1
|
Bouquerel C, Dumas S, Abedelnour E, Simkova E, Gropplero G, Meddahi L, Cinquin B, Tatoulian M, César W, Descroix S. Characterization of the oxygen properties of a hybrid glass chip designed for precise on chip oxygen control. LAB ON A CHIP 2025; 25:2449-2461. [PMID: 40277173 DOI: 10.1039/d4lc01017e] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/26/2025]
Abstract
Despite its relevance in several research fields, the regulation of dissolved gas concentration in microfluidic chips remains overlooked. Precise control of dissolved oxygen levels is of importance for life science applications, especially for faithfully replicating in vivo tissue conditions in organ-on-chips. The current methods to control oxygen on-chip rely on the use of chemical scavengers, on the integration of an additional gas channel or on the perfusion of a liquid pre-equilibrated at a set oxygen level. However, for precise oxygen control, these microfluidic devices must be made from gas-impermeable materials. In this regard, glass is a material of choice due to its complete impermeability, but its microfabrication often requires specific clean room processes. Here, we report a low-tech fabrication method for a hybrid glass chip, which involves assembling glass components using an adhesion process. To evaluate this chip's suitability for use under highly controlled oxygen conditions, we developed a two-step assessment protocol. This involved determining the time needed to reach a target oxygen level during perfusion and measuring the reoxygenation time following the cessation of flow. Based on a dual approach of simulations and experiments, we emphasized crucial adhesive properties such as oxygen diffusion and solubility and proposed a range of well-suited adhesive materials. Finally, we demonstrated the interest of this hybrid glass chip for on-chip cell culture and cell respiration measurements. This work paves the way for broader accessibility in producing low tech gas-tight microfluidic chips for diverse applications.
Collapse
Affiliation(s)
- Charlotte Bouquerel
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
- Stress et Cancer, Inserm, U830, Institut Curie, Equipe labelisée par la Ligue Nationale Contre le Cancer, PSL Research University, 26 rue d'Ulm, 75005, Paris, France
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Simon Dumas
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
| | - Elias Abedelnour
- Chimie ParisTech, PSL University Paris, CNRS, Institut de Recherche de Chimie Paris, UMR8247, 11 rue Pierre et Marie Curie, 75005 Paris, France
- Sanofi, Integrated Drug Discovery - IDD France, 1 Avenue Pierre Brossolette, Chilly Mazarin, France
| | - Ester Simkova
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
| | - Giacomo Gropplero
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
| | - Linda Meddahi
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
| | - Bertrand Cinquin
- UMS 3750, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France
| | - Michael Tatoulian
- Chimie ParisTech, PSL University Paris, CNRS, Institut de Recherche de Chimie Paris, UMR8247, 11 rue Pierre et Marie Curie, 75005 Paris, France
| | - William César
- Fluigent, 67 avenue de Fontainebleau, 94270, Le Kremlin-Bicêtre, France
| | - Stephanie Descroix
- Macromolécules et Microsystèmes en Biologie et Médecine, UMR 168, Institut Curie, Institut Pierre Gilles de Gennes, 6 rue Jean Calvin, 75005, Paris, France.
| |
Collapse
|
2
|
Francis T, Soendenbroe C, Lazarus NR, Mackey AL, Harridge SDR. Insights into human muscle biology from human primary skeletal muscle cell culture. J Muscle Res Cell Motil 2025:10.1007/s10974-025-09696-w. [PMID: 40346328 DOI: 10.1007/s10974-025-09696-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Accepted: 04/28/2025] [Indexed: 05/11/2025]
Abstract
This review arises from the symposium held in honour of Prof Jenny Morgan at UCL in 2024 and the authors would like to acknowledge the outstanding contribution that Prof Morgan has made to the field of translational muscle cell biology. Prof Morgan published a review article in 2010 entitled: Are human and mice satellite cells really the same? In which the authors highlighted differences between species which are still pertinent to skeletal muscle cell culture studies today. To our knowledge there are no comprehensive reviews which outline the considerable work that has been undertaken using human primary skeletal muscle origin cells as the main model system. This review highlights the multitude of muscle biology that has been investigated using human primary cells, as well as discussing the advantages and disadvantages over other cell models. We also discuss future directions for primary cell culture models utilising the latest technologies in cell type specificity and culture systems.
Collapse
Affiliation(s)
- Thomas Francis
- Centre for Human & Applied Physiological Sciences, Basic & Medical Biosciences, Faculty of Life Science & Medicine, King's College London, London, UK.
| | - Casper Soendenbroe
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Norman R Lazarus
- Centre for Human & Applied Physiological Sciences, Basic & Medical Biosciences, Faculty of Life Science & Medicine, King's College London, London, UK
| | - Abigail L Mackey
- Institute of Sports Medicine Copenhagen, Department of Orthopaedic Surgery, Copenhagen University Hospital-Bispebjerg and Frederiksberg, Copenhagen, Denmark
- Center for Healthy Aging, Department of Clinical Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Stephen D R Harridge
- Centre for Human & Applied Physiological Sciences, Basic & Medical Biosciences, Faculty of Life Science & Medicine, King's College London, London, UK
| |
Collapse
|
3
|
Wang W, Pan L, He H, Xue H, Huang H, Samosir AM, Fu X, Shen Y. Systematic Engineering for Efficient Uric Acid-Degrading Activity in Probiotic Yeast Saccharomyces boulardii. ACS Synth Biol 2025. [PMID: 40340401 DOI: 10.1021/acssynbio.4c00831] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/10/2025]
Abstract
Hyperuricemia, caused by uric acid disequilibrium, is a prevalent metabolic disease that most commonly manifests as gout and is closely associated with a spectrum of other comorbidities such as renal disorders and cardiovascular diseases. While natural and engineered probiotics that promote catabolism of uric acid in the intestine have shown promise in relieving hyperuricemia, limitations in strain efficiency and the requirements for achieving high performance remain major hurdles in the practical application of probiotic-mediated prevention and management. Here, we employed a systematic strategy to engineer a high-efficiency uric acid catabolism pathway in S. cerevisiae. An uricase from Vibrio vulnificus, exhibiting high-level activity in S. cerevisiae, was identified as the uric acid-degrading component. The expression level and stability of urate transporter UapA were improved by constructing a chimera, enabling reliable uric acid import in S. cerevisiae. Additionally, constitutive promoters were selected and combinatorially assembled with the two functional components, creating a collection of pathways that confer varied levels of uric acid catabolic activity to S. cerevisiae. The best-performing pathway can express uric acid-degrading activity up to 365.32 ± 20.54 μmol/h/OD, requiring only simple cultivation steps. Eventually, we took advantage of the genetic similarity between model organism S. cerevisiae and probiotic S. boulardii and integrated the optimized pathway into identified high-expression integration loci in the S. boulardii genome. The activity can be stably maintained under high-density fermentation conditions. Overall, this study provided a high-potential hyperuricemia-managing yeast probiotic strain, demonstrating the capabilities of developing recombinant probiotics.
Collapse
Affiliation(s)
- Wenzhuo Wang
- School of Biology and Biological Engineering, South China University of Technology, Guangzhou Higher Education Mega Center, Panyu District, Guangzhou, Guangdong 510006, PR China
- BGI Research, Hangzhou 310030, China
| | - Lei Pan
- BGI Research, Hangzhou 310030, China
- State Key Laboratory of Genome and Multi-omics Technologies, BGI Research, Hangzhou 310030, China
| | | | | | - He Huang
- BGI Research, Changzhou 213299, China
| | | | - Xian Fu
- BGI Research, Changzhou 213299, China
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China
| | - Yue Shen
- BGI Research, Changzhou 213299, China
- BGI Research, Shenzhen 518083, China
- Guangdong Provincial Key Laboratory of Genome Read and Write, BGI Research, Shenzhen 518083, China
| |
Collapse
|
4
|
Shang KM, Suzuki T, Kato H, Toyoda T, Tai YC, Komatsu H. Oxygen dynamics and delivery strategies to enhance beta cell replacement therapy. Am J Physiol Cell Physiol 2025; 328:C1667-C1684. [PMID: 40204281 DOI: 10.1152/ajpcell.00984.2024] [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: 12/13/2024] [Revised: 01/06/2025] [Accepted: 04/01/2025] [Indexed: 04/11/2025]
Abstract
Beta cell replacement therapy via pancreatic islet transplantation offers a promising treatment for type 1 diabetes as an alternative to insulin injections. However, posttransplantation oxygenation remains a critical challenge; isolated islets from donors lose vascularity and rely on slow oxygen diffusion for survival until revascularization occurs in the host tissue. This often results in significant hypoxia-induced acute graft loss. Overcoming the oxygenation barrier is crucial for advancing islet transplantation. This review is structured in three sections: the first examines oxygen dynamics in islet transplantation, focusing on factors affecting oxygen supply, including vascularity. It highlights oxygen dynamics specific to both transplant sites and islet grafts, with particular attention to extrahepatic sites such as subcutaneous tissue. The second section explores current oxygen delivery strategies, categorized into two main approaches: augmenting oxygen supply and enhancing effective oxygen solubility. The final section addresses key challenges, such as the lack of a clearly defined oxygen threshold for islet survival and the limited precision in measuring oxygen levels within small islet constructs. Recent advancements addressing these challenges are introduced. By deepening the understanding of oxygen dynamics and identifying current obstacles, this review aims to guide the development of innovative strategies for future research and clinical applications. These advancements are anticipated to enhance transplantation outcomes and bring us closer to a cure for type 1 diabetes.
Collapse
Affiliation(s)
- Kuang-Ming Shang
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Tomoharu Suzuki
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Hiroyuki Kato
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| | - Taro Toyoda
- Department of Life Science Frontiers, Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yu-Chong Tai
- Department of Medical Engineering, California Institute of Technology, Pasadena, California, United States
| | - Hirotake Komatsu
- Division of Transplant Surgery, Department of Surgery, University of California San Francisco, San Francisco, California, United States
| |
Collapse
|
5
|
Hill M, Meloni GN, Frenguelli BG, Unwin PR. Transient Single Cell Hypoxia Induced by Localized Galvanostatic Oxygen Challenge. ACS MEASUREMENT SCIENCE AU 2025; 5:234-241. [PMID: 40255598 PMCID: PMC12006948 DOI: 10.1021/acsmeasuresciau.4c00100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2024] [Revised: 02/25/2025] [Accepted: 02/25/2025] [Indexed: 04/22/2025]
Abstract
Studying cells exposed to low and controllable oxygen levels is key to investigating various fundamental aspects of pathological states, such as stroke and cancer. At present, available methodologies applied in vitro focus on large groups of cells exposed to low oxygen conditions through slow-time approaches, such as environmental incubators or microfluidic devices. Here, we demonstrate a novel approach for titrating the local oxygen concentration around individual adhered PC12 cells, enabling single cells within a population to be exposed to hypoxic-like conditions. A 25 μm diameter platinum disk microelectrode performing the oxygen reduction reaction (ORR) at constant current (galvanostatic control) is used as a microscale oxygen scavenger that can be positioned precisely over individual cells. By coupling the galvanostatic oxygen challenge with confocal laser scanning microscopy (CLSM) and a commercially available hypoxia dye (Image-iT Green hypoxia reagent), we monitor the response of single cells when exposed to depleted oxygen concentrations over time. Numerical simulations are used to characterize the oxygen and pH gradient imposed by the microelectrode at different cathodic currents, revealing that within seconds, the oxygen depletion zone reaches a steady-state condition, extending a few microelectrode radii into solution, while the corresponding pH gradient is strongly compressed by the buffer solution. Cells under the microelectrode show a marked increase in average fluorescence rate relative to control, reporting their hypoxic conditions and demonstrating the effectiveness of the proposed method. Heterogenous cell response in a challenged group is also observed, highlighting the ability of this approach to investigate the natural heterogeneity in cell populations. This work provides a platform and roadmap for future studies of cellular systems where the ability to control and vary oxygen concentration on a rapid time scale would be beneficial.
Collapse
Affiliation(s)
- Marlene
H. Hill
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Molecular
Analytical Science Centre for Doctoral Training (MAS CDT), University of Warwick, Coventry CV4 7AL, U.K.
| | - Gabriel N. Meloni
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
- Institute
of Chemistry, Department of Fundamental Chemistry, University of São Paulo, São Paulo, São Paulo 05508-000, Brazil
| | | | - Patrick R. Unwin
- Department
of Chemistry, University of Warwick, Coventry CV4 7AL, U.K.
| |
Collapse
|
6
|
Sikorová M, Klener P, Tonarová P, Kalbáčová MH. Interactions between leukemia and feeders in co-cultivation under hypoxia. BMC Cancer 2025; 25:678. [PMID: 40229651 PMCID: PMC11995666 DOI: 10.1186/s12885-025-13988-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2025] [Accepted: 03/20/2025] [Indexed: 04/16/2025] Open
Abstract
BACKGROUND Leukemia is driven by complex interactions within the inherently hypoxic bone marrow microenvironment, impacting both disease progression and therapeutic resistance. Co-cultivation of leukemic cells with feeder cells has emerged as a valuable tool to mimic the bone marrow niche. This study explores the interplay between human commercial SD-1 and patient-derived UPF26K leukemic cell lines with feeders - human fibroblasts (NHDF) and mesenchymal stem cells (hMSCs) under normoxic and hypoxic conditions. RESULTS Co-cultivation with feeders significantly enhances proliferation and glycolytic activity in the SD-1 cells, improving their viability, while this interaction inhibits the growth and glucose metabolism of the feeders, particularly NHDF. In contrast, UPF26K cells show reduced proliferation when co-cultivated with the feeders while this interaction stimulates NHDF and hMSCs proliferation and glycolysis but reduce their mitochondrial metabolism with hypoxia amplifying these effects. CONCLUSIONS Cells that switch to glycolysis during co-cultivation, particularly under hypoxia, benefit most from these low oxygen conditions. Due to this leukemic cells' response heterogeneity, targeting microenvironmental interactions and oxygen levels is crucial for personalized leukemia therapy. Advancing co-cultivation models, particularly through innovations like spheroids, can further enhance in vitro studies of primary leukemic cells and support the testing of novel therapies.
