effects, and its mechanisms of action include its antioxidant effect, anti-inflammatory effect, and anti-apoptotic effect. In addition, gastrodin has many unique advantages in promoting bone healing in tissue engineering, such as inducing high hydrophilicity in the material surface, its anti-inflammatory effect, and pro-vascular regeneration. Therefore, this paper summarized the effects and mechanisms of gastrodin on osteoporosis and bone regeneration in the current research. Here we propose an assumption that the use of gastrodin in the surface loading of oral implants may greatly promote the osseointegration of implants and increase the success rate of implants. In addition, we speculated on the potential mechanisms of gastrodin against osteoporosis, by affecting actin filament polymerization, renin-angiotensin system (RAS) and ferroptosis, and proposed that the potential combination of gastrodin with Mg2+, angiotensin type 2 receptor blockers or artemisinin may greatly inhibit osteoporosis. The purpose of this review is to provide a reference for more in-depth research and application of gastrodin in the treatment of osteoporosis and implant osseointegration in the future.
Collapse
Affiliation(s)
| | - Fenglan Li
- Department of Prosthodontics, Shanxi Provincial People’s Hospital, Shanxi Medical University, Taiyuan 030000, China
| |
Collapse
|
16
|
Scirè A, Cianfruglia L, Minnelli C, Romaldi B, Laudadio E, Galeazzi R, Antognelli C, Armeni T. Glyoxalase 2: Towards a Broader View of the Second Player of the Glyoxalase System. Antioxidants (Basel) 2022; 11:2131. [PMID: 36358501 PMCID: PMC9686547 DOI: 10.3390/antiox11112131] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2022] [Revised: 10/25/2022] [Accepted: 10/26/2022] [Indexed: 07/30/2023] Open
Abstract
Glyoxalase 2 is a mitochondrial and cytoplasmic protein belonging to the metallo-β-lactamase family encoded by the hydroxyacylglutathione hydrolase (HAGH) gene. This enzyme is the second enzyme of the glyoxalase system that is responsible for detoxification of the α-ketothaldehyde methylglyoxal in cells. The two enzymes glyoxalase 1 (Glo1) and glyoxalase 2 (Glo2) form the complete glyoxalase pathway, which utilizes glutathione as cofactor in eukaryotic cells. The importance of Glo2 is highlighted by its ubiquitous distribution in prokaryotic and eukaryotic organisms. Its function in the system has been well defined, but in recent years, additional roles are emerging, especially those related to oxidative stress. This review focuses on Glo2 by considering its genetics, molecular and structural properties, its involvement in post-translational modifications and its interaction with specific metabolic pathways. The purpose of this review is to focus attention on an enzyme that, from the most recent studies, appears to play a role in multiple regulatory pathways that may be important in certain diseases such as cancer or oxidative stress-related diseases.
Collapse
Affiliation(s)
- Andrea Scirè
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Laura Cianfruglia
- Department of Clinical Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Cristina Minnelli
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Brenda Romaldi
- Department of Clinical Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| | - Emiliano Laudadio
- Department of Science and Engineering of Materials, Environment and Urban Planning, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Roberta Galeazzi
- Department of Life and Environmental Sciences, Polytechnic University of Marche, 60131 Ancona, Italy
| | - Cinzia Antognelli
- Department of Medicine and Surgery, University of Perugia, 06132 Perugia, Italy
| | - Tatiana Armeni
- Department of Clinical Sciences, Polytechnic University of Marche, 60126 Ancona, Italy
| |
Collapse
|
17
|
Meng Z, Li Z, Xie M, Yu H, Jiang L, Yao X. TM9SF4 is an F-actin disassembly factor that promotes tumor progression and metastasis. Nat Commun 2022; 13:5728. [PMID: 36175399 PMCID: PMC9522921 DOI: 10.1038/s41467-022-33276-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2020] [Accepted: 09/09/2022] [Indexed: 11/29/2022] Open
Abstract
F-actin dynamics is crucial for many fundamental properties of cancer cells, from cell-substrate adhesion to migration, invasion and metastasis. However, the regulatory mechanisms of actin dynamics are still incompletely understood. In this study, we demonstrate the function of a protein named TM9SF4 in regulating actin dynamics and controlling cancer cell motility and metastasis. We show that an N-terminal fragment (NTF) cleaved from TM9SF4 can directly bind to F-actin to induce actin oxidation at Cys374, consequently enhancing cofilin-mediated F-actin disassembly. Knockdown of TM9SF4 reduces cell migration and invasion in ovarian cancer cells A2780, SKOV3 and several high grade serous ovarian cancer lines (HGSOCs). In vivo, knockdown of TM9SF4 completely abolishes the tumor growth and metastasis in athymic nude mice. These data provide mechanistic insights into TM9SF4-mediated regulation of actin dynamics in ovarian cancer cells. F-actin dynamics influence cancer cell motility. Here the authors show that TM9SF4 facilitates the cofilin-induced disassembly of F-actin to promote cancer cell migration and metastasis.
Collapse
Affiliation(s)
- Zhaoyue Meng
- School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China
| | - Zhichao Li
- School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China.,Key Laboratory of Medical Reprogramming Technology, Shenzhen Second People's Hospital, The First Affiliated Hospital of Shenzhen University, Shenzhen, China
| | - Mingxu Xie
- School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Hongyan Yu
- School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China
| | - Liwen Jiang
- Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| | - Xiaoqiang Yao
- School of Biomedical Sciences and Li Ka Shing Institute of Health Sciences, Faculty of Medicine, The Chinese University of Hong Kong, Hong Kong, China. .,Centre for Cell & Developmental Biology and State Key Laboratory of Agrobiotechnology, School of Life Sciences, The Chinese University of Hong Kong, Hong Kong, China.
| |
Collapse
|
18
|
Gao L, Jin N, Ye Z, Ma T, Huang Y, Li H, Du J, Li Z. A possible connection between reactive oxygen species and the unfolded protein response in lens development: From insight to foresight. Front Cell Dev Biol 2022; 10:820949. [PMID: 36211466 PMCID: PMC9535091 DOI: 10.3389/fcell.2022.820949] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2021] [Accepted: 08/31/2022] [Indexed: 11/28/2022] Open
Abstract
The lens is a relatively special and simple organ. It has become an ideal model to study the common developmental characteristics among different organic systems. Lens development is a complex process influenced by numerous factors, including signals from the intracellular and extracellular environment. Reactive oxygen species (ROS) are a group of highly reactive and oxygen-containing molecules that can cause endoplasmic reticulum stress in lens cells. As an adaptive response to ER stress, lens cells initiate the unfolded protein response (UPR) to maintain normal protein synthesis by selectively increasing/decreasing protein synthesis and increasing the degradation of misfolded proteins. Generally, the UPR signaling pathways have been well characterized in the context of many pathological conditions. However, recent studies have also confirmed that all three UPR signaling pathways participate in a variety of developmental processes, including those of the lens. In this review, we first briefly summarize the three stages of lens development and present the basic profiles of ROS and the UPR. We then discuss the interconnections between lens development and these two mechanisms. Additionally, the potential adoption of human pluripotent stem-cell-based lentoids in lens development research is proposed to provide a novel perspective on future developmental studies.
Collapse
Affiliation(s)
- Lixiong Gao
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Ni Jin
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- Department of Endocrinology, The Second Medical Center and National Clinical Research Center for Geriatric Diseases, The Chinese PLA General Hospital, Beijing, China
| | - Zi Ye
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Tianju Ma
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Yang Huang
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Hongyu Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Jinlin Du
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
| | - Zhaohui Li
- Senior Department of Ophthalmology, The Third Medical Center of Chinese PLA General Hospital, Beijing, China
- *Correspondence: Zhaohui Li,
| |
Collapse
|
19
|
Rouyère C, Serrano T, Frémont S, Echard A. Oxidation and reduction of actin: Origin, impact in vitro and functional consequences in vivo. Eur J Cell Biol 2022; 101:151249. [PMID: 35716426 DOI: 10.1016/j.ejcb.2022.151249] [Citation(s) in RCA: 44] [Impact Index Per Article: 14.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2022] [Revised: 05/13/2022] [Accepted: 06/06/2022] [Indexed: 11/15/2022] Open
Abstract
Actin is among the most abundant proteins in eukaryotic cells and assembles into dynamic filamentous networks regulated by many actin binding proteins. The actin cytoskeleton must be finely tuned, both in space and time, to fulfill key cellular functions such as cell division, cell shape changes, phagocytosis and cell migration. While actin oxidation by reactive oxygen species (ROS) at non-physiological levels are known for long to impact on actin polymerization and on the cellular actin cytoskeleton, growing evidence shows that direct and reversible oxidation/reduction of specific actin amino acids plays an important and physiological role in regulating the actin cytoskeleton. In this review, we describe which actin amino acid residues can be selectively oxidized and reduced in many different ways (e.g. disulfide bond formation, glutathionylation, carbonylation, nitration, nitrosylation and other oxidations), the cellular enzymes at the origin of these post-translational modifications, and the impact of actin redox modifications both in vitro and in vivo. We show that the regulated balance of oxidation and reduction of key actin amino acid residues contributes to the control of actin filament polymerization and disassembly at the subcellular scale and highlight how improper redox modifications of actin can lead to pathological conditions.
