1
|
Twigger SA, Dominguez B, Porto V, Hacker L, Chalmers AJ, Breckenridge R, Treder M, Sedgwick AC, Dominguez F, Hammond EM. The activity of therapeutic molecular cluster Ag5 is dependent on oxygen level and HIF-1 mediated signalling. Redox Biol 2024; 76:103326. [PMID: 39180984 PMCID: PMC11388176 DOI: 10.1016/j.redox.2024.103326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024] Open
Abstract
Regions of hypoxia occur in most solid tumours and are known to significantly impact therapy response and patient prognosis. Ag5 is a recently reported silver molecular cluster which inhibits both glutathione and thioredoxin signalling therefore limiting cellular antioxidant capacity. Ag5 treatment significantly reduces cell viability in a range of cancer cell lines with little to no impact on non-transformed cells. Characterisation of redox homeostasis in hypoxia demonstrated an increase in reactive oxygen species and glutathione albeit with different kinetics. Significant Ag5-mediated loss of viability was observed in a range of hypoxic conditions which mimic the tumour microenvironment however, this effect was reduced compared to normoxic conditions. Reduced sensitivity to Ag5 in hypoxia was attributed to HIF-1 mediated signalling to reduce PDH via PDK1/3 activity and changes in mitochondrial oxygen availability. Importantly, the addition of Ag5 significantly increased radiation-induced cell death in hypoxic conditions associated with radioresistance. Together, these data demonstrate Ag5 is a potent and cancer specific agent which could be used effectively in combination with radiotherapy.
Collapse
Affiliation(s)
- Sophie A Twigger
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | - Blanca Dominguez
- Department of physiology and CIMUS Universidade de Santiago de Compostela, Spain
| | - Vanesa Porto
- Department of physiology and CIMUS Universidade de Santiago de Compostela, Spain
| | - Lina Hacker
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK
| | | | | | | | - Adam C Sedgwick
- Department of Chemistry, King's College London, London, SE1 1DB, UK
| | - Fernando Dominguez
- Department of physiology and CIMUS Universidade de Santiago de Compostela, Spain
| | - Ester M Hammond
- Department of Oncology, University of Oxford, Oxford, OX3 7DQ, UK.
| |
Collapse
|
2
|
Chettouh-Hammas N, Grillon C. Physiological skin oxygen levels: An important criterion for skin cell functionality and therapeutic approaches. Free Radic Biol Med 2024; 222:259-274. [PMID: 38908804 DOI: 10.1016/j.freeradbiomed.2024.06.015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/29/2024] [Revised: 06/15/2024] [Accepted: 06/18/2024] [Indexed: 06/24/2024]
Abstract
The skin is made up of different layers with various gradients, which maintain a complex microenvironment, particularly in terms of oxygen levels. However, all types of skin cells are cultured in conventional incubators that do not reproduce physiological oxygen levels. Instead, they are cultured at atmospheric oxygen levels, a condition that is far removed from physiology and may lead to the generation of free radicals known to induce skin ageing. This review aims to summarize the current literature on the effect of physiological oxygen levels on skin cells, highlight the shortcomings of current in vitro models, and demonstrate the importance of respecting skin oxygen levels. We begin by clarifying the terminology used about oxygen levels and describe the specific distribution of oxygen in the skin. We review and discuss how skin cells adapt their oxygen consumption and metabolism to oxygen levels environment, as well as the changes that are induced, particularly, their redox state, life cycle and functions. We examine the effects of oxygen on both simple culture models and more complex reconstructed skin models. Finally, we present the implications of oxygen modulation for a more therapeutic approach.
Collapse
Affiliation(s)
- Nadira Chettouh-Hammas
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| | - Catherine Grillon
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071, Orléans, Cedex 2, France.
| |
Collapse
|
3
|
Alva R, Wiebe JE, Stuart JA. Revisiting reactive oxygen species production in hypoxia. Pflugers Arch 2024; 476:1423-1444. [PMID: 38955833 DOI: 10.1007/s00424-024-02986-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2024] [Revised: 06/20/2024] [Accepted: 06/24/2024] [Indexed: 07/04/2024]
Abstract
Cellular responses to hypoxia are crucial in various physiological and pathophysiological contexts and have thus been extensively studied. This has led to a comprehensive understanding of the transcriptional response to hypoxia, which is regulated by hypoxia-inducible factors (HIFs). However, the detailed molecular mechanisms of HIF regulation in hypoxia remain incompletely understood. In particular, there is controversy surrounding the production of mitochondrial reactive oxygen species (ROS) in hypoxia and how this affects the stabilization and activity of HIFs. This review examines this controversy and attempts to shed light on its origin. We discuss the role of physioxia versus normoxia as baseline conditions that can affect the subsequent cellular response to hypoxia and highlight the paucity of data on pericellular oxygen levels in most experiments, leading to variable levels of hypoxia that might progress to anoxia over time. We analyze the different outcomes reported in isolated mitochondria, versus intact cells or whole organisms, and evaluate the reliability of various ROS-detecting tools. Finally, we examine the cell-type and context specificity of oxygen's various effects. We conclude that while recent evidence suggests that the effect of hypoxia on ROS production is highly dependent on the cell type and the duration of exposure, efforts should be made to conduct experiments under carefully controlled, physiological microenvironmental conditions in order to rule out potential artifacts and improve reproducibility in research.
Collapse
Affiliation(s)
- Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| | - Jacob E Wiebe
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada
| | - Jeffrey A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, L2S 3A1, Canada.
| |
Collapse
|
4
|
Fan S, Gao H, Zhang Y, Nie L, Bártolo R, Bron R, Santos HA, Schirhagl R. Quantum Sensing of Free Radical Generation in Mitochondria of Single Heart Muscle Cells during Hypoxia and Reoxygenation. ACS NANO 2024; 18:2982-2991. [PMID: 38235677 PMCID: PMC10832053 DOI: 10.1021/acsnano.3c07959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/23/2023] [Revised: 12/31/2023] [Accepted: 01/05/2024] [Indexed: 01/19/2024]
Abstract
Cells are damaged during hypoxia (blood supply deprivation) and reoxygenation (oxygen return). This damage occurs in conditions such as cardiovascular diseases, cancer, and organ transplantation, potentially harming the tissue and organs. The role of free radicals in cellular metabolic reprogramming under hypoxia is under debate, but their measurement is challenging due to their short lifespan and limited diffusion range. In this study, we employed a quantum sensing technique to measure the real-time production of free radicals at the subcellular level. We utilize fluorescent nanodiamonds (FNDs) that exhibit changes in their optical properties based on the surrounding magnetic noise. This way, we were able to detect the presence of free radicals. To specifically monitor radical generation near mitochondria, we coated the FNDs with an antibody targeting voltage-dependent anion channel 2 (anti-VDAC2), which is located in the outer membrane of mitochondria. We observed a significant increase in the radical load on the mitochondrial membrane when cells were exposed to hypoxia. Subsequently, during reoxygenation, the levels of radicals gradually decreased back to the normoxia state. Overall, by applying a quantum sensing technique, the connections among hypoxia, free radicals, and the cellular redox status has been revealed.
