1
|
Chakraborty S, Choudhuri A, Mishra A, Sengupta R. S-nitrosylation and S-glutathionylation: Lying at the forefront of redox dichotomy or a visible synergism? Biochem Biophys Res Commun 2025; 761:151734. [PMID: 40179738 DOI: 10.1016/j.bbrc.2025.151734] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2024] [Revised: 03/06/2025] [Accepted: 03/29/2025] [Indexed: 04/05/2025]
Abstract
The discovery of novel oxidoreductases and their specific functional revelations as cellular disulfide reductants, S-denitrosylases, or S-deglutathionylases, alongside the well-established major redoxins/antioxidant systems comprising thioredoxin and glutaredoxin, enlarges the spectrum of redox players in the intracellular milieu as well as pushes us to stand at the crossroads concerning the choice of antioxidants that can serve the benefit of catalyzing their cognate protein/non-protein substrates with better efficiencies than the rest. The complexity is extended to exploring the redundancy amongst the redoxin systems and identifying their overlapping or unique substrate preferences to intervene with oxidative or nitrosative stress-induced reversible protein posttranslational modifications such as S-nitrosylation and S-glutathionylation. Contrary to popular expectations of reiterating the theoretical and evidence-based existence of these modifications, the current review aims to take the first leap in delineating the logical reasons behind the competing susceptibility of reactive cysteine thiols toward either or both redox modifications and their subsequent extent of stability in the presence of cellular reductants (thioredoxin, glutaredoxin, thioredoxin-like mimetic or lipoic acid, dihydrolipoic acid, and glutathione), thus rebuilding the underpinnings of a 'redox-interactome' that can further pave the way for the global mapping of ideal substrates exhibiting stringencies or synergism in the context of translational redox research.
Collapse
Affiliation(s)
- Surupa Chakraborty
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Ankita Choudhuri
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Akansha Mishra
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India
| | - Rajib Sengupta
- Amity Institute of Biotechnology Kolkata, Amity University Kolkata, Action Area II, Rajarhat, Newtown, Kolkata, West Bengal, 700135, India.
| |
Collapse
|
2
|
Kabir A, Mukilarasi B, Manohar A, Gadani M, Sinha AK, Sharma P, Verma A, Selvaraj V, Sudhakar S. Protein bioactive complexes promote osteogenesis under microgravity environment. Int J Biol Macromol 2025; 303:140483. [PMID: 39904451 DOI: 10.1016/j.ijbiomac.2025.140483] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2024] [Revised: 01/25/2025] [Accepted: 01/28/2025] [Indexed: 02/06/2025]
Abstract
The space microgravity environment and cosmic radiation pose a significant threat to musculoskeletal health, particularly bone mass. However, the critical mechanism underlying space-induced bone loss and its relation to cellular oxidative stress remains unclear. Currently used bone-loss-reversing drugs face limitations like poor efficacy and metabolic defects. Herein, we revealed that simulated microgravity (SMG) induces reactive oxygen species (ROS), negatively impacting osteoblasts, causing cytoskeletal damage, and downregulating osteogenic genes. To combat this, we designed protein-zein nanocages loaded with a chimeric non-enzymatic cocktail (ZNAC) containing ascorbic acid, resveratrol, luteolin, coenzyme Q, and glutathione. These nanocages (~200 nm) demonstrated excellent stability, biocompatibility, and antioxidant properties compared to free drugs. We investigated the effects of ZNAC under SMG using two experimental models: MC3T3 pre-osteoblast/MG63 osteoblasts and regenerating zebrafish scales that represent compositional and physiological/pathophysiological analogy with mammalian system. ZNAC effectively reduced SMG-induced ROS, preserved cytoskeletal integrity, and enhanced alkaline phosphatase (ALP) activity along with the expression of osteogenic genes such as RUNX2 and Col1A1. In zebrafish scales, it increased osteogenic gene expression, calcification, and the calcium/phosphorus ratio, indicating enhanced scale regeneration. These findings suggest that ZNAC is a promising candidate for bone regeneration, offering potential solutions for maintaining astronaut health during extended space missions.
Collapse
Affiliation(s)
- Anisha Kabir
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - B Mukilarasi
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Anagha Manohar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Maulesh Gadani
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Kumar Sinha
- Human Space Flight Centre, Antariksh Bhavan, New BEL Road, Bengaluru 560 094, India.
| | - Payal Sharma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Anurag Verma
- Space Applications Centre, Indian Space Research Organisation, Ahmedabad, Gujarat 380015, India.
| | - Vimalraj Selvaraj
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| | - Swathi Sudhakar
- Department of Applied Mechanics and Biomedical Engineering, Indian Institute of Technology Madras, Chennai, Tamil Nadu 600036, India.
| |
Collapse
|
3
|
Huang HL, Suchenko A, Grandinetti G, Balasubramanian MK, Chinthalapudi K, Heissler SM. Cryo-EM structures of cardiac muscle α-actin mutants M305L and A331P give insights into the structural mechanisms of hypertrophic cardiomyopathy. Eur J Cell Biol 2024; 103:151460. [PMID: 39393252 PMCID: PMC11611453 DOI: 10.1016/j.ejcb.2024.151460] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2024] [Revised: 09/28/2024] [Accepted: 09/29/2024] [Indexed: 10/13/2024] Open
Abstract
Cardiac muscle α-actin is a key protein of the thin filament in the muscle sarcomere that, together with myosin thick filaments, produce force and contraction important for normal heart function. Missense mutations in cardiac muscle α-actin can cause hypertrophic cardiomyopathy, a complex disorder of the heart characterized by hypercontractility at the molecular scale that leads to diverse clinical phenotypes. While the clinical aspects of hypertrophic cardiomyopathy have been extensively studied, the molecular mechanisms of missense mutations in cardiac muscle α-actin that cause the disease remain largely elusive. Here we used cryo-electron microscopy to reveal the structures of hypertrophic cardiomyopathy-associated actin mutations M305L and A331P in the filamentous state. We show that the mutations have subtle impacts on the overall architecture of the actin filament with mutation-specific changes in the nucleotide binding cleft active site, interprotomer interfaces, and local changes around the mutation site. This suggests that structural changes induced by M305L and A331P have implications for the positioning of the thin filament protein tropomyosin and the interaction with myosin motors. Overall, this study supports a structural model whereby altered interactions between thick and thin filament proteins contribute to disease mechanisms in hypertrophic cardiomyopathy.
Collapse
Affiliation(s)
- Hsiang-Ling Huang
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA
| | - Andrejus Suchenko
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Giovanna Grandinetti
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA; Center for Electron Microscopy and Analysis, College of Engineering, The Ohio State University, Columbus, OH, USA
| | - Mohan K Balasubramanian
- Centre for Mechanochemical Cell Biology and Warwick Medical School, Division of Biomedical Sciences, Coventry, United Kingdom
| | - Krishna Chinthalapudi
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| | - Sarah M Heissler
- Department of Physiology and Cell Biology, Dorothy M. Davis Heart and Lung Research Institute, College of Medicine, The Ohio State University, Columbus, OH, USA.
| |
Collapse
|
4
|
Zheng L, Boeren S, Liu C, Bakker W, Wang H, Rietjens IMCM, Saccenti E. Proteomics-based identification of biomarkers reflecting endogenous and exogenous exposure to the advanced glycation end product precursor methylglyoxal in SH-SY5Y human neuroblastoma cells. Int J Biol Macromol 2024; 272:132859. [PMID: 38838889 DOI: 10.1016/j.ijbiomac.2024.132859] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2024] [Revised: 05/30/2024] [Accepted: 06/01/2024] [Indexed: 06/07/2024]
Abstract
Methylglyoxal (MGO), a highly reactive precursor of advanced glycation end products, is endogenously produced and prevalent in various food products. This study aimed to characterize protein modifications in SH-SY5Y human neuroblastoma cells induced by MGO and identify potential biomarkers for its exposure and toxicity. A shot-gun proteomic analysis was applied to characterize protein modifications in cells incubated with and without exogenous MGO. Seventy-seven proteins were identified as highly susceptible to MGO modification, among which eight, including vimentin and histone H2B type 2-F, showing concentration-dependent modifications by externally added MGO, were defined as biomarkers for exogenous MGO exposure. Remarkably, up to 10 modification sites were identified on vimentin. Myosin light polypeptide 6 emerged as a biomarker for MGO toxicity, with modifications exclusively observed under cytotoxic MGO levels. Additionally, proteins like serine/threonine-protein kinase SIK2 and calcyphosin, exhibiting comparable or even higher modification levels in control compared to exogenous MGO-treated cells, were defined as biomarkers for endogenous exposure. Bioinformatics analysis revealed that motor proteins, cytoskeleton components, and glycolysis proteins were overrepresented among those highly susceptible to MGO modification. These results identify biomarkers for both endogenous and exogenous MGO exposure and provide insights into the cellular effects of endogenously formed versus externally added MGO.
Collapse
Affiliation(s)
- Liang Zheng
- Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, the Netherlands.
| | - Sjef Boeren
- Laboratory of Biochemistry, Wageningen University and Research, 6708 WE Wageningen, the Netherlands
| | - Chen Liu
- Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, the Netherlands; Tea Refining and Innovation Key Laboratory of Sichuan Province, College of Horticulture, Sichuan Agricultural University, Chengdu 611130, China
| | - Wouter Bakker
- Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, the Netherlands
| | - Haomiao Wang
- Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, the Netherlands
| | - Ivonne M C M Rietjens
- Division of Toxicology, Wageningen University and Research, 6708 WE Wageningen, the Netherlands
| | - Edoardo Saccenti
- Laboratory of Systems and Synthetic Biology, Wageningen University & Research, 6708 WE Wageningen, the Netherlands
| |
Collapse
|
5
|
Smith FM, Kosman DJ. Loss of filamentous actin, tight junction protein expression, and paracellular barrier integrity in frataxin-deficient human brain microvascular endothelial cells-implications for blood-brain barrier physiology in Friedreich's ataxia. Front Mol Biosci 2024; 10:1299201. [PMID: 38274097 PMCID: PMC10808331 DOI: 10.3389/fmolb.2023.1299201] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Accepted: 12/22/2023] [Indexed: 01/27/2024] Open
Abstract
Introduction: Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia. FRDA results from loss of Frataxin (FXN), an essential mitochondrial iron trafficking protein. FRDA starts with an early burst of neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by progressive brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both brain and heart homeostasis, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Previous reports have identified cytoskeletal alterations in non-barrier forming FRDA cell models, but physiological consequences are limited. Methods: We investigated brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). We have knocked down FXN in immortalized human brain microvascular endothelial cells (hBMVEC), which compose the microcapillaries of the BBB, by using shRNA. We confirmed known cellular pathophysiologies of FXN-knockdown including decreased energy metabolism, markers of oxidative stress, and increased cell size. Results: We investigated cytoskeletal architecture, identifying decreased filamentous actin and Occludin and Claudin-5 tight junction protein expression in shFXN hBMVECs. This was consistent with decreased transendothelial electrical resistance (TEER) and increased paracellular tracer flux during early barrier formation. shFXN hBMVEC start with only 67% barrier integrity of the controls, and flux a paracellular tracer at 800% of physiological levels. Discussion: We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and tight junction protein abundance, co-incident with increased barrier permeability. Changes in the integrity of the BBB may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Furthermore, our findings implicate other barrier cells, e.g., the cardiac microvasculature, loci of disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M. Smith
- Jacobs School of Medicine and Biomedical Sciences, Department of Biochemistry, The State University of New York at Buffalo, Buffalo, NY, United States
| | | |
Collapse
|
6
|
Elkrief D, Matusovsky O, Cheng YS, Rassier DE. From amino-acid to disease: the effects of oxidation on actin-myosin interactions in muscle. J Muscle Res Cell Motil 2023; 44:225-254. [PMID: 37805961 DOI: 10.1007/s10974-023-09658-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2023] [Accepted: 08/15/2023] [Indexed: 10/10/2023]
Abstract
Actin-myosin interactions form the basis of the force-producing contraction cycle within the sarcomere, serving as the primary mechanism for muscle contraction. Post-translational modifications, such as oxidation, have a considerable impact on the mechanics of these interactions. Considering their widespread occurrence, the explicit contributions of these modifications to muscle function remain an active field of research. In this review, we aim to provide a comprehensive overview of the basic mechanics of the actin-myosin complex and elucidate the extent to which oxidation influences the contractile cycle and various mechanical characteristics of this complex at the single-molecule, myofibrillar and whole-muscle levels. We place particular focus on amino acids shown to be vulnerable to oxidation in actin, myosin, and some of their binding partners. Additionally, we highlight the differences between in vitro environments, where oxidation is controlled and limited to actin and myosin and myofibrillar or whole muscle environments, to foster a better understanding of oxidative modification in muscle. Thus, this review seeks to encompass a broad range of studies, aiming to lay out the multi layered effects of oxidation in in vitro and in vivo environments, with brief mention of clinical muscular disorders associated with oxidative stress.