Collapse
Affiliation(s)
- Clémentine Rouyère
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France; Sorbonne Université, Collège Doctoral, F-75005 Paris, France
| | - Thomas Serrano
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
| | - Stéphane Frémont
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France
| | - Arnaud Echard
- Institut Pasteur, Université Paris Cité, CNRS UMR3691, Membrane Traffic and Cell Division Unit, 25-28 rue du Dr Roux, F-75015 Paris, France.
| |
Collapse
|
20
|
Belyy A, Merino F, Mechold U, Raunser S. Mechanism of actin-dependent activation of nucleotidyl cyclase toxins from bacterial human pathogens. Nat Commun 2021; 12:6628. [PMID: 34785651 PMCID: PMC8595890 DOI: 10.1038/s41467-021-26889-2] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2021] [Accepted: 10/25/2021] [Indexed: 12/19/2022] Open
Abstract
Bacterial human pathogens secrete initially inactive nucleotidyl cyclases that become potent enzymes by binding to actin inside eukaryotic host cells. The underlying molecular mechanism of this activation is, however, unclear. Here, we report structures of ExoY from Pseudomonas aeruginosa and Vibrio vulnificus bound to their corresponding activators F-actin and profilin-G-actin. The structures reveal that in contrast to the apo-state, two flexible regions become ordered and interact strongly with actin. The specific stabilization of these regions results in an allosteric stabilization of the nucleotide binding pocket and thereby to an activation of the enzyme. Differences in the sequence and conformation of the actin-binding regions are responsible for the selective binding to either F- or G-actin. Other nucleotidyl cyclase toxins that bind to calmodulin rather than actin undergo a similar disordered-to-ordered transition during activation, suggesting that the allosteric activation-by-stabilization mechanism of ExoY is conserved in these enzymes, albeit the different activator.
Collapse
Affiliation(s)
- Alexander Belyy
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
| | - Felipe Merino
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany
- Department of Protein Evolution, Max Planck Institute for Developmental Biology, Max-Planck-Ring 5, 72076, Tübingen, Germany
| | - Undine Mechold
- Unité de Biochimie des Interactions Macromoléculaires, Département de Biologie Structurale et Chimie, Institut Pasteur, CNRS UMR 3528, Paris, France
| | - Stefan Raunser
- Department of Structural Biochemistry, Max Planck Institute of Molecular Physiology, Otto-Hahn-Str. 11, 44227, Dortmund, Germany.
| |
Collapse
|
21
|
Jiao W, Bai M, Yin H, Liu J, Sun J, Su X, Zeng H, Wen J. Therapeutic Effects of an Inhibitor of Thioredoxin Reductase on Liver Fibrosis by Inhibiting the Transforming Growth Factor-β1/Smads Pathway. Front Mol Biosci 2021; 8:690170. [PMID: 34540892 PMCID: PMC8440796 DOI: 10.3389/fmolb.2021.690170] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2021] [Accepted: 07/21/2021] [Indexed: 12/24/2022] Open
Abstract
Liver fibrosis is an important stage in the progression of liver injury into cirrhosis or even liver cancer. Hepatic stellate cells (HSCs) are induced by transforming growth factor-β1 (TGF-β1) to produce α-smooth muscle actin (α-SMA) and collagens in liver fibrosis. Butaselen (BS), which was previously synthesized by our group, is an organic selenium compound that exerts antioxidant and tumor cell apoptosis–promoting effects by inhibiting the thioredoxin (Trx)/thioredoxin reductase (TrxR) system. The aim of this study was to investigate the potential effects of BS on liver fibrosis and explore the underlying molecular mechanisms of its action. Liver fibrosis models were established using male BALB/c mice through intraperitoneal injection of CCl4. BS was administered orally once daily at a dose of 36, 90, or 180 mg/kg. Silymarin (Si), which is a drug used for patients with nonalcoholic fatty liver disease and nonalcoholic steatohepatitis, was administered at a dose of 30 mg/kg per day as a control. The action mechanisms of BS against liver fibrosis progression were examined in HSCs. The study revealed that the activity and expression levels of TrxR were elevated in the mouse liver and serum after CCl4-induced liver fibrosis. Oral administration of BS relieved the pathological state of mice with liver fibrosis, showing significant therapeutic effects against liver fibrosis. Moreover, BS not only induced HSC apoptosis but also inhibited the production of α-SMA and collagens by HSCs by downregulating the TGF-β1 expression and blocking the TGF-β1/Smads pathway. The results of the study indicated that BS inhibited liver fibrosis by regulating the TGF-β1/Smads pathway.
Collapse
Affiliation(s)
- Wenxuan Jiao
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Man Bai
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China.,Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Hanwei Yin
- Shanghai Yuanxi Medicine Corp, Shanghai, China
| | - Jiayi Liu
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Jing Sun
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Xiaoxia Su
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| | - Huihui Zeng
- State Key Laboratory of Natural and Biomimetic Drugs, School of Pharmaceutical Sciences, Peking University, Beijing, China
| | - Jinhua Wen
- Department of Cell Biology and Stem Cell Research Center, School of Basic Medical Sciences, Peking University Health Science Center, Beijing, China
| |
Collapse
|
22
|
Erdmann C, Hassoun R, Schmitt S, Kikuti C, Houdusse A, Mazur AJ, Mügge A, Hamdani N, Geyer M, Jaquet K, Mannherz HG. Integration of Cardiac Actin Mutants Causing Hypertrophic (p.A295S) and Dilated Cardiomyopathy (p.R312H and p.E361G) into Cellular Structures. Antioxidants (Basel) 2021; 10:antiox10071082. [PMID: 34356314 PMCID: PMC8301065 DOI: 10.3390/antiox10071082] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2021] [Revised: 06/26/2021] [Accepted: 06/30/2021] [Indexed: 02/03/2023] Open
Abstract
The human mutant cardiac α-actins p.A295S or p.R312H and p.E361G, correlated with hypertrophic or dilated cardiomyopathy, respectively, were expressed by the baculovirus/Sf21 insect cell system and purified to homogeneity. The purified cardiac actins maintained their native state but showed differences in Ca2+-sensitivity to stimulate the myosin-subfragment1 ATPase. Here we analyzed the interactions of these c-actins with actin-binding and -modifying proteins implicated in cardiomyocyte differentiation. We demonstrate that Arp2/3 complex and the formin mDia3 stimulated the polymerization rate and extent of the c-actins, albeit to different degrees. In addition, we tested the effect of the MICAL-1 monooxygenase, which modifies the supramolecular actin organization during development and adaptive processes. MICAL-1 oxidized these c-actin variants and induced their de-polymerization, albeit at different rates. Transfection experiments using MDCK cells demonstrated the preferable incorporation of wild type and p.A295S c-actins into their microfilament system but of p.R312H and p.E361G actins into the submembranous actin network. Transduction of neonatal rat cardiomyocytes with adenoviral constructs coding HA-tagged c-actin variants showed their incorporation into microfilaments after one day in culture and thereafter into thin filaments of nascent sarcomeric structures at their plus ends (Z-lines) except the p.E361G mutant, which preferentially incorporated at the minus ends.
Collapse
Affiliation(s)
- Constanze Erdmann
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, D-44780 Bochum, Germany;
| | - Roua Hassoun
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Sebastian Schmitt
- Institute of Structural Biology, University of Bonn, D-53127 Bonn, Germany; (S.S.); (M.G.)
| | - Carlos Kikuti
- Institut Curie, Structural Motility Team, F-75005 Paris, France; (C.K.); (A.H.)
| | - Anne Houdusse
- Institut Curie, Structural Motility Team, F-75005 Paris, France; (C.K.); (A.H.)
| | - Antonina J. Mazur
- Department of Cell Pathology, Faculty of Biotechnology, University of Wroclaw, Pl-50-383 Wroclaw, Poland;
| | - Andreas Mügge
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Nazha Hamdani
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Matthias Geyer
- Institute of Structural Biology, University of Bonn, D-53127 Bonn, Germany; (S.S.); (M.G.)
| | - Kornelia Jaquet
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
| | - Hans Georg Mannherz
- Department of Anatomy and Molecular Embryology, Medical Faculty, Ruhr-University Bochum, D-44780 Bochum, Germany;
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, Ruhr University Bochum, D-44780 Bochum, Germany; (R.H.); (A.M.); (N.H.); (K.J.)
- Department of Cardiology, St. Josef-Hospital and Bergmannsheil, Ruhr University Bochum, D-44780 Bochum, Germany
- Correspondence: ; Fax: +49-234-3214474
| |
Collapse
|
23
|
MacTaggart B, Kashina A. Posttranslational modifications of the cytoskeleton. Cytoskeleton (Hoboken) 2021; 78:142-173. [PMID: 34152688 DOI: 10.1002/cm.21679] [Citation(s) in RCA: 27] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2021] [Revised: 06/13/2021] [Accepted: 06/16/2021] [Indexed: 12/12/2022]
Abstract
The cytoskeleton plays important roles in many essential processes at the cellular and organismal levels, including cell migration and motility, cell division, and the establishment and maintenance of cell and tissue architecture. In order to facilitate these varied functions, the main cytoskeletal components-microtubules, actin filaments, and intermediate filaments-must form highly diverse intracellular arrays in different subcellular areas and cell types. The question of how this diversity is conferred has been the focus of research for decades. One key mechanism is the addition of posttranslational modifications (PTMs) to the major cytoskeletal proteins. This posttranslational addition of various chemical groups dramatically increases the complexity of the cytoskeletal proteome and helps facilitate major global and local cytoskeletal functions. Cytoskeletal proteins undergo many PTMs, most of which are not well understood. Recent technological advances in proteomics and cell biology have allowed for the in-depth study of individual PTMs and their functions in the cytoskeleton. Here, we provide an overview of the major PTMs that occur on the main structural components of the three cytoskeletal systems-tubulin, actin, and intermediate filament proteins-and highlight the cellular function of these modifications.