Collapse
Affiliation(s)
- Miriama Sikorová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Pavel Klener
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Pavla Tonarová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic
| | - Marie Hubálek Kalbáčová
- Institute of Pathological Physiology, 1st Faculty of Faculty of Medicine, Charles University, U nemocnice 5, Prague, 128 53, Czech Republic.
| |
Collapse
|
7
|
Lotoux A, Caulat L, Martins Alves C, Alves Feliciano C, Morvan C, Folgosa F, Martin-Verstraete I. Defense arsenal of the strict anaerobe Clostridioides difficile against reactive oxygen species encountered during its infection cycle. mBio 2025; 16:e0375324. [PMID: 40111048 PMCID: PMC11980386 DOI: 10.1128/mbio.03753-24] [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: 12/05/2024] [Accepted: 02/18/2025] [Indexed: 03/22/2025] Open
Abstract
Clostridioides difficile, a strict anaerobe, is the major cause of antibiotic-associated diarrhea. This enteropathogen must adapt to oxidative stress mediated by reactive oxygen species (ROS), notably those released by the neutrophils and macrophages recruited to the site of infection or those endogenously produced upon high oxygen (O2) exposure. C. difficile uses a superoxide reductase, Sor, and several peroxidases to detoxify ROS. We showed that Sor has a superoxide reductase activity in vitro and protects the bacterium from exposure to menadione, a superoxide donor. After confirming the peroxidase activity of the rubrerythrin, Rbr, we showed that this enzyme together with the peroxiredoxin, Bcp, plays a central role in the detoxification of H2O2 and promotes the survival of C. difficile in the presence of not only H2O2 but also air or 4% O2. Under high O2 concentrations encountered in the gastrointestinal tract, the bacterium generated endogenous H2O2. The two O2 reductases, RevRbr2 and FdpF, have also a peroxidase activity and participate in H2O2 resistance. The CD0828 gene, which also contributes to H2O2 protection, forms an operon with rbr, sor, and perR encoding a H2O2-sensing repressor. The expression of the genes encoding the ROS reductases and the CD0828 protein was induced upon exposure to either H2O2 or air. We showed that the induction of the rbr operon is mediated not only by PerR but also by OseR, a recently identified O2-responsive regulator of C. difficile, and indirectly by σB, the sigma factor of the stress response, whereas the expression of bcp is only controlled by σB. IMPORTANCE ROS plays a fundamental role in intestinal homeostasis, limiting the proliferation of pathogenic bacteria. Clostridioides difficile is an important enteropathogen that induces an intense immune response, characterized by the massive recruitment of immune cells responsible for secreting ROS, mainly H2O2 and superoxide. We showed in this work that ROS exposure leads to the production of an armada of enzymes involved in ROS detoxification. This includes a superoxide reductase and four peroxidases, Rbr, Bcp, revRbr2, and FdpF. These enzymes likely contribute to the survival of vegetative cells of C. difficile in the colon during the host immune response. Distinct regulations are also observed for the genes encoding the ROS detoxification enzymes allowing a fine tuning of the adaptive response to ROS exposure. Understanding the mechanisms of ROS protection during infection could shed light on how C. difficile survives under conditions of an exacerbated inflammatory response.
Collapse
Affiliation(s)
- Aurélie Lotoux
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, Île-de-France, France
| | - Léo Caulat
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, Île-de-France, France
| | - Catarina Martins Alves
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Lisbon, Portugal
| | - Carolina Alves Feliciano
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Lisbon, Portugal
| | - Claire Morvan
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, Île-de-France, France
| | - Filipe Folgosa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Lisbon, Portugal
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université Paris Cité, UMR CNRS 6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, Île-de-France, France
- Institut Universitaire de France, Paris, Île-de-France, France
| |
Collapse
|
8
|
Chalkias A. On the Intensity of the Microvascular Magnetic Field in Normal State and Septic Shock. J Clin Med 2025; 14:2496. [PMID: 40217945 PMCID: PMC11989563 DOI: 10.3390/jcm14072496] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/03/2025] [Accepted: 04/03/2025] [Indexed: 04/14/2025] Open
Abstract
Background: Capillary tortuosity is a morphological variant of microcirculation. However, the mechanisms by which tortuous vessels meet metabolic requirements in health and disease remain unknown. We recently reported that capillary tortuosity score (CTS) is significantly higher in patients with septic shock than in steady-state individuals, and that CTS is significantly associated with alveolar-to-arterial oxygen (A-a O2) gradient and oxygen debt in septic shock patients. Objective: We aimed to investigate the characteristics of the magnetic fields in the sublingual microcirculation of individuals with normal physiology and patients with septic shock. Methods: Systemic hemodynamics were recorded, and sublingual microcirculation was monitored using sidestream dark field (SDF+) imaging. The number of capillary red blood cells (NRBC), the intensity of the magnetic field of a red blood cell (HRBC), the intensity of the magnetic field of each capillary (HCAP), and the intensity with which the magnetic field of a capillary acts on an RBC (FCAP) were calculated. Results: Significant differences in macro- and microhemodynamic variables were observed between the two groups. Although NRBC was significantly higher in individuals with steady-state physiology [87.4 (87.12) vs. 12.23 (6.9)], HRBC was significantly stronger in patients with septic shock [5.9 × 10-16 (6.9 × 10-16) A m-1 vs. 1.6 × 10-15 (1.4 × 10-15) A m-1]. No significant difference was observed in HCAP [2.16 × 10-14 (2.17 × 10-14) A m-1 vs. 1.34 × 10-14 (1.23 × 10-14) A m-1] and FCAP [1.66 × 10-24 (3.36 × 10-24) A m-1 vs. 6.44 × 10-25 (1.1 × 10-24) A m-1] between the two groups. In patients with septic shock, HRBC was associated with De Backer score (rho = -0.608) and venous-arterial carbon dioxide difference (rho = 0.569). In the same group, HCAP was associated with convective oxygen flow (rho = 0.790) and oxygen extraction ratio (rho = -0.596). Also, FCAP was significantly associated with base deficit (rho = 0.701), A-a O2 gradient (rho = 0.658), and oxygen debt (rho = -0.769). Conclusions: Despite the microcirculatory impairment in patients with septic shock, HRBC was significantly stronger in that group than in steady-state individuals. Also, HCAP and FCAP were comparable between the two groups. Tortuous vessels may function as biomagnetic coils that amplify RBC-induced magnetic fields, enhancing perfusion and oxygenation of adjacent tissues.
Collapse
Affiliation(s)
- Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5158, USA;
- OUTCOMES RESEARCH Consortium, Houston, TX 77030, USA
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| |
Collapse
|
9
|
Chiba N, Ohnishi T, Matsuguchi T. Hypoxia-Inducible Factor 1 Alpha Potentiates Lipopolysaccharide-Induced Expression of IL-13 and IL-33 in Mast Cells Under Hypoxia. Microbiol Immunol 2025; 69:247-255. [PMID: 39945318 DOI: 10.1111/1348-0421.13202] [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: 09/17/2024] [Revised: 01/21/2025] [Accepted: 01/23/2025] [Indexed: 04/08/2025]
Abstract
Lipopolysaccharide (LPS) is an exacerbating factor for allergic airway inflammation at least partly due to the activation of mast cells (MCs). LPS stimulates MCs to express both pro-inflammatory and type 2 cytokines, among which interleukin (IL)-13 is essential for the generation of allergic diseases. LPS also induces the expression of "alarmins" such as IL-25, IL-33, and thymic stromal lymphopoietin (TSLP) from various cell types including epithelial cells, and increased serum IL-33 levels were reported to correlate with disease severity of asthma. MCs reside in peripheral tissues where the oxygen concentration is significantly lower than that in the air and further decreased by inflammation and bronchoconstriction in asthma. However, the effects of hypoxia on LPS-induced cytokine expression in MCs have not been fully elucidated. Here we show that LPS induces Il4, Il6, Il13, Il33, Tnf, and Tslp mRNAs in MCs. Notably, hypoxia robustly enhanced expressions of Il13 and Il33, but not the other cytokines in LPS-stimulated MCs. We also found that this promotive effect is dependent on the presence of hypoxia-inducible factor (HIF) 1α protein. Our study will provide new insight on the role of MCs in the LPS-associated pathogenesis of allergic diseases.
Collapse
Affiliation(s)
- Norika Chiba
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tomokazu Ohnishi
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| | - Tetsuya Matsuguchi
- Department of Oral Biochemistry, Kagoshima University Graduate School of Medical and Dental Sciences, Kagoshima, Japan
| |
Collapse
|
10
|
Sastre J, Pérez S, Sabater L, Rius-Pérez S. Redox signaling in the pancreas in health and disease. Physiol Rev 2025; 105:593-650. [PMID: 39324871 DOI: 10.1152/physrev.00044.2023] [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: 12/04/2023] [Revised: 09/11/2024] [Accepted: 09/17/2024] [Indexed: 09/27/2024] Open
Abstract
This review addresses oxidative stress and redox signaling in the pancreas under healthy physiological conditions as well as in acute pancreatitis, chronic pancreatitis, pancreatic cancer, and diabetes. Physiological redox homeodynamics is maintained mainly by NRF2/KEAP1, NF-κB, protein tyrosine phosphatases, peroxisome proliferator-activated receptor-γ coactivator 1α (PGC1α), and normal autophagy. Depletion of reduced glutathione (GSH) in the pancreas is a hallmark of acute pancreatitis and is initially accompanied by disulfide stress, which is characterized by protein cysteinylation without increased glutathione oxidation. A cross talk between oxidative stress, MAPKs, and NF-κB amplifies the inflammatory cascade, with PP2A and PGC1α as key redox regulatory nodes. In acute pancreatitis, nitration of cystathionine-β synthase causes blockade of the transsulfuration pathway leading to increased homocysteine levels, whereas p53 triggers necroptosis in the pancreas through downregulation of sulfiredoxin, PGC1α, and peroxiredoxin 3. Chronic pancreatitis exhibits oxidative distress mediated by NADPH oxidase 1 and/or CYP2E1, which promotes cell death, fibrosis, and inflammation. Oxidative stress cooperates with mutant KRAS to initiate and promote pancreatic adenocarcinoma. Mutant KRAS increases mitochondrial reactive oxygen species (ROS), which trigger acinar-to-ductal metaplasia and progression to pancreatic intraepithelial neoplasia (PanIN). ROS are maintained at a sufficient level to promote cell proliferation, while avoiding cell death or senescence through formation of NADPH and GSH and activation of NRF2, HIF-1/2α, and CREB. Redox signaling also plays a fundamental role in differentiation, proliferation, and insulin secretion of β-cells. However, ROS overproduction promotes β-cell dysfunction and apoptosis in type 1 and type 2 diabetes.
Collapse
Affiliation(s)
- Juan Sastre
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Salvador Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
| | - Luis Sabater
- Liver, Biliary and Pancreatic Unit, Hospital Clínico, Department of Surgery, Faculty of Medicine, University of Valencia, Valencia, Spain
| | - Sergio Rius-Pérez
- Department of Physiology, Faculty of Pharmacy, University of Valencia, Valencia, Spain
- Department of Cell Biology, Functional Biology and Physical Anthropology, Faculty of Biology, University of Valencia, Valencia, Spain
| |
Collapse
|
11
|
Penjweini R, Link KA, Qazi S, Mattu N, Zuchowski A, Vasta A, Sackett DL, Knutson JR. Low dose Taxol causes mitochondrial dysfunction in actively respiring cancer cells. J Biol Chem 2025; 301:108450. [PMID: 40147771 PMCID: PMC12052845 DOI: 10.1016/j.jbc.2025.108450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2024] [Revised: 03/03/2025] [Accepted: 03/23/2025] [Indexed: 03/29/2025] Open
Abstract
Mitochondrial oxygen consumption, dynamics, and morphology play roles in the occurrence, development, and drug resistance of cancer; thus, they are main targets for many anticancer drugs. Increased mitochondrial oxygen consumption and impaired oxygen delivery creates hypoxia, which influences the balance of metabolic cofactors for biogenesis, disease progression, and response to therapeutics. We therefore investigated the effects of Taxol, a well-known anticancer drug, on mitochondrial respiration (principally via a measure of oxidative phosphorylation versus glycolysis), morphology, and dynamics. The concomitant effects of Taxol on mitochondrial ATP and reactive oxygen species production, mitochondrial membrane potential, radical-induced formation of carbonyl groups, mitochondrial release of cytochrome c, as well as cell cycle were investigated. Cells used in this study include the following: A549 (non-small-cell lung epithelial cancer cell line), A549-ρ0 (mitochondrial DNA-depleted derivative of A549), and BEAS-2B (a noncancer cell line derived from normal bronchial epithelium), as well as PC3 (prostate cancer) and HepG2 (hepatocellular carcinoma); these cell lines are known to have disparate metabolic profiles. Using a multitude of fluorescence-based measurements, we show that Taxol, even at a low dose, still adversely affects mitochondria of actively respiring (aerobic) cancer cells. We find an increase in mitochondrial ROS and cytochrome c release, suppression of ATP production and oxidative phosphorylation, fragmentation of the mitochondrial network, and disruption of mitochondria-microtubule linkage. We find these changes in oxidative, but not glycolytic, cancer cells. Noncancer cells, which are oxidative, do not show these changes.
Collapse
Affiliation(s)
- Rozhin Penjweini
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Katie A Link
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Shureed Qazi
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Nikhil Mattu
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Adam Zuchowski
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Alexandra Vasta
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA
| | - Dan L Sackett
- Cytoskeletal Dynamics Group, Division of Basic and Translational Biophysics, Eunice Kennedy Shriver National Institute of Child Health and Human Development, NIH, Bethesda, Maryland, USA.
| | - Jay R Knutson
- Laboratory of Advanced Microscopy and Biophotonics, National Heart, Lung, and Blood Institute (NHLBI), NIH, Bethesda, Maryland, USA.
| |
Collapse
|
12
|
Schneider L, Begovic M, Zhou X, Hamdani N, Akin I, El-Battrawy I. Catecholaminergic Polymorphic Ventricular Tachycardia: Advancing From Molecular Insights to Preclinical Models. J Am Heart Assoc 2025; 14:e038308. [PMID: 40079282 DOI: 10.1161/jaha.124.038308] [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: 08/14/2024] [Accepted: 12/19/2024] [Indexed: 03/15/2025]
Abstract
Inherited cardiac channelopathies are linked to a heightened risk of sudden cardiac death. Despite evolving knowledge on different genes for these inherited conditions, for certain subtypes, such as catecholaminergic polymorphic ventricular tachycardia syndrome, the specific genetic causes remain unidentified. The research of the pathophysiological mechanisms underlying catecholaminergic polymorphic ventricular tachycardia syndrome has been conducted through different in vitro and in vivo models, including genetically modified animal models, cardiac-specific transgenic models, pharmacological interventions in animal models, human-induced pluripotent stem cell-derived cardiomyocytes in 2- and 3-dimensional cardiac models. Recent research predominantly utilizes human-induced pluripotent stem cell-derived cardiomyocytes, focusing on genotype-phenotype correlations and pharmacological screening. The integration of cutting-edge techniques such as clustered regularly interspaced short palindromic repeats/Cas9 genome editing and 3-dimensional-engineered heart tissues has shed new light on the pathophysiological mechanisms of catecholaminergic polymorphic ventricular tachycardia, potentially enhancing drug therapies as part of personalized medicine approaches. This review emphasizes the diverse insights gained from both in vivo and in vitro studies of catecholaminergic polymorphic ventricular tachycardia, along with the application of these models in various research contexts.