Collapse
Affiliation(s)
- Siyu Fan
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Han Gao
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Yue Zhang
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Linyan Nie
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Raquel Bártolo
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Reinier Bron
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| | - Hélder A. Santos
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
- Drug
Research Program, Division of Pharmaceutical Chemistry and Technology,
Faculty of Pharmacy, University of Helsinki, FI-00014, Helsinki, Finland
| | - Romana Schirhagl
- Department
of Biomaterials and Biomedical Technology, University Medical Center
Groningen, University of Groningen, 9713 AV Groningen, The Netherlands
| |
Collapse
|
5
|
Sgarbi G, Righetti R, Del Dotto V, Grillini S, Giorgio V, Baracca A, Solaini G. The pro-oncogenic protein IF 1 does not contribute to the Warburg effect and is not regulated by PKA in cancer cells. Biochim Biophys Acta Mol Basis Dis 2024; 1870:166879. [PMID: 37689158 DOI: 10.1016/j.bbadis.2023.166879] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2023] [Revised: 08/04/2023] [Accepted: 09/02/2023] [Indexed: 09/11/2023]
Abstract
The endogenous inhibitor of mitochondrial F1Fo-ATPase (ATP synthase), IF1, has been shown to exert pro-oncogenic actions, including reprogramming of cellular energy metabolism (Warburg effect). The latter action of IF1 has been reported to be hampered by its PKA-dependent phosphorylation, but both reprogramming of metabolism and PKA-dependent phosphorylation are intensely debated. To clarify these critical issues, we prepared stably IF1-silenced clones and compared their bioenergetics with that of the three parental IF1-expressing cancer cell lines. All functional parameters: respiration rate, ATP synthesis rate (OXPHOS), and mitochondrial membrane potential were similar in IF1-silenced and control cells, clearly indicating that IF1 cannot inhibit the ATP synthase in cancer cells when the enzyme works physiologically. Furthermore, all cell types exposed to PKA modulators and energized with NAD+-dependent substrates or succinate showed similar OXPHOS rate regardless of the presence or absence of IF1. Therefore, our results rule out that IF1 action is modulated by its PKA-dependent phosphorylated/dephosphorylated state. Notably, cells exposed to a negative PKA modulator and energized with NAD+-dependent substrates showed a significant decrease of the OXPHOS rate matching previously reported inactivation of complex I. Overall, this study definitively demonstrates that IF1 inhibits neither mitochondrial ATP synthase nor OXPHOS in normoxic cancer cells and does not contribute to the Warburg effect. Thus, currently the protection of cancer cells from severe hypoxia/anoxia and apoptosis remain the only unquestionable actions of IF1 as pro-oncogenic factor that may be exploited to develop therapeutic approaches.
Collapse
Affiliation(s)
- Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Riccardo Righetti
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Valentina Del Dotto
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Silvia Grillini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Valentina Giorgio
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| | - Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences, University of Bologna, Via Irnerio 48, 40126 Bologna, Italy.
| |
Collapse
|
6
|
Righetti R, Grillini S, Del Dotto V, Costanzini A, Liuzzi F, Zanna C, Sgarbi G, Solaini G, Baracca A. The Pro-Oncogenic Protein IF 1 Promotes Proliferation of Anoxic Cancer Cells during Re-Oxygenation. Int J Mol Sci 2023; 24:14624. [PMID: 37834071 PMCID: PMC10572598 DOI: 10.3390/ijms241914624] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2023] [Revised: 09/15/2023] [Accepted: 09/23/2023] [Indexed: 10/15/2023] Open
Abstract
Cancer cells overexpress IF1, the endogenous protein that inhibits the hydrolytic activity of ATP synthase when mitochondrial membrane potential (ΔμH+) falls, as in ischemia. Other roles have been ascribed to IF1, but the associated molecular mechanisms are still under debate. We investigated the ability of IF1 to promote survival and proliferation in osteosarcoma and colon carcinoma cells exposed to conditions mimicking ischemia and reperfusion, as occurs in vivo, particularly in solid tumors. IF1-silenced and parental cells were exposed to the FCCP uncoupler to collapse ΔμH+ and the bioenergetics of cell models were validated. All the uncoupled cells preserved mitochondrial mass, but the implemented mechanisms differed in IF1-expressing and IF1-silenced cells. Indeed, the membrane potential collapse and the energy charge preservation allowed an increase in both mitophagy and mitochondrial biogenesis in IF1-expressing cells only. Interestingly, the presence of IF1 also conferred a proliferative advantage to cells highly dependent on oxidative phosphorylation when the uncoupler was washed out, mimicking cell re-oxygenation. Overall, our results indicate that IF1, by allowing energy preservation and promoting mitochondrial renewal, can favor proliferation of anoxic cells and tumor growth. Therefore, hindering the action of IF1 may be promising for the therapy of tumors that rely on oxidative phosphorylation for energy production.
Collapse
Affiliation(s)
- Riccardo Righetti
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Silvia Grillini
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Valentina Del Dotto
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Anna Costanzini
- Department of Translational Medicine, St. Anna University Hospital, University of Ferrara, 44124 Ferrara, Italy;
| | - Francesca Liuzzi
- Department of Medical and Surgical Sciences Maternal-Infantile and Adult, University of Modena and Reggio-Emilia, 41125 Modena, Italy;
| | - Claudia Zanna
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Gianluca Sgarbi
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Giancarlo Solaini
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| | - Alessandra Baracca
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (R.R.); (S.G.); (V.D.D.); (C.Z.); (G.S.); (A.B.)
| |
Collapse
|
7
|
Chettouh-Hammas N, Fasani F, Boileau A, Gosset D, Busco G, Grillon C. Improvement of Antioxidant Defences in Keratinocytes Grown in Physioxia: Comparison of 2D and 3D Models. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2023; 2023:6829931. [PMID: 37360501 PMCID: PMC10290565 DOI: 10.1155/2023/6829931] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 02/06/2023] [Revised: 04/25/2023] [Accepted: 05/27/2023] [Indexed: 06/28/2023]
Abstract
Keratinocytes prevent skin photoaging by ensuring the defence against oxidative stress, an excessive production of reactive oxygen species (ROS). They are localized within the epidermis where the oxygen level (1-3% O2), named physioxia, is low compared to other organs. Oxygen is essential for life but also generates ROS. Most of the in vitro studies on keratinocyte antioxidant capacities are performed under atmospheric oxygen, named normoxia, which is very far from the physiological microenvironment, thus submitting cells to an overoxygenation. The present study is aimed at investigating the antioxidant status of keratinocyte grown under physioxia in both 2D and 3D models. First, we show that the basal antioxidant profiles of keratinocytes display important differences when comparing the HaCaT cell line, primary keratinocytes (NHEK), reconstructed epidermis (RHE), and skin explants. Physioxia was shown to promote a strong proliferation of keratinocytes in monolayers and in RHE, resulting in a thinner epidermis likely due to a slowdown in cell differentiation. Interestingly, cells in physioxia exhibited a lower ROS production upon stress, suggesting a better protection against oxidative stress. To understand this effect, we studied the antioxidant enzymes and reported a lower or equivalent level of mRNA for all enzymes in physioxia conditions compared to normoxia, but a higher activity for catalase and superoxide dismutases, whatever the culture model. The unchanged catalase amount, in NHEK and RHE, suggests an overactivation of the enzyme in physioxia, whereas the higher amount of SOD2 can explain the strong activity. Taken together, our results demonstrate the role of oxygen in the regulation of the antioxidant defences in keratinocytes, topic of particular importance for studying skin aging. Additionally, the present work points out the interest of the choice of both the keratinocyte culture model and the oxygen level to be as close as possible to the in situ skin.