Collapse
Affiliation(s)
- Brittany MacTaggart
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Anna Kashina
- School of Veterinary Medicine, Department of Biomedical Sciences, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
24
|
Bago Á, Íñiguez MA, Serrador JM. Nitric Oxide and Electrophilic Cyclopentenone Prostaglandins in Redox signaling, Regulation of Cytoskeleton Dynamics and Intercellular Communication. Front Cell Dev Biol 2021; 9:673973. [PMID: 34026763 PMCID: PMC8137968 DOI: 10.3389/fcell.2021.673973] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 04/01/2021] [Indexed: 12/16/2022] Open
Abstract
Nitric oxide (NO) and electrophilic cyclopentenone prostaglandins (CyPG) are local mediators that modulate cellular response to oxidative stress in different pathophysiological processes. In particular, there is increasing evidence about their functional role during inflammation and immune responses. Although the mechanistic details about their relationship and functional interactions are still far from resolved, NO and CyPG share the ability to promote redox-based post-translational modification (PTM) of proteins that play key roles in cellular homeostasis, signal transduction and transcription. NO-induced S-nitrosylation and S-glutathionylation as well as cyclopentenone-mediated adduct formation, are a few of the main PTMs by which intra- and inter-cellular signaling are regulated. There is a growing body of evidence indicating that actin and actin-binding proteins are susceptible to covalent PTM by these agents. It is well known that the actin cytoskeleton is key for the establishment of interactions among leukocytes, endothelial and muscle cells, enabling cellular activation and migration. In this review we analyze the current knowledge about the actions exerted by NO and CyPG electrophilic lipids on the regulation of actin dynamics and cytoskeleton organization, and discuss some open questions regarding their functional relevance in the regulation of intercellular communication.
Collapse
Affiliation(s)
- Ángel Bago
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular "Severo Ochoa" (CBMSO), CSIC-UAM, Madrid, Spain
| | - Miguel A Íñiguez
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular "Severo Ochoa" (CBMSO), CSIC-UAM, Madrid, Spain.,Departamento de Biología Molecular, Universidad Autónoma de Madrid, Madrid, Spain
| | - Juan M Serrador
- Interactions with the Environment Program, Immune System Development and Function Unit, Centro de Biología Molecular "Severo Ochoa" (CBMSO), CSIC-UAM, Madrid, Spain
| |
Collapse
|
25
|
Prescher N, Hänsch S, Knobbe-Thomsen CB, Stühler K, Poschmann G. The migration behavior of human glioblastoma cells is influenced by the redox-sensitive human macrophage capping protein CAPG. Free Radic Biol Med 2021; 167:81-93. [PMID: 33711419 DOI: 10.1016/j.freeradbiomed.2021.02.038] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Revised: 02/24/2021] [Accepted: 02/26/2021] [Indexed: 12/26/2022]
Abstract
The macrophage capping protein CAPG belongs to the gelsolin superfamily which modulates actin dynamics by capping the growing end of actin filaments in a Ca2+- and PIP2-dependent manner resulting in polymerization inhibition of actin filaments. In the last years, additional functions for CAPG in transcription regulation were described and higher CAPG amounts have been linked to increased invasiveness and migration behavior in different human tumor entities like e.g. glioblastoma. Nevertheless, there is a lack of knowledge how additional functions of CAPG are regulated. As CAPG contains several cysteine residues which may be accessible to oxidation we were especially interested to investigate how alterations in the cysteine oxidation state may influence the function, localization, and regulation of CAPG. In the present study, we provide strong evidence that CAPG is a redox-sensitive protein and identified two cysteines: C282 and C290 as reversibly oxidized in glioblastoma cell lines. Whereas no evidence could be found that the canonical actin capping function of CAPG is redox-regulated, our results point to a novel role of the identified cysteines in the regulation of cell migration. Along with this, we found a localization shift out of the nucleus of CAPG and RAVER1, a potential interaction partner identified in our study which might explain the observed altered cell migration properties. The newly identified redox sensitive cysteines of CAPG could perspectively be considered as new targets for controlling tumor invasive properties.
Collapse
Affiliation(s)
- Nina Prescher
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Sebastian Hänsch
- Department of Biology, Center for Advanced Imaging (CAi), Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Christiane B Knobbe-Thomsen
- Department of Neuropathology, Heinrich-Heine University Düsseldorf and University Hospital, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Kai Stühler
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany; Molecular Proteomics Laboratory, Biomedical Research Centre (BMFZ), Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany
| | - Gereon Poschmann
- Institute of Molecular Medicine, Proteome Research, Medical Faculty and University Hospital Düsseldorf, Heinrich-Heine University Düsseldorf, Universitätsstraße 1, 40225 Düsseldorf, Germany.
| |
Collapse
|
26
|
Zamorano Cuervo N, Fortin A, Caron E, Chartier S, Grandvaux N. Pinpointing cysteine oxidation sites by high-resolution proteomics reveals a mechanism of redox-dependent inhibition of human STING. Sci Signal 2021; 14:14/680/eaaw4673. [PMID: 33906974 DOI: 10.1126/scisignal.aaw4673] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein function is regulated by posttranslational modifications (PTMs), among which reversible oxidation of cysteine residues has emerged as a key regulatory mechanism of cellular responses. Given the redox regulation of virus-host interactions, the identification of oxidized cysteine sites in cells is essential to understand the underlying mechanisms involved. Here, we present a proteome-wide identification of reversibly oxidized cysteine sites in oxidant-treated cells using a maleimide-based bioswitch method coupled to mass spectrometry analysis. We identified 2720 unique oxidized cysteine sites within 1473 proteins with distinct abundances, locations, and functions. Oxidized cysteine sites were found in numerous signaling pathways, many relevant to virus-host interactions. We focused on the oxidation of STING, the central adaptor of the innate immune type I interferon pathway, which is stimulated in response to the detection of cytosolic DNA by cGAS. We demonstrated the reversible oxidation of Cys148 and Cys206 of STING in cells. Molecular analyses led us to establish a model in which Cys148 oxidation is constitutive, whereas Cys206 oxidation is inducible by oxidative stress or by the natural ligand of STING, 2'3'-cGAMP. Our data suggest that the oxidation of Cys206 prevented hyperactivation of STING by causing a conformational change associated with the formation of inactive polymers containing intermolecular disulfide bonds. This finding should aid the design of therapies targeting STING that are relevant to autoinflammatory disorders, immunotherapies, and vaccines.
Collapse
Affiliation(s)
- Natalia Zamorano Cuervo
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Audray Fortin
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Elise Caron
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Stéfany Chartier
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada
| | - Nathalie Grandvaux
- CRCHUM-Centre Hospitalier de l'Université de Montréal, 900 rue Saint Denis, Montréal, H2X 0A9 Québec, Canada. .,Department of Biochemistry and Molecular Medicine, Faculty of Medicine, Université de Montréal, Montréal, H3C 3J7 Québec, Canada
| |
Collapse
|
27
|
Balta E, Kramer J, Samstag Y. Redox Regulation of the Actin Cytoskeleton in Cell Migration and Adhesion: On the Way to a Spatiotemporal View. Front Cell Dev Biol 2021; 8:618261. [PMID: 33585453 PMCID: PMC7875868 DOI: 10.3389/fcell.2020.618261] [Citation(s) in RCA: 35] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2020] [Accepted: 12/21/2020] [Indexed: 12/12/2022] Open
Abstract
The actin cytoskeleton of eukaryotic cells is a dynamic, fibrous network that is regulated by the concerted action of actin-binding proteins (ABPs). In particular, rapid polarization of cells in response to internal and external stimuli is fundamental to cell migration and invasion. Various isoforms of ABPs in different tissues equip cells with variable degrees of migratory and adhesive capacities. In addition, regulation of ABPs by posttranslational modifications (PTM) is pivotal to the rapid responsiveness of cells. In this context, phosphorylation of ABPs and its functional consequences have been studied extensively. However, the study of reduction/oxidation (redox) modifications of oxidation-sensitive cysteine and methionine residues of actin, ABPs, adhesion molecules, and signaling proteins regulating actin cytoskeletal dynamics has only recently emerged as a field. The relevance of such protein oxidations to cellular physiology and pathophysiology has remained largely elusive. Importantly, studying protein oxidation spatiotemporally can provide novel insights into localized redox regulation of cellular functions. In this review, we focus on the redox regulation of the actin cytoskeleton, its challenges, and recently developed tools to study its physiological and pathophysiological consequences.