Collapse
Affiliation(s)
- Luca Schneider
- Department of Cellular and Translational Physiology, Institute of Physiology Ruhr-University Bochum Bochum Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital Ruhr-University Bochum Bochum Germany
| | - Merima Begovic
- Department of Cellular and Translational Physiology, Institute of Physiology Ruhr-University Bochum Bochum Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital Ruhr-University Bochum Bochum Germany
| | - Xiaobo Zhou
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim Heidelberg University Mannheim Germany
- Key Laboratory of Medical Electrophysiology of Ministry of Education and Medical Electrophysiological Key Laboratory of Sichuan Province Institute of Cardiovascular Research, Southwest Medical University Luzhou Sichuan China
| | - Nazha Hamdani
- Department of Cellular and Translational Physiology, Institute of Physiology Ruhr-University Bochum Bochum Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital Ruhr-University Bochum Bochum Germany
- Department of Physiology Cardiovascular Research Institute, University Maastricht Maastricht The Netherlands
- HCEMM-SU Cardiovascular Comorbidities Research Group, Center for Pharmacology and Drug Research & Development, Department of Pharmacology and Pharmacotherapy Intézet címe Semmelweis University Budapest Hungary
- Department of Cardiology and Rhythmology St. Josef Hospital, Ruhr University Bochum Germany
| | - Ibrahim Akin
- Cardiology, Angiology, Haemostaseology, and Medical Intensive Care, Medical Centre Mannheim, Medical Faculty Mannheim Heidelberg University Mannheim Germany
| | - Ibrahim El-Battrawy
- Department of Cellular and Translational Physiology, Institute of Physiology Ruhr-University Bochum Bochum Germany
- Institut für Forschung und Lehre (IFL), Molecular and Experimental Cardiology, St. Josef Hospital Ruhr-University Bochum Bochum Germany
- Department of Cardiology and Rhythmology St. Josef Hospital, Ruhr University Bochum Germany
| |
Collapse
|
13
|
Chalkias A, Papagiannakis N, Katsifa K, Destounis A, Gravos A, Kanakaki S, Karapiperis G, Koufaki F, Prekates A, Tselioti P. Characterization of Sublingual Microvascular Tortuosity in Steady-State Physiology and Septic Shock. Biomedicines 2025; 13:691. [PMID: 40149667 PMCID: PMC11939869 DOI: 10.3390/biomedicines13030691] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2025] [Revised: 03/02/2025] [Accepted: 03/10/2025] [Indexed: 03/29/2025] Open
Abstract
Background: The characteristics of hemodynamic coherence in healthy states and disease remain unknown. Capillary tortuosity is a morphologic variant of microcirculatory vessels, but its effects have generally not been considered in the assessment of tissue perfusion and oxygenation. We investigated the role of sublingual capillary tortuosity in the hemodynamic coherence of anesthetized adult individuals with steady-state physiology (ASA 1) and patients with septic shock requiring emergency abdominal surgery (ASA 4E and 5E). Methods: Sublingual macro and microcirculatory variables, oxygen transport, metabolic parameters, and the capillary tortuosity score (CTS) were assessed. Results: Mean (SD) CTS was 0.55 (0.76) and 3.31 (0.86) in the steady-state and septic shock group, respectively (p < 0.001). In patients with septic shock, CTS was significantly associated with alveolar-to-arterial oxygen gradient (r = 0.658, p = 0.015) and oxygen debt (r = -0.769, p = 0.002). Significant differences were also observed in Consensus Proportion of Perfused Vessels (PPV; p < 0.001), Consensus PPV (small) (p < 0.001), Microvascular Flow Index (p < 0.001), vessel diameter (p < 0.001) and length (p < 0.001), wall shear stress (p < 0.001), lactate (p < 0.001), oxygen extraction ratio (p = 0.001), arterial oxygen content (p < 0.001), venous oxygen content (p < 0.001), oxygen delivery (p < 0.001), oxygen consumption (p < 0.001), and oxygen debt (p = 0.002) between the two groups. Conclusions: Sublingual tortuosity was essentially absent in individuals with steady-state physiology. In contrast, it was significantly increased and associated with Alveolar-to-arterial oxygen gradient and oxygen debt in critically ill patients with septic shock.
Collapse
Affiliation(s)
- Athanasios Chalkias
- Institute for Translational Medicine and Therapeutics, University of Pennsylvania Perelman School of Medicine, Philadelphia, PA 19104-5158, USA
- OUTCOMES RESEARCH Consortium, Houston, TX 77030, USA
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Nikolaos Papagiannakis
- First Department of Neurology, Eginition University Hospital, Medical School, National and Kapodistrian University of Athens, 11528 Athens, Greece
| | - Konstantina Katsifa
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Antonios Destounis
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Athanasios Gravos
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Sofia Kanakaki
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Georgios Karapiperis
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Faidra Koufaki
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Athanasios Prekates
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| | - Paraskevi Tselioti
- Department of Critical Care Medicine, General Hospital of Piraeus “Tzaneio”, 18536 Piraeus, Greece
| |
Collapse
|
14
|
Mishra S, Singh PR, Hu X, Lopez-Quezada L, Jinich A, Jahn R, Geurts L, Shen N, DeJesus MA, Hartman T, Rhee K, Zimmerman M, Dartois V, Jones RM, Jiang X, Almada-Monter R, Bourouiba L, Nathan C. Candidate transmission survival genome of Mycobacterium tuberculosis. Proc Natl Acad Sci U S A 2025; 122:e2425981122. [PMID: 40053362 PMCID: PMC11912377 DOI: 10.1073/pnas.2425981122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Accepted: 01/29/2025] [Indexed: 03/19/2025] Open
Abstract
Mycobacterium tuberculosis (Mtb), a leading cause of death from infection, completes its life cycle entirely in humans except for transmission through the air. To begin to understand how Mtb survives aerosolization, we mimicked liquid and atmospheric conditions experienced by Mtb before and after exhalation using a model aerosol fluid (MAF) based on the water-soluble, lipidic, and cellular constituents of necrotic tuberculosis lesions. MAF induced drug tolerance in Mtb, remodeled its transcriptome, and protected Mtb from dying in microdroplets desiccating in air. Yet survival was not passive: Mtb appeared to rely on hundreds of genes to survive conditions associated with transmission. Essential genes subserving proteostasis offered most protection. A large number of conventionally nonessential genes appeared to contribute as well, including genes encoding proteins that resemble antidesiccants. The candidate transmission survival genome of Mtb may offer opportunities to reduce transmission of tuberculosis.
Collapse
Affiliation(s)
- Saurabh Mishra
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Prabhat Ranjan Singh
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Xiaoyi Hu
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Landys Lopez-Quezada
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Adrian Jinich
- Department of Chemistry and Biochemistry, Skaggs School of Pharmacy and Pharmaceutical Sciences, University of California San Diego, San Diego, CA92093-0021
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA92093-0021
| | - Robin Jahn
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Luc Geurts
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Naijian Shen
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Michael A. DeJesus
- Laboratory of Host-Pathogen Biology, Rockefeller University, New York, NY10021
| | - Travis Hartman
- Department of Medicine, Weill Cornell Medicine, New York, NY10065
| | - Kyu Rhee
- Department of Medicine, Weill Cornell Medicine, New York, NY10065
| | - Matthew Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Veronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Richard M. Jones
- Department of Microbiology, University of Washington, Seattle, WA98195
| | - Xiuju Jiang
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| | - Ricardo Almada-Monter
- Department of Chemistry and Biochemistry, University of California San Diego, San Diego, CA92093-0021
| | - Lydia Bourouiba
- The Fluid Dynamics of Disease Transmission Laboratory, Fluids and Health Network, Department of Mechanical Engineering, Massachusetts Institute of Technology, Cambridge, MA02139
| | - Carl Nathan
- Department of Microbiology and Immunology, Weill Cornell Medicine, New York, NY10065
| |
Collapse
|
15
|
Mirchandani AS, Sanchez-Garcia MA, Walmsley SR. How oxygenation shapes immune responses: emerging roles for physioxia and pathological hypoxia. Nat Rev Immunol 2025; 25:161-177. [PMID: 39349943 DOI: 10.1038/s41577-024-01087-5] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 08/23/2024] [Indexed: 03/04/2025]
Abstract
Most eukaryotes require oxygen for their survival and, with increasing multicellular complexity, oxygen availability and delivery rates vary across the tissues of complex organisms. In humans, healthy tissues have markedly different oxygen gradients, ranging from the hypoxic environment of the bone marrow (where our haematopoietic stem cells reside) to the lungs and their alveoli, which are among the most oxygenated areas of the body. Immune cells are therefore required to adapt to varying oxygen availability as they move from the bone marrow to peripheral organs to mediate their effector functions. These changing oxygen gradients are exaggerated during inflammation, where oxygenation is often depleted owing to alterations in tissue perfusion and increased cellular activity. As such, it is important to consider the effects of oxygenation on shaping the immune response during tissue homeostasis and disease conditions. In this Review, we address the relevance of both physiological oxygenation (physioxia) and disease-associated hypoxia (where cellular oxygen demand outstrips supply) for immune cell functions, discussing the relevance of hypoxia for immune responses in the settings of tissue homeostasis, inflammation, infection, cancer and disease immunotherapy.
Collapse
Affiliation(s)
- Ananda Shanti Mirchandani
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| | | | - Sarah Ruth Walmsley
- Centre for Inflammation Research, Institute for Regeneration and Repair, University of Edinburgh, Edinburgh, UK.
| |
Collapse
|
16
|
Nash J, Pym D, Davies A, Saunders C, George C, Williams JO, Grinberg OY, James PE. Enhanced oxygen availability and preserved aggregative function in platelet concentrates stored at reduced platelet concentration. Transfusion 2025; 65:575-587. [PMID: 39673310 PMCID: PMC11925141 DOI: 10.1111/trf.18101] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2024] [Revised: 11/20/2024] [Accepted: 11/26/2024] [Indexed: 12/16/2024]
Abstract
BACKGROUND Storage of platelets as platelet concentrates for transfusion is limited to 7 days in the United Kingdom due to deleterious effects on platelet quality and function that occur over time. Oxygen (O2) availability and sufficient gaseous exchange are known to be essential in maintaining the viability and function of platelets stored for transfusion. Despite this, there is a paucity of studies undertaking direct measures of O2 and optimization of conditions throughout storage. We address this and modulate the storage conditions to improve platelet quality and function. STUDY DESIGN AND METHODS Electron paramagnetic resonance oximetry was implemented to directly measure the [O2] experienced by stored platelet concentrates and the O2 consumption rate under standard blood banking conditions. From these direct measures the mathematical modeling was then applied to predict the main parameters contributing to effective O2 distribution throughout the unit. RESULTS This study demonstrates reducing the storage [O2] to reflect near physiological levels significantly alters O2 distribution within the unit and negatively impacts platelet functionality and quality, and therefore is not a viable storage option. DISCUSSION We show the reduction of platelet concentration within a unit improves O2 availability and pH, promotes a more uniform distribution of O2 throughout prolonged storage, and maintains platelet agonist-induced aggregation comparable to 100% platelet concentration. This may be a viable option and could potentially lead to reduced donor demand.
Collapse
Affiliation(s)
- Jamie Nash
- Center for Cardiovascular Health and AgeingCardiff Metropolitan UniversityCardiffUK
- Component Development and Research LaboratoryWelsh Blood Service NHSWalesUK
| | - Dean Pym
- Center for Cardiovascular Health and AgeingCardiff Metropolitan UniversityCardiffUK
| | - A. Davies
- Center for Cardiovascular Health and AgeingCardiff Metropolitan UniversityCardiffUK
| | - Christine Saunders
- Component Development and Research LaboratoryWelsh Blood Service NHSWalesUK
| | - Chloe George
- Component Development and Research LaboratoryWelsh Blood Service NHSWalesUK
| | - J. O. Williams
- Center for Cardiovascular Health and AgeingCardiff Metropolitan UniversityCardiffUK
| | | | - Philip E. James
- Center for Cardiovascular Health and AgeingCardiff Metropolitan UniversityCardiffUK
| |
Collapse
|
17
|
Lu Y, Kobayashi Y, Niki Y, Moriyama K. Possible role of superoxide dismutase 3 in hypoxia-induced developmental defects in murine molars. J Oral Biosci 2025; 67:100611. [PMID: 39746510 DOI: 10.1016/j.job.2024.100611] [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: 08/01/2024] [Revised: 12/29/2024] [Accepted: 12/31/2024] [Indexed: 01/04/2025]
Abstract
OBJECTIVES To investigate the effects of hypoxia on tooth germ development in mice and explore the underlying mechanisms. METHODS Tooth germs were extracted from E14.5 mouse embryos and divided into the control and hypoxia groups for organ culture. The hypoxia group was exposed to hypoxia (0% oxygen) for 3 h, followed by normoxia for 21 h. After 2 or 7 days, samples were collected for morphometric analysis, reverse transcription-quantitative polymerase chain reaction, immunohistochemistry (IHC), and immunofluorescent staining (IF). Additionally, superoxide dismutase 3 (SOD3) expression patterns in mandibular molar tooth germs from C57BL/6 mouse embryos were analyzed using IHC. The SOD inhibitor sodium N, N-diethyldithiocarbamate trihydrate (DETC; 400 μM) was applied under normoxia for 3 days, followed by morphometry, IHC, and IF. RESULTS After 7 days, the hypoxia group exhibited significantly smaller tooth size, fewer cusps, reduced cell proliferation, and increased apoptosis in the epithelium compared to the control group. Sod3 mRNA expression was higher than other Sod family member expressions in the control group. In the hypoxia group, Sod3 mRNA and SOD3 protein expression were significantly decreased, whereas hypoxia-inducible factor-1 expression and reactive oxygen species levels were increased. SOD3 was primarily expressed in the dental epithelium from E12.5 to E17.5. DETC impaired tooth germ development in the control group, resulting in a phenotype similar to that of the hypoxia group, and significantly reduced amelogenin and msh homeobox 2 expression in the epithelium. CONCLUSIONS Hypoxia impairs tooth germ development. SOD3 probably plays a protective role during this process.
Collapse
Affiliation(s)
- Yeming Lu
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Yukiho Kobayashi
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan.
| | - Yuki Niki
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| | - Keiji Moriyama
- Department of Maxillofacial Orthognathics, Division of Maxillofacial and Neck Reconstruction, Graduate School of Medical and Dental Sciences, Institute of Science Tokyo, 1-5-45, Bunkyo-ku, Tokyo, 113-8549, Japan
| |
Collapse
|
18
|
Miri SM, Ata BN, Çimen Ş, Barakat S, Ghaffari Zaki A, Armouch J, Vatandaşlar E, Vilain S, Öztürk G, Eroğlu E. Development of an Oxygen-Insensitive Nrf2 Reporter Reveals Redox Regulation under Physiological Normoxia. ACS Sens 2025. [PMID: 40021628 DOI: 10.1021/acssensors.4c03167] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 03/03/2025]
Abstract
Reactive oxygen species, particularly hydrogen peroxide (H2O2), play crucial roles in cellular signaling, with Nrf2 serving as a key transcription factor in maintaining redox homeostasis. However, the precise influence of H2O2 on Nrf2 activity under physiological normoxia remains unclear due to the limitations of oxygen-sensitive imaging methods. To address this, we developed and validated an oxygen-insensitive Nrf2 reporter named pericellular oxygen-insensitive Nrf2 transcriptional performance reporter (POINTER). We employed this reporter in human cerebral microvascular endothelial cells (hCMEC/D3). Using POINTER, we investigated how varying intracellular H2O2 concentrations affect Nrf2 regulation under normoxia (5 kPa O2) compared to hyperoxia (ambient air, 21 kPa O2). We manipulated intracellular H2O2 levels through exogenous application, chemogenetic production using a modified amino acid oxidase, and pharmacological induction with Auranofin. Our findings reveal that Nrf2 transcriptional activity is significantly lower under normoxia than under hyperoxia, supporting previous literature and expectations. Using POINTER, we found that both antioxidant pathway inhibition and sustained H2O2 elevation are essential for modulating Nrf2 activity. These findings provide new insights into the regulation of Nrf2 by H2O2.