Collapse
Affiliation(s)
- Nadira Chettouh-Hammas
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Fabienne Fasani
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Amandine Boileau
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - David Gosset
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Giovanni Busco
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| | - Catherine Grillon
- Center for Molecular Biophysics UPR4301 CNRS, Rue Charles Sadron, 45071 Orléans Cedex 2, France
| |
Collapse
|
8
|
Sánchez-Argüello P, Franco D, Fernández MD. Combined cytotoxicity of ZnO nanoparticles and chlorpyrifos in the rainbow trout, Oncorhynchus mikyss, gonadal cell line RTG-2. AQUATIC TOXICOLOGY (AMSTERDAM, NETHERLANDS) 2023; 261:106612. [PMID: 37331202 DOI: 10.1016/j.aquatox.2023.106612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/08/2023] [Revised: 06/12/2023] [Accepted: 06/14/2023] [Indexed: 06/20/2023]
Abstract
The toxicity of ZnO nanoparticles (ZnO NPs) in aquatic organisms has been extensively studied, but little information is available on the effects associated with their interaction with other contaminants. In this context, the in vitro effects of co-exposure of chlorpyrifos (CPF) and ZnO NPs on fish-derived cells were investigated. A selection of concentrations was tested in single and binary exposures: CPF (0.312 - 75 mg/L) and ZnO NPs (10 - 100 mg/L). Cytotoxicity was measured using commonly used cellular endpoints: Alamar Blue/CFDA-AM for viability and plasma membrane integrity, NRU for lysosomal disruption and MTT for mitochondrial function. In addition, specific mechanisms of toxicity for CPF and ZnO NPs were tested: acetylcholinesterase (AChE) activity and ROS generation, respectively. AChE was by far the most sensitive assay for single exposure to CPF. There was no concentration-response relationship for ROS after single exposure to ZnO NPs, but 10 mg/L produced significant effects only for this cellular endpoint. Co-exposure of CPF with 10 m/L of ZnO NPs produced significant effects in almost all endpoints tested, which were enhanced by co-exposure with 100 mg/L of ZnO NPs. AChE testing of additional co-exposures with bulk ZnO, together with the application of the Independent Action (IA) prediction model, which allowed us to draw more in-depth conclusions on the toxicological behavior of the mixture. Synergism was observed at 0.625 mg/L CPF concentration and antagonism at 5 mg/L CPF in mixtures containing 100 mg/L of both ZnO NPs and bulk ZnO. However, more cases of synergism between CPF and ZnO NPs occurred at intermediate CPF concentrations, demonstrating that nano-sized particles have a more toxic interaction with CPF than bulk ZnO. Therefore it can be argued that in vitro assays allow the identification of interaction profiles of NP-containing mixtures by achieving multiple endpoints with a large number of concentration combinations.
Collapse
Affiliation(s)
- Paloma Sánchez-Argüello
- Laboratory for Ecotoxicology, Department of Environment and Agronomy, INIA-CSIC (National Institute for Agricultural Research and Food Research and Technology-CSIC), A Coruña, km 7.5. 28040 Madrid, Spain.
| | - Daniel Franco
- Laboratory for Ecotoxicology, Department of Environment and Agronomy, INIA-CSIC (National Institute for Agricultural Research and Food Research and Technology-CSIC), A Coruña, km 7.5. 28040 Madrid, Spain
| | - Mª Dolores Fernández
- Laboratory for Ecotoxicology, Department of Environment and Agronomy, INIA-CSIC (National Institute for Agricultural Research and Food Research and Technology-CSIC), A Coruña, km 7.5. 28040 Madrid, Spain
| |
Collapse
|
9
|
Perego F, Ticozzi RM, Troia A, Prato M, Taramelli D, Basilico N. Dextran-shelled oxygen-loaded nanodroplets modulate macrophages killing and inflammatory response to Enterococcus faecalis. Eur J Pharmacol 2022; 931:175161. [PMID: 35964657 DOI: 10.1016/j.ejphar.2022.175161] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2021] [Revised: 07/13/2022] [Accepted: 07/19/2022] [Indexed: 11/03/2022]
Abstract
Chronic wounds are associated with inflammation, infections, and hypoxic environment. Macrophages play a crucial role in wound healing removing bacteria and secreting signal molecules to coordinate tissue repair. Recently, dextran-shelled Oxygen-Loaded NanoDroplets (OLNDs) have been proposed as new tools to counteract hypoxia in chronic wounds. Here we investigated the effects of OLNDs on Enterococcus faecalis (E. faecalis) killing and the secretion of inflammatory and angiogenic factors by murine (BMDM) and human (dTHP-1, differentiated THP-1) macrophages, in normoxia and hypoxia. Both OLNDs and Oxygen-Free NanoDroplets (OFNDs) significantly increased reactive oxygen species production by BMDM in normoxia (4.1 and 4 fold increase by 10% OLNDs and OFNDs, respectively, after 120 min) and hypoxia (3.8 and 4 fold increase by 10% OLNDs and OFNDs respectively) but not by dTHP-1. Moreover, only OLNDs induced nitric oxide secretion by BMDM in normoxia. Consequently, both nanodroplets improved E. faecalis killing by BMDM in normoxia (% of killing OLNDs = 44.2%; p < 0.01; OFNDs = 41.4%; p < 0.05) and hypoxia (% of killing OLNDs = 43.1%; p < 0.01; OFNDs = 37.7%; p < 0.05), while dTHP-1-mediated killing was not affected. The secretion of the inflammatory cytokines (TNFα, IL-6, IL-1β) induced by E. faecalis infection in dTHP-1 was reduced by both types of nanodroplets, suggesting a novel anti-inflammatory activity of the dextran shell. Instead, the increase of VEGF induced by hypoxia was reduced only by OLNDs. These data provide new knowledge on the effects of OLNDs as innovative adjuvant in chronic wounds healing promoting bacterial killing and reducing inflammation.