Collapse
Affiliation(s)
- Daren Elkrief
- Department of Physiology, McGill University, Montreal, QC, Canada
| | - Oleg Matusovsky
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Yu-Shu Cheng
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada
| | - Dilson E Rassier
- Department of Physiology, McGill University, Montreal, QC, Canada.
- Department of Kinesiology and Physical Education, McGill University, Montreal, QC, Canada.
- Simon Fraser University, Burnaby, BC, Canada.
| |
Collapse
|
7
|
Mahoney SA, Dey AK, Basisty N, Herman AB. Identification and functional analysis of senescent cells in the cardiovascular system using omics approaches. Am J Physiol Heart Circ Physiol 2023; 325:H1039-H1058. [PMID: 37656130 PMCID: PMC10908411 DOI: 10.1152/ajpheart.00352.2023] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/15/2023] [Revised: 08/28/2023] [Accepted: 08/28/2023] [Indexed: 09/02/2023]
Abstract
Cardiovascular disease (CVD) is a leading cause of morbidity and mortality worldwide, and senescent cells have emerged as key contributors to its pathogenesis. Senescent cells exhibit cell cycle arrest and secrete a range of proinflammatory factors, termed the senescence-associated secretory phenotype (SASP), which promotes tissue dysfunction and exacerbates CVD progression. Omics technologies, specifically transcriptomics and proteomics, offer powerful tools to uncover and define the molecular signatures of senescent cells in cardiovascular tissue. By analyzing the comprehensive molecular profiles of senescent cells, omics approaches can identify specific genetic alterations, gene expression patterns, protein abundances, and metabolite levels associated with senescence in CVD. These omics-based discoveries provide insights into the mechanisms underlying senescence-induced cardiovascular damage, facilitating the development of novel diagnostic biomarkers and therapeutic targets. Furthermore, integration of multiple omics data sets enables a systems-level understanding of senescence in CVD, paving the way for precision medicine approaches to prevent or treat cardiovascular aging and its associated complications.
Collapse
Affiliation(s)
- Sophia A Mahoney
- Department of Integrative Physiology, University of Colorado at Boulder, Boulder, Colorado, United States
| | - Amit K Dey
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Nathan Basisty
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| | - Allison B Herman
- Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, Maryland, United States
| |
Collapse
|
8
|
Medvedeva MV, Kleimenov SY, Samygina VR, Muronetz VI, Schmalhausen EV. S-nitrosylation and S-glutathionylation of GAPDH: Similarities, differences, and relationships. Biochim Biophys Acta Gen Subj 2023; 1867:130418. [PMID: 37355052 DOI: 10.1016/j.bbagen.2023.130418] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2023] [Revised: 05/24/2023] [Accepted: 06/20/2023] [Indexed: 06/26/2023]
Abstract
The aim of this work was to compare the effect of reversible post-translational modifications, S-nitrosylation and S-glutathionylation, on the properties of glyceraldehyde-3-phosphate dehydrogenase (GAPDH), and to reveal the mechanism of the relationship between these modifications. Comparison of S-nitrosylated and S-glutathionylated GAPDH showed that both modifications inactivate the enzyme and change its spatial structure, decreasing the thermal stability of the protein and increasing its sensitivity to trypsin cleavage. Both modifications are reversible in the presence of dithiothreitol, however, in the presence of reduced glutathione and glutaredoxin 1, the reactivation of S-glutathionylated GAPDH is much slower (10% in 2 h) compared to S-nitrosylated GAPDH (60% in 10 min). This suggests that S-glutathionylation is a much less reversible modification compared to S-nitrosylation. Incubation of HEK 293 T cells in the presence of H2O2 or with the NO donor diethylamine NONOate results in accumulation of sulfenated GAPDH (by data of Western blotting) and S-glutathionylated GAPDH (by data of immunoprecipitation with anti-GSH antibodies). Besides GAPDH, a protein of 45 kDa was found to be sulfenated and S-glutathionylated in the cells treated with H2O2 or NO. This protein was identified as beta-actin. The results of this study confirm the previously proposed hypothesis based on in vitro investigations, according to which S-nitrosylation of the catalytic cysteine residue (Cys152) of GAPDH with subsequent formation of cysteine sulfenic acid at Cys152 may promote its S-glutathionylation in the presence of cellular GSH. Presumably, the mechanism may be valid in the case of beta-actin.
Collapse
Affiliation(s)
- M V Medvedeva
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia
| | - S Yu Kleimenov
- Bach Institute of Biochemistry, Research Center of Biotechnology of Russian Academy of Sciences, Leninsky prospect 33, bld. 2, Moscow 119071, Russia; Koltzov Institute of Developmental Biology of Russian Academy of Sciences, ul. Vavilova 26, Moscow 119334, Russia
| | - V R Samygina
- Shubnikov Institute of Crystallography of Federal Scientific Research Centre Crystallography and Photonics of Russian Academy of Sciences, Leninsky prospect 59, Moscow 119333, Russia
| | - V I Muronetz
- Faculty of Bioengineering and Bioinformatics, Lomonosov Moscow State University, Moscow 119991, Russia; Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia
| | - E V Schmalhausen
- Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, Moscow 119991, Russia.
| |
Collapse
|
9
|
Smith FM, Kosman DJ. Frataxin-deficient human brain microvascular endothelial cells lose polymerized actin and are paracellularly permeable -implications for blood-brain barrier integrity in Friedreich's Ataxia. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.02.09.527936. [PMID: 36798283 PMCID: PMC9934603 DOI: 10.1101/2023.02.09.527936] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/12/2023]
Abstract
Background Friedreich's Ataxia (FRDA) is the most prevalent inherited ataxia; the disease results from loss of Frataxin, an essential mitochondrial iron trafficking protein. FRDA presents as neurodegeneration of the dorsal root ganglion and cerebellar dentate nuclei, followed by brain iron accumulation in the latter. End stage disease includes cardiac fibrosis that contributes to hypertrophic cardiomyopathy. The microvasculature plays an essential barrier role in both the brain and heart, thus an investigation of this tissue system in FRDA is essential to the delineation of the cellular dysfunction in this genetic disorder. Here, we investigate brain microvascular endothelial cell integrity in FRDA in a model of the blood-brain barrier (BBB). Methods We used lentiviral mediated shRNA delivery to generate a novel FRDA model in immortalized human brain microvascular endothelial cells (hBMVEC) that compose the microcapillaries of the BBB. We verified known cellular pathophysiologies of FXN knockdown including increased oxidative stress, loss of energy metabolism, and increased cell size. Furthermore, we investigated cytoskeletal architecture including the abundance and organization of filamentous actin, and barrier physiology via transendothelial electrical resistance and fluorescent tracer flux. Results shFXN hBMVEC display the known FRDA cell morbidity including increased oxidative stress, decreased energy metabolism, and an increase in cell size. We demonstrate that shFXN hBMVEC have less overall filamentous actin, and that filamentous actin is lost at the cell membrane and cortical actin ring. Consistent with loss of cytoskeletal structure and anchorage, we found decreased barrier strength and increased paracellular tracer flux in the shFXN hBMVEC transwell model. Conclusion We identified that insufficient FXN levels in the hBMVEC BBB model causes changes in cytoskeletal architecture and increased barrier permeability, cell pathologies that may be related to patient brain iron accumulation, neuroinflammation, neurodegeneration, and stroke. Our findings implicate other barrier cells, e.g., the cardiac microvasculature, likely contributory also to disease pathology in FRDA.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| | - Daniel J Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, The University of New York at Buffalo
| |
Collapse
|
10
|
Rosas PC, Solaro RJ. Implications of S-glutathionylation of sarcomere proteins in cardiac disorders, therapies, and diagnosis. Front Cardiovasc Med 2023; 9:1060716. [PMID: 36762302 PMCID: PMC9902711 DOI: 10.3389/fcvm.2022.1060716] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2022] [Accepted: 12/29/2022] [Indexed: 01/25/2023] Open
Abstract
The discovery that cardiac sarcomere proteins are substrates for S-glutathionylation and that this post-translational modification correlates strongly with diastolic dysfunction led to new concepts regarding how levels of oxidative stress affect the heartbeat. Major sarcomere proteins for which there is evidence of S-glutathionylation include cardiac myosin binding protein C (cMyBP-C), actin, cardiac troponin I (cTnI) and titin. Our hypothesis is that these S-glutathionylated proteins are significant factors in acquired and familial disorders of the heart; and, when released into the serum, provide novel biomarkers. We consider the molecular mechanisms for these effects in the context of recent revelations of how these proteins control cardiac dynamics in close collaboration with Ca2+ fluxes. These revelations were made using powerful approaches and technologies that were focused on thin filaments, thick filaments, and titin filaments. Here we integrate their regulatory processes in the sarcomere as modulated mainly by neuro-humoral control of phosphorylation inasmuch evidence indicates that S-glutathionylation and protein phosphorylation, promoting increased dynamics and modifying the Frank-Starling relation, may be mutually exclusive. Earlier studies demonstrated that in addition to cTnI as a well-established biomarker for cardiac disorders, serum levels of cMyBP-C are also a biomarker for cardiac disorders. We describe recent studies approaching the question of whether serum levels of S-glutathionylated-cMyBP-C could be employed as an important clinical tool in patient stratification, early diagnosis in at risk patients before HFpEF, determination of progression, effectiveness of therapeutic approaches, and as a guide in developing future therapies.
