201
|
Rigual-González Y, Gómez L, Núñez J, Vergara M, Díaz-Barrera A, Berrios J, Altamirano C. Application of a new model based on oxygen balance to determine the oxygen uptake rate in mammalian cell chemostat cultures. Chem Eng Sci 2016. [DOI: 10.1016/j.ces.2016.06.051] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
202
|
Oomen PE, Skolimowski MD, Verpoorte E. Implementing oxygen control in chip-based cell and tissue culture systems. LAB ON A CHIP 2016; 16:3394-414. [PMID: 27492338 DOI: 10.1039/c6lc00772d] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/23/2023]
Abstract
Oxygen is essential in the energy metabolism of cells, as well as being an important regulatory parameter influencing cell differentiation and function. Interest in precise oxygen control for in vitro cultures of tissues and cells continues to grow, especially with the emergence of the organ-on-a-chip and the desire to emulate in vivo conditions. This was recently discussed in this journal in a Critical Review by Brennan et al. (Lab Chip (2014). DOI: ). Microfluidics can be used to introduce flow to facilitate nutrient supply to and waste removal from in vitro culture systems. Well-defined oxygen gradients can also be established. However, cells can quickly alter the oxygen balance in their vicinity. In this Tutorial Review, we expand on the Brennan paper to focus on the implementation of oxygen analysis in these systems to achieve continuous monitoring. Both electrochemical and optical approaches for the integration of oxygen monitoring in microfluidic tissue and cell culture systems will be discussed. Differences in oxygen requirements from one organ to the next are a challenging problem, as oxygen delivery is limited by its uptake into medium. Hence, we discuss the factors determining oxygen concentrations in solutions and consider the possible use of artificial oxygen carriers to increase dissolved oxygen concentrations. The selection of device material for applications requiring precise oxygen control is discussed in detail, focusing on oxygen permeability. Lastly, a variety of devices is presented, showing the diversity of approaches that can be employed to control and monitor oxygen concentrations in in vitro experiments.
Collapse
Affiliation(s)
- Pieter E Oomen
- Pharmaceutical Analysis, Groningen Research Institute of Pharmacy, University of Groningen, Antonius Deusinglaan 1 (XB20), 9713 AV Groningen, The Netherlands.
| | | | | |
Collapse
|
203
|
Littlejohn NK, Keen HL, Weidemann BJ, Claflin KE, Tobin KV, Markan KR, Park S, Naber MC, Gourronc FA, Pearson NA, Liu X, Morgan DA, Klingelhutz AJ, Potthoff MJ, Rahmouni K, Sigmund CD, Grobe JL. Suppression of Resting Metabolism by the Angiotensin AT2 Receptor. Cell Rep 2016; 16:1548-1560. [PMID: 27477281 DOI: 10.1016/j.celrep.2016.07.003] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2016] [Revised: 06/09/2016] [Accepted: 07/01/2016] [Indexed: 11/15/2022] Open
Abstract
Activation of the brain renin-angiotensin system (RAS) stimulates energy expenditure through increasing of the resting metabolic rate (RMR), and this effect requires simultaneous suppression of the circulating and/or adipose RAS. To identify the mechanism by which the peripheral RAS opposes RMR control by the brain RAS, we examined mice with transgenic activation of the brain RAS (sRA mice). sRA mice exhibit increased RMR through increased energy flux in the inguinal adipose tissue, and this effect is attenuated by angiotensin II type 2 receptor (AT2) activation. AT2 activation in inguinal adipocytes opposes norepinephrine-induced uncoupling protein-1 (UCP1) production and aspects of cellular respiration, but not lipolysis. AT2 activation also opposes inguinal adipocyte function and differentiation responses to epidermal growth factor (EGF). These results highlight a major, multifaceted role for AT2 within inguinal adipocytes in the control of RMR. The AT2 receptor may therefore contribute to body fat distribution and adipose depot-specific effects upon cardio-metabolic health.