Collapse
Affiliation(s)
- Federica Perego
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milano, Italy.
| | - Rosalia Maria Ticozzi
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milano, Italy.
| | - Adriano Troia
- Istituto Nazionale di Ricerca Metrologica (INRIM), Strada delle Cacce 91, 10135, Torino, Italy.
| | - Mauro Prato
- Dipartimento di Neuroscienze, Università di Torino, Corso Raffaello 30, 10125, Torino, Italy.
| | - Donatella Taramelli
- Dipartimento di Scienze Farmacologiche e Biomolecolari, Università degli Studi di Milano, via Pascal 36, 20133, Milano, Italy.
| | - Nicoletta Basilico
- Dipartimento di Scienze Biomediche, Chirurgiche e Odontoiatriche, Università degli Studi di Milano, via Pascal 36, 20133, Milano, Italy.
| |
Collapse
|
10
|
Gatto C, Grandi M, Solaini G, Baracca A, Giorgio V. The F1Fo-ATPase inhibitor protein IF1 in pathophysiology. Front Physiol 2022; 13:917203. [PMID: 35991181 PMCID: PMC9389554 DOI: 10.3389/fphys.2022.917203] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2022] [Accepted: 06/27/2022] [Indexed: 12/15/2022] Open
Abstract
The endogenous inhibitor of ATP synthase is a protein of about 10 kDa, known as IF1 which binds to the catalytic domain of the enzyme during ATP hydrolysis. The main role of IF1 consists of limiting ATP dissipation under condition of severe oxygen deprivation or in the presence of dysfunctions of mitochondrial respiratory complexes, causing a collapse in mitochondrial membrane potential and therefore ATP hydrolysis. New roles of IF1 are emerging in the fields of cancer and neurodegeneration. Its high expression levels in tumor tissues have been associated with different roles favouring tumor formation, progression and evasion. Since discordant mechanisms of action have been proposed for IF1 in tumors, it is of the utmost importance to clarify them in the prospective of defining novel approaches for cancer therapy. Other IF1 functions, including its involvement in mitophagy, may be protective for neurodegenerative and aging-related diseases. In the present review we aim to clarify and discuss the emerging mechanisms in which IF1 is involved, providing a critical view of the discordant findings in the literature.
Collapse
|
11
|
Probing Cell Redox State and Glutathione-Modulating Factors Using a Monochlorobimane-Based Microplate Assay. Antioxidants (Basel) 2022; 11:antiox11020391. [PMID: 35204274 PMCID: PMC8869332 DOI: 10.3390/antiox11020391] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/05/2022] [Accepted: 02/11/2022] [Indexed: 12/10/2022] Open
Abstract
Thiol compounds including predominantly glutathione (GSH) are key components of redox homeostasis, which are involved in the protection and regulation of mammalian cells. The assessment of cell redox status by means of in situ analysis of GSH in living cells is often preferable over established assays in cell lysates due to fluctuations of the GSH pool. For this purpose, we propose a microplate assay with monochlorobimane (MCB) as an available fluorescent probe for GSH, although poorly detected in the microplate format. In addition to the new procedure for improved MCB-assisted GSH detection in plate-grown cells and its verification with GSH modulators, this study provides a useful methodology for the evaluation of cell redox status probed through relative GSH content and responsiveness to both supplemented thiols and variation in oxygen pressure. The roles of extracellular interactions of thiols and natural variability of cellular glutathione on the assay performance were emphasized and discussed. The results are of broad interest in cell biology research and should be particularly useful for the characterization of pathological cells with decreased GSH status and increased oxidative status as well as redox-modulating factors.
Collapse
|
12
|
Ullah A, Leong SW, Wang J, Wu Q, Ghauri MA, Sarwar A, Su Q, Zhang Y. Cephalomannine inhibits hypoxia-induced cellular function via the suppression of APEX1/HIF-1α interaction in lung cancer. Cell Death Dis 2021; 12:490. [PMID: 33990544 PMCID: PMC8121842 DOI: 10.1038/s41419-021-03771-z] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2021] [Revised: 03/29/2021] [Accepted: 03/30/2021] [Indexed: 12/12/2022]
Abstract
Lung cancer (LC) is one of the leading causes of cancer-related death. As one of the key features of tumor microenvironment, hypoxia conditions are associated with poor prognosis in LC patients. Upregulation of hypoxic-induced factor-1α (HIF-1α) leads to the activation of various factors that contribute to the increased drug resistance, proliferation, and migration of tumor cells. Apurinic/apyrimidinic endonuclease-1 (APEX1) is a multi-functional protein that regulates several transcription factors, including HIF-1α, that contribute to tumor growth, oxidative stress responses, and DNA damage. In this study, we explored the mechanisms underlying cell responses to hypoxia and modulation of APEX1, which regulate HIF-1α and downstream pathways. We found that hypoxia-induced APEX1/HIF-1α pathways regulate several key cellular functions, including reactive oxygen species (ROS) production, carbonic anhydrase 9 (CA9)-mediated intracellular pH, migration, and angiogenesis. Cephalomannine (CPM), a natural compound, exerted inhibitory effects in hypoxic LC cells via the inhibition of APEX1/HIF-1α interaction in vitro and in vivo. CPM can significantly inhibit cell viability, ROS production, intracellular pH, and migration in hypoxic LC cells as well as angiogenesis of HUVECs under hypoxia through the inhibition of APEX1/HIF-1α interaction. Taken together, CPM could be considered as a promising compound for LC treatment.
Collapse
Affiliation(s)
- Asmat Ullah
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Sze Wei Leong
- Department of Microbiology, Faculty of Biotechnology and Biomolecular Sciences, Universiti Putra Malaysia, Seri Kembangan, Malaysia
| | - Jingjing Wang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qing Wu
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Mohsin Ahmad Ghauri
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Ammar Sarwar
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China
| | - Qi Su
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| | - Yanmin Zhang
- School of Pharmacy, Health Science Center, Xi'an Jiaotong University, Xi'an, Shaanxi, 710061, P.R. China.
| |
Collapse
|
13
|
Solaini G, Sgarbi G, Baracca A. The F1Fo-ATPase inhibitor, IF1, is a critical regulator of energy metabolism in cancer cells. Biochem Soc Trans 2021; 49:815-827. [PMID: 33929490 DOI: 10.1042/bst20200742] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Revised: 03/25/2021] [Accepted: 03/26/2021] [Indexed: 12/17/2022]
Abstract
In the last two decades, IF1, the endogenous inhibitor of the mitochondrial F1Fo-ATPase (ATP synthase) has assumed greater and ever greater interest since it has been found to be overexpressed in many cancers. At present, several findings indicate that IF1 is capable of playing a central role in cancer cells by promoting metabolic reprogramming, proliferation and resistance to cell death. However, the mechanism(s) at the basis of this pro-oncogenic action of IF1 remains elusive. Here, we recall the main features of the mechanism of the action of IF1 when the ATP synthase works in reverse, and discuss the experimental evidence that support its relevance in cancer cells. In particular, a clear pro-oncogenic action of IF1 is to avoid wasting of ATP when cancer cells are exposed to anoxia or near anoxia conditions, therefore favoring cell survival and tumor growth. However, more recently, various papers have described IF1 as an inhibitor of the ATP synthase when it is working physiologically (i.e. synthethizing ATP), and therefore reprogramming cell metabolism to aerobic glycolysis. In contrast, other studies excluded IF1 as an inhibitor of ATP synthase under normoxia, providing the basis for a hot debate. This review focuses on the role of IF1 as a modulator of the ATP synthase in normoxic cancer cells with the awareness that the knowledge of the molecular action of IF1 on the ATP synthase is crucial in unravelling the molecular mechanism(s) responsible for the pro-oncogenic role of IF1 in cancer and in developing related anticancer strategies.