Collapse
Affiliation(s)
- Paola C. Rosas
- Department of Pharmacy Practice, College of Pharmacy, Chicago, IL, United States
| | - R. John Solaro
- Department of Physiology and Biophysics, College of Medicine, University of Illinois at Chicago, Chicago, IL, United States
| |
Collapse
|
11
|
Vrettou S, Wirth B. S-Glutathionylation and S-Nitrosylation in Mitochondria: Focus on Homeostasis and Neurodegenerative Diseases. Int J Mol Sci 2022; 23:15849. [PMID: 36555492 PMCID: PMC9779533 DOI: 10.3390/ijms232415849] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2022] [Revised: 11/24/2022] [Accepted: 12/12/2022] [Indexed: 12/15/2022] Open
Abstract
Redox post-translational modifications are derived from fluctuations in the redox potential and modulate protein function, localization, activity and structure. Amongst the oxidative reversible modifications, the S-glutathionylation of proteins was the first to be characterized as a post-translational modification, which primarily protects proteins from irreversible oxidation. However, a growing body of evidence suggests that S-glutathionylation plays a key role in core cell processes, particularly in mitochondria, which are the main source of reactive oxygen species. S-nitrosylation, another post-translational modification, was identified >150 years ago, but it was re-introduced as a prototype cell-signaling mechanism only recently, one that tightly regulates core processes within the cell’s sub-compartments, especially in mitochondria. S-glutathionylation and S-nitrosylation are modulated by fluctuations in reactive oxygen and nitrogen species and, in turn, orchestrate mitochondrial bioenergetics machinery, morphology, nutrients metabolism and apoptosis. In many neurodegenerative disorders, mitochondria dysfunction and oxidative/nitrosative stresses trigger or exacerbate their pathologies. Despite the substantial amount of research for most of these disorders, there are no successful treatments, while antioxidant supplementation failed in the majority of clinical trials. Herein, we discuss how S-glutathionylation and S-nitrosylation interfere in mitochondrial homeostasis and how the deregulation of these modifications is associated with Alzheimer’s, Parkinson’s, amyotrophic lateral sclerosis and Friedreich’s ataxia.
Collapse
Affiliation(s)
- Sofia Vrettou
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
| | - Brunhilde Wirth
- Institute of Human Genetics, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
- Center for Molecular Medicine Cologne, University of Cologne, 50931 Cologne, Germany
- Institute for Genetics, University of Cologne, 50674 Cologne, Germany
- Center for Rare Diseases, University Hospital of Cologne, University of Cologne, 50931 Cologne, Germany
| |
Collapse
|
12
|
A. HP, Diwakar L, Ravindranath V. Protein Glutathionylation and Glutaredoxin: Role in Neurodegenerative Diseases. Antioxidants (Basel) 2022; 11:antiox11122334. [PMID: 36552543 PMCID: PMC9774553 DOI: 10.3390/antiox11122334] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 11/14/2022] [Accepted: 11/15/2022] [Indexed: 11/29/2022] Open
Abstract
Oxidative stress has been implicated in the pathogenesis and progression of many neurodegenerative disorders including Parkinson's disease and Alzheimer's disease. One of the major enzyme systems involved in the defense against reactive oxygen species are the tripeptide glutathione and oxidoreductase glutaredoxin. Glutathione and glutaredoxin system are very important in the brain because of the oxidative modification of protein thiols to protein glutathione mixed disulfides with the concomitant formation of oxidized glutathione during oxidative stress. Formation of Pr-SSG acts as a sink in the brain and is reduced back to protein thiols during recovery, thus restoring protein functions. This is unlike in the liver, which has a high turnover of glutathione, and formation of Pr-SSG is very minimal as liver is able to quickly quench the prooxidant species. Given the important role glutathione and glutaredoxin play in the brain, both in normal and pathologic states, it is necessary to study ways to augment the system to help maintain the protein thiol status. This review details the importance of glutathione and glutaredoxin systems in several neurodegenerative disorders and emphasizes the potential augmentation of this system as a target to effectively protect the brain during aging.
Collapse
Affiliation(s)
- Haseena P. A.
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
- Manipal Academy of Higher Education (MAHE), Manipal 576104, India
| | - Latha Diwakar
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
| | - Vijayalakshmi Ravindranath
- Centre for Brain Research, Indian Institute of Science, Bangalore 560012, India
- Correspondence: ; Tel.: +91-80-22933433; Fax: +91-80-23603323
| |
Collapse
|
13
|
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: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [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
|
14
|
Hadpech S, Peerapen P, Thongboonkerd V. Alpha-tubulin relocalization is involved in calcium oxalate-induced tight junction disruption in renal epithelial cells. Chem Biol Interact 2022; 368:110236. [DOI: 10.1016/j.cbi.2022.110236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Revised: 10/14/2022] [Accepted: 10/21/2022] [Indexed: 11/03/2022]
|
15
|
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
|
16
|
Jeyavani J, Sibiya A, Gopi N, Mahboob S, Riaz MN, Vaseeharan B. Dietary consumption of polypropylene microplastics alter the biochemical parameters and histological response in freshwater benthic mollusc Pomacea paludosa. ENVIRONMENTAL RESEARCH 2022; 212:113370. [PMID: 35504343 DOI: 10.1016/j.envres.2022.113370] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 09/28/2021] [Revised: 03/27/2022] [Accepted: 04/22/2022] [Indexed: 05/06/2023]
Abstract
One of the most common environmental pollutant in aquatic ecosystems are polypropylene microplastics and their impacts on aquatic organisms are still scarce. The study aimed to prepare polypropylene microplastics using organic solvent (spherical and 11.86-44.62 μm) and then test their toxicity on the freshwater benthic mollusc grazer Pomaceae paludosa. The present study investigated chronic (28 days) exposure of polypropylene microplastics via dietary supplements (250 mg kg-1, 500 mg kg-1 & 750 mg kg-1) in P. paludosa, and the toxic effect was evaluated in digestive gland tissue. The FTIR results revealed no change in polypropylene microplastics during ingestion or after egestion. On the other hand, Ingestion causes accumulation in their bodies and disrupts redox homeostasis. Meanwhile, alteration occurs in oxidative stress-related biomarkers such as increased reactive oxygen species level (ROS), impaired the biochemical parameters of antioxidant system catalase (CAT), glutathione peroxidase (GPx), reduced glutathione (GSH), and glutathione - S- transferase (GST), deterioration of oxidative stress effects in lipid peroxidation (LPO) and carbonyl protein (CP) and changed the digestive enzymes such as amylase, pepsin, esterase and alkaline phosphatase that are measured in hepatopancreas tissue. The histology results revealed that ingesting these microplastics caused severe damage to the digestive gland cells. According to the findings, ingestion of polypropylene microplastics in benthic freshwater mollusc causes more serious harm and impacts energy acquisition. This finding represents the ecological risk of polypropylene microplastic pollution in the freshwater ecosystem.
Collapse
Affiliation(s)
- Jeyaraj Jeyavani
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630003, Tamil Nadu, India
| | - Ashokkumar Sibiya
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630003, Tamil Nadu, India
| | - Narayanan Gopi
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630003, Tamil Nadu, India
| | - Shahid Mahboob
- Department of Zoology, College of Science, King Saud University, Riyadh, 11451, Saudi Arabia
| | | | - Baskaralingam Vaseeharan
- Biomaterials and Biotechnology in Animal Health Lab, Department of Animal Health and Management, Alagappa University, Karaikudi, 630003, Tamil Nadu, India.
| |
Collapse
|
17
|
Morellon-Sterling R, Bolivar JM, Fernandez-Lafuente R. Switch off/switch on of a cysteinyl protease as a way to preserve the active catalytic group by modification with a reversible covalent thiol modifier: Immobilization of ficin on vinyl-sulfone activated supports. Int J Biol Macromol 2022; 220:1155-1162. [PMID: 36037909 DOI: 10.1016/j.ijbiomac.2022.08.155] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2022] [Revised: 08/11/2022] [Accepted: 08/23/2022] [Indexed: 11/05/2022]
Abstract
The immobilization of ficin (a cysteinyl proteases) on vinyl sulfone agarose produced its almost full inactivation. It was observed that the incubation of the free and immobilized enzyme in β-mercaptoethanol produced a 20 % of enzyme activity recovery, suggesting that the inactivation due to the immobilization could be a consequence of the modification of the catalytic Cys. To prevent the enzyme inactivation during the immobilization, switching off of ficin via Cys reaction with dipyridyl-disulfide was implemented, giving a reversible disulfide bond that produced a fully inactive enzyme. The switch on of ficin activity was implemented by incubation in 1 M β-mercaptoethanol. Using this strategy to immobilize the enzyme on vinyl sulfone agarose beads, the expressed activity of the immobilized ficin could be boosted up to 80 %. The immobilized enzyme presented a thermal stabilization similar to that obtained using ficin-glyoxyl-agarose beads. This procedure may be extended to many enzymes containing critical Cys, to permit their immobilization or chemical modification.
Collapse
Affiliation(s)
- Roberto Morellon-Sterling
- Departamento de Biocatálisis, ICP-CSIC, Marie Curie 2, Campus UAM-CSIC Cantoblanco, 28049 Madrid, Spain; Student of Departamento de Biología Molecular, Universidad Autónoma de Madrid, Darwin 2, Campus UAM-CSIC, Cantoblanco, 28049 Madrid, Spain
| | - Juan M Bolivar
- FQPIMA Group, Chemical and Materials Engineering Department, Faculty of Chemical Sciences, Complutense University of Madrid, Complutense Ave., Madrid 28040, Spain
| | - Roberto Fernandez-Lafuente
- Departamento de Biocatálisis, ICP-CSIC, Marie Curie 2, Campus UAM-CSIC Cantoblanco, 28049 Madrid, Spain; Center of Excellence in Bionanoscience Research, External Scientific Advisory Academics, King Abdulaziz University, Jeddah 21589, Saudi Arabia.
| |
Collapse
|
18
|
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: 37] [Impact Index Per Article: 12.3] [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
|
19
|
Massai L, Messori L, Carpentieri A, Amoresano A, Melchiorre C, Fiaschi T, Modesti A, Gamberi T, Magherini F. The effects of two gold-N-heterocyclic carbene (NHC) complexes in ovarian cancer cells: a redox proteomic study. Cancer Chemother Pharmacol 2022; 89:809-823. [PMID: 35543764 PMCID: PMC9135895 DOI: 10.1007/s00280-022-04438-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 04/15/2022] [Indexed: 01/04/2023]
Abstract
PURPOSE Ovarian cancer is the fifth leading cause of cancer-related deaths in women. Standard treatment consists of tumor debulking surgery followed by platinum and paclitaxel chemotherapy; yet, despite the initial response, about 70-75% of patients develop resistance to chemotherapy. Gold compounds represent a family of very promising anticancer drugs. Among them, we previously investigated the cytotoxic and pro-apoptotic properties of Au(NHC) and Au(NHC)2PF6, i.e., a monocarbene gold(I) complex and the corresponding bis(carbene) complex. Gold compounds are known to alter the redox state of cells interacting with free cysteine and selenocysteine residues of several proteins. Herein, a redox proteomic study has been carried out to elucidate the mechanisms of cytotoxicity in A2780 human ovarian cancer cells. METHODS A biotinylated iodoacetamide labeling method coupled with mass spectrometry was used to identify oxidation-sensitive protein cysteines. RESULTS Gold carbene complexes cause extensive oxidation of several cellular proteins; many affected proteins belong to two major functional classes: carbohydrate metabolism, and cytoskeleton organization/cell adhesion. Among the affected proteins, Glyceraldehyde-3-phosphate dehydrogenase inhibition was proved by enzymatic assays and by ESI-MS studies. We also found that Au(NHC)2PF6 inhibits mitochondrial respiration impairing complex I function. Concerning the oxidized cytoskeletal proteins, gold binding to the free cysteines of actin was demonstrated by ESI-MS analysis. Notably, both gold compounds affected cell migration and invasion. CONCLUSIONS In this study, we deepened the mode of action of Au(NHC) and Au(NHC)2PF6, identifying common cellular targets but confirming their different influence on the mitochondrial function.