Collapse
Affiliation(s)
| | - Henry L Keen
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Kristin E Claflin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kevin V Tobin
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Kathleen R Markan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Sungmi Park
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Meghan C Naber
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | | | - Nicole A Pearson
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Xuebo Liu
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Donald A Morgan
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA
| | - Aloysius J Klingelhutz
- Department of Microbiology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA
| | - Matthew J Potthoff
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA
| | - Kamal Rahmouni
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA
| | - Curt D Sigmund
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| | - Justin L Grobe
- Department of Pharmacology, University of Iowa, Iowa City, IA 52242, USA; Fraternal Order of Eagles' Diabetes Research Center, University of Iowa, Iowa City, IA 52242, USA; Obesity Research and Education Initiative, University of Iowa, Iowa City, IA 52242, USA; François M. Abboud Cardiovascular Research Center, University of Iowa, Iowa City, IA 52242, USA; Center for Hypertension Research, University of Iowa, Iowa City, IA 52242, USA.
| |
Collapse
|
204
|
Coleman MC, Ramakrishnan PS, Brouillette MJ, Martin JA. Injurious Loading of Articular Cartilage Compromises Chondrocyte Respiratory Function. Arthritis Rheumatol 2016; 68:662-71. [PMID: 26473613 DOI: 10.1002/art.39460] [Citation(s) in RCA: 64] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Accepted: 09/29/2015] [Indexed: 01/11/2023]
Abstract
OBJECTIVE To determine whether repeatedly overloading healthy cartilage disrupts mitochondrial function in a manner similar to that associated with osteoarthritis (OA) pathogenesis. METHODS We exposed normal articular cartilage on bovine osteochondral explants to 1 day or 7 consecutive days of cyclic axial compression (0.25 MPa or 1.0 MPa at 0.5 Hz for 3 hours) and evaluated the effects on chondrocyte viability, ATP concentration, reactive oxygen species (ROS) production, indicators of oxidative stress, respiration, and mitochondrial membrane potential. RESULTS Neither 0.25 MPa nor 1.0 MPa of cyclic compression caused extensive chondrocyte death, macroscopic tissue damage, or overt changes in stress-strain behavior. After 1 day of loading, differences in respiratory activities between the 0.25 MPa and 1.0 MPa groups were minimal; however, after 7 days of loading, respiratory activity and ATP levels were suppressed in the 1.0 MPa group relative to the 0.25 MPa group, an effect prevented by pretreatment with 10 mM N-acetylcysteine. These changes were accompanied by increased proton leakage and decreased mitochondrial membrane potential, as well as by increased ROS formation, as indicated by dihydroethidium staining and glutathione oxidation. CONCLUSION Repeated overloading leads to chondrocyte oxidant-dependent mitochondrial dysfunction. This mitochondrial dysfunction may contribute to destabilization of cartilage during various stages of OA in distinct ways by disrupting chondrocyte anabolic responses to mechanical stimuli.
Collapse
|
205
|
Vacaflores A, Chapman NM, Harty JT, Richer MJ, Houtman JCD. Exposure of Human CD4 T Cells to IL-12 Results in Enhanced TCR-Induced Cytokine Production, Altered TCR Signaling, and Increased Oxidative Metabolism. PLoS One 2016; 11:e0157175. [PMID: 27280403 PMCID: PMC4900534 DOI: 10.1371/journal.pone.0157175] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Accepted: 05/25/2016] [Indexed: 12/24/2022] Open
Abstract
Human CD4 T cells are constantly exposed to IL-12 during infections and certain autoimmune disorders. The current paradigm is that IL-12 promotes the differentiation of naïve CD4 T cells into Th1 cells, but recent studies suggest IL-12 may play a more complex role in T cell biology. We examined if exposure to IL-12 alters human CD4 T cell responses to subsequent TCR stimulation. We found that IL-12 pretreatment increased TCR-induced IFN-γ, TNF-α, IL-13, IL-4 and IL-10 production. This suggests that prior exposure to IL-12 potentiates the TCR-induced release of a range of cytokines. We observed that IL-12 mediated its effects through both transcriptional and post-transcriptional mechanisms. IL-12 pretreatment increased the phosphorylation of AKT, p38 and LCK following TCR stimulation without altering other TCR signaling molecules, potentially mediating the increase in transcription of cytokines. In addition, the IL-12-mediated enhancement of cytokines that are not transcriptionally regulated was partially driven by increased oxidative metabolism. Our data uncover a novel function of IL-12 in human CD4 T cells; specifically, it enhances the release of a range of cytokines potentially by altering TCR signaling pathways and by enhancing oxidative metabolism.