Collapse
Affiliation(s)
- Seyed Mohammad Miri
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Büşra N Ata
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| | - Şeyma Çimen
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Department of Nutrition and Dietetics, Institution of Health Sciences, Istanbul Medipol University, Istanbul 34810, Turkey
| | - Sarah Barakat
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Asal Ghaffari Zaki
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Joudi Armouch
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Emre Vatandaşlar
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Sven Vilain
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
| | - Gürkan Öztürk
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Department of Physiology, School of Medicine, Bolu Abant Izzet Baysal University, Bolu 14030, Turkey
| | - Emrah Eroğlu
- Regenerative and Restorative Medicine Research Center (REMER), Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, Istanbul 34810, Turkey
- Molecular Biology, Genetics and Bioengineering Program, Faculty of Engineering and Natural Sciences, Sabanci University, Istanbul 34956, Turkey
| |
Collapse
|
19
|
Gdesz-Birula K, Drobczyński S, Sarat K, Duś-Szachniewicz K. Sonidegib Inhibits the Adhesion of Acute Myeloid Leukemia to the Bone Marrow in Hypoxia: An Optical Tweezer Study. Biomedicines 2025; 13:578. [PMID: 40149555 PMCID: PMC11940413 DOI: 10.3390/biomedicines13030578] [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: 11/15/2024] [Revised: 02/11/2025] [Accepted: 02/13/2025] [Indexed: 03/29/2025] Open
Abstract
Background: Acute myeloid leukemia (AML) is a heterogeneous disease highly resistant to chemotherapeutic agents. Leukemia stem cells (LSCs) can enter a dormant state and avoid apoptosis in the protective niche of the bone marrow (BM) microenvironment. Moreover, bone marrow stromal cells protect leukemia cells by promoting pro-survival signaling pathways and drug resistance. Therefore, attenuating interactions between leukemia cells and BM cells may have a positive therapeutic effect. Objectives: In this work, we hypothesized that sondages may inhibit the adhesion of leukemia cells to the bone marrow by inhibiting the Hedgehog (Hh) signaling pathway. The Hedgehog pathway is a key therapeutic target in AML due to its role in leukemic cell growth and survival. Methods: We investigated the effects of sonidegib on the adhesion of individual OCI-AML3 cells to a bone marrow stromal spheroid derived from the HS-5 cell line. For this purpose, we precisely determined the minimum cell-to-cell adhesion time using optical tweezers under normoxic (21% of O2) and hypoxic (1% of O2) conditions. Results: Our results demonstrated that sonidegib significantly increased the minimum cell-to-cell adhesion time necessary for leukemic cells to establish adhesive bonds with bone marrow stromal cells, thereby indicating a reduction in their adhesive properties. Additionally, we showed that sonidegib is particularly effective at hypoxic oxygen concentrations. Conclusions: The results obtained in this study suggest that sonidegib, through its modulation of the Hedgehog signaling pathway, holds promise as a potential therapeutic approach to target leukemic cell adhesion within the bone marrow microenvironment.
Collapse
Affiliation(s)
- Katarzyna Gdesz-Birula
- Department of Clinical and Experimental Pathology, Institute of General and Experimental Pathology, Wrocław Medical University, 50-368 Wrocław, Poland
| | - Sławomir Drobczyński
- Department of Optics and Photonics, Faculty of Fundamental Problems of Technology, Wrocław University of Science and Technology, 50-370 Wrocław, Poland;
| | - Krystian Sarat
- Laboratory of Genetics and Epigenetics of Human Diseases, Department of Experimental Therapy, Ludwik Hirszfeld Institute of Immunology and Experimental Therapy, Polish Academy of Sciences, 53-114 Wrocław, Poland;
| | - Kamila Duś-Szachniewicz
- Department of Clinical and Experimental Pathology, Institute of General and Experimental Pathology, Wrocław Medical University, 50-368 Wrocław, Poland
| |
Collapse
|
20
|
Li W, McLeod D, Antonevich S, Li Z, Entcheva E. Pericellular oxygen dynamics in human cardiac fibroblasts and iPSC-cardiomyocytes in high-throughput plates: insights from experiments and modeling. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.02.19.639086. [PMID: 40060418 PMCID: PMC11888188 DOI: 10.1101/2025.02.19.639086] [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: 03/17/2025]
Abstract
Adequate oxygen supply is crucial for proper cellular function. The emergence of high-throughput (HT) expansion of human stem-cell-derived cells and HT in vitro cellular assays for drug testing necessitate monitoring and understanding of the oxygenation conditions, yet virtually no data exists for such settings. For metabolically active cells like cardiomyocytes, variations in oxygenation may significantly impact their maturation and function; conversely, electromechanical activity can drive oxygen demands. We used HT label-free optical measurements and computational modeling to gain insights about oxygen availability (peri-cellular oxygen dynamics) in syncytia of human induced pluripotent stem cell derived cardiomyocytes (hiPSC-CM) and human cardiac fibroblasts (cFB) grown in glass-bottom 96-well plates under static conditions. Our experimental results highlight the critical role of cell density and solution height (oxygen delivery path) in peri-cellular oxygen dynamics. The developed 3D reaction-diffusion model with Michaelis-Menten kinetics, trained on the obtained comprehensive data set, revealed that time-variant maximum oxygen consumption rate, Vmax, is needed to faithfully capture the complex peri-cellular oxygen dynamics in the excitable hiPSC-CMs, but not in the cFB. For the latter, accounting for cell proliferation was needed. Interestingly, we found both hypoxic (< 2%) and hyperoxic (> 7%) conditions can easily emerge in these standard HT plates in static culture and that peri-cellular oxygen dynamics evolves with days in culture. Our results and the developed computational model can directly be used to optimize cardiac cell growth in HT plates to achieve desired physiological conditions, important in cellular assays for cardiotoxicity, drug development, personalized medicine and heart regeneration applications.
Collapse
Affiliation(s)
- Weizhen Li
- Department of Biomedical Engineering, The George Washington University, Washington DC 20052, USA
| | - David McLeod
- Department of Biomedical Engineering, The George Washington University, Washington DC 20052, USA
| | - Sarah Antonevich
- Department of Biomedical Engineering, The George Washington University, Washington DC 20052, USA
| | - Zhenyu Li
- Department of Biomedical Engineering, The George Washington University, Washington DC 20052, USA
| | - Emilia Entcheva
- Department of Biomedical Engineering, The George Washington University, Washington DC 20052, USA
| |
Collapse
|
21
|
Valeros J, Jerome M, Tseyang T, Vo P, Do T, Fajardo Palomino D, Grotehans N, Kunala M, Jerrett AE, Hathiramani NR, Mireku M, Magesh RY, Yenilmez B, Rosen PC, Mann JL, Myers JW, Kunchok T, Manning TL, Boercker LN, Carr PE, Munim MB, Lewis CA, Sabatini DM, Kelly M, Xie J, Czech MP, Gao G, Shepherd JN, Walker AK, Kim H, Watson EV, Spinelli JB. Rhodoquinone carries electrons in the mammalian electron transport chain. Cell 2025; 188:1084-1099.e27. [PMID: 39909039 PMCID: PMC11845293 DOI: 10.1016/j.cell.2024.12.007] [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: 05/09/2024] [Revised: 08/19/2024] [Accepted: 12/09/2024] [Indexed: 02/07/2025]
Abstract
Ubiquinone (UQ), the only known electron carrier in the mammalian electron transport chain (ETC), preferentially delivers electrons to the terminal electron acceptor oxygen (O2). In hypoxia, ubiquinol (UQH2) diverts these electrons onto fumarate instead. Here, we identify rhodoquinone (RQ), an electron carrier detected in mitochondria purified from certain mouse and human tissues that preferentially delivers electrons to fumarate through the reversal of succinate dehydrogenase, independent of environmental O2 levels. The RQ/fumarate ETC is strictly present in vivo and is undetectable in cultured mammalian cells. Using genetic and pharmacologic tools that reprogram the ETC from the UQ/O2 to the RQ/fumarate pathway, we establish that these distinct ETCs support unique programs of mitochondrial function and that RQ confers protection upon hypoxia exposure in vitro and in vivo. Thus, in discovering the presence of RQ in mammals, we unveil a tractable therapeutic strategy that exploits flexibility in the ETC to ameliorate hypoxia-related conditions.
Collapse
Affiliation(s)
- Jonathan Valeros
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Madison Jerome
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Tenzin Tseyang
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paula Vo
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Thang Do
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Diana Fajardo Palomino
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nils Grotehans
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Manisha Kunala
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Alexandra E Jerrett
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Nicolai R Hathiramani
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Morningside Graduate School of Biomedical Sciences, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael Mireku
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Rayna Y Magesh
- Department of Systems Biology, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Batuhan Yenilmez
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Paul C Rosen
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jessica L Mann
- Massachusetts Institute of Technology, Cambridge, MA 02142, USA
| | - Jacob W Myers
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | | | - Tanner L Manning
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Lily N Boercker
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Paige E Carr
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | | | - Caroline A Lewis
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - David M Sabatini
- Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences, Flemingovo n. 2, 166 10 Prague, Czech Republic; Institute of Organic Chemistry and Biochemistry of the Czech Academy of Sciences Boston Branch, 840 Memorial Drive, Cambridge, MA 02139, USA
| | - Mark Kelly
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Division of Cardiovascular Medicine, Department of Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jun Xie
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Michael P Czech
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA; Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Chan Medical School, Worcester, MA 01605, USA; Li Weibo Institute for Rare Disease Research, UMass Chan Medical School, Worcester, MA 01655, USA; Department of Microbiology and Physiological Systems, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jennifer N Shepherd
- Department of Chemistry and Biochemistry, Gonzaga University, Spokane, WA 99258, USA
| | - Amy K Walker
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Hahn Kim
- Small Molecule Screening Center, Princeton University, Princeton, NJ 08544, USA; Department of Chemistry, Princeton University, Princeton, NJ 08544, USA
| | - Emma V Watson
- Diabetes Center of Excellence, UMass Chan Medical School, Worcester, MA 01605, USA
| | - Jessica B Spinelli
- Program in Molecular Medicine, UMass Chan Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
22
|
Peter A, Berneman ZN, Cools N. Cellular respiration in dendritic cells: Exploring oxygen-dependent pathways for potential therapeutic interventions. Free Radic Biol Med 2025; 227:536-556. [PMID: 39643130 DOI: 10.1016/j.freeradbiomed.2024.12.014] [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: 08/06/2024] [Revised: 12/01/2024] [Accepted: 12/03/2024] [Indexed: 12/09/2024]
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells crucial for initiating and regulating adaptive immune responses, making them promising candidates for therapeutic interventions in various immune-mediated diseases. Increasing evidence suggests that the microenvironment in which cells are cultured, as well as the milieu in which they perform their functions, significantly impact their immunomodulatory properties. Among these environmental factors, the role of oxygen in DC biology and its significance for both their in vitro generation and in vivo therapeutic application require investigation. Unlike the atmospheric oxygen level of 21 % commonly used in in vitro assays, physiological oxygen levels are much lower (3-9 %), and hypoxia (<1.3 %) is a prevalent condition of both healthy tissues and disease states. This mismatch between laboratory and physiological conditions underscores the critical need to culture and evaluate therapeutic cells under physiologically relevant oxygen levels to improve their translational relevance and clinical outcomes. This review explores the characteristic hallmarks of human DCs that are influenced by oxygen-dependent pathways, including metabolism, phenotype, cytokine secretion, and migration. Furthermore, we discuss the potential of manipulating oxygen levels to refine the generation and functionality of DCs for therapeutic purposes.
Collapse
Affiliation(s)
- Antonia Peter
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium.
| | - Zwi N Berneman
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| | - Nathalie Cools
- Laboratory of Experimental Hematology, Vaccine & Infectious Disease Institute (VAXINFECTIO), Faculty of Medicine and Health Sciences, University of Antwerp, 2610 Antwerp, Belgium; Center for Cell Therapy and Regenerative Medicine, Antwerp University Hospital, 2650 Edegem, Belgium
| |
Collapse
|
23
|
Sergeeva AD, Panova AS, Ivanova AD, Khramova YV, Morozova KI, Kotova DA, Guryleva AV, Khokhlov DD, Kelmanson IV, Vasilev AV, Kostyuk AI, Semyanov AV, Oleinikov VA, Belousov VV, Machikhin AS, Brazhe NA, Bilan DS. Where in the Tissues of Danio rerio Is More H 2O 2 Produced During Acute Hypoxia? Antioxid Redox Signal 2025; 42:292-300. [PMID: 39086238 DOI: 10.1089/ars.2024.0563] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 08/02/2024]
Abstract
The lack of oxygen (O2) causes changes in the cell functioning. Modeling hypoxic conditions in vitro is challenging given that different cell types exhibit different sensitivities to tissue O2 levels. We present an effective in vivo platform for assessing various tissue and organ parameters in Danio rerio larvae under acute hypoxic conditions. Our system allows simultaneous positioning of multiple individuals within a chamber where O2 level in the water can be precisely and promptly regulated, all while conducting microscopy. We applied this approach in combination with a genetically encoded pH-biosensor SypHer3s and a highly H2O2-sensitive HyPer7 biosensor. Hypoxia causes H2O2 production in areas of brain, heart, and skeletal muscles, exclusively in the mitochondrial matrix; it is noteworthy that H2O2 does not penetrate into the cytosol and is neutralized in the matrix upon reoxygenation. Hypoxia causes pronounced tissue acidosis, expressed by a decrease in pH by 0.4-0.6 units everywhere. Using imaging photoplethysmography, we measured in D. rerio larvae real-time heart rate decrease under conditions of hypoxia and subsequent reoxygenation. Our observations in this experimental system lead to the hypothesis that mitochondria are the only source of H2O2 in cells of D. rerio under hypoxia. Antioxid. Redox Signal. 42, 292-300.
Collapse
Affiliation(s)
- Anastasia D Sergeeva
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
| | - Anastasiya S Panova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexandra D Ivanova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Yulia V Khramova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Ksenia I Morozova
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Daria A Kotova
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Anastasia V Guryleva
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Demid D Khokhlov
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Ilya V Kelmanson
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Aleksandr V Vasilev
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Alexander I Kostyuk
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexey V Semyanov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
- Sechenov First Moscow State Medical University, Moscow, Russia
- College of Medicine, Jiaxing University, Jiaxing, China
| | - Vladimir A Oleinikov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
| | - Vsevolod V Belousov
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| | - Alexander S Machikhin
- Scientific and Technological Centre of Unique Instrumentation, Russian Academy of Sciences, Moscow, Russia
| | - Nadezda A Brazhe
- Faculty of Biology, M.V. Lomonosov Moscow State University, Moscow, Russia
| | - Dmitry S Bilan
- M.M. Shemyakin and Yu.A. Ovchinnikov Institute of Bioorganic Chemistry, Russian Academy of Sciences, Moscow, Russia
- Federal Center of Brain Research and Neurotechnologies, Federal Medical Biological Agency, Moscow, Russia
- Pirogov Russian National Research Medical University, Moscow, Russia
| |
Collapse
|
24
|
Sridhar A, Bakke I, Gopalakrishnan S, Osoble NMM, Hammarqvist EP, Pettersen HPS, Sandvik AK, Østvik AE, Hansen MD, Bruland T. Tofacitinib and budesonide treatment affect stemness and chemokine release in IBD patient-derived colonoids. Sci Rep 2025; 15:3753. [PMID: 39885201 PMCID: PMC11782514 DOI: 10.1038/s41598-025-86314-2] [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: 10/22/2024] [Accepted: 01/09/2025] [Indexed: 02/01/2025] Open
Abstract
Restoration of the intestinal epithelial barrier is crucial for achieving mucosal healing, the therapeutic goal for inflammatory bowel disease (IBD). During homeostasis, epithelial renewal is maintained by crypt stem cells and progenitors that cease to divide as they differentiate into mature colonocytes. Inflammation is a major effector of mucosal damage in IBD and has been found to affect epithelial stemness, regeneration and cellular functions. However, the impact of immune cell-modulating IBD drugs on epithelial homeostasis and repair is poorly understood. It is likely that these drugs will have distinct mechanisms of action (MOA) in intestinal epithelium relevant for homeostasis that will vary among patients. We investigated cellular effects of pan-Janus Kinase (JAK) inhibitor tofacitinib and the corticosteroid budesonide on uninflamed and TNF + Poly(I:C) stimulated human colon organoids (colonoids) from healthy donors and IBD-patients. Our findings reveal that although both tofacitinib and budesonide exhibit anti-inflammatory effects, tofacitinib increased colonoid size and proliferation during differentiation, and promoted epithelial stemness. In contrast, budesonide decreased colonoid size and showed no consistent effect on proliferation or stemness. Our study demonstrates the value of employing human colonoids to investigate how IBD drugs affect intestinal epithelial cells and inter-individual variations relevant to mucosal healing and personalized IBD treatment.