Collapse
Affiliation(s)
- Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| |
Collapse
|
14
|
Fu L, Zhang L, Zhang X, Chen L, Cai Q, Yang X. Roles of oxygen level and hypoxia-inducible factor signaling pathway in cartilage, bone and osteochondral tissue engineering. Biomed Mater 2021; 16:022006. [PMID: 33440367 DOI: 10.1088/1748-605x/abdb73] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The repair and treatment of articular cartilage injury is a huge challenge of orthopedics. Currently, most of the clinical methods applied in treating cartilage injuries are mainly to relieve pains rather than to cure them, while the strategy of tissue engineering is highly expected to achieve the successful repair of osteochondral defects. Clear understandings of the physiological structures and mechanical properties of cartilage, bone and osteochondral tissues have been established, but the understanding of their physiological heterogeneity still needs further investigation. Apart from the gradients in the micromorphology and composition of cartilage-to-bone extracellular matrixes, an oxygen gradient also exists in natural osteochondral tissue. The response of hypoxia-inducible factor (HIF)-mediated cells to oxygen would affect the differentiation of stem cells and the maturation of osteochondral tissue. This article reviews the roles of oxygen level and HIF signaling pathway in the development of articular cartilage tissue, and their prospective applications in bone and cartilage tissue engineering. The strategies for regulating HIF signaling pathway and how these strategies finding their potential applications in the regeneration of integrated osteochondral tissue are also discussed.
Collapse
Affiliation(s)
- Lei Fu
- State Key Laboratory of Organic-Inorganic Composites, Beijing Laboratory of Biomedical Materials, Beijing University of Chemical Technology, Beijing 100029, People's Republic of China
| | | | | | | | | | | |
Collapse
|
15
|
Yao Q, Parvez-Khan M, Schipani E. In vivo survival strategies for cellular adaptation to hypoxia: HIF1α-dependent suppression of mitochondrial oxygen consumption and decrease of intracellular hypoxia are critical for survival of hypoxic chondrocytes. Bone 2020; 140:115572. [PMID: 32768687 DOI: 10.1016/j.bone.2020.115572] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Revised: 07/29/2020] [Accepted: 07/30/2020] [Indexed: 12/15/2022]
Abstract
Hypoxia occurs not only in pathological conditions like cancer and ischemia and in a variety of physiological settings in the adult organism, but also during normal embryonic development. In the inner portion of the fetal growth plate, which is an avascular tissue originating from mesenchymal progenitor cells, chondrocytes experience physiological hypoxia. Hypoxia-Inducible Transcription Factor-1α (HIF1α), a crucial mediator of cellular adaptation to hypoxia, is an essential survival factor for fetal growth plate chondrocytes. This brief review summarizes our current understanding of the survival function of HIF1α during endochondral bone development.
Collapse
Affiliation(s)
- Qing Yao
- Guangdong Provincial Key Laboratory of Cell Microenvironment and Disease Research, Shenzhen Key Laboratory of Cell Microenvironment and School of Medicine, Southern University of Science and Technology, 1088 Xue Yuan Road, Shenzhen, Guangdong 518055, China
| | - Mohd Parvez-Khan
- Departments of Orthopaedic Surgery, Medicine, and Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA
| | - Ernestina Schipani
- Departments of Orthopaedic Surgery, Medicine, and Cell and Developmental Biology, University of Michigan School of Medicine, Ann Arbor, MI 48109, USA.
| |
Collapse
|
16
|
Stachon T, Latta L, Seitz B, Szentmáry N. Hypoxic stress increases NF-κB and iNOS mRNA expression in normal, but not in keratoconus corneal fibroblasts. Graefes Arch Clin Exp Ophthalmol 2020; 259:449-458. [PMID: 32886165 PMCID: PMC7843574 DOI: 10.1007/s00417-020-04900-8] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2020] [Revised: 07/30/2020] [Accepted: 08/16/2020] [Indexed: 12/18/2022] Open
Abstract
Background Keratoconus (KC) is associated with oxidative stress and hypoxia and as several times discussed, potentially with inflammatory components. Inflammation, hypoxia, and oxidative stress may result in metabolic dysfunction and are directly linked to each other. In the current study, we investigate the effect of hypoxia through NF-κB signaling pathways on iNOS, hypoxia-induced factors (HIF), ROS, and proliferation of normal and KC human corneal fibroblasts (HCFs), in vitro. Methods Primary human KC-HCFs and normal HCFs were isolated and cultured in DMEM/Ham’s F12 medium supplemented with 5% fetal calf serum. Hypoxic conditions were generated and quantitative PCR and Western blot analysis were performed to examine NF-κB, iNOS, HIF, and PHD2 expression in KC and normal HCFs. ROS level was analyzed using flow cytometry and proliferation by BrdU-ELISA. Results Hypoxia increased NF-κB mRNA and protein expression in normal HCFs, but in KC-HCFs NF-κB mRNA and protein expression remained unchanged. Hypoxic conditions upregulated iNOS mRNA expression of normal HCFs, but iNOS mRNA expression of KC-HCFs and iNOS protein expression of both HCF types remained unchanged. Hypoxia downregulated HIF-1α and HIF-2α mRNA expression in normal and KC-HCFs. PHD2 mRNA expression is upregulated under hypoxia in KC-HCFs, but not in normal HCFs. PHD2 protein expression was upregulated by hypoxia in both HCF types. Total ROS concentration is downregulated in normal and KC-HCFs under hypoxic conditions. Proliferation rate of KC-HCFs was upregulated through hypoxia, but did not change in normal HCFs. Conclusions Hypoxia increases NF-κB and iNOS mRNA expression in normal HCFs, but there does not seem to be enough capacity in KC-HCFs to increase NF-κB and iNOS mRNA expression under hypoxia, maybe due to the preexisting oxidative stress. HIF and PHD2 do not show altered iNOS regulation under hypoxic conditions in KC-HCFs, and therefore do not seem to play a role in keratoconus pathogenesis. An increased proliferation of cells may refer to compensatory mechanisms under hypoxia in KC. Understanding the mechanism of the altered regulation of NF-κB and iNOS in KC-HCFs will provide better insight into the potential inflammatory component of the KC pathogenesis.![]()
Collapse
Affiliation(s)
- Tanja Stachon
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany. .,Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg/Saar, Germany.
| | - Lorenz Latta
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg/Saar, Germany
| | - Berthold Seitz
- Department of Ophthalmology, Saarland University Medical Center, Homburg, Saar, Germany
| | - Nóra Szentmáry
- Dr. Rolf. M. Schwiete Center for Limbal Stem Cell and Aniridia Research, Saarland University, Homburg/Saar, Germany.,Department of Ophthalmology, Semmelweis University, Budapest, Hungary
| |
Collapse
|
17
|
Dikova V, Vorhauser J, Geng A, Pelster B, Sandbichler AM. Metabolic interaction of hydrogen peroxide and hypoxia in zebrafish fibroblasts. Free Radic Biol Med 2020; 152:469-481. [PMID: 31740229 DOI: 10.1016/j.freeradbiomed.2019.11.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/13/2019] [Revised: 11/12/2019] [Accepted: 11/12/2019] [Indexed: 11/26/2022]
Abstract
Cells require oxygen for aerobic metabolism, which may also result in the production of reactive oxygen species (ROS) as a by-product. Under low oxygen conditions, ROS formation has been reported to either increase or decrease. We addressed this physiological response for the first time in zebrafish embryonic fibroblasts (Z3) and used a hydrogen peroxide (H2O2)-specific fluorescent protein (roGFP2-Orp1) either targeted to the mitochondria or expressed in the cytosol. Microfluidic live-cell imaging measurements showed that oxygen deprivation in Z3 cells results in decreased or stable H2O2 levels within the mitochondria or the cytosol, respectively, and that the reductive shift recorded in the mitochondrial matrix is directly dependent on oxygen concentration. The response was accompanied by a transient increase in extracellular acidification rate (ECAR) and a lower cellular reducing potential as assessed by the viability stain alamarBlue. Complex I and III inhibition with Rotenone and Antimycin A led to H2O2 production under normoxia but these inhibitors were not able to avert the reductive shift under hypoxia. Only by system-wide inhibition of flavin-containing oxidases with Diphenyleneiodonium (DPI) were we able to decrease the reductive shift, while selective inhibition of NADPH oxidases with the inhibitor Apocynin had no effect on the hypoxia response. Since DPI also led to a strong increase in ECAR we found that, in order to keep the cytosolic H2O2 levels stable, glycolytic metabolism was of fundamental importance. According to our experiments with the glucose-6-phosphate dehydrogenase inhibitor 6-Aminonicotinamide, this was attributable to the pentose phosphate pathway producing reducing equivalents required for ROS degradation.