Collapse
Affiliation(s)
- Aldo Vacaflores
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - Nicole M. Chapman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
| | - John T. Harty
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Pathology, University of Iowa, Iowa City, Iowa, United States of America
| | - Martin J. Richer
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
| | - Jon C. D. Houtman
- Interdisciplinary Graduate Program in Immunology, University of Iowa, Iowa City, Iowa, United States of America
- Department of Microbiology, Carver College of Medicine, University of Iowa, Iowa City, Iowa, United States of America
- Department of Internal Medicine, Division of Immunology, University of Iowa, Iowa City, Iowa, United States of America
- * E-mail:
| |
Collapse
|
206
|
Yilmaz A, Utz M. Characterisation of oxygen permeation into a microfluidic device for cell culture by in situ NMR spectroscopy. LAB ON A CHIP 2016; 16:2079-2085. [PMID: 27149932 DOI: 10.1039/c6lc00396f] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/05/2023]
Abstract
A compact microfluidic device for perfusion culture of mammalian cells under in situ metabolomic observation by NMR spectroscopy is presented. The chip is made from poly(methyl methacrylate) (PMMA), and uses a poly(dimethyl siloxane) (PDMS) membrane to allow gas exchange. It is integrated with a generic micro-NMR detector developed recently by our group [J. Magn. Reson., 2016, 262, 73-80]. While PMMA is an excellent material in the context of NMR, PDMS is known to produce strong background signals. To mitigate this, the device keeps the PDMS away from the detection area. The oxygen permeation into the device is quantified using a flow chemistry approach. A solution of glucose is mixed on the chip with a solution of glucose oxidase, before flowing through the gas exchanger. The resulting concentration of gluconate is measured by (1)H NMR spectroscopy as a function of flow rate. An oxygen equilibration rate constant of 2.4 s(-1) is found for the device, which is easily sufficient to maintain normoxic conditions in a cell culture at low perfusion flow rates.
Collapse
Affiliation(s)
- Ali Yilmaz
- School of Chemistry, University of Southampton, SO17 1BJ, UK.
| | - Marcel Utz
- School of Chemistry, University of Southampton, SO17 1BJ, UK.
| |
Collapse
|
207
|
McMurtrey RJ. Analytic Models of Oxygen and Nutrient Diffusion, Metabolism Dynamics, and Architecture Optimization in Three-Dimensional Tissue Constructs with Applications and Insights in Cerebral Organoids. Tissue Eng Part C Methods 2016; 22:221-249. [PMID: 26650970 PMCID: PMC5029285 DOI: 10.1089/ten.tec.2015.0375] [Citation(s) in RCA: 137] [Impact Index Per Article: 15.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2015] [Accepted: 12/02/2015] [Indexed: 12/12/2022] Open
Abstract
Diffusion models are important in tissue engineering as they enable an understanding of gas, nutrient, and signaling molecule delivery to cells in cell cultures and tissue constructs. As three-dimensional (3D) tissue constructs become larger, more intricate, and more clinically applicable, it will be essential to understand internal dynamics and signaling molecule concentrations throughout the tissue and whether cells are receiving appropriate nutrient delivery. Diffusion characteristics present a significant limitation in many engineered tissues, particularly for avascular tissues and for cells whose viability, differentiation, or function are affected by concentrations of oxygen and nutrients. This article seeks to provide novel analytic solutions for certain cases of steady-state and nonsteady-state diffusion and metabolism in basic 3D construct designs (planar, cylindrical, and spherical forms), solutions that would otherwise require mathematical approximations achieved through numerical methods. This model is applied to cerebral organoids, where it is shown that limitations in diffusion and organoid size can be partially overcome by localizing metabolically active cells to an outer layer in a sphere, a regionalization process that is known to occur through neuroglial precursor migration both in organoids and in early brain development. The given prototypical solutions include a review of metabolic information for many cell types and can be broadly applied to many forms of tissue constructs. This work enables researchers to model oxygen and nutrient delivery to cells, predict cell viability, study dynamics of mass transport in 3D tissue constructs, design constructs with improved diffusion capabilities, and accurately control molecular concentrations in tissue constructs that may be used in studying models of development and disease or for conditioning cells to enhance survival after insults like ischemia or implantation into the body, thereby providing a framework for better understanding and exploring the characteristics and behaviors of engineered tissue constructs.