Collapse
Affiliation(s)
- Arun Sridhar
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Ingunn Bakke
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway.
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway.
| | - Shreya Gopalakrishnan
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
| | - Nimo Mukhtar Mohamud Osoble
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
| | - Emilie Prytz Hammarqvist
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
| | - Henrik P Sahlin Pettersen
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
- Department of Pathology, St. Olav's University Hospital, Trondheim, Norway
| | - Arne Kristian Sandvik
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Ann Elisabet Østvik
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Marianne Doré Hansen
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine (IKOM), Faculty of Medicine and Health Sciences, NTNU - Norwegian University of Science and Technology, Prinsesse Kristinas gt. 1, Trondheim, 7030, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| |
Collapse
|
25
|
Taba R, Põlluaed M, Tein K, Puurand M, Käämbre T, Terasmaa A. Effect of Media Composition and Oxygen Tension on Cellular Stress Response and Nrf2 Activation in HepG2ARE Cells. Antioxidants (Basel) 2025; 14:137. [PMID: 40002325 PMCID: PMC11851573 DOI: 10.3390/antiox14020137] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2024] [Revised: 01/20/2025] [Accepted: 01/20/2025] [Indexed: 02/27/2025] Open
Abstract
Cell models play a central role in preclinical research aimed at the mechanism of disease and drug discovery. The outside environment of the cells, including levels of nutrients and oxygen tension, regulates cellular stress response pathways. Routinely used in vitro disease models often overlook cell growth conditions. This study aimed to evaluate the effect of substituting classic cell media (DMEM) with media matching the nutrient composition of human plasma (Plasmax) on cell viability, the activation of nuclear factor erythroid 2-related factor 2 (Nrf2), glutathione (GSH), and malondialdehyde (MDA) levels by different pharmacological inducers of cell stress. The cells were grown at ambient (~19%) and reduced (5%) oxygen levels. The activation of Nrf2 by ferroptosis activators (erastin and RSL3) was dependent on cell media and oxygen tension. The induction of Nrf2 by an inducer of endoplasmic reticulum stress, thapsigargin, was observable only in cells grown in DMEM and at low oxygen tension. GSH and MDA levels were elevated in Plasmax media. Results indicate that stress tolerance and the activation of Nrf2 in the HepG2ARE cell line depend on the growth conditions, including cell media and oxygen. Cell culture conditions should be critically considered when designing in vitro models of diseases involving oxidative stress.
Collapse
Affiliation(s)
- Rutt Taba
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; (M.P.); (K.T.); (M.P.); (T.K.)
| | | | | | | | | | - Anton Terasmaa
- Laboratory of Chemical Biology, National Institute of Chemical Physics and Biophysics, Akadeemia tee 23, 12618 Tallinn, Estonia; (M.P.); (K.T.); (M.P.); (T.K.)
| |
Collapse
|
26
|
Rogers RS, Mootha VK. Hypoxia as a medicine. Sci Transl Med 2025; 17:eadr4049. [PMID: 39841808 DOI: 10.1126/scitranslmed.adr4049] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2024] [Accepted: 12/31/2024] [Indexed: 01/24/2025]
Abstract
Oxygen is essential for human life, yet a growing body of preclinical research is demonstrating that chronic continuous hypoxia can be beneficial in models of mitochondrial disease, autoimmunity, ischemia, and aging. This research is revealing exciting new and unexpected facets of oxygen biology, but translating these findings to patients poses major challenges, because hypoxia can be dangerous. Overcoming these barriers will require integrating insights from basic science, high-altitude physiology, clinical medicine, and sports technology. Here, we explore the foundations of this nascent field and outline a path to determine how chronic continuous hypoxia can be safely, effectively, and practically delivered to patients.
Collapse
Affiliation(s)
- Robert S Rogers
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute, Cambridge, MA 02142, USA
| | - Vamsi K Mootha
- Department of Molecular Biology, Massachusetts General Hospital, Boston, MA 02114, USA
- Broad Institute, Cambridge, MA 02142, USA
- Department of Systems Biology, Harvard Medical School, Boston, MA 02115, USA
- Howard Hughes Medical Institute, Boston, MA 02114, USA
| |
Collapse
|
27
|
Pinho SA, Gardner GL, Alva R, Stuart JA, Cunha-Oliveira T. Creating Physiological Cell Environments In Vitro: Adjusting Cell Culture Media Composition and Oxygen Levels to Investigate Mitochondrial Function and Cancer Metabolism. Methods Mol Biol 2025; 2878:163-199. [PMID: 39546262 DOI: 10.1007/978-1-0716-4264-1_9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2024]
Abstract
In vitro and ex vivo studies are crucial for mitochondrial research, offering valuable insights into cellular mechanisms and aiding in diagnostic and therapeutic strategies. Accurate in vitro models rely on adequate cell culture conditions, such as the composition of culture media and oxygenation levels. These conditions can influence energy metabolism and mitochondrial activities, thus impacting studies involving mitochondrial components, such as the effectiveness of anticancer drugs. This chapter focuses on practical guidance for creating setups that replicate in vivo microenvironments, capturing the original metabolic context of cells. We explore protocols to better mimic the physiological cell environment, promote cellular reconfiguration, and prime cells according to the modeled context. The first part is dedicated to the use of human dermal fibroblasts, which are a promising model for pre-clinical mitochondrial research due to their adaptability and relevance to human mitochondrial physiology. We present an optimized protocol for gradually adjusting extracellular glucose levels, which demonstrated significant mitochondrial, metabolic, and redox remodeling in normal adult dermal fibroblasts. The second part is dedicated to replication of tumor microenvironments, which are relevant for studies targeting cellular energy metabolism to inhibit tumor growth. Currently available physiological media can mimic blood plasma metabolome but not the specific tumor microenvironment. To address this, we describe optimized media formulation and oxygenation protocols, which can simulate the tumor microenvironment in cell culture experiments. Replicating in vivo microenvironments in in vitro and ex vivo studies can enhance our understanding of cellular processes, facilitate drug development, and advance personalized therapeutics in mitochondrial medicine.
Collapse
Affiliation(s)
- Sónia A Pinho
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal
- PDBEB - Doctoral Programme in Experimental Biology and Biomedicine, Institute of Interdisciplinary Research, University of Coimbra, Coimbra, Portugal
| | - Georgina L Gardner
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Ricardo Alva
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Jeff A Stuart
- Department of Biological Sciences, Brock University, St. Catharines, ON, Canada
| | - Teresa Cunha-Oliveira
- CNC-UC, Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- CIBB, Center for Innovative Biomedicine and Biotechnology, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
28
|
Rogers ZJ, Flood D, Bencherif SA, Taylor CT. Oxygen control in cell culture - Your cells may not be experiencing what you think! Free Radic Biol Med 2025; 226:279-287. [PMID: 39577817 DOI: 10.1016/j.freeradbiomed.2024.11.036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/23/2024] [Revised: 10/30/2024] [Accepted: 11/19/2024] [Indexed: 11/24/2024]
Abstract
Oxygen (O2)-controlled cell culture has been pivotal in studying mammalian mechanisms of O2 sensing, regulation, and utilization. We posit, however, that O2-controlled cell culture is paradoxically not controlling O2. There is overwhelming evidence that the pericellular O2 is lower than the surrounding gas phase due to cellular O2 consumption. Standard hypoxic cell culture is at high risk of inducing pericellular anoxia. We discuss the implications of poor O2 control for cellular O2 regulation mechanisms, bioenergetics, and redox signaling. We also highlight the evidence of frequent under-oxygenation in standard (i.e., normoxic) cell culture. This issue has been largely overlooked because strategies to control pericellular O2 have been lacking. Here, we propose a framework to control pericellular O2 based on our recent investigation into the nature of the gas/pericellular O2 gradient. Implementing this framework into standard practice will unlock quantitative O2 control in vitro, improving our ability to understand the role of O2 in biology.
Collapse
Affiliation(s)
- Zachary J Rogers
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Darragh Flood
- Conway Institute of Biomolecular and Biomedical Research and School of Medicine, University College Dublin, Belfield, Dublin, Ireland
| | - Sidi A Bencherif
- Department of Chemical Engineering, Northeastern University, Boston, MA, 02115, USA
| | - Cormac T Taylor
- Conway Institute of Biomolecular and Biomedical Research and School of Medicine, University College Dublin, Belfield, Dublin, Ireland.
| |
Collapse
|
29
|
Fan X, Li B, Chai S, Zhang R, Cai C, Ge R. Hypoxia Promotes Osteoclast Differentiation by Weakening USP18-Mediated Suppression on the NF-κB Signaling Pathway. Int J Mol Sci 2024; 26:10. [PMID: 39795869 PMCID: PMC11719700 DOI: 10.3390/ijms26010010] [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: 12/11/2024] [Revised: 12/17/2024] [Accepted: 12/19/2024] [Indexed: 01/13/2025] Open
Abstract
Osteoporosis, a prevalent metabolic bone disorder, is characterized by reduced bone density and increased fracture risk. The pathogenesis of osteoporosis is closely associated with an imbalance in bone remodeling, in which the resorption function of osteoclasts exceeds the formation function of osteoblasts. Hypoxia has been implicated in the promotion of osteoclast differentiation and the subsequent development of osteoporosis. The ubiquitin-proteasome system (UPS) and its regulatory enzymes, deubiquitinating enzymes (DUBs), play a significant role in bone homeostasis. In this study, we investigated the contribution and mechanism of Ubiquitin-specific protease 18 (USP18), a DUB, in osteoclast differentiation under hypoxic conditions. BMDMs and RAW264.7 cells were treated with RANKL to induce osteoclastogenesis and were subjected to overexpression or knockdown of USP18 under normoxic or hypoxia conditions. Osteoclast formation was assessed using TRAP staining, and the expression of osteoclast marker genes was determined using qRT-PCR. The activation of the NF-κB signaling pathway was evaluated using immunoblotting. We found that hypoxia significantly enhanced the differentiation of BMDMs and RAW264.7 cells into osteoclasts, accompanied by a notable downregulation of USP18 expression. The overexpression of USP18 inhibited RANKL-induced osteoclast differentiation, while the knockdown of USP18 promoted that process, unveiling the inhibitory effect of USP18 in osteoclastogenesis. Furthermore, the overexpression of USP18 rescued the hypoxia-induced increase in osteoclast differentiation. Mechanistic insights revealed that USP18 inhibits osteoclastogenesis by suppressing the NF-κB signaling pathway, with a potential target on TAK1 or its upstream molecules. This study indicates that hypoxia promotes osteoclast differentiation through the downregulation of USP18, which, in turn, relieves the suppression of the activation of the NF-κB signaling pathway. The USP18 emerges as a potential therapeutic target for osteoporosis treatment, highlighting the importance of the hypoxia-DUB axis in the pathogenesis of the disease.
Collapse
Affiliation(s)
- Xiaoxia Fan
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Botong Li
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Shengjun Chai
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Rong Zhang
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Chunmei Cai
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| | - Rili Ge
- Research Center for High Altitude Medicine, Qinghai University, Xining 810001, China; (X.F.); (B.L.); (S.C.); (R.Z.)
- Key Laboratory of the Ministry of High Altitude Medicine, Qinghai University, Xining 810001, China
- Key Laboratory of Applied Fundamentals of High Altitude Medicine, Qinghai-Utah Joint Key Laboratory of Plateau Medicine, Qinghai University, Xining 810001, China
- Laboratory for High Altitude Medicine of Qinghai Province, Qinghai University, Xining 810001, China
| |
Collapse
|
30
|
Nowicki C, Ganse B. Near-Infrared Spectroscopy Allows for Monitoring of Bone Fracture Healing via Changes in Oxygenation. J Funct Biomater 2024; 15:384. [PMID: 39728184 DOI: 10.3390/jfb15120384] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2024] [Revised: 12/06/2024] [Accepted: 12/17/2024] [Indexed: 12/28/2024] Open
Abstract
Bone fractures are associated with hypoxia, but no longitudinal studies of perfusion measurements in human patients have been reported despite the clinical and research potential. In this longitudinal observational cohort study, the near-infrared spectroscopy (NIRS) device PortaMon was used to assess oxy-(O2Hb), deoxy-(HHb) and total (tHb) haemoglobin, as well as the differences between O2Hb and HHb (HbDiff) and the tissue saturation index (TSI) at three different depths in the fracture gap. Linear mixed effect models were fitted to analyse time effects. One-way ANOVAs were conducted to compare groups. The time points corresponding to minima were calculated via linear regression. In this study, 11 patients with tibial shaft fractures underwent longitudinal measurements. Additionally, 9 patients with diagnosed tibial shaft nonunion and 23 age-matched controls were measured once. In the longitudinal group, all fractures healed, and decreases in O2Hb and HbDiff (all p < 0.05) were observed, with minima occurring 19-21 days after fracture. O2Hb values in nonunion patients did not differ from the minima in longitudinally measured union patients, whereas differences in HHb and tHb were significant (all p < 0.05). Previously, the onset of hypoxia has been assumed to be much faster. The characteristic trajectories of the NIRS parameters O2Hb and HbDiff can be used to fulfil the need for a non-invasive method to monitor fracture healing. These results suggest that NIRS could supplement radiographs and clinical impressions in daily clinical practice and may enable earlier diagnosis of nonunion.