Collapse
Affiliation(s)
- Valentina Dikova
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Julia Vorhauser
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Anne Geng
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | - Bernd Pelster
- Institute of Zoology and Center for Molecular Biosciences (CMBI), University of Innsbruck, Austria
| | | |
Collapse
|
18
|
Hamon MP, Gergondey R, L'honoré A, Friguet B. Mitochondrial Lon protease - depleted HeLa cells exhibit proteome modifications related to protein quality control, stress response and energy metabolism. Free Radic Biol Med 2020; 148:83-95. [PMID: 31904544 DOI: 10.1016/j.freeradbiomed.2019.12.039] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2019] [Revised: 12/23/2019] [Accepted: 12/24/2019] [Indexed: 12/20/2022]
Abstract
The ATP-dependent Lon protease is located in the mitochondrial matrix and oxidized proteins are among its primary targets for their degradation. Impairment of mitochondrial morphology and function together with apoptosis were observed in lung fibroblasts depleted for Lon expression while accumulation of carbonylated mitochondrial proteins has been reported for yeast and HeLa Lon deficient cells. In addition, age-related mitochondrial dysfunction has been associated with an impairment of Lon expression. Using a HeLa cell line stably transfected with an inducible shRNA directed against Lon, we have previously observed that Lon depletion results in a mild phenotype characterized by an increase of both production of reactive oxygen species and level of oxidized proteins (Bayot et al., 2014, Biochimie, 100: 38-47). In this study using the same cell line, we now show that Lon knockdown leads to modifications of the expression of a number of specific proteins involved in protein quality control, stress response and energy metabolism, as evidenced using a 2D gel-based proteomic approach, and to alteration of the mitochondrial network morphology. We also show that these effects are associated with decreased proliferation and can be modulated by culture conditions in galactose versus glucose containing medium.
Collapse
Affiliation(s)
- Marie-Paule Hamon
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Rachel Gergondey
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Aurore L'honoré
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France
| | - Bertrand Friguet
- Sorbonne Université, CNRS, INSERM, Institut de Biologie Paris-Seine, Biological Adaptation and Aging, B2A-IBPS, F-75005, Paris, France.
| |
Collapse
|
19
|
Costanzini A, Sgarbi G, Maresca A, Del Dotto V, Solaini G, Baracca A. Mitochondrial Mass Assessment in a Selected Cell Line under Different Metabolic Conditions. Cells 2019; 8:cells8111454. [PMID: 31752092 PMCID: PMC6912592 DOI: 10.3390/cells8111454] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2019] [Revised: 09/27/2019] [Accepted: 11/14/2019] [Indexed: 01/14/2023] Open
Abstract
Changes of quantity and/or morphology of cell mitochondria are often associated with metabolic modulation, pathology, and apoptosis. Exogenous fluorescent probes used to investigate changes in mitochondrial content and dynamics are strongly dependent, for their internalization, on the mitochondrial membrane potential and composition, thus limiting the reliability of measurements. To overcome this limitation, genetically encoded recombinant fluorescent proteins, targeted to different cellular districts, were used as reporters. Here, we explored the potential use of mitochondrially targeted red fluorescent probe (mtRFP) to quantify, by flow cytometry, mitochondrial mass changes in cells exposed to different experimental conditions. We first demonstrated that the mtRFP fluorescence intensity is stable during cell culture and it is related with the citrate synthase activity, an established marker of the mitochondrial mass. Incidentally, the expression of mtRFP inside mitochondria did not alter the oxygen consumption rate under both state 3 and 4 respiration conditions. In addition, using this method, we showed for the first time that different inducers of mitochondrial mass change, such as hypoxia exposure or resveratrol treatment of cells, could be consistently detected. We suggest that transfection and selection of stable clones expressing mtRFP is a reliable method to monitor mitochondrial mass changes, particularly when pathophysiological or experimental conditions change ΔΨm, as it occurs during mitochondrial uncoupling or hypoxia/anoxia conditions.
Collapse
Affiliation(s)
- Anna Costanzini
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (A.C.); (G.S.)
- Department of Medical Sciences, University of Ferrara, 44121 Ferrara, Italy
| | - Gianluca Sgarbi
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (A.C.); (G.S.)
| | - Alessandra Maresca
- UOC Clinica Neurologica, IRCCS Istituto delle Scienze Neurologiche di Bologna, 40126 Bologna, Italy;
| | - Valentina Del Dotto
- Unit of Neurology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40139 Bologna, Italy;
| | - Giancarlo Solaini
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (A.C.); (G.S.)
- Correspondence: (G.S.); (A.B.); Tel.: +39-051-2091215 (G.S.); Tel.: +39-051-2091244 (A.B.)
| | - Alessandra Baracca
- Laboratory of Biochemistry and Mitochondrial Pathophysiology, Department of Biomedical and Neuromotor Sciences, University of Bologna, 40126 Bologna, Italy; (A.C.); (G.S.)
- Correspondence: (G.S.); (A.B.); Tel.: +39-051-2091215 (G.S.); Tel.: +39-051-2091244 (A.B.)