Collapse
Affiliation(s)
- Lara Massai
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Luigi Messori
- Department of Chemistry 'Ugo Schiff', University of Florence, via della Lastruccia 3-13, Sesto Fiorentino, 50019, Firenze, Italy
| | - Andrea Carpentieri
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Angela Amoresano
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Chiara Melchiorre
- Department of Chemical Sciences, University of Naples Federico II, Naples, Italy
| | - Tania Fiaschi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Alessandra Modesti
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy
| | - Tania Gamberi
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| | - Francesca Magherini
- Department of Experimental and Clinical Biomedical Sciences, Mario Serio" University of Florence Viale G.B. Morgagni 50, 50134, Florence, Italy.
| |
Collapse
|
20
|
Ogata FT, Simões Sato AY, Coppo L, Arai RJ, Stern AI, Pequeno Monteiro H. Thiol-Based Antioxidants and the Epithelial/Mesenchymal Transition in Cancer. Antioxid Redox Signal 2022; 36:1037-1050. [PMID: 34541904 DOI: 10.1089/ars.2021.0199] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Significance: The epithelial/mesenchymal transition (EMT) is commonly associated with tumor metastasis. Oxidative and nitrosative stress is maintained in cancer cells and is involved in the EMT. Cancer cells are endowed with high levels of enzymatic and nonenzymatic antioxidants, which counteract the effects of oxidative and nitrosative stress. Thiol-based antioxidant systems such as the thioredoxin/thioredoxin reductase (Trx/TrxR) and glutathione/glutaredoxin (GSH/Grx) are continually active in cancer cells, while the thioredoxin-interacting protein (Txnip), the negative regulator of the Trx/TrxR system, is downregulated. Recent Advances: Trx/TrxR and GSH/Grx systems play a major role in maintaining EMT signaling and cancer cell progression. Critical Issues: Enhanced stress conditions stimulated in cancer cells inhibit EMT signaling. The elevated expression levels of the Trx/TrxR and GSH/Grx systems in these cells provide the antioxidant protection necessary to guarantee the occurrence of the EMT. Future Directions: Elevation of the intracellular reactive oxygen species and nitric oxide concentrations in cancer cells has been viewed as a promising strategy for elimination of these cells. The development of inhibitors of GSH synthesis and of the Trx/TrxR system together with genetic-based strategies to enhance Txnip levels may provide the necessary means to achieve this goal. Antioxid. Redox Signal. 36, 1037-1050.
Collapse
Affiliation(s)
- Fernando Toshio Ogata
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Alex Yuri Simões Sato
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil.,Department of Morphology and Genetics, Universidade Federal de São Paulo, São Paulo, Brazil
| | - Lucia Coppo
- Department of Medical Biochemistry and Biophysics, Karolinska Institutet, Stockholm, Sweden
| | - Roberto Jun Arai
- Department of Oncology and Radiology, Instituto do Câncer do Estado de São Paulo, Faculdade de Medicina-Universidade de São Paulo, São Paulo, Brazil
| | - Arnold Ira Stern
- Grossman School of Medicine, New York University, New York, New York, USA
| | - Hugo Pequeno Monteiro
- Department of Biochemistry, Center for Cellular and Molecular Therapy-CTCMol, Universidade Federal de São Paulo, São Paulo, Brazil
| |
Collapse
|
21
|
Rosenblum SL, Kosman DJ. Aberrant Cerebral Iron Trafficking Co-morbid With Chronic Inflammation: Molecular Mechanisms and Pharmacologic Intervention. Front Neurol 2022; 13:855751. [PMID: 35370907 PMCID: PMC8964494 DOI: 10.3389/fneur.2022.855751] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2022] [Accepted: 02/14/2022] [Indexed: 12/24/2022] Open
Abstract
The redox properties that make iron an essential nutrient also make iron an efficient pro-oxidant. Given this nascent cytotoxicity, iron homeostasis relies on a combination of iron transporters, chaperones, and redox buffers to manage the non-physiologic aqueous chemistry of this first-row transition metal. Although a mechanistic understanding of the link between brain iron accumulation (BIA) and neurodegenerative diseases is lacking, BIA is co-morbid with the majority of cognitive and motor function disorders. The most prevalent neurodegenerative disorders, including Alzheimer's Disease (AD), Parkinson's Disease (PD), Multiple System Atrophy (MSA), and Multiple Sclerosis (MS), often present with increased deposition of iron into the brain. In addition, ataxias that are linked to mutations in mitochondrial-localized proteins (Friedreich's Ataxia, Spinocerebellar Ataxias) result in mitochondrial iron accumulation and degradation of proton-coupled ATP production leading to neuronal degeneration. A comorbidity common in the elderly is a chronic systemic inflammation mediated by primary cytokines released by macrophages, and acute phase proteins (APPs) released subsequently from the liver. Abluminal inflammation in the brain is found downstream as a result of activation of astrocytes and microglia. Reasonably, the iron that accumulates in the brain comes from the cerebral vasculature via the microvascular capillary endothelial cells whose tight junctions represent the blood-brain barrier. A premise amenable to experimental interrogation is that inflammatory stress alters both the trans- and para-cellular flux of iron at this barrier resulting in a net accumulation of abluminal iron over time. This review will summarize the evidence that lends support to this premise; indicate the mechanisms that merit delineation; and highlight possible therapeutic interventions based on this model.
Collapse
Affiliation(s)
| | - Daniel J. Kosman
- Department of Biochemistry, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
22
|
The Oxidative Balance Orchestrates the Main Keystones of the Functional Activity of Cardiomyocytes. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:7714542. [PMID: 35047109 PMCID: PMC8763515 DOI: 10.1155/2022/7714542] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/16/2021] [Revised: 11/03/2021] [Accepted: 12/15/2021] [Indexed: 12/11/2022]
Abstract
This review is aimed at providing an overview of the key hallmarks of cardiomyocytes in physiological and pathological conditions. The main feature of cardiac tissue is the force generation through contraction. This process requires a conspicuous energy demand and therefore an active metabolism. The cardiac tissue is rich of mitochondria, the powerhouses in cells. These organelles, producing ATP, are also the main sources of ROS whose altered handling can cause their accumulation and therefore triggers detrimental effects on mitochondria themselves and other cell components thus leading to apoptosis and cardiac diseases. This review highlights the metabolic aspects of cardiomyocytes and wanders through the main systems of these cells: (a) the unique structural organization (such as different protein complexes represented by contractile, regulatory, and structural proteins); (b) the homeostasis of intracellular Ca2+ that represents a crucial ion for cardiac functions and E-C coupling; and (c) the balance of Zn2+, an ion with a crucial impact on the cardiovascular system. Although each system seems to be independent and finely controlled, the contractile proteins, intracellular Ca2+ homeostasis, and intracellular Zn2+ signals are strongly linked to each other by the intracellular ROS management in a fascinating way to form a "functional tetrad" which ensures the proper functioning of the myocardium. Nevertheless, if ROS balance is not properly handled, one or more of these components could be altered resulting in deleterious effects leading to an unbalance of this "tetrad" and promoting cardiovascular diseases. In conclusion, this "functional tetrad" is proposed as a complex network that communicates continuously in the cardiomyocytes and can drive the switch from physiological to pathological conditions in the heart.
Collapse
|
23
|
Hu S, Mahadevan A, Elysee IF, Choi J, Souchet NR, Bae GH, Taboada AK, Sanketi B, Duhamel GE, Sevier CS, Tao G, Kurpios NA. The asymmetric Pitx2 gene regulates gut muscular-lacteal development and protects against fatty liver disease. Cell Rep 2021; 37:110030. [PMID: 34818545 PMCID: PMC8650168 DOI: 10.1016/j.celrep.2021.110030] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2020] [Revised: 08/19/2021] [Accepted: 10/29/2021] [Indexed: 12/25/2022] Open
Abstract
Intestinal lacteals are essential lymphatic channels for absorption and transport of dietary lipids and drive the pathogenesis of debilitating metabolic diseases. However, organ-specific mechanisms linking lymphatic dysfunction to disease etiology remain largely unknown. In this study, we uncover an intestinal lymphatic program that is linked to the left-right (LR) asymmetric transcription factor Pitx2. We show that deletion of the asymmetric Pitx2 enhancer ASE alters normal lacteal development through the lacteal-associated contractile smooth muscle lineage. ASE deletion leads to abnormal muscle morphogenesis induced by oxidative stress, resulting in impaired lacteal extension and defective lymphatic system-dependent lipid transport. Surprisingly, activation of lymphatic system-independent trafficking directs dietary lipids from the gut directly to the liver, causing diet-induced fatty liver disease. Our study reveals the molecular mechanism linking gut lymphatic function to the earliest symmetry-breaking Pitx2 and highlights the important relationship between intestinal lymphangiogenesis and the gut-liver axis.