Collapse
Affiliation(s)
- Cedric Nowicki
- Innovative Implant Development (Fracture Healing), Departments and Institutes of Surgery, Saarland University, 66421 Homburg, Germany
| | - Bergita Ganse
- Innovative Implant Development (Fracture Healing), Departments and Institutes of Surgery, Saarland University, 66421 Homburg, Germany
- Department of Trauma, Hand and Reconstructive Surgery, Departments and Institutes of Surgery, Saarland University, 66421 Homburg, Germany
| |
Collapse
|
31
|
Hossain T, Secor JT, Eckmann DM. Hyperbaric oxygen rapidly produces intracellular bioenergetics dysfunction in human pulmonary cells. Chem Biol Interact 2024; 404:111266. [PMID: 39426659 DOI: 10.1016/j.cbi.2024.111266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2024] [Revised: 08/30/2024] [Accepted: 10/11/2024] [Indexed: 10/21/2024]
Abstract
Hyperoxic exposure lasting days alters mitochondrial bioenergetic and dynamic functions in pulmonary cells as indices of oxygen toxicity. The aim of this study was to examine effects of short duration hyperbaric and hyperoxic exposures to induce oxygen toxicity similarly. Cultured human lung microvascular endothelial cells, human pulmonary artery endothelial cells and A549 cells were exposed to hyperoxia (∼5 % CO2 equivalent, balance O2) under hyperbaric conditions (4.8 ATA) for 1 and 4 h. Measures of mitochondrial dynamics, inner membrane potential, mitochondrial respiration, the intracellular distribution of bioenergetic capacity and respiration complex protein levels were then quantified. Exposures resulted in altered mitochondrial motility, presence of inhomogeneities in respiration parameters, loss of inner membrane potential, and changes in intracellular partitioning of ATP-linked respiration. Changes in the levels of respiration complex protein levels were also found. The combination of hyperoxic exposure with hyperbaric conditions rapidly produced changes in mitochondrial dynamics and bioenergetics in pulmonary cells. These changes are consistent with the onset of pulmonary oxygen toxicity previously known to result from long duration exposure to hyperoxia alone. These findings suggest health caution is warranted in environmental settings in which both hyperoxic and hyperbaric conditions are present. The synergism of hyperoxia and hyperbaria for rapid induction of oxygen toxicity in cellular models has utility for the study of mechanistic determinants of oxygen toxicity, testing of putative therapeutics, and associated investigations of mitochondrial dysfunction.
Collapse
Affiliation(s)
- Tanvir Hossain
- Department of Anesthesiology, The Ohio State University, Columbus, OH, 43210, USA
| | - Jackson T Secor
- Department of Anesthesiology, The Ohio State University, Columbus, OH, 43210, USA
| | - David M Eckmann
- Department of Anesthesiology, The Ohio State University, Columbus, OH, 43210, USA; Center for Medical and Engineering Innovation, The Ohio State University, Columbus, OH, 43210, USA.
| |
Collapse
|
32
|
Ghaffari-Bohlouli P, Jafari H, Nie L, Kakkar A, Shavandi A. Enzymes in Addressing Hypoxia for Biomaterials Engineering. Adv Healthc Mater 2024; 13:e2401713. [PMID: 39183514 DOI: 10.1002/adhm.202401713] [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: 05/09/2024] [Revised: 08/05/2024] [Indexed: 08/27/2024]
Abstract
Oxygen is essential for normal cellular functions. Hypoxia impacts various cellular processes, such as metabolism, growth, proliferation, angiogenesis, metastasis, tumorigenesis, microbial infection, and immune response, mediated by hypoxia-inducible factors (HIFs). Hypoxia contributes to the progression and development of cancer, cardiovascular diseases, metabolic disorders, kidney diseases, and infections. The potential alleviation of hypoxia has been explored through the enzymatic in situ decomposition of hydrogen peroxide, leading to the generation of oxygen. However, challenges such as limited stability restrict the effectiveness of enzymes such as catalase in biomedical and in vivo applications. To overcome these limitations, targeted delivery of the enzymes has been proposed. This review offers a critical comparison of i) current approaches to enhance the in vivo stability of catalase; and ii) the structure, mechanism of action, and kinetics of catalase and catalase-like nanozymes.
Collapse
Affiliation(s)
- Pejman Ghaffari-Bohlouli
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Hafez Jafari
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| | - Lei Nie
- College of Life Sciences, Xinyang Normal University, Xinyang, 464000, China
| | - Ashok Kakkar
- Department of Chemistry, McGill University, 801 Sherbrooke Street West, Montréal, Québec, H3A 0B8, Canada
| | - Amin Shavandi
- 3BIO-BioMatter, École Polytechnique de Bruxelles, Université Libre de Bruxelles, Avenue F.D. Roosevelt, 50-CP 165/61, Brussels, 1050, Belgium
| |
Collapse
|
33
|
Li M, Zhou S, Dong G, Wang C. Emergence of FLASH‑radiotherapy across the last 50 years (Review). Oncol Lett 2024; 28:602. [PMID: 39493433 PMCID: PMC11529378 DOI: 10.3892/ol.2024.14735] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2024] [Accepted: 09/02/2024] [Indexed: 11/05/2024] Open
Abstract
A novel radiotherapy (RT) approach termed FLASH-RT, which irradiates areas at ultra-high dose rates, is of current interest to medical researchers. FLASH-RT can maintain equivalent antitumor effects while sparing healthy tissue compared with conventional RT (CONV-RT), which uses low dose rates. The sparing effect on healthy tissue after FLASH-RT is known as the FLASH effect. Owing to the FLASH effect, FLASH-RT can raise the maximum tolerable dose to control tumor growth or eradicate the tumor and provide a new strategy for clinical RT. However, definitive irradiation conditions for reproducing the FLASH effect and the biological mechanism of the FLASH effect have not yet been fully elucidated. The efficacy of FLASH-RT is controversial despite its successful application in clinical RT. The present review recapitulates the progression of FLASH-RT and critically comments on the hypothesis of the FLASH effect. In addition, the review expounds on the current issues with regard to the differential phenomena between in vitro and in vivo studies, and elaborates on the challenges for the application of FLASH-RT that need to be addressed in the future.
Collapse
Affiliation(s)
- Menghua Li
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Sen Zhou
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
| | - Guofu Dong
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Changzhen Wang
- Beijing Key Laboratory for Radiobiology, Beijing Institute of Radiation Medicine, Beijing 100850, P.R. China
- Department of Biochemistry, Basic Medical College of Anhui Medical University, Hefei, Anhui 230032, P.R. China
| |
Collapse
|
34
|
Crugeiras J, Calls A, Contreras E, Alemany M, Navarro X, Yuste VJ, Casanovas O, Udina E, Bruna J. Oxygen matters: Unraveling the role of oxygen in the neuronal response to cisplatin. J Peripher Nerv Syst 2024; 29:528-536. [PMID: 39329299 PMCID: PMC11625991 DOI: 10.1111/jns.12659] [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: 07/19/2024] [Revised: 08/31/2024] [Accepted: 09/11/2024] [Indexed: 09/28/2024]
Abstract
BACKGROUND AND AIMS Cell culture is a fundamental experimental tool for understanding cell physiology. However, translating these findings to in vivo settings has proven challenging. Replicating donor tissue conditions, including oxygen levels, is crucial for achieving meaningful results. Nevertheless, oxygen culture conditions are often overlooked, particularly in the context of chemotherapy-induced neurotoxicity. METHODS In this study, we investigated the role of oxygen levels in primary neuronal cultures by comparing neuronal performance under cisplatin exposure (1 μg/mL) in supraphysiological normoxia (representing atmospheric conditions in a standard incubator; 18.5% O2) and physioxia (representing physiologic oxygen conditions in nervous tissue; 5% O2). Experiments were also conducted to assess survival, neurite development, senescence marker expression, and proinflammatory cytokine secretion. RESULTS Under control conditions, both oxygen concentration conditions exhibited similar behaviors. However, after cisplatin administration, sensory neurons cultured under supraphysiological normoxic conditions show higher mortality, exhibit an evolutionarily proinflammatory cytokine profile over time, and activate apoptotic-regulated neuron death markers. In contrast, under physiological conditions, neurons treated with cisplatin exhibited senescence marker expression and an attenuated inflammatory secretome. INTERPRETATION These results underscore the critical role of oxygen in neuronal culture, particularly in studying compounds where neuronal damage is mechanistically linked to oxidative stress. Even at identical doses of evaluated neurotoxic drugs, distinct cellular phenotypic fates can emerge, impacting translatability to the in vivo setting.
Collapse
Affiliation(s)
- Jose Crugeiras
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Aina Calls
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Estefanía Contreras
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Montse Alemany
- Unit of Neuro‐Oncology, Hospital Universitari de BellvitgeBellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
| | - Xavier Navarro
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Victor J. Yuste
- Department of BiochemistryInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
| | - Oriol Casanovas
- Tumor Angiogenesis Group, ProCURE ProgramCatalan Institute of Oncology, OncoBell Program, IDIBELLBarcelonaSpain
| | - Esther Udina
- Department of Cell Biology, Physiology, and ImmunologyInstitute of Neuroscience, Autonomous University of BarcelonaBellaterraSpain
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
| | - Jordi Bruna
- Biomedical Research Center Network on Neurodegenerative Diseases (CIBERNED)BellaterraSpain
- Unit of Neuro‐Oncology, Hospital Universitari de BellvitgeBellvitge Institute for Biomedical Research (IDIBELL)BarcelonaSpain
| |
Collapse
|
35
|
Fernandez E, Warde M, Manjarres-Raza I, Bobo-Jimenez V, Martinez-Luna M, Vicente-Gutierrez C, Garcia-Rodriguez D, Jimenez-Blasco D, Almeida A, Bolaños JP. Transcriptomic and metabolic signatures of neural cells cultured under a physiologic-like environment. J Biol Chem 2024; 300:107937. [PMID: 39476959 PMCID: PMC11742299 DOI: 10.1016/j.jbc.2024.107937] [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: 09/22/2024] [Revised: 10/08/2024] [Accepted: 10/22/2024] [Indexed: 11/21/2024] Open
Abstract
Cultured brain cells are used conventionally to investigate fundamental neurobiology and identify therapeutic targets against neural diseases. However, standard culture conditions do not simulate the natural cell microenvironment, thus hampering in vivo translational insight. Major weaknesses include atmospheric (21%) O2 tension and lack of intercellular communication, the two factors likely impacting metabolism and signaling. Here, we addressed this issue in mouse neurons and astrocytes in primary culture. We found that the signs of cellular and mitochondrial integrity were optimal when these cells were acclimated to grow in coculture, to emulate intercellular coupling, under physiologic (5%) O2 tension. Transcriptomic scrutiny, performed to elucidate the adaptive mechanism involved, revealed that the vast majority of differentially expressed transcripts were downregulated in both astrocytes and neurons. Gene ontology evaluation unveiled that the largest group of altered transcripts was glycolysis, which was experimentally validated by metabolic flux analyses. This protocol and database resource for neural cells grown under in vivo-like microenvironment may move forward the translation of basic into applied neurobiological research.
Collapse
Affiliation(s)
- Emilio Fernandez
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Moussa Warde
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Israel Manjarres-Raza
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Veronica Bobo-Jimenez
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Maria Martinez-Luna
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain
| | | | - Dario Garcia-Rodriguez
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Daniel Jimenez-Blasco
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain
| | - Angeles Almeida
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics, University of Salamanca, CSIC, Salamanca, Spain; Institute of Biomedical Research of Salamanca, University Hospital of Salamanca, University of Salamanca, CSIC, Salamanca, Spain; Centro de Investigación Biomédica en Red sobre Fragilidad y Envejecimiento Saludable (CIBERFES), Instituto de Salud Carlos III, Madrid, Spain.
| |
Collapse
|
36
|
Boix J, Knuever J, Niehoff N, Sen A, Pla-Martin D, Baris OR, Etich J, Brachvogel B, Kaul H, Isbrandt D, Soroka E, Bazzi H, Wenger RH, Giavalisco P, Wiesner RJ. Constitutive HIF-1α Expression in the Epidermis Fuels Proliferation and Is Essential for Effective Barrier Formation. J Invest Dermatol 2024:S0022-202X(24)02951-8. [PMID: 39580109 DOI: 10.1016/j.jid.2024.09.022] [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: 06/29/2023] [Revised: 09/15/2024] [Accepted: 09/30/2024] [Indexed: 11/25/2024]
Abstract
Epidermis is one of the most rapidly proliferating tissues in the body with high demands for adenosine triphosphate and cellular building blocks. In this study, we show that to meet these requirements, keratinocytes constitutively express HIF-1α, even in the presence of oxygen levels sufficient for HIF-1α hydroxylation. We previously reported that mice with severe epidermal mitochondrial dysfunction actually showed a hyperproliferative epidermis but rapidly died of systemic lactic acidosis and hypoglycemia, indicating excessive glycolysis. In this work, we interrogated HIF-1α function in glycolysis by its epidermal ablation combined with mitochondrial dysfunction, which resulted in decreased proliferation but even earlier lethality due to a severe barrier defect. Our data demonstrate that HIF-1α is indispensable for maintaining a high aerobic glycolytic flux necessary for supplying energy but also for synthetizing cellular building blocks such as lipids, which are both essential for proliferation as well as barrier formation. HIF-1α is stabilized in keratinocytes in the presence of oxygen by high levels of HIF-1α transcripts, low levels of prolyl-4-hydroxylases (PHD2 and PHD3), and a low cellular a-ketoglutarate/lactate ratio, likely inhibiting prolyl-4-hydroxylase activity. Our data suggest a key role for constitutive HIF-1α expression allowing a Warburg-like metabolism in healthy, highly proliferative keratinocytes, similar to that in tumor cells.
Collapse
Affiliation(s)
- Julia Boix
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany; Center for Molecular Medicine Cologne, University of Köln, Köln, Germany
| | - Jana Knuever
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany; Department of Dermatology and Venereology, University Hospital of Köln, Köln, Germany.
| | - Nadine Niehoff
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany
| | - Ayesha Sen
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany
| | - David Pla-Martin
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany; Center for Molecular Medicine Cologne, University of Köln, Köln, Germany; Institute of Biochemistry and Molecular Biology I, University Hospital Düssledorf, Heinrich Heine University Düsseldorf, Düsseldorf, Germany
| | - Olivier R Baris
- University of Angers, MitoLab, Unité MitoVasc, UMR CNRS 6015, INSERM U1083, SFR ICAT, Angers, France
| | - Julia Etich
- Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany
| | - Bent Brachvogel
- Center for Molecular Medicine Cologne, University of Köln, Köln, Germany; Experimental Neonatology, Department of Pediatrics and Adolescent Medicine, Faculty of Medicine and University Hospital Cologne, University of Cologne, Cologne, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Köln, Köln, Germany
| | - Harshita Kaul
- Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Köln, Köln, Germany
| | - Dirk Isbrandt
- Center for Molecular Medicine Cologne, University of Köln, Köln, Germany; Institute for Molecular and Behavioral Neuroscience, Faculty of Medicine, University of Köln, Köln, Germany; German Center for Neurodegenerative Diseases (DZNE), Bonn, Germany
| | - Ekaterina Soroka
- Department of Dermatology and Venereology, University Hospital of Köln, Köln, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Köln, Köln, Germany
| | - Hisham Bazzi
- Center for Molecular Medicine Cologne, University of Köln, Köln, Germany; Department of Dermatology and Venereology, University Hospital of Köln, Köln, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Köln, Köln, Germany
| | - Roland H Wenger
- Institute of Physiology, University of Zurich, Zurich, Switzerland
| | | | - Rudolf J Wiesner
- Center for Physiology and Pathophysiology, Institute of Systems Physiology, University of Köln, Köln, Germany; Center for Molecular Medicine Cologne, University of Köln, Köln, Germany; Cologne Excellence Cluster on Cellular Stress Responses in Aging-associated Diseases (CECAD), University of Köln, Köln, Germany
| |
Collapse
|
37
|
Margaritelis NV, Cobley JN, Nastos GG, Papanikolaou K, Bailey SJ, Kritsiligkou P, Nikolaidis MG. Evidence-based sports supplements: A redox analysis. Free Radic Biol Med 2024; 224:62-77. [PMID: 39147071 DOI: 10.1016/j.freeradbiomed.2024.08.012] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/31/2024] [Revised: 07/30/2024] [Accepted: 08/09/2024] [Indexed: 08/17/2024]
Abstract
Despite the overwhelming number of sports supplements on the market, only seven are currently recognized as effective. Biological functions are largely regulated through redox reactions, yet no comprehensive analysis of the redox properties of these supplements has been compiled. Here, we analyze the redox characteristics of these seven supplements: bicarbonates, beta-alanine, caffeine, creatine, nitrates, carbohydrates, and proteins. Our findings suggest that all sports supplements exhibit some degree of redox activity. However, the precise physiological implications of these redox properties remain unclear. Future research, employing unconventional perspectives and methodologies, will reveal new redox pixels of the exercise physiology and sports nutrition picture.