Collapse
Affiliation(s)
- Richard J. McMurtrey
- Institute of Neural Regeneration & Tissue Engineering, Highland, Utah, United States
- Department of Engineering Science, Institute of Biomedical Engineering, University of Oxford, Oxford, United Kingdom
| |
Collapse
|
208
|
Wiest J, Namias A, Pfister C, Wolf P, Demmel F, Brischwein M. Data Processing in Cellular Microphysiometry. IEEE Trans Biomed Eng 2016; 63:2368-2375. [PMID: 26929025 DOI: 10.1109/tbme.2016.2533868] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
GOAL This contribution points out the need for well-defined and documented data processing protocols in microphysiometry, an evolving field of label-free cell assays. The sensitivity of the obtained cell metabolic rates toward different routines of raw data processing is evaluated. METHODS A standard microphysiometric experiment structured in discrete measurement intervals was performed on a platform with a pH- and O 2-sensor readout. It is evaluated using three different data evaluation protocols, based on A) fast Fourier transformation of such dynamics, B) linear regression (LIN) of pH(t) and O2(t) dynamics, and C) numerical simulation (SIM) with a subsequent fitting of dynamics for parameter estimation. RESULTS We propose a sequence of well documented steps for an organized processing of raw sensor data. Figures of merit for the quality of raw data and the performance of data processing are provided. To estimate metabolic rates, a reaction-diffusion modeling approach is recommended if the necessary model input parameters such as the distribution of the active biomass, sensor response time, and material properties are available. CONCLUSION The information about cellular metabolic activity contained by measured sensor data dynamics is superimposed by manifold sources of error. Careful consideration of data processing is necessary to eliminate these errors as much as possible and to avoid an incorrect interpretation of data.
Collapse
|
209
|
Jagannathan L, Cuddapah S, Costa M. Oxidative stress under ambient and physiological oxygen tension in tissue culture. ACTA ACUST UNITED AC 2016; 2:64-72. [PMID: 27034917 DOI: 10.1007/s40495-016-0050-5] [Citation(s) in RCA: 88] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Oxygen (O2) levels range from 2-9% in vivo. However, cell culture experiments are performed at atmospheric O2 levels (21%). Oxidative stress due to generation of reactive oxygen species (ROS) in cells cultured at higher than physiological levels is implicated in multitude of deleterious effects including DNA damage, genomic instability and senescence. In addition, oxidative stress activates redox sensitive transcription factors related to inflammatory signaling and apoptotic signaling. Furthermore, several chromatin-modifying enzymes are affected by ROS, potentially impacting epigenetic regulation of gene expression. While primary cells are cultured at lower O2 levels due to their inability to grow at higher O2, the immortalized cells, which display no such apparent growth difficulties, are typically cultured at 21% O2. This review will provide an overview of issues associated with increased oxygen levels in in vitro cell culture and point out the benefits of using lower levels of oxygen tension even for immortalized cells.
Collapse
Affiliation(s)
- Lakshmanan Jagannathan
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987
| | - Suresh Cuddapah
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, NY 10987
| |
Collapse
|
210
|
Kučera O, Cifra M. Radiofrequency and microwave interactions between biomolecular systems. J Biol Phys 2016; 42:1-8. [PMID: 26174548 PMCID: PMC4713408 DOI: 10.1007/s10867-015-9392-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2015] [Accepted: 06/18/2015] [Indexed: 11/30/2022] Open
Abstract
The knowledge of mechanisms underlying interactions between biological systems, be they biomacromolecules or living cells, is crucial for understanding physiology, as well as for possible prevention, diagnostics and therapy of pathological states. Apart from known chemical and direct contact electrical signaling pathways, electromagnetic phenomena were proposed by some authors to mediate non-chemical interactions on both intracellular and intercellular levels. Here, we discuss perspectives in the research of nanoscale electromagnetic interactions between biosystems on radiofrequency and microwave wavelengths. Based on our analysis, the main perspectives are in (i) the micro and nanoscale characterization of both passive and active radiofrequency properties of biomacromolecules and cells, (ii) experimental determination of viscous damping of biomacromolecule structural vibrations and (iii) detailed analysis of energetic circumstances of electromagnetic interactions between oscillating polar biomacromolecules. Current cutting-edge nanotechnology and computational techniques start to enable such studies so we can expect new interesting insights into electromagnetic aspects of molecular biophysics of cell signaling.