Collapse
Affiliation(s)
- Shing Hu
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Aparna Mahadevan
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Isaac F Elysee
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Joseph Choi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Nathan R Souchet
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gloria H Bae
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Alessandra K Taboada
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Bhargav Sanketi
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Gerald E Duhamel
- Department of Biomedical Sciences, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Carolyn S Sevier
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA
| | - Ge Tao
- Department of Regenerative Medicine and Cell Biology, Medical University of South Carolina, Charleston, SC 29425, USA
| | - Natasza A Kurpios
- Department of Molecular Medicine, College of Veterinary Medicine, Cornell, Ithaca, NY 14853, USA.
| |
Collapse
|
24
|
Bamburg JR, Minamide LS, Wiggan O, Tahtamouni LH, Kuhn TB. Cofilin and Actin Dynamics: Multiple Modes of Regulation and Their Impacts in Neuronal Development and Degeneration. Cells 2021; 10:cells10102726. [PMID: 34685706 PMCID: PMC8534876 DOI: 10.3390/cells10102726] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2021] [Revised: 10/06/2021] [Accepted: 10/07/2021] [Indexed: 02/06/2023] Open
Abstract
Proteins of the actin depolymerizing factor (ADF)/cofilin family are ubiquitous among eukaryotes and are essential regulators of actin dynamics and function. Mammalian neurons express cofilin-1 as the major isoform, but ADF and cofilin-2 are also expressed. All isoforms bind preferentially and cooperatively along ADP-subunits in F-actin, affecting the filament helical rotation, and when either alone or when enhanced by other proteins, promotes filament severing and subunit turnover. Although self-regulating cofilin-mediated actin dynamics can drive motility without post-translational regulation, cells utilize many mechanisms to locally control cofilin, including cooperation/competition with other proteins. Newly identified post-translational modifications function with or are independent from the well-established phosphorylation of serine 3 and provide unexplored avenues for isoform specific regulation. Cofilin modulates actin transport and function in the nucleus as well as actin organization associated with mitochondrial fission and mitophagy. Under neuronal stress conditions, cofilin-saturated F-actin fragments can undergo oxidative cross-linking and bundle together to form cofilin-actin rods. Rods form in abundance within neurons around brain ischemic lesions and can be rapidly induced in neurites of most hippocampal and cortical neurons through energy depletion or glutamate-induced excitotoxicity. In ~20% of rodent hippocampal neurons, rods form more slowly in a receptor-mediated process triggered by factors intimately connected to disease-related dementias, e.g., amyloid-β in Alzheimer’s disease. This rod-inducing pathway requires a cellular prion protein, NADPH oxidase, and G-protein coupled receptors, e.g., CXCR4 and CCR5. Here, we will review many aspects of cofilin regulation and its contribution to synaptic loss and pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- James R. Bamburg
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Correspondence: ; Tel.: +1-970-988-9120; Fax: +1-970-491-0494
| | - Laurie S. Minamide
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - O’Neil Wiggan
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
| | - Lubna H. Tahtamouni
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Biology and Biotechnology, The Hashemite University, Zarqa 13115, Jordan
| | - Thomas B. Kuhn
- Department of Biochemistry and Molecular Biology, Colorado State University, Fort Collins, CO 80523, USA; (L.S.M.); (O.W.); (L.H.T.); (T.B.K.)
- Department of Chemistry and Biochemistry, University of Alaska, Fairbanks, AK 99775, USA
| |
Collapse
|
25
|
Hebbar S, Knust E. Reactive oxygen species (ROS) constitute an additional player in regulating epithelial development. Bioessays 2021; 43:e2100096. [PMID: 34260754 DOI: 10.1002/bies.202100096] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2021] [Revised: 06/09/2021] [Accepted: 06/09/2021] [Indexed: 12/18/2022]
Abstract
Reactive oxygen species (ROS) are highly reactive molecules produced in cells. So far, they have mostly been connected to diseases and pathological conditions. More recent results revealed a somewhat unexpected role of ROS in control of developmental processes. In this review, we elaborate on ROS in development, focussing on their connection to epithelial tissue morphogenesis. After briefly summarising unique characteristics of epithelial cells, we present some characteristic features of ROS species, their production and targets, with a focus on proteins important for epithelial development and function. Finally, we provide examples of regulation of epithelial morphogenesis by ROS, and also of developmental genes that regulate the overall redox status. We conclude by discussing future avenues of research that will further elucidate ROS regulation in epithelial development.
Collapse
Affiliation(s)
- Sarita Hebbar
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| | - Elisabeth Knust
- Max-Planck-Institute of Molecular Cell Biology and Genetics, Dresden, Germany
| |
Collapse
|
26
|
Ogata FT, Branco V, Vale FF, Coppo L. Glutaredoxin: Discovery, redox defense and much more. Redox Biol 2021; 43:101975. [PMID: 33932870 PMCID: PMC8102999 DOI: 10.1016/j.redox.2021.101975] [Citation(s) in RCA: 77] [Impact Index Per Article: 19.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2020] [Revised: 04/07/2021] [Accepted: 04/10/2021] [Indexed: 01/15/2023] Open
Abstract
Glutaredoxin, Grx, is a small protein containing an active site cysteine pair and was discovered in 1976 by Arne Holmgren. The Grx system, comprised of Grx, glutathione, glutathione reductase, and NADPH, was first described as an electron donor for Ribonucleotide Reductase but, from the first discovery in E.coli, the Grx family has impressively grown, particularly in the last two decades. Several isoforms have been described in different organisms (from bacteria to humans) and with different functions. The unique characteristic of Grxs is their ability to catalyse glutathione-dependent redox regulation via glutathionylation, the conjugation of glutathione to a substrate, and its reverse reaction, deglutathionylation. Grxs have also recently been enrolled in iron sulphur cluster formation. These functions have been implied in various physiological and pathological conditions, from immune defense to neurodegeneration and cancer development thus making Grx a possible drug target. This review aims to give an overview on Grxs, starting by a phylogenetic analysis of vertebrate Grxs, followed by an analysis of the mechanisms of action, the specific characteristics of the different human isoforms and a discussion on aspects related to human physiology and diseases.
Collapse
Affiliation(s)
- Fernando T Ogata
- Department of Biochemistry/Molecular Biology, CTCMol, Universidade Federal de São Paulo, Rua Mirassol, 207. 04044-010, São Paulo - SP, Brazil
| | - Vasco Branco
- Research Institute for Medicines (iMed.ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Av. Prof. Gama Pinto, 1649-003, Lisboa, Portugal
| | - Filipa F Vale
- Host-Pathogen Interactions Unit, Research Institute for Medicines (iMed-ULisboa), Faculty of Pharmacy, Universidade de Lisboa, Lisboa, Portugal
| | - Lucia Coppo
- Division of Biochemistry, Department of Medical Biochemistry and Biophysics, Karolinska Institute, Solnavägen 9, SE-17165, Stockholm, Sweden.
| |
Collapse
|
27
|
Giustarini D, Milzani A, Dalle-Donne I, Rossi R. Measurement of S-glutathionylated proteins by HPLC. Amino Acids 2021; 54:675-686. [PMID: 34129091 PMCID: PMC9117368 DOI: 10.1007/s00726-021-03015-6] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2021] [Accepted: 05/29/2021] [Indexed: 11/28/2022]
Abstract
S-glutathionylated proteins (GSSP), i.e., protein-mixed disulfides with glutathione (GSH), are considered a suitable biomarker of oxidative stress. In fact, they occur within cells at low level and their concentration increases markedly under pro-oxidant conditions. Plasma is something different, since it is physiologically rich in S-thiolated proteins (RSSP), i.e., protein-mixed disulfides with various types of low molecular mass thiols (LMM-SH). However, albumin, which is largely the most abundant plasma protein, possesses a cysteine residue at position 34 that is mostly reduced (about 60%) under physiological conditions, but easily involved in the formation of additional RSSP in the presence of oxidants. The quantification of GSSP requires special attention to sample handling, since their level can be overestimated as a result of artefactual oxidation of GSH. We have developed the present protocol to avoid this methodological problem. Samples should be treated as soon as possible after their collection with the alkylating agent N-ethylmaleimide that masks –SH groups and prevents their oxidation. The GSH released from mixed disulfides by reduction with dithiothreitol is then labeled with the fluorescent probe monobromobimane and quantified by HPLC. The method can be applied to many different biological samples, comprising blood components, red blood cell plasma membrane, cultured cells, and solid organs from animal models.
Collapse
Affiliation(s)
- Daniela Giustarini
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018-2022), Laboratory of Pharmacology and Toxicology, University of Siena, Via A. Moro 4, 53100, Siena, Italy.
| | - Aldo Milzani
- Department of Biosciences (Department of Excellence 2018-2022), Università Degli Studi Di Milano, via Celoria 26, I-20133, Milan, Italy
| | - Isabella Dalle-Donne
- Department of Biosciences (Department of Excellence 2018-2022), Università Degli Studi Di Milano, via Celoria 26, I-20133, Milan, Italy
| | - Ranieri Rossi
- Department of Biotechnology, Chemistry and Pharmacy (Department of Excellence 2018-2022), Laboratory of Pharmacology and Toxicology, University of Siena, Via A. Moro 4, 53100, Siena, Italy
| |
Collapse
|
28
|
van de Wetering C, Elko E, Berg M, Schiffers CHJ, Stylianidis V, van den Berge M, Nawijn MC, Wouters EFM, Janssen-Heininger YMW, Reynaert NL. Glutathione S-transferases and their implications in the lung diseases asthma and chronic obstructive pulmonary disease: Early life susceptibility? Redox Biol 2021; 43:101995. [PMID: 33979767 PMCID: PMC8131726 DOI: 10.1016/j.redox.2021.101995] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/04/2020] [Revised: 04/23/2021] [Accepted: 04/24/2021] [Indexed: 01/01/2023] Open
Abstract
Our lungs are exposed daily to airborne pollutants, particulate matter, pathogens as well as lung allergens and irritants. Exposure to these substances can lead to inflammatory responses and may induce endogenous oxidant production, which can cause chronic inflammation, tissue damage and remodeling. Notably, the development of asthma and Chronic Obstructive Pulmonary Disease (COPD) is linked to the aforementioned irritants. Some inhaled foreign chemical compounds are rapidly absorbed and processed by phase I and II enzyme systems critical in the detoxification of xenobiotics including the glutathione-conjugating enzymes Glutathione S-transferases (GSTs). GSTs, and in particular genetic variants of GSTs that alter their activities, have been found to be implicated in the susceptibility to and progression of these lung diseases. Beyond their roles in phase II metabolism, evidence suggests that GSTs are also important mediators of normal lung growth. Therefore, the contribution of GSTs to the development of lung diseases in adults may already start in utero, and continues through infancy, childhood, and adult life. GSTs are also known to scavenge oxidants and affect signaling pathways by protein-protein interaction. Moreover, GSTs regulate reversible oxidative post-translational modifications of proteins, known as protein S-glutathionylation. Therefore, GSTs display an array of functions that impact the pathogenesis of asthma and COPD. In this review we will provide an overview of the specific functions of each class of mammalian cytosolic GSTs. This is followed by a comprehensive analysis of their expression profiles in the lung in healthy subjects, as well as alterations that have been described in (epithelial cells of) asthmatics and COPD patients. Particular emphasis is placed on the emerging evidence of the regulatory properties of GSTs beyond detoxification and their contribution to (un)healthy lungs throughout life. By providing a more thorough understanding, tailored therapeutic strategies can be designed to affect specific functions of particular GSTs.
Collapse
Affiliation(s)
- Cheryl van de Wetering
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Evan Elko
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Marijn Berg
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Caspar H J Schiffers
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA
| | - Vasili Stylianidis
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands
| | - Maarten van den Berge
- Pulmonology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Martijn C Nawijn
- Pathology and Medical Biology, GRIAC Research Institute, University of Groningen, University Medical Center Groningen (UMCG), Groningen, the Netherlands
| | - Emiel F M Wouters
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands; Ludwig Boltzmann Institute for Lung Health, Vienna, Austria
| | - Yvonne M W Janssen-Heininger
- Department of Pathology and Laboratory Medicine, University of Vermont College of Medicine, Burlington, VT, USA.
| | - Niki L Reynaert
- Department of Respiratory Medicine, Maastricht University Medical Center+, Maastricht, the Netherlands.
| |
Collapse
|
29
|
Glutathione S-Transferases in Cancer. Antioxidants (Basel) 2021; 10:antiox10050701. [PMID: 33946704 PMCID: PMC8146591 DOI: 10.3390/antiox10050701] [Citation(s) in RCA: 101] [Impact Index Per Article: 25.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2021] [Revised: 04/23/2021] [Accepted: 04/27/2021] [Indexed: 02/06/2023] Open
Abstract
In humans, the glutathione S-transferases (GST) protein family is composed of seven members that present remarkable structural similarity and some degree of overlapping functionalities. GST proteins are crucial antioxidant enzymes that regulate stress-induced signaling pathways. Interestingly, overactive GST proteins are a frequent feature of many human cancers. Recent evidence has revealed that the biology of most GST proteins is complex and multifaceted and that these proteins actively participate in tumorigenic processes such as cell survival, cell proliferation, and drug resistance. Structural and pharmacological studies have identified various GST inhibitors, and these molecules have progressed to clinical trials for the treatment of cancer and other diseases. In this review, we discuss recent findings in GST protein biology and their roles in cancer development, their contribution in chemoresistance, and the development of GST inhibitors for cancer treatment.