Collapse
Affiliation(s)
- Nikos V Margaritelis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece.
| | - James N Cobley
- School of Life Sciences, The University of Dundee, Dundee, Scotland, UK
| | - George G Nastos
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece
| | | | - Stephen J Bailey
- School of Sport, Exercise and Health Sciences, Loughborough University, Loughborough, UK
| | - Paraskevi Kritsiligkou
- Department of Biochemistry, Cell and Systems Biology, Institute of Systems, Molecular and Integrative Biology, University of Liverpool, Liverpool, UK
| | - Michalis G Nikolaidis
- Department of Physical Education and Sports Science at Serres, Aristotle University of Thessaloniki, Serres, Greece
| |
Collapse
|
38
|
Rey-Keim S, Schito L. Origins and molecular effects of hypoxia in cancer. Semin Cancer Biol 2024; 106-107:166-178. [PMID: 39427969 DOI: 10.1016/j.semcancer.2024.10.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Revised: 10/02/2024] [Accepted: 10/09/2024] [Indexed: 10/22/2024]
Abstract
Hypoxia (insufficient O2) is a pivotal factor in cancer progression, triggering genetic, transcriptional, translational and epigenetic adaptations associated to therapy resistance, metastasis and patient mortality. In this review, we outline the microenvironmental origins and molecular mechanisms responsible for hypoxic cancer cell adaptations in situ and in vitro, whilst outlining current approaches to stratify, quantify and therapeutically target hypoxia in the context of precision oncology.
Collapse
Affiliation(s)
- Sergio Rey-Keim
- UCD School of Medicine, University College Dublin, Belfield, Dublin D04 C7X2, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D04 C7X2, Ireland.
| | - Luana Schito
- UCD School of Medicine, University College Dublin, Belfield, Dublin D04 C7X2, Ireland; UCD Conway Institute of Biomolecular and Biomedical Research, University College Dublin, Belfield, Dublin D04 C7X2, Ireland.
| |
Collapse
|
39
|
Pridgeon CS, Airavaara K, Monola J, Jokela A, Palmer D, Yliperttula M, Harjumäki R. Chronic hypoxia for the adaptation of extracellular vesicle phenotype. Sci Rep 2024; 14:25189. [PMID: 39448620 PMCID: PMC11502752 DOI: 10.1038/s41598-024-73453-1] [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: 06/25/2024] [Accepted: 09/17/2024] [Indexed: 10/26/2024] Open
Abstract
Variations in oxygen level affect the phenotype of cells and extracellular vesicles (EVs). Depending on the metabolic oxygen demand of cells, hypoxic cell culture can produce conditions more like those found in vivo, and with appropriate oxygen levels, mimic hypoxic tumours. However, most previous experiments studying both EVs and the effects of hypoxia on cells use periods of 72 h or less of hypoxia. We hypothesised that this was insufficient time for adaptation to hypoxic conditions both for EVs and cells which may skew the results of such studies. In this study, the effects of acute (72 h) and chronic hypoxia (> 2 weeks) on the phenotype of HepG2 and PC3 cells and their EVs were examined. Cells could be cultured normally under chronic hypoxic conditions and cryopreserved and recovered. The effects of hypoxia on EV phenotype are slow to establish and dependent on cell line. In PC3 cells, the greatest change in phenotype and increase in EV production occurred only with chronic hypoxic culture. In HepG2 cells, the number of EVs produced was insensitive to hypoxic culture and the greatest changes in protein expression were observed after acute hypoxic culture. Nonetheless, biphasic changes in EV phenotype were detected in both cell types in response to either acute or chronic hypoxia. These results indicate that for cells which do not induce consumptive oxygen depletion, prolonged hypoxic culture is required for complete adaptation.
Collapse
Affiliation(s)
- Chris S Pridgeon
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Kerttu Airavaara
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Julia Monola
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Alisa Jokela
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Daniel Palmer
- Institut für Biostatistik und Informatik in Medizin und Alternsforschung, University of Rostock, Rostock, Germany
| | - Marjo Yliperttula
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Riina Harjumäki
- Division of Pharmaceutical Biosciences, Drug Research Program, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland.
| |
Collapse
|
40
|
Caulat LC, Lotoux A, Martins MC, Kint N, Anjou C, Teixeira M, Folgosa F, Morvan C, Martin-Verstraete I. Physiological role and complex regulation of O 2-reducing enzymes in the obligate anaerobe Clostridioides difficile. mBio 2024; 15:e0159124. [PMID: 39189748 PMCID: PMC11481553 DOI: 10.1128/mbio.01591-24] [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: 06/05/2024] [Accepted: 07/22/2024] [Indexed: 08/28/2024] Open
Abstract
Clostridioides difficile, the major cause of antibiotic-associated diarrhea, is a strict anaerobic, sporulating Firmicutes. However, during its infectious cycle, this anaerobe is exposed to low oxygen (O2) tensions, with a longitudinal decreasing gradient along the gastrointestinal tract and a second lateral gradient with higher O2 tensions in the vicinity of the cells. A plethora of enzymes involved in oxidative stress detoxication has been identified in C. difficile, including four O2-reducing enzymes: two flavodiiron proteins (FdpA and FdpF) and two reverse rubrerythrins (revRbr1 and revRbr2). Here, we investigated the role of the four O2-reducing enzymes in the tolerance to increasing physiological O2 tensions and air. The four enzymes have different, yet overlapping, spectra of activity. revRbr2 is specific to low O2 tensions (<0.4%), FdpA to low and intermediate O2 tensions (0.4%-1%), revRbr1 has a wider spectrum of activity (0.1%-4%), and finally FdpF is more specific to tensions > 4% and air. These different O2 ranges of action partly arise from differences in regulation of expression of the genes encoding those enzymes. Indeed, we showed that revrbr2 is under the dual control of σA and σB. We also identified a regulator of the Spx family that plays a role in the induction of fdp and revrbr genes upon O2 exposure. Finally, fdpF is regulated by Rex, a regulator sensing the NADH/NAD+ ratio. Our results demonstrate that the multiplicity of O2-reducing enzymes of C. difficile is associated with different roles depending on the environmental conditions, stemming from a complex multi-leveled network of regulation. IMPORTANCE The gastrointestinal tract is a hypoxic environment, with the existence of two gradients of O2 along the gut, one longitudinal anteroposterior decreasing gradient and one proximodistal increasing from the lumen to the epithelial cells. O2 is a major source of stress for an obligate anaerobe such as the enteropathogen C. difficile. This bacterium possesses a plethora of enzymes capable of scavenging O2 and reducing it to H2O. In this work, we identified the role of the four O2-reducing enzymes in the tolerance to the physiological O2 tensions faced by C. difficile during its infectious cycle. These four enzymes have different spectra of action and protect the vegetative cells over a large range of O2 tensions. These differences are associated with a distinct regulation of each gene encoding those enzymes. The complex network of regulation is crucial for C. difficile to adapt to the various O2 tensions encountered during infection.
Collapse
Affiliation(s)
- Léo C. Caulat
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Aurélie Lotoux
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Maria C. Martins
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Nicolas Kint
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Cyril Anjou
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Miguel Teixeira
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Filipe Folgosa
- Instituto de Tecnologia Química e Biológica António Xavier, Universidade Nova de Lisboa, Oeiras, Portugal
| | - Claire Morvan
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
| | - Isabelle Martin-Verstraete
- Institut Pasteur, Université de Paris, CNRS UMR6047, Laboratoire Pathogenèse des Bactéries Anaérobies, Paris, France
- Institut Universitaire de France, Paris, France
| |
Collapse
|
41
|
Gardner GL, Stuart JA. Tumor microenvironment-like conditions alter pancreatic cancer cell metabolism and behavior. Am J Physiol Cell Physiol 2024; 327:C959-C978. [PMID: 39183564 DOI: 10.1152/ajpcell.00452.2024] [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: 07/03/2024] [Revised: 08/19/2024] [Accepted: 08/20/2024] [Indexed: 08/27/2024]
Abstract
The tumor microenvironment is complex and dynamic, characterized by poor vascularization, limited nutrient availability, hypoxia, and an acidic pH. This environment plays a critical role in driving cancer progression. However, standard cell culture conditions used to study cancer cell biology in vitro fail to replicate the in vivo environment of tumors. Recently, "physiological" cell culture media that closely resemble human plasma have been developed (e.g., Plasmax, HPLM), along with more frequent adoption of physiological oxygen conditions (1%-8% O2). Nonetheless, further refinement of tumor-specific culture conditions may be needed. In this study, we describe the development of a tumor microenvironment medium (TMEM) based on murine pancreatic ductal adenocarcinoma (PDAC) tumor interstitial fluid. Using RNA-sequencing, we show that murine PDAC cells (KPCY) cultured in tumor-like conditions (TMEM, pH 7.0, 1.5% O2) exhibit profound differences in gene expression compared with plasma-like conditions (mouse plasma medium, pH 7.4, 5% O2). Specifically, the expression of genes and pathways associated with cell migration, biosynthesis, angiogenesis, and epithelial-to-mesenchymal transition were altered, suggesting tumor-like conditions promote metastatic phenotypes and metabolic remodeling. Using functional assays to validate RNA-seq data, we confirmed increased motility at 1.5% O2/TMEM, despite reduced cell proliferation. Moreover, a hallmark shift to glycolytic metabolism was identified via measurement of glucose uptake/lactate production and mitochondrial respiration. Taken together, these findings demonstrate that growth in 1.5% O2/TMEM alters several biological responses in ways relevant to cancer biology, and more closely models hallmark cancerous phenotypes in culture. This highlights the importance of establishing tumor microenvironment-like conditions in standard cancer research. NEW & NOTEWORTHY Standard cell culture conditions do not replicate the complex tumor microenvironment experienced by cells in vivo. Although currently available plasma-like media are superior to traditional supraphysiological media, they fail to model tumor-like conditions. Using RNA-seq analysis and functional metabolic and migratory assays, we show that tumor microenvironment medium (TMEM), used with representative tumor hypoxia, better models cancerous phenotypes in culture. This emphasizes the critical importance of accurately modeling the tumor microenvironment in cancer research.
Collapse
Affiliation(s)
| | - Jeffrey Alan Stuart
- Department of Biological Sciences, Brock University, St. Catharines, Ontario, Canada
| |
Collapse
|
42
|
Mirabelli M, Misiti R, Sicilia L, Brunetti FS, Chiefari E, Brunetti A, Foti DP. Hypoxia in Human Obesity: New Insights from Inflammation towards Insulin Resistance-A Narrative Review. Int J Mol Sci 2024; 25:9802. [PMID: 39337290 PMCID: PMC11432683 DOI: 10.3390/ijms25189802] [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: 08/02/2024] [Revised: 09/06/2024] [Accepted: 09/09/2024] [Indexed: 09/30/2024] Open
Abstract
Insulin resistance (IR), marked by reduced cellular responsiveness to insulin, and obesity, defined by the excessive accumulation of adipose tissue, are two intertwined conditions that significantly contribute to the global burden of cardiometabolic diseases. Adipose tissue, beyond merely storing triglycerides, acts as an active producer of biomolecules. In obesity, as adipose tissue undergoes hypertrophy, it becomes dysfunctional, altering the release of adipocyte-derived factors, known as adipokines. This dysfunction promotes low-grade chronic inflammation, exacerbates IR, and creates a hyperglycemic, proatherogenic, and prothrombotic environment. However, the fundamental cause of these phenomena remains unclear. This narrative review points to hypoxia as a critical trigger for the molecular changes associated with fat accumulation, particularly within visceral adipose tissue (VAT). The activation of hypoxia-inducible factor-1 (HIF-1), a transcription factor that regulates homeostatic responses to low oxygen levels, initiates a series of molecular events in VAT, leading to the aberrant release of adipokines, many of which are still unexplored, and potentially affecting peripheral insulin sensitivity. Recent discoveries have highlighted the role of hypoxia and miRNA-128 in regulating the insulin receptor in visceral adipocytes, contributing to their dysfunctional behavior, including impaired glucose uptake. Understanding the complex interplay between adipose tissue hypoxia, dysfunction, inflammation, and IR in obesity is essential for developing innovative, targeted therapeutic strategies.
Collapse
Affiliation(s)
- Maria Mirabelli
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Roberta Misiti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| | - Luciana Sicilia
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Francesco S. Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
| | - Eusebio Chiefari
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Antonio Brunetti
- Department of Health Sciences, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy; (M.M.)
- Operative Unit of Endocrinology, “Renato Dulbecco” University Hospital, 88100 Catanzaro, Italy
| | - Daniela P. Foti
- Department of Experimental and Clinical Medicine, University “Magna Græcia” of Catanzaro, 88100 Catanzaro, Italy;
- Operative Unit of Clinical Pathology, “Renato Dulbecco” Hospital, 88100 Catanzaro, Italy
| |
Collapse
|
43
|
Reis A, Rocha BS, Laranjinha J, de Freitas V. Dietary (poly)phenols as modulators of the biophysical properties in endothelial cell membranes: its impact on nitric oxide bioavailability in hypertension. FEBS Lett 2024; 598:2190-2210. [PMID: 38281810 DOI: 10.1002/1873-3468.14812] [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: 10/13/2023] [Revised: 12/18/2023] [Accepted: 12/27/2023] [Indexed: 01/30/2024]
Abstract
Hypertension is a major contributor to premature death, owing to the associated increased risk of damage to the heart, brain and kidneys. Although hypertension is manageable by medication and lifestyle changes, the risk increases with age. In an increasingly aged society, the incidence of hypertension is escalating, and is expected to increase the prevalence of (cerebro)vascular events and their associated mortality. Adherence to plant-based diets improves blood pressure and vascular markers in individuals with hypertension. Food flavonoids have an inhibitory effect towards angiotensin-converting enzyme (ACE1) and although this effect is greatly diminished upon metabolization, their microbial metabolites have been found to improve endothelial nitric oxide synthase (eNOS) activity. Considering the transmembrane location of ACE1 and eNOS, the ability of (poly)phenols to interact with membrane lipids modulate the cell membrane's biophysical properties and impact on nitric oxide (·NO) synthesis and bioavailability, remain poorly studied. Herein, we provide an overview of the current knowledge on the lipid remodeling of endothelial membranes with age, its impact on the cell membrane's biophysical properties and ·NO permeability across the endothelial barrier. We also discuss the potential of (poly)phenols and other plant-based compounds as key players in hypertension management, and address the caveats and challenges in adopted methodologies.