Collapse
Affiliation(s)
- Ondřej Kučera
- Institute of Photonics and Electronics, The Czech Academy of Sciences, Chaberska 57, 182 00, Prague, Czechia
| | - Michal Cifra
- Institute of Photonics and Electronics, The Czech Academy of Sciences, Chaberska 57, 182 00, Prague, Czechia.
| |
Collapse
|
211
|
Rabovsky AB, Buettner GR, Fink B. In vivo imaging of free radicals produced by multivitamin-mineral supplements. BMC Nutr 2015; 1. [PMID: 26705481 PMCID: PMC4687973 DOI: 10.1186/s40795-015-0025-7] [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] [Indexed: 11/16/2022] Open
Abstract
Background Redox active minerals in dietary supplements can catalyze unwanted
and potentially harmful oxidations. Methods To determine if this occurs in vivo we employed electron paramagnetic
(EPR) imaging. We used 1-hydroxy-3-carboxy-2,2,5,5-tetramethylpyrrolidine
(CPH) as a reporter for one-electron oxidations, e.g. free
radical-mediated oxidations; the one-electron oxidation product of CPH,
3-carboxy-2,2,5,5-tetramethyl-1-pyrrolidinyloxy (CP•), is
a nitroxide free radical that is relatively persistent in vivo and
detectable by EPR. As model systems, we used research formulations of
vitamin mineral supplements (RVM) that are typical of commercial
products. Results In in vitro experiments, upon suspension of RVM in aqueous solution,
we observed: (1) the uptake of oxygen in the solution, consistent with
oxidation of the components in the RVM; (2) the ascorbate free radical, a
real-time indicator of ongoing oxidations; and (3) when amino
acid/oligosaccharide (AAOS; glycinate or aspartate with non-digestible
oligofructose) served as the matrix in the RVM, the rate of oxidation was
significantly slowed. In a murine model, EPR imaging showed that the
ingestion of RVM along with CPH results in the one-electron oxidation of CPH
by RVM in the digestive system. The resulting CP•
distributes throughout the body. Inclusion of AAOS in the RVM formulation
diminished the oxidation of CPH to CP• in vivo. Conclusions These data demonstrate that typical formulations of
multivitamin/multimineral dietary supplements can initiate the oxidation of
bystander substances and that AAOS-complexes of essential redox active
metals, e.g. copper and iron, have reduced ability to
catalyze free radical formation and associated detrimental oxidations when a
part of a multivitamin/multimineral formulation.
Collapse
Affiliation(s)
- Alexander B Rabovsky
- Research & Development, Melaleuca Inc, 4609 West 65th South, Idaho Falls, ID 83402, USA
| | - Garry R Buettner
- Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA, USA
| | - Bruno Fink
- Noxygen Science Transfer & Diagnostics GmbH, Elzach, Germany
| |
Collapse
|
212
|
Kaiser SC, Kraume M, Eibl D. Development of the Travelling Wave Bioreactor. Part I: Design Studies Based on Numerical Models. CHEM-ING-TECH 2015. [DOI: 10.1002/cite.201500092] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
213
|
Kagawa Y, Miyahara H, Ota Y, Tsuneda S. System for measuring oxygen consumption rates of mammalian cells in static culture under hypoxic conditions. Biotechnol Prog 2015; 32:189-97. [PMID: 26558344 DOI: 10.1002/btpr.2202] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2015] [Revised: 10/28/2015] [Indexed: 01/30/2023]
Abstract
Estimating the oxygen consumption rates (OCRs) of mammalian cells in hypoxic environments is essential for designing and developing a three-dimensional (3-D) cell culture system. However, OCR measurements under hypoxic conditions are infrequently reported in the literature. Here, we developed a system for measuring OCRs at low oxygen levels. The system injects nitrogen gas into the environment and measures the oxygen concentration by an optical oxygen microsensor that consumes no oxygen. The developed system was applied to HepG2 cells in static culture. Specifically, we measured the spatial profiles of the local dissolved oxygen concentration in the medium, then estimated the OCRs of the cells. The OCRs, and also the pericellular oxygen concentrations, decreased nonlinearly as the oxygen partial pressure in the environment decreased from 19% to 1%. The OCRs also depended on the culture period and the matrix used for coating the dish surface. Using this system, we can precisely estimate the OCRs of various cell types under environments that mimic 3-D culture conditions, contributing crucial data for an efficient 3-D culture system design.