Collapse
|
30
|
Chen M, Wang J, Yang Y, Zhong T, Zhou P, Ma H, Li J, Li D, Zhou J, Xie S, Liu M. Redox-dependent regulation of end-binding protein 1 activity by glutathionylation. SCIENCE CHINA. LIFE SCIENCES 2021; 64:575-583. [PMID: 32737853 DOI: 10.1007/s11427-020-1765-6] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/29/2020] [Accepted: 06/23/2020] [Indexed: 12/21/2022]
Abstract
Cytoskeletal proteins are susceptible to glutathionylation under oxidizing conditions, and oxidative damage has been implicated in several neurodegenerative diseases. End-binding protein 1 (EB1) is a master regulator of microtubule plus-end tracking proteins (+TIPs) and is critically involved in the control of microtubule dynamics and cellular processes. However, the impact of glutathionylation on EB1 functions remains unknown. Here we reveal that glutathionylation is important for controlling EB1 activity and protecting EB1 from irreversible oxidation. In vitro biochemical and cellular assays reveal that EB1 is glutathionylated. Diamide, a mild oxidizing reagent, reduces EB1 comet number and length in cells, indicating the impairment of microtubule dynamics. Three cysteine residues of EB1 are glutathionylated, with mutations of these three cysteines to serines attenuating microtubule dynamics but buffering diamide-induced decrease in microtubule dynamics. In addition, glutaredoxin 1 (Grx1) deglutathionylates EB1, and Grx1 depletion suppresses microtubule dynamics and leads to defects in cell division orientation and cell migration, suggesting a critical role of Grx1-mediated deglutathionylation in maintaining EB1 activity. Collectively, these data reveal that EB1 glutathionylation is an important protective mechanism for the regulation of microtubule dynamics and microtubule-based cellular activities.
Collapse
Affiliation(s)
- Miao Chen
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Jian Wang
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Yang Yang
- Department of Translational Medicine Center, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, 450052, China
| | - Tao Zhong
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Peng Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Huixian Ma
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Jingrui Li
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
| | - Dengwen Li
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Jun Zhou
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China
- State Key Laboratory of Medicinal Chemical Biology, Key Laboratory of Bioactive Materials of the Ministry of Education, Tianjin Key Laboratory of Protein Science, College of Life Sciences, Nankai University, Tianjin, 300071, China
| | - Songbo Xie
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China.
| | - Min Liu
- Shandong Provincial Key Laboratory of Animal Resistance Biology, College of Life Sciences, Collaborative Innovation Center of Cell Biology in Universities of Shandong, Institute of Biomedical Sciences, Shandong Normal University, Jinan, 250014, China.
| |
Collapse
|
31
|
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
|
32
|
Vitamin C Recycling Regulates Neurite Growth in Neurospheres Differentiated In Vitro. Antioxidants (Basel) 2020; 9:antiox9121276. [PMID: 33327638 PMCID: PMC7765149 DOI: 10.3390/antiox9121276] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2020] [Revised: 12/09/2020] [Accepted: 12/10/2020] [Indexed: 12/13/2022] Open
Abstract
The reduced form of vitamin C, ascorbic acid (AA), has been related with gene expression and cell differentiation in the cerebral cortex. In neurons, AA is mainly oxidized to dehydroascorbic acid (DHA); however, DHA cannot accumulate intracellularly because it induces metabolic changes and cell death. In this context, it has been proposed that vitamin C recycling via neuron–astrocyte coupling maintains AA levels and prevents DHA parenchymal accumulation. To date, the role of this mechanism during the outgrowth of neurites is unknown. To stimulate neuronal differentiation, adhered neurospheres treated with AA and retinoic acid (RA) were used. Neuritic growth was analyzed by confocal microscopy, and the effect of vitamin C recycling (bystander effect) in vitro was studied using different cells. AA stimulates neuritic growth more efficiently than RA. However, AA is oxidized to DHA in long incubation periods, generating a loss in the formation of neurites. Surprisingly, neurite growth is maintained over time following co-incubation of neurospheres with cells that efficiently capture DHA. In this sense, astrocytes have high capacity to recycle DHA and stimulate the maintenance of neurites. We demonstrated that vitamin C recycling in vitro regulates the morphology of immature neurons during the differentiation and maturation processes.
Collapse
|
33
|
Smith FM, Kosman DJ. Molecular Defects in Friedreich's Ataxia: Convergence of Oxidative Stress and Cytoskeletal Abnormalities. Front Mol Biosci 2020; 7:569293. [PMID: 33263002 PMCID: PMC7686857 DOI: 10.3389/fmolb.2020.569293] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 09/10/2020] [Indexed: 01/18/2023] Open
Abstract
Friedreich’s ataxia (FRDA) is a multi-faceted disease characterized by progressive sensory–motor loss, neurodegeneration, brain iron accumulation, and eventual death by hypertrophic cardiomyopathy. FRDA follows loss of frataxin (FXN), a mitochondrial chaperone protein required for incorporation of iron into iron–sulfur cluster and heme precursors. After the discovery of the molecular basis of FRDA in 1996, over two decades of research have been dedicated to understanding the temporal manifestations of disease both at the whole body and molecular level. Early research indicated strong cellular iron dysregulation in both human and yeast models followed by onset of oxidative stress. Since then, the pathophysiology due to dysregulation of intracellular iron chaperoning has become central in FRDA relative to antioxidant defense and run-down in energy metabolism. At the same time, limited consideration has been given to changes in cytoskeletal organization, which was one of the first molecular defects noted. These alterations include both post-translational oxidative glutathionylation of actin monomers and differential DNA processing of a cytoskeletal regulator PIP5K1β. Currently unknown in respect to FRDA but well understood in the context of FXN-deficient cell physiology is the resulting impact on the cytoskeleton; this disassembly of actin filaments has a particularly profound effect on cell–cell junctions characteristic of barrier cells. With respect to a neurodegenerative disorder such as FRDA, this cytoskeletal and tight junction breakdown in the brain microvascular endothelial cells of the blood–brain barrier is likely a component of disease etiology. This review serves to outline a brief history of this research and hones in on pathway dysregulation downstream of iron-related pathology in FRDA related to actin dynamics. The review presented here was not written with the intent of being exhaustive, but to instead urge the reader to consider the essentiality of the cytoskeleton and appreciate the limited knowledge on FRDA-related cytoskeletal dysfunction as a result of oxidative stress. The review examines previous hypotheses of neurodegeneration with brain iron accumulation (NBIA) in FRDA with a specific biochemical focus.
Collapse
Affiliation(s)
- Frances M Smith
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| | - Daniel J Kosman
- Department of Biochemistry, State University of New York at Buffalo, Buffalo, NY, United States
| |
Collapse
|
34
|
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
|
35
|
Rashdan NA, Shrestha B, Pattillo CB. S-glutathionylation, friend or foe in cardiovascular health and disease. Redox Biol 2020; 37:101693. [PMID: 32912836 PMCID: PMC7767732 DOI: 10.1016/j.redox.2020.101693] [Citation(s) in RCA: 32] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2020] [Revised: 08/12/2020] [Accepted: 08/16/2020] [Indexed: 12/27/2022] Open
Abstract
Glutathione is a low molecular weight thiol that is present at high levels in the cell. The high levels of glutathione in the cell make it one of the most abundant antioxidants contributing to cellular redox homeostasis. As a general rule, throughout cardiovascular disease and progression there is an imbalance in redox homeostasis characterized by reactive oxygen species overproduction and glutathione underproduction. As research into these imbalances continues, glutathione concentrations are increasingly being observed to drive various physiological and pathological signaling responses. Interestingly in addition to acting directly as an antioxidant, glutathione is capable of post translational modifications (S-glutathionylation) of proteins through both chemical interactions and enzyme mediated events. This review will discuss both the chemical and enzyme-based S-glutathionylation of proteins involved in cardiovascular pathologies and angiogenesis.
Collapse
Affiliation(s)
- N A Rashdan
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA
| | - B Shrestha
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA
| | - C B Pattillo
- Department of Cellular and Molecular Physiology, Louisiana State Health Science Center, Shreveport, LA, USA.
| |
Collapse
|
36
|
Mullen L, Mengozzi M, Hanschmann EM, Alberts B, Ghezzi P. How the redox state regulates immunity. Free Radic Biol Med 2020; 157:3-14. [PMID: 31899344 DOI: 10.1016/j.freeradbiomed.2019.12.022] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 12/05/2019] [Accepted: 12/19/2019] [Indexed: 12/30/2022]
Abstract
Oxidative stress is defined as an imbalance between the levels of reactive oxygen species (ROS) and antioxidant defences. The view of oxidative stress as a cause of cell damage has evolved over the past few decades to a much more nuanced view of the role of oxidative changes in cell physiology. This is no more evident than in the field of immunity, where oxidative changes are now known to regulate many aspects of the immune response, and inflammatory pathways in particular. Our understanding of redox regulation of immunity now encompasses not only increases in reactive oxygen and nitrogen species, but also changes in the activities of oxidoreductase enzymes. These enzymes are important regulators of immune pathways both via changes in their redox activity, but also via other more recently identified cytokine-like functions. The emerging picture of redox regulation of immune pathways is one of increasing complexity and while therapeutic targeting of the redox environment to treat inflammatory disease is a possibility, any such strategy is likely to be more nuanced than simply inhibiting ROS production.
Collapse
Affiliation(s)
- Lisa Mullen
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | | | - Eva-Maria Hanschmann
- Department of Neurology, Medical Faculty, Heinrich-Heine University Düsseldorf, Germany
| | - Ben Alberts
- Brighton and Sussex Medical School, Falmer, Brighton, UK
| | - Pietro Ghezzi
- Brighton and Sussex Medical School, Falmer, Brighton, UK.
| |
Collapse
|
37
|
Terzi A, Suter DM. The role of NADPH oxidases in neuronal development. Free Radic Biol Med 2020; 154:33-47. [PMID: 32370993 DOI: 10.1016/j.freeradbiomed.2020.04.027] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/15/2020] [Revised: 04/20/2020] [Accepted: 04/27/2020] [Indexed: 12/15/2022]
Abstract
Reactive oxygen species (ROS) are critical for maintaining cellular homeostasis and function when produced in physiological ranges. Important sources of cellular ROS include NADPH oxidases (Nox), which are evolutionary conserved multi-subunit transmembrane proteins. Nox-mediated ROS regulate variety of biological processes including hormone synthesis, calcium signaling, cell migration, and immunity. ROS participate in intracellular signaling by introducing post-translational modifications to proteins and thereby altering their functions. The central nervous system (CNS) expresses different Nox isoforms during both development and adulthood. Here, we review the role of Nox-mediated ROS during CNS development. Specifically, we focus on how individual Nox isoforms contribute to signaling in neural stem cell maintenance and neuronal differentiation, as well as neurite outgrowth and guidance. We also discuss how ROS regulates the organization and dynamics of the actin cytoskeleton in the neuronal growth cone. Finally, we review recent evidence that Nox-derived ROS modulate axonal regeneration upon nervous system injury.