Collapse
Affiliation(s)
- Ana Reis
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal
| | - Barbara S Rocha
- Faculty of Pharmacy and Center for Neuroscience and Cell Biology, University of Coimbra, Polo das Ciências da Saúde, Portugal
| | - João Laranjinha
- Faculty of Pharmacy and Center for Neuroscience and Cell Biology, University of Coimbra, Polo das Ciências da Saúde, Portugal
| | - Victor de Freitas
- REQUIMTE/LAQV, Departamento de Química e Bioquímica, Faculdade de Ciências, Universidade do Porto, Portugal
| |
Collapse
|
44
|
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
|
45
|
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
|
46
|
Altun HY, Secilmis M, Yang F, Akgul Caglar T, Vatandaslar E, Toy MF, Vilain S, Mann GE, Öztürk G, Eroglu E. Visualizing hydrogen peroxide and nitric oxide dynamics in endothelial cells using multispectral imaging under controlled oxygen conditions. Free Radic Biol Med 2024; 221:89-97. [PMID: 38735541 DOI: 10.1016/j.freeradbiomed.2024.05.021] [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: 01/12/2024] [Revised: 04/04/2024] [Accepted: 05/10/2024] [Indexed: 05/14/2024]
Abstract
The complex interplay between hydrogen peroxide (H2O2) and nitric oxide (NO) in endothelial cells presents challenges due to technical limitations in simultaneous measurement, hindering the elucidation of their direct relationship. Previous studies have yielded conflicting findings regarding the impact of H2O2 on NO production. To address this problem, we employed genetically encoded biosensors, HyPer7 for H2O2 and geNOps for NO, allowing simultaneous imaging in single endothelial cells. Optimization strategies were implemented to enhance biosensor performance, including camera binning, temperature regulation, and environmental adjustments to mimic physiological normoxia. Our results demonstrate that under ambient oxygen conditions, H2O2 exhibited no significant influence on NO production. Subsequent exploration under physiological normoxia (5 kPa O2) revealed distinct oxidative stress levels characterized by reduced basal HyPer7 signals, enhanced H2O2 scavenging kinetics, and altered responses to pharmacological treatment. Investigation of the relationship between H2O2 and NO under varying oxygen conditions revealed a lack of NO response to H2O2 under hyperoxia (18 kPa O2) but a modest NO response under physiological normoxia (5 kPa O2). Importantly, the NO response was attenuated by l-NAME, suggesting activation of eNOS by endogenous H2O2 generation upon auranofin treatment. Our study highlights the intricate interplay between H2O2 and NO within the endothelial EA.hy926 cell line, emphasizing the necessity for additional research within physiological contexts due to differential response observed under physiological normoxia (5 kPa O2). This further investigation is essential for a comprehensive understanding of the H2O2 and NO signaling considering the physiological effects of ambient O2 levels involved.
Collapse
Affiliation(s)
- Hamza Yusuf Altun
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Melike Secilmis
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Fan Yang
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK
| | - Tuba Akgul Caglar
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey
| | - Emre Vatandaslar
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Muhammed Fatih Toy
- School of Engineering and Natural Science, Istanbul Medipol University, 34810, Istanbul, Turkey
| | - Sven Vilain
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey.
| | - Giovanni E Mann
- King's British Heart Foundation Centre of Research Excellence, School of Cardiovascular and Metabolic Medicine & Sciences, Faculty of Life Sciences & Medicine, King's College London, 150 Stamford Street, London, SE1 9NH, UK.
| | - Gürkan Öztürk
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Physiology Department, School of Medicine, Bolu Abant Izzet Baysal University (BAIBU), 14200, Bolu, Türkiye.
| | - Emrah Eroglu
- Research Institute for Health Sciences and Technologies (SABITA), Istanbul Medipol University, 34810, Istanbul, Turkey; Molecular Biology, Genetics, and Bioengineering Program, Sabanci University, 34956, Istanbul, Turkey.
| |
Collapse
|
47
|
Sæterstad S, Østvik AE, Hansen MD, Bruland T, van Beelen Granlund A. The effect of rs2910686 on ERAP2 expression in IBD and epithelial inflammatory response. J Transl Med 2024; 22:750. [PMID: 39123229 PMCID: PMC11316291 DOI: 10.1186/s12967-024-05532-w] [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: 05/03/2024] [Accepted: 07/23/2024] [Indexed: 08/12/2024] Open
Abstract
BACKGROUND ERAP2 is an aminopeptidase involved in antigen processing and presentation, and harbor genetic variants linked to several inflammatory diseases such as Inflammatory Bowel Disease (IBD). The lack of an ERAP2 gene homologue in mice has hampered functional studies, and most human studies have focused on cells of hematopoietic origin. Using an IBD biobank as vantage point, this study explores how genetic variation in ERAP2 affects gene expression in human-derived epithelial organoids upon proinflammatory stimulation. METHODS An IBD patient cohort was genotyped with regards to two single nucleotide polymorphisms (SNP) (rs2910686/rs2248374) associated with ERAP2 expression levels, and we examined the correlation between colon gene expression and genotype, specifically aiming to establish a relationship with ERAP2 expression proficiency. Human-derived colon organoids (colonoids) with known ERAP2 genotype were established and used to explore differences in whole genome gene expression between ERAP2-deficient (n = 4) and -proficient (n = 4) donors upon pro-inflammatory encounter. RESULTS When taking rs2910686 genotype into account, ERAP2 gene expression is upregulated in the inflamed colon of IBD patients. Colonoids upregulate ERAP2 upon IFNɣ stimulation, and ERAP2 expression proficiency is dependent on rs2910686 genotype. Colonoid genotyping confirms that mechanisms independent of the frequently studied SNP rs2248374 can cause ERAP2-deficiency. A total of 586 genes involved in various molecular mechanisms are differentially expressed between ERAP2 proficient- and deficient colonoids upon proinflammatory stimulation, including genes encoding proteins with the following molecular function: catalytic activity (AOC1, CPE, ANPEP and MEP1A), regulator activity (TNFSF9, MDK, GDF15, ILR6A, LGALS3 and FLNA), transmembrane transporter activity (SLC40A1 and SLC5A1), and extracellular matrix structural constituents (FGL2, HMCN2, and MUC17). CONCLUSIONS ERAP2 is upregulated in the inflamed IBD colon mucosa, and expression proficiency is highly correlated with genotype of rs2910686. While the SNP rs2248374 is commonly used to determine ERAP2 expressional proficiency, our data confirms that mechanisms independent of this SNP can lead to ERAP2 deficiency. Our data demonstrates that epithelial ERAP2 presence affects the inflammatory response in colonoids, suggesting a pleiotropic role of ERAP2 beyond MHC class I antigen processing.
Collapse
Affiliation(s)
- Siri Sæterstad
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
| | - Ann Elisabeth Østvik
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Marianne Doré Hansen
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Clinic of Laboratory Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Torunn Bruland
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway
| | - Atle van Beelen Granlund
- Department of Clinical and Molecular Medicine, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
- Department of Gastroenterology and Hepatology, Clinic of Medicine, St. Olav's University Hospital, Trondheim, Norway.
- Department of Pathology, St. Olav's University Hospital, Trondheim, Norway.
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology (NTNU), Trondheim, Norway.
| |
Collapse
|
48
|
Rogers ZJ, Colombani T, Khan S, Bhatt K, Nukovic A, Zhou G, Woolston BM, Taylor CT, Gilkes DM, Slavov N, Bencherif SA. Controlling Pericellular Oxygen Tension in Cell Culture Reveals Distinct Breast Cancer Responses to Low Oxygen Tensions. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024; 11:e2402557. [PMID: 38874400 PMCID: PMC11321643 DOI: 10.1002/advs.202402557] [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: 03/14/2024] [Revised: 04/11/2024] [Indexed: 06/15/2024]
Abstract
In oxygen (O2)-controlled cell culture, an indispensable tool in biological research, it is presumed that the incubator setpoint equals the O2 tension experienced by cells (i.e., pericellular O2). However, it is discovered that physioxic (5% O2) and hypoxic (1% O2) setpoints regularly induce anoxic (0% O2) pericellular tensions in both adherent and suspension cell cultures. Electron transport chain inhibition ablates this effect, indicating that cellular O2 consumption is the driving factor. RNA-seq analysis revealed that primary human hepatocytes cultured in physioxia experience ischemia-reperfusion injury due to cellular O2 consumption. A reaction-diffusion model is developed to predict pericellular O2 tension a priori, demonstrating that the effect of cellular O2 consumption has the greatest impact in smaller volume culture vessels. By controlling pericellular O2 tension in cell culture, it is found that hypoxia vs. anoxia induce distinct breast cancer transcriptomic and translational responses, including modulation of the hypoxia-inducible factor (HIF) pathway and metabolic reprogramming. Collectively, these findings indicate that breast cancer cells respond non-monotonically to low O2, suggesting that anoxic cell culture is not suitable for modeling hypoxia. Furthermore, it is shown that controlling atmospheric O2 tension in cell culture incubators is insufficient to regulate O2 in cell culture, thus introducing the concept of pericellular O2-controlled cell culture.
Collapse
Affiliation(s)
- Zachary J. Rogers
- Department of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Thibault Colombani
- Department of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Saad Khan
- Department of BioengineeringNortheastern UniversityBostonMA02115USA
| | - Khushbu Bhatt
- Department of Pharmaceutical SciencesNortheastern UniversityBostonMA02115USA
| | - Alexandra Nukovic
- Department of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | - Guanyu Zhou
- Department of Chemical EngineeringNortheastern UniversityBostonMA02115USA
| | | | - Cormac T. Taylor
- Conway Institute of Biomolecular and Biomedical Research and School of MedicineUniversity College DublinBelfieldDublinD04 V1W8Ireland
| | - Daniele M. Gilkes
- Department of OncologyThe Sidney Kimmel Comprehensive Cancer CenterThe Johns Hopkins University School of MedicineBaltimoreMD21321USA
- Cellular and Molecular Medicine ProgramThe Johns Hopkins University School of MedicineBaltimoreMD21321USA
- Department of Chemical and Biomolecular EngineeringThe Johns Hopkins UniversityBaltimoreMD21218USA
- Johns Hopkins Institute for NanoBioTechnologyThe Johns Hopkins UniversityBaltimoreMD21218USA
| | - Nikolai Slavov
- Department of BioengineeringNortheastern UniversityBostonMA02115USA
- Departments of BioengineeringBiologyChemistry and Chemical BiologySingle Cell Center and Barnett InstituteNortheastern UniversityBostonMA02115USA
- Parallel Squared Technology InstituteWatertownMA02472USA
| | - Sidi A. Bencherif
- Department of Chemical EngineeringNortheastern UniversityBostonMA02115USA
- Department of BioengineeringNortheastern UniversityBostonMA02115USA
- Harvard John A. Paulson School of Engineering and Applied SciencesHarvard UniversityCambridgeMA02138USA
- Biomechanics and Bioengineering (BMBI)UTC CNRS UMR 7338University of Technology of CompiègneSorbonne UniversityCompiègne60203France
| |
Collapse
|
49
|
Kochanowsky R, Carothers K, Roxas BAP, Anwar F, Viswanathan VK, Vedantam G. Clostridioides difficile superoxide reductase mitigates oxygen sensitivity. J Bacteriol 2024; 206:e0017524. [PMID: 38953644 PMCID: PMC11270899 DOI: 10.1128/jb.00175-24] [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: 04/24/2024] [Accepted: 06/12/2024] [Indexed: 07/04/2024] Open
Abstract
Clostridioides difficile causes a serious diarrheal disease and is a common healthcare-associated bacterial pathogen. Although it has a major impact on human health, the mechanistic details of C. difficile intestinal colonization remain undefined. C. difficile is highly sensitive to oxygen and requires anaerobic conditions for in vitro growth. However, the mammalian gut is not devoid of oxygen, and C. difficile tolerates moderate oxidative stress in vivo. The C. difficile genome encodes several antioxidant proteins, including a predicted superoxide reductase (SOR) that is upregulated upon exposure to antimicrobial peptides. The goal of this study was to establish SOR enzymatic activity and assess its role in protecting C. difficile against oxygen exposure. Insertional inactivation of sor rendered C. difficile more sensitive to superoxide, indicating that SOR contributes to antioxidant defense. Heterologous C. difficile sor expression in Escherichia coli conferred protection against superoxide-dependent growth inhibition, and the corresponding cell lysates showed superoxide scavenging activity. Finally, a C. difficile SOR mutant exhibited global proteome changes under oxygen stress when compared to the parent strain. Collectively, our data establish the enzymatic activity of C. difficile SOR, confirm its role in protection against oxidative stress, and demonstrate SOR's broader impacts on the C. difficile vegetative cell proteome.IMPORTANCEClostridioides difficile is an important pathogen strongly associated with healthcare settings and capable of causing severe diarrheal disease. While considered a strict anaerobe in vitro, C. difficile has been shown to tolerate low levels of oxygen in the mammalian host. Among other well-characterized antioxidant proteins, the C. difficile genome encodes a predicted superoxide reductase (SOR), an understudied component of antioxidant defense in pathogens. The significance of the research reported herein is the characterization of SOR's enzymatic activity, including confirmation of its role in protecting C. difficile against oxidative stress. This furthers our understanding of C. difficile pathogenesis and presents a potential new avenue for targeted therapies.
Collapse
Affiliation(s)
- Rebecca Kochanowsky
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Katelyn Carothers
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Bryan Angelo P. Roxas
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
| | - Farhan Anwar
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
| | - V. K. Viswanathan
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
- BIO5 Institute for Collaborative Research, The University of Arizona, Tucson, Arizona, USA
| | - Gayatri Vedantam
- School of Animal and Comparative Biomedical Sciences, The University of Arizona, Tucson, Arizona, USA
- BIO5 Institute for Collaborative Research, The University of Arizona, Tucson, Arizona, USA
- Southern Arizona VA Healthcare System, Tucson, Arizona, USA
| |
Collapse
|
50
|
Cobley JN, Margaritelis NV, Chatzinikolaou PN, Nikolaidis MG, Davison GW. Ten "Cheat Codes" for Measuring Oxidative Stress in Humans. Antioxidants (Basel) 2024; 13:877. [PMID: 39061945 PMCID: PMC11273696 DOI: 10.3390/antiox13070877] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2024] [Revised: 07/17/2024] [Accepted: 07/18/2024] [Indexed: 07/28/2024] Open
Abstract
Formidable and often seemingly insurmountable conceptual, technical, and methodological challenges hamper the measurement of oxidative stress in humans. For instance, fraught and flawed methods, such as the thiobarbituric acid reactive substances assay kits for lipid peroxidation, rate-limit progress. To advance translational redox research, we present ten comprehensive "cheat codes" for measuring oxidative stress in humans. The cheat codes include analytical approaches to assess reactive oxygen species, antioxidants, oxidative damage, and redox regulation. They provide essential conceptual, technical, and methodological information inclusive of curated "do" and "don't" guidelines. Given the biochemical complexity of oxidative stress, we present a research question-grounded decision tree guide for selecting the most appropriate cheat code(s) to implement in a prospective human experiment. Worked examples demonstrate the benefits of the decision tree-based cheat code selection tool. The ten cheat codes define an invaluable resource for measuring oxidative stress in humans.
Collapse
Affiliation(s)
- James N. Cobley
- The University of Dundee, Dundee DD1 4HN, UK
- Ulster University, Belfast BT15 1ED, Northern Ireland, UK;
| | - Nikos V. Margaritelis
- Aristotle University of Thessaloniki, 62122 Serres, Greece; (N.V.M.); (P.N.C.); (M.G.N.)
| | | | - Michalis G. Nikolaidis
- Aristotle University of Thessaloniki, 62122 Serres, Greece; (N.V.M.); (P.N.C.); (M.G.N.)
| | | |
Collapse
|