Collapse
Affiliation(s)
- Emre Balta
- Section Molecular Immunology, Institute of Immunology, Heidelberg University, Heidelberg, Germany
| | - Johanna Kramer
- Section Molecular Immunology, Institute of Immunology, Heidelberg University, Heidelberg, Germany
| | - Yvonne Samstag
- Section Molecular Immunology, Institute of Immunology, Heidelberg University, Heidelberg, Germany
| |
Collapse
|
28
|
Martínez-Banaclocha M. N-acetyl-cysteine in Schizophrenia: Potential Role on the Sensitive Cysteine Proteome. Curr Med Chem 2021; 27:6424-6439. [PMID: 33115390 DOI: 10.2174/0929867326666191015091346] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 09/11/2019] [Accepted: 10/02/2019] [Indexed: 12/14/2022]
Abstract
BACKGROUND N-acetyl-cysteine (NAC) has shown widespread utility in different psychiatric disorders, including a beneficial role in schizophrenic patients. Although the replenishment of glutathione and the antioxidant activity of NAC have been suggested as the mechanisms that improve such a wide range of disorders, none seems to be sufficiently specific to explain these intriguing effects. A sensitive cysteine proteome is emerging as a functional and structural network of interconnected Sensitive Cysteine-containing Proteins (SCCPs) that together with reactive species and the cysteine/ glutathione cycles can regulate the bioenergetic metabolism, the redox homeostasis and the cellular growth, differentiation and survival, acting through different pathways that are regulated by the same thiol radical in cysteine residues. OBJECTIVE Since this sensitive cysteine network has been implicated in the pathogenesis of Parkinson's and Alzheimer's diseases, I have reviewed if the proteins that play a role in schizophrenia can be classified as SCCPs. RESULTS The results show that the principal proteins playing a role in schizophrenia can be classified as SCCPs, suggesting that the sensitive cysteine proteome (cysteinet) is defective in this type of psychosis. CONCLUSION The present review proposes that there is a deregulation of the sensitive cysteine proteome in schizophrenia as the consequence of a functional imbalance among different SCCPs, which play different functions in neurons and glial cells. In this context, the role of NAC to restore and prevent schizophrenic disorders is discussed.
Collapse
|
29
|
Sciortino G, Aureliano M, Garribba E. Rationalizing the Decavanadate(V) and Oxidovanadium(IV) Binding to G-Actin and the Competition with Decaniobate(V) and ATP. Inorg Chem 2021; 60:334-344. [PMID: 33253559 PMCID: PMC8016201 DOI: 10.1021/acs.inorgchem.0c02971] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Indexed: 02/07/2023]
Abstract
The experimental data collected over the past 15 years on the interaction of decavanadate(V) (V10O286-; V10), a polyoxometalate (POM) with promising anticancer and antibacterial action, with G-actin, were rationalized by using several computational approaches (docking, density functional theory (DFT), and molecular dynamics (MD)). Moreover, a comparison with the isostructural and more stable decaniobate(V) (Nb10O286-; Nb10) was carried out. Four binding sites were identified, named α, β, γ, and δ, the site α being the catalytic nucleotide site located in the cleft of the enzyme at the interface of the subdomains II and IV. It was observed that the site α is preferred by V10, whereas Nb10 is more stable at the site β; this indicates that, differently from other proteins, G-actin could contemporaneously bind the two POMs, whose action would be synergistic. Both decavanadate and decaniobate induce conformational rearrangements in G-actin, larger for V10 than Nb10. Moreover, the binding mode of oxidovanadium(IV) ion, VIVO2+, formed upon the reduction of decavanadate(V) by the -SH groups of accessible cysteine residues, is also found in the catalytic site α with (His161, Asp154) coordination; this adduct overlaps significantly with the region where ATP is bound, accounting for the competition between V10 and its reduction product VIVO2+ with ATP, as previously observed by EPR spectroscopy. Finally, the competition with ATP was rationalized: since decavanadate prefers the nucleotide site α, Ca2+-ATP displaces V10 from this site, while the competition is less important for Nb10 because this POM shows a higher affinity for β than for site α. A relevant consequence of this paper is that other metallodrug-protein systems, in the absence or presence of eventual inhibitors and/or competition with molecules of the organism, could be studied with the same approach, suggesting important elements for an explanation of the biological data and a rational drug design.
Collapse
Affiliation(s)
- Giuseppe Sciortino
- Dipartimento
di Chimica e Farmacia, Università
di Sassari, Via Vienna 2, I-07100 Sassari, Italy
- Institute
of Chemical Research of Catalonia (ICIQ), Avgda. Països Catalans, 16, 43007 Tarragona, Spain
| | - Manuel Aureliano
- CCMar,
FCT, Faculdade de Ciências e Tecnologia, Universidade do Algarve, 8000-139 Faro, Portugal
| | - Eugenio Garribba
- Dipartimento
di Chimica e Farmacia, Università
di Sassari, Via Vienna 2, I-07100 Sassari, Italy
| |
Collapse
|
30
|
Stimulating Embryo Polarization with Mitochondrial Peroxide. Dev Cell 2020; 53:261-262. [PMID: 32369741 DOI: 10.1016/j.devcel.2020.04.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Centrosomes break symmetry in the C. elegans one-cell embryo, triggering its anterior-posterior polarization and initiating segregation of somatic and germline cell lineages. In this issue of Developmental Cell, De Henau et al. show that mitochondria also contribute to symmetry breaking by producing hydrogen peroxide at the egg's future posterior pole.
Collapse
|
31
|
NaveenKumar SK, Hemshekhar M, Jagadish S, Manikanta K, Vishalakshi GJ, Kemparaju K, Girish KS. Melatonin restores neutrophil functions and prevents apoptosis amid dysfunctional glutathione redox system. J Pineal Res 2020; 69:e12676. [PMID: 32597503 DOI: 10.1111/jpi.12676] [Citation(s) in RCA: 52] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 06/08/2020] [Accepted: 06/21/2020] [Indexed: 12/26/2022]
Abstract
Melatonin is a chronobiotic hormone, which can regulate human diseases like cancer, atherosclerosis, respiratory disorders, and microbial infections by regulating redox system. Melatonin exhibits innate immunomodulation by communicating with immune system and influencing neutrophils to fight infections and inflammation. However, sustaining redox homeostasis and reactive oxygen species (ROS) generation in neutrophils are critical during chemotaxis, oxidative burst, phagocytosis, and neutrophil extracellular trap (NET) formation. Therefore, endogenous antioxidant glutathione (GSH) redox cycle is highly vital in regulating neutrophil functions. Reduced intracellular GSH levels and glutathione reductase (GR) activity in the neutrophils during clinical conditions like autoimmune disorders, neurological disorders, diabetes, and microbial infections lead to dysfunctional neutrophils. Therefore, we hypothesized that redox modulators like melatonin can protect neutrophil health and functions under GSH and GR activity-deficient conditions. We demonstrate the dual role of melatonin, wherein it protects neutrophils from oxidative stress-induced apoptosis by reducing ROS generation; in contrast, it restores neutrophil functions like phagocytosis, degranulation, and NETosis in GSH and GR activity-deficient neutrophils by regulating ROS levels both in vitro and in vivo. Melatonin mitigates LPS-induced neutrophil dysfunctions by rejuvenating GSH redox system, specifically GR activity by acting as a parallel redox system. Our results indicate that melatonin could be a potential auxiliary therapy to treat immune dysfunction and microbial infections, including virus, under chronic disease conditions by restoring neutrophil functions. Further, melatonin could be a promising immune system booster to fight unprecedented pandemics like the current COVID-19. However, further studies are indispensable to address the clinical usage of melatonin.
Collapse
Affiliation(s)
| | | | - Swamy Jagadish
- Department of Studies in Biochemistry, University of Mysore, Mysore, India
| | | | | | - Kempaiah Kemparaju
- Department of Studies in Biochemistry, University of Mysore, Mysore, India
| | - Kesturu S Girish
- Department of Studies in Biochemistry, University of Mysore, Mysore, India
- Department of Studies and Research in Biochemistry, Tumkur University, Tumakuru, India
| |
Collapse
|
32
|
Sokolik CG, Qassem N, Chill JH. The Disordered Cellular Multi-Tasker WIP and Its Protein-Protein Interactions: A Structural View. Biomolecules 2020; 10:biom10071084. [PMID: 32708183 PMCID: PMC7407642 DOI: 10.3390/biom10071084] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2020] [Revised: 07/16/2020] [Accepted: 07/18/2020] [Indexed: 01/21/2023] Open
Abstract
WASp-interacting protein (WIP), a regulator of actin cytoskeleton assembly and remodeling, is a cellular multi-tasker and a key member of a network of protein-protein interactions, with significant impact on health and disease. Here, we attempt to complement the well-established understanding of WIP function from cell biology studies, summarized in several reviews, with a structural description of WIP interactions, highlighting works that present a molecular view of WIP's protein-protein interactions. This provides a deeper understanding of the mechanisms by which WIP mediates its biological functions. The fully disordered WIP also serves as an intriguing example of how intrinsically disordered proteins (IDPs) exert their function. WIP consists of consecutive small functional domains and motifs that interact with a host of cellular partners, with a striking preponderance of proline-rich motif capable of interactions with several well-recognized binding partners; indeed, over 30% of the WIP primary structure are proline residues. We focus on the binding motifs and binding interfaces of three important WIP segments, the actin-binding N-terminal domain, the central domain that binds SH3 domains of various interaction partners, and the WASp-binding C-terminal domain. Beyond the obvious importance of a more fundamental understanding of the biology of this central cellular player, this approach carries an immediate and highly beneficial effect on drug-design efforts targeting WIP and its binding partners. These factors make the value of such structural studies, challenging as they are, readily apparent.