Collapse
Affiliation(s)
- Yuki Kagawa
- Inst. for Nanoscience and Nanotechnology, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku, Tokyo, 162-8480, Japan
| | - Hirotaka Miyahara
- Dept. of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku, Tokyo, 162-8480, Japan
| | - Yuri Ota
- Dept. of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku, Tokyo, 162-8480, Japan
| | - Satoshi Tsuneda
- Inst. for Nanoscience and Nanotechnology, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku, Tokyo, 162-8480, Japan.,Dept. of Life Science and Medical Bioscience, Waseda University, 2-2 Wakamatsu-Cho, Shinjuku, Tokyo, 162-8480, Japan
| |
Collapse
|
214
|
Woldman YY, Eubank TD, Mock AJ, Stevens NC, Varadharaj S, Turco J, Gavrilin MA, Branchini BR, Khramtsov VV. Detection of nitric oxide production in cell cultures by luciferin-luciferase chemiluminescence. Biochem Biophys Res Commun 2015; 465:232-8. [PMID: 26253471 DOI: 10.1016/j.bbrc.2015.08.001] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2015] [Accepted: 08/01/2015] [Indexed: 11/24/2022]
Abstract
A chemiluminescent method is proposed for quantitation of NO generation in cell cultures. The method is based on activation of soluble guanylyl cyclase by NO. The product of the guanylyl cyclase reaction, pyrophosphate, is converted to ATP by ATP sulfurylase and ATP is detected in a luciferin-luciferase system. The method has been applied to the measurement of NO generated by activated murine macrophages (RAW 264.7) and bovine aortic endothelial cells. For macrophages activated by lipopolysaccharide and γ-interferon, the rate of NO production is about 100 amol/(cell·min). The rate was confirmed by the measurements of nitrite, the product of NO oxidation. For endothelial cells, the basal rate of NO generation is 5 amol/(cell·min); the rate approximately doubles upon activation by bradykinin, Ca(2+) ionophore A23187 or mechanical stress. For both types of cells the measured rate of NO generation is strongly affected by inhibitors of NO synthase. The sensitivity of the method is about 50 pM/min, allowing the registration of NO generated by 10(2)-10(4) cells. The enzyme-linked chemiluminescent method is two orders of magnitude more sensitive than fluorescent detection using 4-amino-5-methylamino-2',7'-difluorofluorescein (DAF-FM).
Collapse
Affiliation(s)
- Yakov Y Woldman
- Department of Chemistry, Valdosta State University, Valdosta, GA 31698, USA.
| | - Tim D Eubank
- Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Andrew J Mock
- Department of Biology, Valdosta State University, Valdosta, GA 31698, USA
| | - Natalia C Stevens
- Department of Biology, Valdosta State University, Valdosta, GA 31698, USA
| | - Saradhadevi Varadharaj
- Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Jenifer Turco
- Department of Biology, Valdosta State University, Valdosta, GA 31698, USA
| | - Mikhail A Gavrilin
- Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| | - Bruce R Branchini
- Department of Chemistry, Connecticut College, New London, CT 06320, USA
| | - Valery V Khramtsov
- Davis Heart & Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
215
|
Doskey CM, van ‘t Erve TJ, Wagner BA, Buettner GR. Moles of a Substance per Cell Is a Highly Informative Dosing Metric in Cell Culture. PLoS One 2015; 10:e0132572. [PMID: 26172833 PMCID: PMC4501792 DOI: 10.1371/journal.pone.0132572] [Citation(s) in RCA: 42] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2014] [Accepted: 06/16/2015] [Indexed: 01/24/2023] Open
Abstract
Background The biological consequences upon exposure of cells in culture to a dose of xenobiotic are not only dependent on biological variables, but also the physical aspects of experiments e.g. cell number and media volume. Dependence on physical aspects is often overlooked due to the unrecognized ambiguity in the dominant metric used to express exposure, i.e. initial concentration of xenobiotic delivered to the culture medium over the cells. We hypothesize that for many xenobiotics, specifying dose as moles per cell will reduce this ambiguity. Dose as moles per cell can also provide additional information not easily obtainable with traditional dosing metrics. Methods Here, 1,4-benzoquinone and oligomycin A are used as model compounds to investigate moles per cell as an informative dosing metric. Mechanistic insight into reactions with intracellular molecules, differences between sequential and bolus addition of xenobiotic and the influence of cell volume and protein content on toxicity are also investigated. Results When the dose of 1,4-benzoquinone or oligomycin A was specified as moles per cell, toxicity was independent of the physical conditions used (number of cells, volume of medium). When using moles per cell as a dose-metric, direct quantitative comparisons can be made between biochemical or biological endpoints and the dose of xenobiotic applied. For example, the toxicity of 1,4-benzoquinone correlated inversely with intracellular volume for all five cell lines exposed (C6, MDA-MB231, A549, MIA PaCa-2, and HepG2). Conclusions Moles per cell is a useful and informative dosing metric in cell culture. This dosing metric is a scalable parameter that: can reduce ambiguity between experiments having different physical conditions; provides additional mechanistic information; allows direct comparison between different cells; affords a more uniform platform for experimental design; addresses the important issue of repeatability of experimental results, and could increase the translatability of information gained from in vitro experiments.