Collapse
Affiliation(s)
- Aslihan Terzi
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA
| | - Daniel M Suter
- Department of Biological Sciences, Purdue University, West Lafayette, IN, 47907, USA; Purdue Institute for Integrative Neuroscience, Purdue University, West Lafayette, IN, 47907, USA; Bindley Bioscience Center, Purdue University, West Lafayette, IN, 47907, USA.
| |
Collapse
|
38
|
Purroy R, Medina-Carbonero M, Ros J, Tamarit J. Frataxin-deficient cardiomyocytes present an altered thiol-redox state which targets actin and pyruvate dehydrogenase. Redox Biol 2020; 32:101520. [PMID: 32279039 PMCID: PMC7152683 DOI: 10.1016/j.redox.2020.101520] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2019] [Revised: 03/10/2020] [Accepted: 03/21/2020] [Indexed: 01/11/2023] Open
Abstract
Friedreich ataxia (FA) is a cardioneurodegenerative disease caused by deficient frataxin expression. This mitochondrial protein has been related to iron homeostasis, energy metabolism, and oxidative stress. Previously, we set up a cardiac cellular model of FA based on neonatal rat cardiac myocytes (NRVM) and lentivirus-mediated frataxin RNA interference. These frataxin-deficient NRVMs presented lipid droplet accumulation, mitochondrial swelling and signs of oxidative stress. Therefore, we decided to explore the presence of protein thiol modifications in this model. With this purpose, reduced glutathione (GSH) levels were measured and the presence of glutathionylated proteins was analyzed. We observed decreased GSH content and increased presence of glutahionylated actin in frataxin-deficient NRVMs. Moreover, the presence of oxidized cysteine residues was investigated using the thiol-reactive fluorescent probe iodoacetamide-Bodipy and 2D-gel electrophoresis. With this approach, we identified two proteins with altered redox status in frataxin-deficient NRVMs: electron transfer flavoprotein-ubiquinone oxidoreductase and dihydrolipoyl dehydrogenase (DLDH). As DLDH is involved in protein-bound lipoic acid redox cycling, we analyzed the redox state of this cofactor and we observed that lipoic acid from pyruvate dehydrogenase was more oxidized in frataxin-deficient cells. Also, by targeted proteomics, we observed a decreased content on the PDH A1 subunit from pyruvate dehydrogenase. Finally, we analyzed the consequences of supplementing frataxin-deficient NRVMs with the PDH cofactors thiamine and lipoic acid, the PDH activator dichloroacetate and the antioxidants N-acetyl cysteine and Tiron. Both dichloroacetate and Tiron were able to partially prevent lipid droplet accumulation in these cells. Overall, these results indicate that frataxin-deficient NRVMs present an altered thiol-redox state which could contribute to the cardiac pathology.
Collapse
Affiliation(s)
- Rosa Purroy
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Marta Medina-Carbonero
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Joaquim Ros
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain
| | - Jordi Tamarit
- Dept. Ciències Mèdiques Bàsiques, Fac. Medicina, IRBLLeida, Universitat de Lleida, Lleida, Spain.
| |
Collapse
|
39
|
|
40
|
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: 12] [Impact Index Per Article: 2.0] [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
|
41
|
Duarte S, Melo T, Domingues R, de Dios Alché J, Pérez-Sala D. Insight into the cellular effects of nitrated phospholipids: Evidence for pleiotropic mechanisms of action. Free Radic Biol Med 2019; 144:192-202. [PMID: 31199965 DOI: 10.1016/j.freeradbiomed.2019.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 03/06/2019] [Revised: 05/26/2019] [Accepted: 06/03/2019] [Indexed: 12/20/2022]
Abstract
Nitrated phospholipids have been recently identified in biological systems and showed to display anti-oxidant and anti-inflammatory potential in models of inflammation in vitro. Here, we have explored the effects of nitrated 1-palmitoyl-2-oleyl-phosphatidyl choline (NO2-POPC) in cellular models. We have observed that NO2-POPC, but not POPC, induces cellular changes consisting in cytoskeletal rearrangement and cell shrinking, and ultimately, loss of cell adhesion or impaired cell attachment. NO2-POPC releases NO in vitro and induces accumulation of NO in cells. Nevertheless, the effects of NO2-POPC are not superimposable with those of NO donors, which points to distinctive mechanisms of action. Notably, they show a stronger parallelism, although not complete overlap, with the effects of nitrated fatty acids. Interestingly, redistribution of vimentin by NO2-POPC is attenuated in a C328S mutant, thus indicating that this residue may be a target for direct or indirect modification in NO2-POPC-treated cells. Additionally, NO2-POPC interacts with several typical lipoxidation targets in vitro, including vimentin and PPARγ constructs, likely through cysteine residues. Therefore, nitrated phospholipids emerge as potential novel electrophilic lipid mediators with selective actions.
Collapse
Affiliation(s)
- Sofia Duarte
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040, Madrid, Spain
| | - Tânia Melo
- Mass Spectrometry Center & QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; Department of Chemistry & CESAM& ECOMARE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Rosário Domingues
- Mass Spectrometry Center & QOPNA, Department of Chemistry, University of Aveiro, 3810-193 Aveiro, Portugal; Department of Chemistry & CESAM& ECOMARE, University of Aveiro, 3810-193, Aveiro, Portugal
| | - Juan de Dios Alché
- Plant Reproductive Biology and Advanced Imaging Laboratory, Estación Experimental del Zaidín, Consejo Superior de Investigaciones Científicas, 18008, Granada, Spain
| | - Dolores Pérez-Sala
- Department of Structural and Chemical Biology, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, 28040, Madrid, Spain.
| |
Collapse
|
42
|
Uemura T, Tsaprailis G, Gerner EW. GSTΠ stimulates caveolin-1-regulated polyamine uptake via actin remodeling. Oncotarget 2019; 10:5713-5723. [PMID: 31620246 PMCID: PMC6779281 DOI: 10.18632/oncotarget.27192] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2019] [Accepted: 08/16/2019] [Indexed: 12/31/2022] Open
Abstract
Polyamines spermidine and spermine, and their diamine precursor putrescine, are essential for normal cellular functions in both pro- and eukaryotes. Cellular polyamine levels are regulated by biosynthesis, degradation and transport. Transport of dietary and luminal bacterial polyamines in gastrointestinal (GI) tissues plays a significant role in tissue polyamine homeostasis. We have reported that caveolin-1 play an inhibitory role in polyamine uptake in GI tissues. We investigated the mechanism of caveolin-1-regulated polyamine transport. We found that glutathione S-transferase Π(GSTΠ) was secreted from caveolin-1 knockdown cells and stimulated spermidine transport in human colon-derived HCT116 cells. GSTΠ secreted in the medium increased S-glutathionylated protein level in the plasma membrane fraction. Proteomic analysis revealed that actin was S-glutathionylated by GSTΠ. Immunofluorescence microscopy demonstrated that actin filaments around plasma membrane were S-glutathionylated in caveolin-1 knockdown cells. Inhibition of actin remodeling by jasplakinolide caused a decrease in polyamine uptake activity. These data support a model in which caveolin-1 negatively regulates polyamine uptake by inhibiting GSTΠ secretion, which stimulates actin remodeling and endocytosis.
Collapse
Affiliation(s)
- Takeshi Uemura
- Amine Pharma Research Institute, Chuo-ku, Chiba 260-0856, Japan
| | - George Tsaprailis
- Center for Toxicology, College of Pharmacy, Tucson, Arizona 85721, USA
| | - Eugene W Gerner
- Cancer Prevention Pharmaceuticals, Tucson, Arizona 85718, USA
| |
Collapse
|
43
|
Griffith AW, Harke MJ, DePasquale E, Berry DL, Gobler CJ. The harmful algae, Cochlodinium polykrikoides and Aureococcus anophagefferens, elicit stronger transcriptomic and mortality response in larval bivalves ( Argopecten irradians) than climate change stressors. Ecol Evol 2019; 9:4931-4948. [PMID: 31031955 PMCID: PMC6476759 DOI: 10.1002/ece3.5100] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2019] [Revised: 02/26/2019] [Accepted: 03/04/2019] [Indexed: 12/04/2022] Open
Abstract
Global ocean change threatens marine life, yet a mechanistic understanding of how organisms are affected by specific stressors is poorly understood. Here, we identify and compare the unique and common transcriptomic responses of an organism experiencing widespread fisheries declines, Argopecten irradians (bay scallop) exposed to multiple stressors including high pCO2, elevated temperature, and two species of harmful algae, Cochlodinium (aka Margalefidinium) polykrikoides and Aureococcus anophagefferens using high-throughput sequencing (RNA-seq). After 48 hr of exposure, scallop transcriptomes revealed distinct expression profiles with larvae exposed to harmful algae (C. polykrikoides and A. anophagefferens) displaying broader responses in terms of significantly and differentially expressed (DE) transcripts (44,922 and 4,973; respectively) than larvae exposed to low pH or elevated temperature (559 and 467; respectively). Patterns of expression between larvae exposed to each harmful algal treatment were, however, strikingly different with larvae exposed to A. anophagefferens displaying large, significant declines in the expression of transcripts (n = 3,615; 87% of DE transcripts) whereas exposure to C. polykrikoides increased the abundance of transcripts, more than all other treatments combined (n = 43,668; 97% of DE transcripts). Larvae exposed to each stressor up-regulated a common set of 21 genes associated with protein synthesis, cellular metabolism, shell growth, and membrane transport. Larvae exposed to C. polykrikoides displayed large increases in antioxidant-associated transcripts, whereas acidification-exposed larvae increased abundance of transcripts associated with shell formation. After 10 days of exposure, each harmful algae caused declines in survival that were significantly greater than all other treatments. Collectively, this study reveals the common and unique transcriptional responses of bivalve larvae to stressors that promote population declines within coastal zones, providing insight into the means by which they promote mortality as well as traits possessed by bay scallops that enable potential resistance.