Collapse
|
33
|
Kanou A, Nishimura S, Tabuchi T, Matsuyama A, Yoshida M, Kato T, Kakeya H. Serine catabolism produces ROS, sensitizes cells to actin dysfunction, and suppresses cell growth in fission yeast. J Antibiot (Tokyo) 2020; 73:574-580. [PMID: 32313168 DOI: 10.1038/s41429-020-0305-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Revised: 03/12/2020] [Accepted: 03/23/2020] [Indexed: 12/30/2022]
Abstract
Serine is an essential component in organisms as a building block of biomolecules, a precursor of metabolites, an allosteric regulator of an enzyme, etc. This amino acid is thought to be a key metabolite in human diseases including cancers and infectious diseases. To understand the consequence of serine catabolism, we screened natural products to identify a fungal metabolite chaetoglobosin D (ChD) as a specific inhibitor of fission yeast cell growth when cultivated with serine as a sole nitrogen source. ChD targets actin, and actin mutant cells showed severe growth defect on serine medium. ROS accumulated in cells when cultivated in serine medium, while actin mutant cells showed increased sensitivity to oxidative stress. ROS production is a new aspect of serine metabolism, which might be involved in disease progression, and actin could be the drug target for curing serine-dependent symptoms.
Collapse
Affiliation(s)
- Akihiko Kanou
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan
| | - Shinichi Nishimura
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan. .,Department of Biotechnology, The University of Tokyo, Tokyo, 113-8657, Japan. .,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, 113-8657, Japan. .,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan.
| | - Toshitsugu Tabuchi
- Department of Biotechnology, The University of Tokyo, Tokyo, 113-8657, Japan
| | - Akihisa Matsuyama
- Department of Biotechnology, The University of Tokyo, Tokyo, 113-8657, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan
| | - Minoru Yoshida
- Department of Biotechnology, The University of Tokyo, Tokyo, 113-8657, Japan.,Collaborative Research Institute for Innovative Microbiology, The University of Tokyo, Tokyo, 113-8657, Japan.,Chemical Genomics Research Group, RIKEN Center for Sustainable Resource Science, Saitama, 351-0198, Japan
| | - Taira Kato
- Research & Development Division, MicroBiopharm Japan Co., Ltd., 156 Nakagawara, Kiyosu-shi, Aichi, 452-0915, Japan
| | - Hideaki Kakeya
- Department of System Chemotherapy and Molecular Sciences, Division of Bioinformatics and Chemical Genomics, Graduate School of Pharmaceutical Sciences, Kyoto University, Kyoto, 606-8501, Japan.
| |
Collapse
|
34
|
De Henau S, Pagès-Gallego M, Pannekoek WJ, Dansen TB. Mitochondria-Derived H 2O 2 Promotes Symmetry Breaking of the C. elegans Zygote. Dev Cell 2020; 53:263-271.e6. [PMID: 32275886 DOI: 10.1016/j.devcel.2020.03.008] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 02/04/2020] [Accepted: 03/11/2020] [Indexed: 12/21/2022]
Abstract
Symmetry breaking is an essential step in cell differentiation and early embryonic development. However, the molecular cues that trigger symmetry breaking remain largely unknown. Here, we show that mitochondrial H2O2 acts as a symmetry-breaking cue in the C. elegans zygote. We find that symmetry breaking is marked by a local H2O2 increase and coincides with a relocation of mitochondria to the cell cortex. Lowering endogenous H2O2 levels delays the onset of symmetry breaking, while artificially targeting mitochondria to the cellular cortex using a light-induced heterodimerization technique is sufficient to initiate symmetry breaking in a H2O2-dependent manner. In wild-type development, both sperm and maternal mitochondria contribute to symmetry breaking. Our findings reveal that mitochondrial H2O2-signaling promotes the onset of polarization, a fundamental process in development and cell differentiation, and this is achieved by both mitochondrial redistribution and differential H2O2-production.
Collapse
Affiliation(s)
- Sasha De Henau
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Marc Pagès-Gallego
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Willem-Jan Pannekoek
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands
| | - Tobias B Dansen
- Center for Molecular Medicine, Molecular Cancer Research, University Medical Center Utrecht, Heidelberglaan 100, 3584 CG Utrecht, the Netherlands.
| |
Collapse
|
35
|
Mild depolarization of the inner mitochondrial membrane is a crucial component of an anti-aging program. Proc Natl Acad Sci U S A 2020; 117:6491-6501. [PMID: 32152094 PMCID: PMC7104298 DOI: 10.1073/pnas.1916414117] [Citation(s) in RCA: 109] [Impact Index Per Article: 21.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Abstract
The mitochondria, organelles that produce the largest amounts of ATP and reactive oxygen species (mROS) in living cells, are equipped with a universal mechanism that can completely prevent mROS production. This mechanism consists of mild depolarization of the inner mitochondrial membrane to decrease the membrane potential to a level sufficient to form ATP but insufficient to generate mROS. In short-lived mice, aging is accompanied by inactivation of the mild depolarization mechanism, resulting in chronic poisoning of the organism with mROS. However, mild depolarization still functions for many years in long-lived naked mole rats and bats. The mitochondria of various tissues from mice, naked mole rats (NMRs), and bats possess two mechanistically similar systems to prevent the generation of mitochondrial reactive oxygen species (mROS): hexokinases I and II and creatine kinase bound to mitochondrial membranes. Both systems operate in a manner such that one of the kinase substrates (mitochondrial ATP) is electrophoretically transported by the ATP/ADP antiporter to the catalytic site of bound hexokinase or bound creatine kinase without ATP dilution in the cytosol. One of the kinase reaction products, ADP, is transported back to the mitochondrial matrix via the antiporter, again through an electrophoretic process without cytosol dilution. The system in question continuously supports H+-ATP synthase with ADP until glucose or creatine is available. Under these conditions, the membrane potential, ∆ψ, is maintained at a lower than maximal level (i.e., mild depolarization of mitochondria). This ∆ψ decrease is sufficient to completely inhibit mROS generation. In 2.5-y-old mice, mild depolarization disappears in the skeletal muscles, diaphragm, heart, spleen, and brain and partially in the lung and kidney. This age-dependent decrease in the levels of bound kinases is not observed in NMRs and bats for many years. As a result, ROS-mediated protein damage, which is substantial during the aging of short-lived mice, is stabilized at low levels during the aging of long-lived NMRs and bats. It is suggested that this mitochondrial mild depolarization is a crucial component of the mitochondrial anti-aging system.
Collapse
|
36
|
Urmey AR, Zondlo NJ. Structural preferences of cysteine sulfinic acid: The sulfinate engages in multiple local interactions with the peptide backbone. Free Radic Biol Med 2020; 148:96-107. [PMID: 31883974 DOI: 10.1016/j.freeradbiomed.2019.12.030] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/22/2019] [Revised: 12/16/2019] [Accepted: 12/19/2019] [Indexed: 02/06/2023]
Abstract
Cysteine sulfinic acid (Cys-SO2-) is a non-enzymatic oxidative post-translational modification (PTM) that has been identified in hundreds of proteins. However, the effects of cysteine sulfination are in most cases poorly understood. Cys-SO2- is structurally distinctive, with long sulfur-carbon and sulfur-oxygen bonds, and with tetrahedral geometry around sulfur due to its lone pair. Cys-SO2- thus has a unique range of potential interactions with the protein backbone which could facilitate protein structural changes. Herein, the structural effects of cysteine oxidation to the sulfinic acid were investigated in model peptides and folded proteins using NMR spectroscopy, circular dichroism, bioinformatics, and computational studies. In the PDB, Cys-SO2- shows a greater preference for α-helix than Cys. In addition, Cys-SO2- is more commonly found in structures with φ > 0, including in multiple types of β-turn. Sulfinate oxygens engage in hydrogen bonds with adjacent (i or i + 1) amide hydrogens. Over half of sulfinates have at least one hydrogen bond with an adjacent amide, and several structures have hydrogen bonds with both adjacent amides. Alternately, sulfur or either oxygen can act as an electron donor for n→π* interactions with the backbone carbonyl of the same residue, as indicated by frequent S⋯CO or O⋯CO distances below the sums of their van der Waals radii in protein structures. In peptides, Cys-SO2- favored α-helical structure at the N-terminus, consistent with helix dipole effects and backbone hydrogen bonds with the sulfinate promoting α-helix. Cys-SO2- has only modestly greater polyproline II helix propensity than Cys-SH, likely due to competition from multiple side chain-backbone interactions. Cys-SO2- stabilizes the i+1 position of a β-turn relative to Cys-SH. Within proteins, the range of side chain-main chain interactions available to Cys-SO2- compared to Cys-SH provides a basis for potential changes in protein structure and function due to cysteine oxidation to the sulfinic acid.