| |
Collapse
|
20
|
Koo MA, Hee Hong S, Hee Lee M, Kwon BJ, Mi Seon G, Sung Kim M, Kim D, Chang Nam K, Park JC. Effective stacking and transplantation of stem cell sheets using exogenous ROS-producing film for accelerated wound healing. Acta Biomater 2019; 95:418-426. [PMID: 30660002 DOI: 10.1016/j.actbio.2019.01.019] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2018] [Revised: 01/10/2019] [Accepted: 01/11/2019] [Indexed: 12/26/2022]
Abstract
Extensive skin loss caused by burns or diabetic ulcers may lead to major disability or even death. Therefore, cell-based therapies that enhance skin regeneration are clinically needed. Previous approaches have been applied the injections of cell suspensions and the implantation of biodegradable three-dimensional scaffolds seeded cells. However, these treatments have limits due to poor localization of the injected cells and insufficient delivery of oxygen and nutrients to cells. Recently, cell sheet-based tissue engineering has been developed to transplant cell sheets, which are cell-dense tissues without scaffolds. Because cell density is one of the important factors for improving the therapeutic effect of cell transplantation, transplanting layered cell sheet constructs can promote the recovery of tissue function and tissue regeneration compared with a single cell sheet. Thus, this study designed ROS-induced cell sheet stacking method with newly fabricated hematoporphyrin-incorporated polyketone film (Hp-PK film) to enhance cell sheet delivery efficiency and application in wound healing. We have demonstrated the therapeutic effect of a multi-layered mesenchymal stem cell sheets onto a full-thickness wound defect in nude mice. Consequentially, three-layered cell sheets transplanted and stacked by ROS-induced method promoted angiogenesis and skin regeneration at the wound site. Thus, our strategy based on Hp-PK film, which allows for easy stacking and transplantation of cell sheets, could be applied to enhance tissue regeneration. STATEMENT OF SIGNIFICANCE: We herein report exogenous ROS-induced cell sheet stacking method with newly fabricated hematoporphyrin-incorporated polyketone film (Hp-PK film) to enhance cell sheet transplantation efficiency and application in wound healing. Although there are several ways to stack-up cell sheets, all of these methods have limitations in transplanting the cell sheet directly to the target site. The method is simple and takes a relatively short time compared to previously reported methods for stacking and transplanting cell sheets. Thus, our study will provide a scientific impact because the method of applying exogenous ROS generated from Hp-PK film on cell detachment can transplant the cell sheet through a process of putting a cell sheet-cultured film on the lesion, irradiating with light, and then removing only the film.
Collapse
Affiliation(s)
- Min-Ah Koo
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Seung Hee Hong
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Mi Hee Lee
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Byeong-Ju Kwon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Gyeung Mi Seon
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Min Sung Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Dohyun Kim
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea
| | - Ki Chang Nam
- Department of Medical Engineering, Dongguk University College of Medicine, Gyeonggi-do 10326, Republic of Korea
| | - Jong-Chul Park
- Department of Medical Engineering, Yonsei University College of Medicine, Seoul 03722, Republic of Korea; Brain Korea 21 PLUS Project for Medical Science, Yonsei University College of Medicine, Seoul 03722, Republic of Korea.
| |
Collapse
|
21
|
Gorini G, Gamberi T, Fiaschi T, Mannelli M, Modesti A, Magherini F. Irreversible plasma and muscle protein oxidation and physical exercise. Free Radic Res 2018; 53:126-138. [PMID: 30513020 DOI: 10.1080/10715762.2018.1542141] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
The imbalance between the reactive oxygen (ROS) and nitrogen (RNS) species production and their handling by the antioxidant machinery (low molecular weight antioxidant molecules and antioxidant enzymes), also known as oxidative stress, is a condition caused by physiological and pathological processes. Moreover, oxidative stress may be due to an overproduction of free radicals during physical exercise. Excess of radical species leads to the modification of molecules, such as proteins - the most susceptible to oxidative modification - lipids and DNA. With regard to the oxidation of proteins, carbonylation is an oxidative modification that has been widely described. Several studies have detected changes in the total amount of protein carbonyls following different types of physical exercise, but only few of these identified the specific amino acidic residues targets of such oxidation. In this respect, proteomic approaches allow to identify the proteins susceptible to carbonylation and in many cases, it is also possible to identify the specific protein carbonylation sites. This review focuses on the role of protein oxidation, and specifically carbonyl formation, for plasma and skeletal muscle proteins, following different types of physical exercise performed at different intensities. Furthermore, we focused on the proteomic strategies used to identify the specific protein targets of carbonylation. Overall, our analysis suggests that regular physical activity promotes a protection against protein carbonylation, due to the activation of the antioxidant defence or of the turnover of protein carbonyls. However, we can conclude that from the comprehensive bibliography analysed, there is no clearly defined specific physiological role about this post-translational modification of proteins.
Collapse
Affiliation(s)
- Giulia Gorini
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Tania Gamberi
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Tania Fiaschi
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Michele Mannelli
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Alessandra Modesti
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| | - Francesca Magherini
- a Department of Biomedical, Experimental and Clinical Sciences "Mario Serio" , University of Florence , Florence , Italy
| |
Collapse
|
22
|
Sgarbi G, Liuzzi F, Baracca A, Solaini G. Resveratrol preserves mitochondrial function in a human post-mitotic cell model. J Nutr Biochem 2018; 62:9-17. [PMID: 30216747 DOI: 10.1016/j.jnutbio.2018.07.017] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Revised: 06/18/2018] [Accepted: 07/25/2018] [Indexed: 02/06/2023]
Abstract
Dysfunctions caused by genetic defects in the mitochondrial DNA (mtDNA) of humans are called mitochondrial diseases; however, mtDNA mutations are also associated with aging and age-related diseases. Here, we present an original cellular model that allows gathering information on molecules that might contrast or prevent mitochondrial dysfunctions and their related diseases. This model allowed us to show that resveratrol (RSV), a phytochemical present in food, exerts protective effects at low concentrations on resting human fibroblasts carrying dysfunctional respiratory chain Complex I. Cells were maintained both in resting condition, to mimic the high energy demanding post-mitotic tissues (serum absence and gramicidin presence), and under glucose deficiency to push the synthesis of ATP via oxidative phosphorylation. Pre-incubation with RSV prolonged the viability of the fibroblasts exposed to rotenone, a well-known specific inhibitor of the respiratory chain Complex I, and decreased mitochondrial fragmentation. It significantly prevented the oxidative phosphorylation impairment indirectly caused by the rotenone-mediated Complex I inhibition, allowing for an almost complete preservation of the cellular ATP level. Indeed, RSV limited the rotenone-induced reactive oxygen species increase, allowing for the maintenance of a functional mitochondrial membrane potential. These findings indicate the potential usage of resveratrol to prevent or possibly treat many disorders, in which the bioenergetic defects and oxidative stress are the primary (mitochondrial encephalomyopathy), or the secondary (age-related diseases) causes of the pathology; and to also assist cell senescence during aging.
Collapse
Affiliation(s)
- Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - Francesca Liuzzi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy.
| | - Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy.
| |
Collapse
|
23
|
Zhang M, Wu Y, Wang M, Wang Y, Tausif R, Yang Y. Genistein rescues hypoxia-induced pulmonary arterial hypertension through estrogen receptor and β-adrenoceptor signaling. J Nutr Biochem 2018; 58:110-118. [DOI: 10.1016/j.jnutbio.2018.04.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2017] [Revised: 04/20/2018] [Accepted: 04/21/2018] [Indexed: 12/29/2022]
|
24
|
Siques P, Brito J, Pena E. Reactive Oxygen Species and Pulmonary Vasculature During Hypobaric Hypoxia. Front Physiol 2018; 9:865. [PMID: 30050455 PMCID: PMC6052911 DOI: 10.3389/fphys.2018.00865] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2017] [Accepted: 06/18/2018] [Indexed: 12/12/2022] Open
Abstract
An increasing number of people are living or working at high altitudes (hypobaric hypoxia) and therefore suffering several physiological, biochemical, and molecular changes. Pulmonary vasculature is one of the main and first responses to hypoxia. These responses imply hypoxic pulmonary vasoconstriction (HPV), remodeling, and eventually pulmonary hypertension (PH). These events occur according to the type and extension of the exposure. There is also increasing evidence that these changes in the pulmonary vascular bed could be mainly attributed to a homeostatic imbalance as a result of increased levels of reactive oxygen species (ROS). The increase in ROS production during hypobaric hypoxia has been attributed to an enhanced activity and expression of nicotinamide adenine dinucleotide phosphate oxidase (NADPH oxidase), though there is some dispute about which subunit is involved. This enzymatic complex may be directly induced by hypoxia-inducible factor-1α (HIF-1α). ROS has been found to be related to several pathways, cells, enzymes, and molecules in hypoxic pulmonary vasculature responses, from HPV to inflammation, and structural changes, such as remodeling and, ultimately, PH. Therefore, we performed a comprehensive review of the current evidence on the role of ROS in the development of pulmonary vasculature changes under hypoxic conditions, with a focus on hypobaric hypoxia. This review provides information supporting the role of oxidative stress (mainly ROS) in the pulmonary vasculature’s responses under hypobaric hypoxia and depicting possible future therapeutics or research targets. NADPH oxidase-produced oxidative stress is highlighted as a major source of ROS. Moreover, new molecules, such as asymmetric dimethylarginine, and critical inflammatory cells as fibroblasts, could be also involved. Several controversies remain regarding the role of ROS and the mechanisms involved in hypoxic responses that need to be elucidated.