Collapse
Affiliation(s)
- Claire M. Doskey
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Thomas J. van ‘t Erve
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Brett A. Wagner
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa, 52242, United States of America
| | - Garry R. Buettner
- Interdisciplinary Graduate Program in Human Toxicology, The University of Iowa, Iowa City, Iowa, 52242, United States of America
- Free Radical and Radiation Biology Program, Department of Radiation Oncology, The University of Iowa, Iowa City, Iowa, 52242, United States of America
- * E-mail:
| |
Collapse
|
216
|
Transcription regulates HIF-1α expression in CD4(+) T cells. Immunol Cell Biol 2015; 94:109-13. [PMID: 26150319 DOI: 10.1038/icb.2015.64] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2015] [Accepted: 06/03/2015] [Indexed: 12/11/2022]
Abstract
The transcription factor hypoxia inducible factor-1α (HIF-1α) mediates the metabolic adaptation of cells to hypoxia and T-helper cell fate. However, HIF-1α regulation in CD4(+) T cells (T cells) remains elusive. Here we observed that depletion of oxygen (O2⩽2%) alone was not sufficient to induce HIF-1α expression in T cells. However, when hypoxic T cells were stimulated, HIF-1α was expressed and this was dependent on nuclear factor-κB- and nuclear factor of activated T cell (NFAT)-mediated transcriptional upregulation of Hif-1α mRNA. HIF-1α upregulation could be blocked by drugs inhibiting NF-κB, NFAT or mammalian target of rapamycin precluding CD4(+) T-cell stimulation or translation in T cells, as well as by blocking transcription. CD3, CD28, phorbol-12-myristat-13-acetat (PMA) or ionomycin-stimulated T cells did not express HIF-1α under normoxic conditions. In conclusion, regulation of HIF-1α expression in CD4(+) T cells in hypoxia gravely relies on its transcriptional upregulation and subsequent enhanced protein stabilization.
Collapse
|
217
|
Chandrasekaran V, Lea C, Sosa JC, Higgins D, Lein PJ. Reactive oxygen species are involved in BMP-induced dendritic growth in cultured rat sympathetic neurons. Mol Cell Neurosci 2015; 67:116-25. [PMID: 26079955 PMCID: PMC4550485 DOI: 10.1016/j.mcn.2015.06.007] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2014] [Revised: 05/26/2015] [Accepted: 06/12/2015] [Indexed: 12/28/2022] Open
Abstract
Previous studies have shown that bone morphogenetic proteins (BMPs) promote dendritic growth in sympathetic neurons; however, the downstream signaling molecules that mediate the dendrite promoting activity of BMPs are not well characterized. Here we test the hypothesis that reactive oxygen species (ROS)-mediated signaling links BMP receptor activation to dendritic growth. In cultured rat sympathetic neurons, exposure to any of the three mechanistically distinct antioxidants, diphenylene iodinium (DPI), nordihydroguaiaretic acid (NGA) or desferroxamine (DFO), blocked de novo BMP-induced dendritic growth. Addition of DPI to cultures previously induced with BMP to extend dendrites caused dendritic retraction while DFO and NGA prevented further growth of dendrites. The inhibition of the dendrite promoting activity of BMPs by antioxidants was concentration-dependent and occurred without altering axonal growth or neuronal cell survival. Antioxidant treatment did not block BMP activation of SMAD 1,5 as determined by nuclear localization of these SMADs. While BMP treatment did not cause a detectable increase in intracellular ROS in cultured sympathetic neurons as assessed using fluorescent indicator dyes, BMP treatment increased the oxygen consumption rate in cultured sympathetic neurons as determined using the Seahorse XF24 Analyzer, suggesting increased mitochondrial activity. In addition, BMPs upregulated expression of NADPH oxidase 2 (NOX2) and either pharmacological inhibition or siRNA knockdown of NOX2 significantly decreased BMP-7 induced dendritic growth. Collectively, these data support the hypothesis that ROS are involved in the downstream signaling events that mediate BMP7-induced dendritic growth in sympathetic neurons, and suggest that ROS-mediated signaling positively modulates dendritic complexity in peripheral neurons.