Collapse
Affiliation(s)
- Andrew R Urmey
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, United States
| | - Neal J Zondlo
- Department of Chemistry and Biochemistry, University of Delaware, Newark, DE, 19716, United States.
| |
Collapse
|
37
|
Rambacher KM, Moniri NH. Cysteine redox state regulates human β2-adrenergic receptor binding and function. Sci Rep 2020; 10:2934. [PMID: 32076070 PMCID: PMC7031529 DOI: 10.1038/s41598-020-59983-4] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2019] [Accepted: 02/05/2020] [Indexed: 01/08/2023] Open
Abstract
Bronchoconstrictive airway disorders such as asthma are characterized by inflammation and increases in reactive oxygen species (ROS), which produce a highly oxidative environment. β2-adrenergic receptor (β2AR) agonists are a mainstay of clinical therapy for asthma and provide bronchorelaxation upon inhalation. We have previously shown that β2AR agonism generates intracellular ROS, an effect that is required for receptor function, and which post-translationally oxidizes β2AR cysteine thiols to Cys-S-sulfenic acids (Cys-S-OH). Furthermore, highly oxidative environments can irreversibly oxidize Cys-S-OH to Cys-S-sulfinic (Cys-SO2H) or S-sulfonic (Cys-SO3H) acids, which are incapable of further participating in homeostatic redox reactions (i.e., redox-deficient). The aim of this study was to examine the vitality of β2AR-ROS interplay and the resultant functional consequences of β2AR Cys-redox in the receptors native, oxidized, and redox-deficient states. Here, we show for the first time that β2AR can be oxidized to Cys-S-OH in situ, moreover, using both clonal cells and a human airway epithelial cell line endogenously expressing β2AR, we show that receptor redox state profoundly influences β2AR orthosteric ligand binding and downstream function. Specifically, homeostatic β2AR redox states are vital toward agonist-induced cAMP formation and subsequent CREB and G-protein-dependent ERK1/2 phosphorylation, in addition to β-arrestin-2 recruitment and downstream arrestin-dependent ERK1/2 phosphorylation and internalization. On the contrary, redox-deficient β2AR states exhibit decreased ability to signal via either Gαs or β-arrestin. Together, our results demonstrate a β2AR-ROS redox axis, which if disturbed, interferes with proper receptor function.
Collapse
Affiliation(s)
- Kalyn M Rambacher
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA30341, United States
| | - Nader H Moniri
- Department of Pharmaceutical Sciences, College of Pharmacy, Mercer University Health Sciences Center, Mercer University, Atlanta, GA30341, United States.
| |
Collapse
|
38
|
Kommaddi RP, Tomar DS, Karunakaran S, Bapat D, Nanguneri S, Ray A, Schneider BL, Nair D, Ravindranath V. Glutaredoxin1 Diminishes Amyloid Beta-Mediated Oxidation of F-Actin and Reverses Cognitive Deficits in an Alzheimer's Disease Mouse Model. Antioxid Redox Signal 2019; 31:1321-1338. [PMID: 31617375 DOI: 10.1089/ars.2019.7754] [Citation(s) in RCA: 32] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aims: Reactive oxygen species (ROS) generated during Alzheimer's disease (AD) pathogenesis through multiple sources are implicated in synaptic pathology observed in the disease. We have previously shown F-actin disassembly in dendritic spines in early AD (34). The actin cytoskeleton can be oxidatively modified resulting in altered F-actin dynamics. Therefore, we investigated whether disruption of redox signaling could contribute to actin network disassembly and downstream effects in the amyloid precursor protein/presenilin-1 double transgenic (APP/PS1) mouse model of AD. Results: Synaptosomal preparations from 1-month-old APP/PS1 mice showed an increase in ROS levels, coupled with a decrease in the reduced form of F-actin and increase in glutathionylated synaptosomal actin. Furthermore, synaptic glutaredoxin 1 (Grx1) and thioredoxin levels were found to be lowered. Overexpressing Grx1 in the brains of these mice not only reversed F-actin loss seen in APP/PS1 mice but also restored memory recall after contextual fear conditioning. F-actin levels and F-actin nanoarchitecture in spines were also stabilized by Grx1 overexpression in APP/PS1 primary cortical neurons, indicating that glutathionylation of F-actin is a critical event in early pathogenesis of AD, which leads to spine loss. Innovation: Loss of thiol/disulfide oxidoreductases in the synapse along with increase in ROS can render F-actin nanoarchitecture susceptible to oxidative modifications in AD. Conclusions: Our findings provide novel evidence that altered redox signaling in the form of S-glutathionylation and reduced Grx1 levels can lead to synaptic dysfunction during AD pathogenesis by directly disrupting the F-actin nanoarchitecture in spines. Increasing Grx1 levels is a potential target for novel disease-modifying therapies for AD.
Collapse
Affiliation(s)
| | | | | | - Deepti Bapat
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | | | - Ajit Ray
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Bernard L Schneider
- Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne, Lausanne, Switzerland
| | - Deepak Nair
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India
| | - Vijayalakshmi Ravindranath
- Centre for Neuroscience, Indian Institute of Science, Bangalore, India.,Centre for Brain Research, Indian Institute of Science, Bangalore, India
| |
Collapse
|
39
|
A new bioluminescence-based tool for modulating target proteins in live cells. Sci Rep 2019; 9:18239. [PMID: 31796796 PMCID: PMC6890795 DOI: 10.1038/s41598-019-54712-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/18/2019] [Indexed: 11/08/2022] Open
Abstract
We have developed a new genetically encoded tool designed to generate reactive oxygen species (ROS) at target proteins in cultured cells; it is designed using firefly luciferase and photosensitiser protein KillerRed. Targeting this fusion protein, KillerFirefly, to F-actin in live cells and treatment with luciferin induced a characteristic structure, previously reported as a cofilin-actin rod, which is seen in patients with Alzheimer's disease. This structural change is considered to be elicited by the consistent generation of very low-level ROS by KillerFirefly in the vicinity of F-actin. Moreover, our results suggest the presence of an actin-regulating system, controlled by very low levels of endogenously generated ROS.
Collapse
|
40
|
Vukelic S, Xu Q, Seidel-Rogol B, Faidley EA, Dikalova AE, Hilenski LL, Jorde U, Poole LB, Lassègue B, Zhang G, Griendling KK. NOX4 (NADPH Oxidase 4) and Poldip2 (Polymerase δ-Interacting Protein 2) Induce Filamentous Actin Oxidation and Promote Its Interaction With Vinculin During Integrin-Mediated Cell Adhesion. Arterioscler Thromb Vasc Biol 2019; 38:2423-2434. [PMID: 30354218 DOI: 10.1161/atvbaha.118.311668] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Objective- Actin cytoskeleton assembly and organization, as a result of focal adhesion (FA) formation during cell adhesion, are dependent on reactive oxygen species and the cellular redox environment. Poldip2 (polymerase δ-interacting protein 2), a novel regulator of NOX4 (NADPH oxidase 4), plays a significant role in reactive oxygen species production and cytoskeletal remodeling. Thus, we hypothesized that endogenous reactive oxygen species derived from Poldip2/NOX4 contribute to redox regulation of actin and cytoskeleton assembly during integrin-mediated cell adhesion. Approach and Results- Using vascular smooth muscle cells, we verified that hydrogen peroxide (H2O2) levels increase during integrin-mediated cell attachment as a result of activation of NOX4. Filamentous actin (F-actin) was oxidized by sulfenylation during cell attachment, with a peak at 3 hours (0.80±0.04 versus 0.08±0.13 arbitrary units at time zero), which was enhanced by overexpression of Poldip2. Depletion of Poldip2 or NOX4 using siRNA, or scavenging of endogenous H2O2 with catalase, inhibited F-actin oxidation by 78±26%, 99±1%, and 98±1%, respectively. To determine the consequence of F-actin oxidation, we examined the binding of F-actin to vinculin, a protein involved in FA complexes that regulates FA maturation. Vinculin binding during cell adhesion as well as migration capacity were inhibited after transfection with actin containing 2 oxidation-resistant point mutations (C272A and C374A). Silencing of Poldip2 or NOX4 also impaired actin-vinculin interaction, which disturbed maturation of FAs and inhibited cell migration. Conclusions- These results suggest that integrin engagement during cell attachment activates Poldip2/Nox4 to oxidize actin, which modulates FA assembly.