Collapse
Affiliation(s)
- Patricia Siques
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Julio Brito
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| | - Eduardo Pena
- Institute of Health Studies, Arturo Prat University, Iquique, Chile
| |
Collapse
|
25
|
Sgarbi G, Gorini G, Liuzzi F, Solaini G, Baracca A. Hypoxia and IF₁ Expression Promote ROS Decrease in Cancer Cells. Cells 2018; 7:E64. [PMID: 29933600 PMCID: PMC6071258 DOI: 10.3390/cells7070064] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2018] [Revised: 06/15/2018] [Accepted: 06/19/2018] [Indexed: 12/11/2022] Open
Abstract
The role of reactive oxygen species (ROS) in the metabolic reprogramming of cells adapted to hypoxia and the interplay between ROS and hypoxia in malignancy is under debate. Here, we examined how ROS levels are modulated by hypoxia in human cancer compared to untransformed cells. Short time exposure (20 min) of either fibroblasts or 143B osteosarcoma cells to low oxygen tension down to 0.5% induced a significant decrease of the cellular ROS level, as detected by the CellROX fluorescent probe (−70%). Prolonging the cells’ exposure to hypoxia for 24 h, ROS decreased further, reaching nearly 20% of the normoxic value. In this regard, due to the debated role of the endogenous inhibitor protein (IF₁) of the ATP synthase complex in cancer cell bioenergetics, we investigated whether IF₁ is involved in the control of ROS generation under severe hypoxic conditions. A significant ROS content decrease was observed in hypoxia in both IF₁-expressing and IF₁- silenced cells compared to normoxia. However, IF₁-silenced cells showed higher ROS levels compared to IF1-containing cells. In addition, the MitoSOX Red-measured superoxide level of all the hypoxic cells was significantly lower compared to normoxia; however, the decrease was milder than the marked drop of ROS content. Accordingly, the difference between IF₁-expressing and IF₁-silenced cells was smaller but significant in both normoxia and hypoxia. In conclusion, the interplay between ROS and hypoxia and its modulation by IF₁ have to be taken into account to develop therapeutic strategies against cancer.
Collapse
Affiliation(s)
- Gianluca Sgarbi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Giulia Gorini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
- Department of Biomedical, Experimental, and Clinical Sciences "Mario Serio", University of Florence, Florence 50121, Italy.
| | - Francesca Liuzzi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Giancarlo Solaini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| | - Alessandra Baracca
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, Bologna 40126, Italy.
| |
Collapse
|
26
|
Sgarbi G, Barbato S, Costanzini A, Solaini G, Baracca A. The role of the ATPase inhibitor factor 1 (IF 1) in cancer cells adaptation to hypoxia and anoxia. BIOCHIMICA ET BIOPHYSICA ACTA. BIOENERGETICS 2018; 1859:99-109. [PMID: 29097244 DOI: 10.1016/j.bbabio.2017.10.007] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Revised: 10/19/2017] [Accepted: 10/27/2017] [Indexed: 12/23/2022]
Abstract
The physiological role of the mitochondrial ATP synthase complex is to generate ATP through oxidative phosphorylation. Indeed, the enzyme can reverse its activity and hydrolyze ATP under ischemic conditions, as shown in isolated mitochondria and in mammalian heart and liver. However, what occurs when cancer cells experience hypoxia or anoxia has not been well explored. In the present study, we investigated the bioenergetics of cancer cells under hypoxic/anoxic conditions with particular emphasis on ATP synthase, and the conditions driving it to work in reverse. In this context, we further examined the role exerted by its endogenous inhibitor factor, IF1, that it is overexpressed in cancer cells. Metabolic and bioenergetic analysis of cancer cells exposed to severe hypoxia (down to 0.1% O2) unexpectedly showed that Δψm is preserved independently of the presence of IF1 and that ATP synthase still phosphorylates ADP though at a much lower rate than in normoxia. However, when we induced an anoxia-mimicking condition by collapsing ΔμΗ+ with the FCCP uncoupler, the IF1-silenced clones only reversed the ATP synthase activity hydrolyzing ATP in order to reconstitute the electrochemical proton gradient. Notably, in cancer cells IF1 overexpression fully prevents ATP synthase hydrolytic activity activation under uncoupling conditions. Therefore, our results suggest that IF1 overexpression promotes cancer cells survival under temporary anoxic conditions by preserving cellular ATP despite mitochondria dysfunction.
Collapse
Affiliation(s)
- G Sgarbi
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - S Barbato
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - A Costanzini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy
| | - G Solaini
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy.
| | - A Baracca
- Department of Biomedical and Neuromotor Sciences, Laboratory of Biochemistry and Mitochondrial Pathophysiology, University of Bologna, via Irnerio, 48, 40126 Bologna, Italy.
| |
Collapse
|
27
|
Bordt EA. The importance of controlling in vitro oxygen tension to accurately model in vivo neurophysiology. Neurotoxicology 2017; 66:213-220. [PMID: 29102646 DOI: 10.1016/j.neuro.2017.10.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2017] [Revised: 10/27/2017] [Accepted: 10/31/2017] [Indexed: 12/13/2022]
Abstract
The majority of in vitro studies modeling in vivo conditions are performed on the lab bench in atmospheric air. However, the oxygen tension (pO2) present in atmospheric air (160mm Hg, ∼21% O2) is in great excess to the pO2 that permeates tissues within the brain (5-45mm Hg, ∼1-6% O2). This review will discuss the differentiation between pO2 in the in vivo environment and the pO2 commonly used during in vitro experiments, and how this could affect assay outcomes. Also highlighted are studies linking changes in pO2 to changes in cellular function, particularly the role of pO2 in mitochondrial function, reactive oxygen species production, and cellular growth and differentiation. The role of hypoxia inducible factor 1 and oxygen sensing is also presented. Finally, emerging literature exploring sex differences in tissue oxygenation is discussed.
Collapse
Affiliation(s)
- Evan A Bordt
- Department of Pediatrics, Lurie Center for Autism, Massachusetts General Hospital for Children, Harvard Medical School, Boston, MA, 02129, USA.
| |
Collapse
|