Collapse
Affiliation(s)
- Andrew W. Griffith
- School of Marine and Atmospheric SciencesStony Brook UniversitySouthamptonNew York
- Department of Biological SciencesUniversity of Southern CaliforniaLos AngelesCalifornia
| | - Matthew J. Harke
- Lamont‐Doherty Earth ObservatoryColumbia UniversityPalisadesNew York
| | - Elizabeth DePasquale
- School of Marine and Atmospheric SciencesStony Brook UniversitySouthamptonNew York
| | - Dianna L. Berry
- School of Marine and Atmospheric SciencesStony Brook UniversitySouthamptonNew York
| | | |
Collapse
|
44
|
Ranieri M, Di Mise A, Difonzo G, Centrone M, Venneri M, Pellegrino T, Russo A, Mastrodonato M, Caponio F, Valenti G, Tamma G. Green olive leaf extract (OLE) provides cytoprotection in renal cells exposed to low doses of cadmium. PLoS One 2019; 14:e0214159. [PMID: 30897184 PMCID: PMC6428325 DOI: 10.1371/journal.pone.0214159] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 03/07/2019] [Indexed: 02/07/2023] Open
Abstract
Cadmium (Cd) is a heavy and highly toxic metal that contaminates air, food and water. Cadmium accumulates in several organs altering normal functions. The kidney is the major organ at risk of damage from chronic exposure to cadmium as a contaminant in food and water. This study aims to investigate the beneficial effects of OLE in renal collecting duct MCD4 cells exposed to a low dose cadmium (1 μM). In MCD4 cells cadmium caused an increase in ROS production, as well as generation of lipid droplets and reduced cell viability. Moreover, cadmium exposure led to a remarkable increase in the frequency of micronuclei and DNA double-strand breaks, assessed using the alkaline comet assay. In addition, cadmium dramatically altered cell cytoskeleton architecture and caused S-glutathionylation of actin. Notably, all cadmium-induced cellular deregulations were prevented by co-treatment with OLE, possibly due to its antioxidant action and to the presence of bioactive phytocompounds. Indeed, OLE treatment attenuated Cd-induced actin S-glutathionylation, thereby stabilizing actin filaments. Taken together, these observations provide a novel insight into the biological action of OLE in renal cells and support the notion that OLE may serve as a potential adjuvant against cadmium-induced nephrotoxicity.
Collapse
Affiliation(s)
- Marianna Ranieri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Annarita Di Mise
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Graziana Difonzo
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Mariangela Centrone
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Maria Venneri
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Tommaso Pellegrino
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | - Annamaria Russo
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
| | | | - Francesco Caponio
- Department of Soil, Plant and Food Sciences, University of Bari Aldo Moro, Bari, Italy
| | - Giovanna Valenti
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.), Bari, Italy
- Center of Excellence in Comparative Genomics (CEGBA), University of Bari, Bari, Italy
| | - Grazia Tamma
- Department of Biosciences, Biotechnologies and Biopharmaceutics, University of Bari Aldo Moro, Bari, Italy
- Istituto Nazionale di Biostrutture e Biosistemi (I.N.B.B.), Bari, Italy
- * E-mail:
| |
Collapse
|
45
|
Dökümcü K, Simonian M, Farahani RM. miR4673 improves fitness profile of neoplastic cells by induction of autophagy. Cell Death Dis 2018; 9:1068. [PMID: 30341280 PMCID: PMC6195512 DOI: 10.1038/s41419-018-1088-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 06/16/2018] [Accepted: 06/18/2018] [Indexed: 12/18/2022]
Abstract
Therapeutic resistance of neoplasms is mainly attributed to gradual evolution of mutational profile1. Here, we demonstrate a microRNA-mediated mechanism that effectively improves fitness of SKBR3 mammary carcinoma cells by cytoplasmic reprogramming. The reprogramming is triggered by endogenous miR4673 transcribed from notch-1 locus. The miRNA downregulates cdk-18, a cyclin-dependent kinase that regulates M-G1 transition in cycling cells2,3. Suppression of cdk-18 triggers mitophagy and autophagy. Due to high autophagic flux, oestrogen receptor-1+/progesterone receptor+/p53+ (Esr1+/Pr+/p53+) SKBR3 cells are coerced into an Esr1-/Prlow/p53-profile. Increased mitophagy in combination with proteasomal degradation of p53 transiently arrests the cycling cells at G0 and enhances radio-resistance of the SKBR3 population. These findings highlight the impact on cancer therapy of non-encoded neoplastic resistance, arising as a consequence of miRNA-mediated autophagic reprogramming that uncouples phenotype and genotype.
Collapse
Affiliation(s)
- Kağan Dökümcü
- Institute of Dental Research, Westmead Institute for Medical Research and Westmead Centre for Oral Health, Westmead, NSW, Australia
- Department of Life Sciences, The University of Sydney Dental School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia
| | - Mary Simonian
- Institute of Dental Research, Westmead Institute for Medical Research and Westmead Centre for Oral Health, Westmead, NSW, Australia
| | - Ramin M Farahani
- Institute of Dental Research, Westmead Institute for Medical Research and Westmead Centre for Oral Health, Westmead, NSW, Australia.
- Department of Life Sciences, The University of Sydney Dental School, Faculty of Medicine and Health, University of Sydney, Sydney, NSW, 2006, Australia.
| |
Collapse
|
46
|
Cuello F, Wittig I, Lorenz K, Eaton P. Oxidation of cardiac myofilament proteins: Priming for dysfunction? Mol Aspects Med 2018; 63:47-58. [PMID: 30130564 DOI: 10.1016/j.mam.2018.08.003] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2018] [Revised: 08/13/2018] [Accepted: 08/17/2018] [Indexed: 02/07/2023]
Abstract
Oxidants are produced endogenously and can react with and thereby post-translationally modify target proteins. They have been implicated in the redox regulation of signal transduction pathways conferring protection, but also in mediating oxidative stress and causing damage. The difference is that in scenarios of injury the amount of oxidants generated is higher and/or the duration of oxidant exposure sustained. In the cardiovascular system, oxidants are important for blood pressure homeostasis, for unperturbed cardiac function and also contribute to the observed protection during ischemic preconditioning. In contrast, oxidative stress accompanies all major cardiovascular pathologies and has been attributed to mediate contractile dysfunction in part by inducing oxidative modifications in myofilament proteins. However, the proportion to which oxidative modifications of contractile proteins are beneficial or causatively mediate disease progression needs to be carefully reconsidered. These antithetical aspects will be discussed in this review with special focus on direct oxidative post-translational modifications of myofilament proteins that have been described to occur in vivo and to regulate actin-myosin interactions in the cardiac myocyte sarcomere, the methodologies for detection of oxidative post-translational modifications in target proteins and the feasibility of antioxidant therapy strategies as a potential treatment for cardiac disorders.
Collapse
Affiliation(s)
- Friederike Cuello
- Institute of Experimental Pharmacology and Toxicology, Cardiovascular Research Center, University Medical Center Hamburg-Eppendorf, Hamburg, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Hamburg/Kiel/Lübeck, Germany.
| | - Ilka Wittig
- Functional Proteomics, SFB 815 Core Unit, Faculty of Medicine, Johann Wolfgang Goethe University, Frankfurt am Main, Germany; DZHK (German Center for Cardiovascular Research), Partner Site Rhine-Main, Germany
| | - Kristina Lorenz
- Comprehensive Heart Failure Center, Würzburg, Leibniz-Institut für Analytische Wissenschaften-ISAS-e.V. Dortmund, West German Heart and Vascular Center, Essen, Germany
| | - Philip Eaton
- King's British Heart Foundation Centre, King's College London, UK
| |
Collapse
|
47
|
Guo J, Kim HS, Asmis R, Ludueña RF. Interactions of β tubulin isotypes with glutathione in differentiated neuroblastoma cells subject to oxidative stress. Cytoskeleton (Hoboken) 2018; 75:283-289. [PMID: 29663696 DOI: 10.1002/cm.21447] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2018] [Revised: 03/16/2018] [Accepted: 04/11/2018] [Indexed: 01/23/2023]
Abstract
Microtubules are a major component of the neuronal cytoskeleton. Tubulin, the subunit protein of microtubules, is an α/β heterodimer. Both α and β exist as families of isotypes, whose members are encoded by different genes and have different amino acid sequences. The βII and βIII isotypes are very prominent in the nervous system. Our previous work has suggested that βII may play a role in neuronal differentiation, but the role of βIII in neurons is not well understood. In the work reported here, we examined the roles of the different β-tubulin isotypes in response to glutamate/glycine treatment, and found that both βII and βIII bind to glutathione in the presence of ROS, especially βIII. In contrast, βI did not bind to glutathione. Our results suggest that βII and βIII, but especially βIII, may play an important role in the response of neuronal cells to stress. In view of the high levels of βII and βIII expressed in the nervous system it is conceivable that these tubulin isotypes may use their sulfhydryl groups to scavenge ROS and protect neuronal cells against oxidative stress.
Collapse
Affiliation(s)
- Jiayan Guo
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas 72290-3900
| | - Hong Seok Kim
- Department of Molecular Medicine, College of Medicine, Inha University, Incheon 22212, Republic of Korea
| | - Reto Asmis
- Clinical Laboratory Science, University of Texas Health Science Center at San Antonio, San Antonio, Texas 72290-3900
| | - Richard F Ludueña
- Department of Biochemistry, University of Texas Health Science Center at San Antonio, San Antonio, Texas 72290-3900
| |
Collapse
|
48
|
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: 143] [Impact Index Per Article: 20.4] [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
|
49
|
Liu Y, Xiao W, Shinde M, Field J, Templeton DM. Cadmium favors F-actin depolymerization in rat renal mesangial cells by site-specific, disulfide-based dimerization of the CAP1 protein. Arch Toxicol 2018; 92:1049-1064. [PMID: 29222746 PMCID: PMC6925060 DOI: 10.1007/s00204-017-2142-3] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2017] [Accepted: 12/05/2017] [Indexed: 12/30/2022]
Abstract
Cadmium is a toxic metal that produces oxidative stress and has been shown to disrupt the actin cytoskeleton in rat renal mesangial cells (RMC). In a survey of proteins that might undergo Cd2+-dependent disulfide crosslinking, we identified the adenylyl cyclase-associated protein, CAP1, as undergoing a dimerization in response to Cd2+ (5-40 µM) that was sensitive to disulfide reducing agents, was reproduced by the disulfide crosslinking agent diamide, and was shown by site-directed mutagenesis to involve the Cys29 residue of the protein. Reactive oxygen species are not involved in the thiol oxidation, and glutathione modulates background levels of dimer. CAP1 is known to enhance cofilin's F-actin severing activity through binding to F-actin and cofilin. F-actin sedimentation and GST-cofilin pulldown studies of CAP1 demonstrated enrichment of the CAP1 dimer's association with cofilin, and in the cofilin-F-actin pellet, suggesting that Cd2+-induced dimer increases the formation of a CAP1-cofilin-F-actin complex. Both siRNA-based silencing of CAP1 and overexpression of a CAP1 mutant lacking Cys29 (and therefore, incapable of dimerization in response to Cd2+) increased RMC viability and provided some protection of F-actin structures against Cd2+. It is concluded that Cd2+ brings about disruption of the RMC cytoskeleton in part through formation of a CAP1 dimer that increases recruitment of cofilin to F-actin filaments.
Collapse
Affiliation(s)
- Ying Liu
- Laboratory Medicine and Pathobiology 1 King's College Circle, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Weiqun Xiao
- Laboratory Medicine and Pathobiology 1 King's College Circle, University of Toronto, Toronto, ON, M5S 1A8, Canada
| | - Manasi Shinde
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Jeffrey Field
- Department of Systems Pharmacology and Translational Therapeutics, University of Pennsylvania, Philadelphia, PA, 19104, USA
| | - Douglas M Templeton
- Laboratory Medicine and Pathobiology 1 King's College Circle, University of Toronto, Toronto, ON, M5S 1A8, Canada.
| |
Collapse
|
50
|
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
|