Collapse
Affiliation(s)
- Sasa Vukelic
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.).,Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (S.V., U.J.)
| | - Qian Xu
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.).,Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (Q.X., G.Z.)
| | - Bonnie Seidel-Rogol
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Elizabeth A Faidley
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Anna E Dikalova
- Department of Medicine, Vanderbilt University, Nashville, TN (A.E.D.)
| | - Lula L Hilenski
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Ulrich Jorde
- Division of Cardiology, Department of Medicine, Albert Einstein College of Medicine, Bronx, NY (S.V., U.J.)
| | - Leslie B Poole
- Department of Biochemistry, Center for Molecular Signaling, Center for Redox Biology and Medicine, Wake Forest School of Medicine, Winston-Salem, NC (L.B.P.)
| | - Bernard Lassègue
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| | - Guogang Zhang
- Department of Cardiovascular Medicine, Xiangya Hospital, Central South University, Changsha, China (Q.X., G.Z.)
| | - Kathy K Griendling
- From the Division of Cardiology, Department of Medicine, Emory University, Atlanta, GA (S.V., Q.X., B.S.-R., E.A.F., L.L.H., B.L., K.K.G.)
| |
Collapse
|
41
|
Paracrine Mechanisms of Redox Signalling for Postmitotic Cell and Tissue Regeneration. Trends Cell Biol 2019; 29:514-530. [DOI: 10.1016/j.tcb.2019.01.006] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2018] [Revised: 01/10/2019] [Accepted: 01/18/2019] [Indexed: 01/08/2023]
|
42
|
Lagousi T, Basdeki P, Routsias J, Spoulou V. Novel Protein-Based Pneumococcal Vaccines: Assessing the Use of Distinct Protein Fragments Instead of Full-Length Proteins as Vaccine Antigens. Vaccines (Basel) 2019; 7:vaccines7010009. [PMID: 30669439 PMCID: PMC6466302 DOI: 10.3390/vaccines7010009] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2018] [Revised: 01/15/2019] [Accepted: 01/16/2019] [Indexed: 12/20/2022] Open
Abstract
Non-serotype-specific protein-based pneumococcal vaccines have received extensive research focus due to the limitations of polysaccharide-based vaccines. Pneumococcal proteins (PnPs), universally expressed among serotypes, may induce broader immune responses, stimulating humoral and cellular immunity, while being easier to manufacture and less expensive. Such an approach has raised issues mainly associated with sequence/level of expression variability, chemical instability, as well as possible undesirable reactogenicity and autoimmune properties. A step forward employs the identification of highly-conserved antigenic regions within PnPs with the potential to retain the benefits of protein antigens. Besides, their low-cost and stable construction facilitates the combination of several antigenic regions or peptides that may impair different stages of pneumococcal disease offering even wider serotype coverage and more efficient protection. This review discusses the up-to-date progress on PnPs that are currently under clinical evaluation and the challenges for their licensure. Focus is given on the progress on the identification of antigenic regions/peptides within PnPs and their evaluation as vaccine candidates, accessing their potential to overcome the issues associated with full-length protein antigens. Particular mention is given of the use of newer delivery system technologies including conjugation to Toll-like receptors (TLRs) and reformulation into nanoparticles to enhance the poor immunogenicity of such antigens.
Collapse
Affiliation(s)
- Theano Lagousi
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| | - Paraskevi Basdeki
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| | - John Routsias
- Department of Microbiology, Athens Medical School, 11527 Athens, Greece.
| | - Vana Spoulou
- First Department of Paediatrics, "Aghia Sophia" Children's Hospital, Immunobiology Research Laboratory and Infectious Diseases Department "MAKKA," Athens Medical School, 11527 Athens, Greece.
| |
Collapse
|
43
|
Non-enzymatic cleavage of Hsp90 by oxidative stress leads to actin aggregate formation: A novel gain-of-function mechanism. Redox Biol 2019; 21:101108. [PMID: 30660959 PMCID: PMC6348241 DOI: 10.1016/j.redox.2019.101108] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2018] [Revised: 01/04/2019] [Accepted: 01/10/2019] [Indexed: 01/03/2023] Open
Abstract
Aging is accompanied by the accumulation of oxidized proteins. To remove them, cells employ the proteasomal and autophagy-lysosomal systems; however, if the clearance rate is inferior to its formation, protein aggregates form as a hallmark of proteostasis loss. In cells, during stress conditions, actin aggregates accumulate leading to impaired proliferation and reduced proteasomal activity, as observed in cellular senescence. The heat shock protein 90 (Hsp90) is a molecular chaperone that binds and protects the proteasome from oxidative inactivation. We hypothesized that in oxidative stress conditions a malfunction of Hsp90 occurs resulting in the aforementioned protein aggregates. Here, we demonstrate that upon oxidative stress Hsp90 loses its function in a highly specific non-enzymatic iron-catalyzed oxidation event and its breakdown product, a cleaved form of Hsp90 (Hsp90cl), acquires a new function in mediating the accumulation of actin aggregates. Moreover, the prevention of Hsp90 cleavage reduces oxidized actin accumulation, whereas transfection of the cleaved form of Hsp90 leads to an enhanced accumulation of oxidized actin. This indicates a clear role of the Hsp90cl in the aggregation of oxidized proteins.
Collapse
|
44
|
Olthoff JT, Lindsay A, Abo-Zahrah R, Baltgalvis KA, Patrinostro X, Belanto JJ, Yu DY, Perrin BJ, Garry DJ, Rodney GG, Lowe DA, Ervasti JM. Loss of peroxiredoxin-2 exacerbates eccentric contraction-induced force loss in dystrophin-deficient muscle. Nat Commun 2018; 9:5104. [PMID: 30504831 PMCID: PMC6269445 DOI: 10.1038/s41467-018-07639-3] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Accepted: 11/14/2018] [Indexed: 12/28/2022] Open
Abstract
Force loss in skeletal muscle exposed to eccentric contraction is often attributed to injury. We show that EDL muscles from dystrophin-deficient mdx mice recover 65% of lost force within 120 min of eccentric contraction and exhibit minimal force loss when the interval between contractions is increased from 3 to 30 min. A proteomic screen of mdx muscle identified an 80% reduction in the antioxidant peroxiredoxin-2, likely due to proteolytic degradation following hyperoxidation by NADPH Oxidase 2. Eccentric contraction-induced force loss in mdx muscle was exacerbated by peroxiredoxin-2 ablation, and improved by peroxiredoxin-2 overexpression or myoglobin knockout. Finally, overexpression of γcyto- or βcyto-actin protects mdx muscle from eccentric contraction-induced force loss by blocking NADPH Oxidase 2 through a mechanism dependent on cysteine 272 unique to cytoplasmic actins. Our data suggest that eccentric contraction-induced force loss may function as an adaptive circuit breaker that protects mdx muscle from injurious contractions.
Collapse
Affiliation(s)
- John T Olthoff
- Molecular, Cellular, Developmental Biology, and Genetics Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Angus Lindsay
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Reem Abo-Zahrah
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Kristen A Baltgalvis
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Xiaobai Patrinostro
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Joseph J Belanto
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA
| | - Dae-Yeul Yu
- Aging Research Center, Korea Research Institute of Bioscience and Biotechnology (KRIBB), Daejeon, Republic of Korea
| | - Benjamin J Perrin
- Department of Biology, Indiana University-Purdue University Indianapolis, Indianapolis, IN, 46022, USA
| | - Daniel J Garry
- Lillehei Heart Institute and Department of Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - George G Rodney
- Department of Molecular Physiology and Biophysics, Baylor College of Medicine, Houston, TX, 77030, USA
| | - Dawn A Lowe
- Divisions of Rehabilitation Science and Physical Therapy, Department of Rehabilitation Medicine, University of Minnesota, Minneapolis, MN, 55455, USA
| | - James M Ervasti
- Molecular, Cellular, Developmental Biology, and Genetics Graduate Program, University of Minnesota, Minneapolis, MN, 55455, USA.
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota, Minneapolis, MN, 55455, USA.
| |
Collapse
|
45
|
Cheriyath V, Kaur J, Davenport A, Khalel A, Chowdhury N, Gaddipati L. G1P3 (IFI6), a mitochondrial localised antiapoptotic protein, promotes metastatic potential of breast cancer cells through mtROS. Br J Cancer 2018; 119:52-64. [PMID: 29899394 PMCID: PMC6035266 DOI: 10.1038/s41416-018-0137-3] [Citation(s) in RCA: 52] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2017] [Revised: 04/27/2018] [Accepted: 05/09/2018] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Redox deregulations are ubiquitous in cancer cells. However, the role of mitochondrial redox deregulation in metastasis remains unclear. In breast cancer, upregulation of mitochondrial antiapoptotic protein G1P3 (IFI6) was associated with poor distance metastasis-free survival (DMFS). Therefore, we tested the hypothesis that G1P3-induced mitochondrial redox deregulation confers metastatic potentials in breast cancer cells. METHODS Cell migration and invasion assays; confocal and immunofluorescence microscopy; and Illumina HumanHT-12 BeadChip to assess gene expression. RESULTS Consequent to its localisation on inner-mitochondrial membrane, mtROS were higher in G1P3-expressing cells (MCF-7G1P3). G1P3-overexpressing cells migrated and invaded faster than the vector controls with increased number of filopodia and F-actin bundles (p ≤ 0.05). mtROS suppression with H2O2 scavengers and mitochondrial-specific antioxidants significantly decreased migratory structures and reversed G1P3-induced migration and invasion (p ≤ 0.05). Knocking down G1P3 decreased both migration and migratory structures in MCF-7G1P3 cells. Moreover, gene networks involved in redox regulation, metastasis and actin remodelling were upregulated in MCF-7G1P3 cells. CONCLUSIONS G1P3-induced mtROS have a direct role in migratory structure formation and nuclear gene expression to promote breast cancer cell metastasis. Therefore, interrupting mitochondrial functions of G1P3 may improve clinical outcomes in breast cancer patients.
Collapse
Affiliation(s)
- Venugopalan Cheriyath
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA.
| | - Jaspreet Kaur
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA
| | - Anne Davenport
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA.,Department of Biology, Texas Woman's University, Denton, TX, 76204, USA
| | - Ashjan Khalel
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA
| | - Nobel Chowdhury
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA
| | - Lalitha Gaddipati
- Department of Biological and Environmental Sciences, Texas A&M University-Commerce, Commerce, TX, 75429, USA
| |
Collapse
|
46
|
Ishizuka Y, Koshinaga T, Hirano T, Nagasaki-Maeoka E, Watanabe Y, Hoshi R, Yoshizawa S, Sugito K, Kawashima H, Uekusa S, Fukuda N, Soma M, Fujiwara K. NRP1 knockdown promotes the migration and invasion of human neuroblastoma-derived SK‑N‑AS cells via the activation of β1 integrin expression. Int J Oncol 2018; 53:159-166. [PMID: 29750423 DOI: 10.3892/ijo.2018.4397] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2018] [Accepted: 04/24/2018] [Indexed: 11/06/2022] Open
Abstract
Neuropilin 1 (NRP1) is a transmembrane glycoprotein, which regulates many aspects of cellular function by functioning as co-receptor of various ligands. Recent studies have suggested that NRP1 promotes tumorigenesis, not only by activating the growth of tumor vessels, but also by activating the growth or migration of tumor cells themselves. The present study was performed to elucidate the roles of NRP1 in the development and/or progression of neuroblastoma (NB). In contrast to previous observations in various types of cancer, the analysis of public datasets indicated that lower levels of NRP1 expression were significantly associated with a shorter survival period of patients with NB. Consistent with this finding, wound-healing assay and Matrigel invasion assay revealed that NB cells in which NRP1 was knocked down exhibited increased migratory and invasive abilities. Further analyses indicated that β1 integrin expression was markedly increased in NB cells in which NRP1 was knocked down, and NB cells in which β1 integrin was knocked down exhibited decreased migratory and invasive abilities. The results presented herein indicate that NRP1 exerts tumor suppressive effects in NB, at least in part by regulating the expression of β1 integrin.
Collapse
Affiliation(s)
- Yoshiaki Ishizuka
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Tsugumichi Koshinaga
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Takayuki Hirano
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Eri Nagasaki-Maeoka
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Yosuke Watanabe
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Reina Hoshi
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shinsuke Yoshizawa
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kiminobu Sugito
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Hiroyuki Kawashima
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Shota Uekusa
- Department of Pediatric Surgery, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Noboru Fukuda
- Division of Nephrology, Hypertension and Endocrinology, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Masayoshi Soma
- Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan
| | - Kyoko Fujiwara
- Division of General Medicine, Department of Medicine, Nihon University School of Medicine, Tokyo 173-8610, Japan
| |
Collapse
|
47
|
Oswald MCW, Garnham N, Sweeney ST, Landgraf M. Regulation of neuronal development and function by ROS. FEBS Lett 2018; 592:679-691. [PMID: 29323696 PMCID: PMC5888200 DOI: 10.1002/1873-3468.12972] [Citation(s) in RCA: 147] [Impact Index Per Article: 21.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2017] [Accepted: 01/02/2018] [Indexed: 12/20/2022]
Abstract
Reactive oxygen species (ROS) have long been studied as destructive agents in the context of nervous system ageing, disease and degeneration. Their roles as signalling molecules under normal physiological conditions is less well understood. Recent studies have provided ample evidence of ROS-regulating neuronal development and function, from the establishment of neuronal polarity to growth cone pathfinding; from the regulation of connectivity and synaptic transmission to the tuning of neuronal networks. Appreciation of the varied processes that are subject to regulation by ROS might help us understand how changes in ROS metabolism and buffering could progressively impact on neuronal networks with age and disease.
Collapse
Affiliation(s)
| | - Nathan Garnham
- Department of BiologyUniversity of YorkHeslington YorkUK
| | | | | |
Collapse
|
48
|
Liang D. A Salutary Role of Reactive Oxygen Species in Intercellular Tunnel-Mediated Communication. Front Cell Dev Biol 2018; 6:2. [PMID: 29503816 PMCID: PMC5821100 DOI: 10.3389/fcell.2018.00002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 01/18/2018] [Indexed: 12/17/2022] Open
Abstract
The reactive oxygen species, generally labeled toxic due to high reactivity without target specificity, are gradually uncovered as signaling molecules involved in a myriad of biological processes. But one important feature of ROS roles in macromolecule movement has not caught attention until recent studies with technique advance and design elegance have shed lights on ROS signaling for intercellular and interorganelle communication. This review begins with the discussions of genetic and chemical studies on the regulation of symplastic dye movement through intercellular tunnels in plants (plasmodesmata), and focuses on the ROS regulatory mechanisms concerning macromolecule movement including small RNA-mediated gene silencing movement and protein shuttling between cells. Given the premise that intercellular tunnels (bridges) in mammalian cells are the key physical structures to sustain intercellular communication, movement of macromolecules and signals is efficiently facilitated by ROS-induced membrane protrusions formation, which is analogously applied to the interorganelle communication in plant cells. Although ROS regulatory differences between plant and mammalian cells exist, the basis for ROS-triggered conduit formation underlies a unifying conservative theme in multicellular organisms. These mechanisms may represent the evolutionary advances that have enabled multicellularity to gain the ability to generate and utilize ROS to govern material exchanges between individual cells in oxygenated environment.
Collapse
Affiliation(s)
- Dacheng Liang
- Hubei Collaborative Innovation Center for Grain Industry, School of Agriculture, Yangtze University, Jingzhou, China.,Engineering Research Center of Ecology and Agricultural Use of Wetland, Ministry of Education, Yangtze University, Jingzhou, China
| |
Collapse
|
49
|
Horn A, Van der Meulen JH, Defour A, Hogarth M, Sreetama SC, Reed A, Scheffer L, Chandel NS, Jaiswal JK. Mitochondrial redox signaling enables repair of injured skeletal muscle cells. Sci Signal 2017; 10:eaaj1978. [PMID: 28874604 PMCID: PMC5949579 DOI: 10.1126/scisignal.aaj1978] [Citation(s) in RCA: 114] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Strain and physical trauma to mechanically active cells, such as skeletal muscle myofibers, injures their plasma membranes, and mitochondrial function is required for their repair. We found that mitochondrial function was also needed for plasma membrane repair in myoblasts as well as nonmuscle cells, which depended on mitochondrial uptake of calcium through the mitochondrial calcium uniporter (MCU). Calcium uptake transiently increased the mitochondrial production of reactive oxygen species (ROS), which locally activated the guanosine triphosphatase (GTPase) RhoA, triggering F-actin accumulation at the site of injury and facilitating membrane repair. Blocking mitochondrial calcium uptake or ROS production prevented injury-triggered RhoA activation, actin polymerization, and plasma membrane repair. This repair mechanism was shared between myoblasts, nonmuscle cells, and mature skeletal myofibers. Quenching mitochondrial ROS in myofibers during eccentric exercise ex vivo caused increased damage to myofibers, resulting in a greater loss of muscle force. These results suggest a physiological role for mitochondria in plasma membrane repair in injured cells, a role that highlights a beneficial effect of ROS.
Collapse
Affiliation(s)
- Adam Horn
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010-2970, USA
| | - Jack H Van der Meulen
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Aurelia Defour
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Marshall Hogarth
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Sen Chandra Sreetama
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Aaron Reed
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Luana Scheffer
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA
| | - Navdeep S Chandel
- Department of Medicine, Feinberg School of Medicine, Northwestern University, Chicago, IL 60611, USA
| | - Jyoti K Jaiswal
- Children's National Health System, Center for Genetic Medicine Research, 111 Michigan Avenue Northwest, Washington, DC 20010-2970, USA.
- Department of Integrative Systems Biology, George Washington University School of Medicine and Health Sciences, Washington, DC 20010-2970, USA
| |
Collapse
|
50
|
Redox regulation in tumor cell epithelial-mesenchymal transition: molecular basis and therapeutic strategy. Signal Transduct Target Ther 2017; 2:17036. [PMID: 29263924 PMCID: PMC5661624 DOI: 10.1038/sigtrans.2017.36] [Citation(s) in RCA: 141] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2016] [Revised: 04/25/2017] [Accepted: 04/25/2017] [Indexed: 02/05/2023] Open
Abstract
Epithelial–mesenchymal transition (EMT) is recognized as a driving force of cancer cell metastasis and drug resistance, two leading causes of cancer recurrence and cancer-related death. It is, therefore, logical in cancer therapy to target the EMT switch to prevent such cancer metastasis and recurrence. Previous reports have indicated that growth factors (such as epidermal growth factor and fibroblast growth factor) and cytokines (such as the transforming growth factor beta (TGF-β) family) are major stimulators of EMT. However, the mechanisms underlying EMT initiation and progression remain unclear. Recently, emerging evidence has suggested that reactive oxygen species (ROS), important cellular secondary messengers involved in diverse biological events in cancer cells, play essential roles in the EMT process in cancer cells by regulating extracellular matrix (ECM) remodeling, cytoskeleton remodeling, cell–cell junctions, and cell mobility. Thus, targeting EMT by manipulating the intracellular redox status may hold promise for cancer therapy. Herein, we will address recent advances in redox biology involved in the EMT process in cancer cells, which will contribute to the development of novel therapeutic strategies by targeting redox-regulated EMT for cancer treatment.
Collapse
|