Collapse
Affiliation(s)
| | - Charlotte Lea
- Department of Biology, Saint Mary's College of California, Moraga, CA, USA
| | - Jose Carlo Sosa
- Department of Biology, Saint Mary's College of California, Moraga, CA, USA
| | - Dennis Higgins
- Department of Pharmacology and Toxicology, University of Buffalo, Buffalo, NY, USA
| | - Pamela J Lein
- Department of Molecular Biosciences, University of California, Davis, CA, USA
| |
Collapse
|
218
|
Du J, Wagner BA, Buettner GR, Cullen JJ. Role of labile iron in the toxicity of pharmacological ascorbate. Free Radic Biol Med 2015; 84:289-295. [PMID: 25857216 PMCID: PMC4739508 DOI: 10.1016/j.freeradbiomed.2015.03.033] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/10/2014] [Revised: 03/25/2015] [Accepted: 03/27/2015] [Indexed: 11/17/2022]
Abstract
Pharmacological ascorbate has been shown to induce toxicity in a wide range of cancer cell lines. Pharmacological ascorbate in animal models has shown promise for use in cancer treatment. At pharmacological concentrations the oxidation of ascorbate produces a high flux of H2O2 via the formation of ascorbate radical (Asc(•-)). The rate of oxidation of ascorbate is principally a function of the level of catalytically active metals. Iron in cell culture media contributes significantly to the rate of H2O2 generation. We hypothesized that increasing intracellular iron would enhance ascorbate-induced cytotoxicity and that iron chelators could modulate the catalytic efficiency with respect to ascorbate oxidation. Treatment of cells with the iron-chelators deferoxamine (DFO) or dipyridyl (DPD) in the presence of 2mM ascorbate decreased the flux of H2O2 generated by pharmacological ascorbate and reversed ascorbate-induced toxicity. Conversely, increasing the level of intracellular iron by preincubating cells with Fe-hydroxyquinoline (HQ) increased ascorbate toxicity and decreased clonogenic survival. These findings indicate that redox metal metals, e.g., Fe(3+)/Fe(2+), have an important role in ascorbate-induced cytotoxicity. Approaches that increase catalytic iron could potentially enhance the cytotoxicity of pharmacological ascorbate in vivo.
Collapse
Affiliation(s)
- Juan Du
- Department of Surgery, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Brett A Wagner
- Department of Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa College of Medicine, Iowa City, IA 52242, USA
| | - Garry R Buettner
- Department of Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa College of Medicine, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Iowa City, IA, USA
| | - Joseph J Cullen
- Department of Surgery, University of Iowa College of Medicine, Iowa City, IA 52242, USA; Department of Radiation Oncology, Free Radical and Radiation Biology Program, University of Iowa College of Medicine, Iowa City, IA 52242, USA; Holden Comprehensive Cancer Center, Iowa City, IA, USA; Veterans Affairs Medical Center, Iowa City, IA, USA.
| |
Collapse
|
219
|
Du J, Cieslak JA, Welsh JL, Sibenaller ZA, Allen BG, Wagner BA, Kalen AL, Doskey CM, Strother RK, Button AM, Mott SL, Smith B, Tsai S, Mezhir J, Goswami PC, Spitz DR, Buettner GR, Cullen JJ. Pharmacological Ascorbate Radiosensitizes Pancreatic Cancer. Cancer Res 2015; 75:3314-26. [PMID: 26081808 DOI: 10.1158/0008-5472.can-14-1707] [Citation(s) in RCA: 82] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2014] [Accepted: 05/20/2015] [Indexed: 02/03/2023]
Abstract
The toxicity of pharmacologic ascorbate is mediated by the generation of H2O2 via the oxidation of ascorbate. Because pancreatic cancer cells are sensitive to H2O2 generated by ascorbate, they would also be expected to become sensitized to agents that increase oxidative damage such as ionizing radiation. The current study demonstrates that pharmacologic ascorbate enhances the cytotoxic effects of ionizing radiation as seen by decreased cell viability and clonogenic survival in all pancreatic cancer cell lines examined, but not in nontumorigenic pancreatic ductal epithelial cells. Ascorbate radiosensitization was associated with an increase in oxidative stress-induced DNA damage, which was reversed by catalase. In mice with established heterotopic and orthotopic pancreatic tumor xenografts, pharmacologic ascorbate combined with ionizing radiation decreased tumor growth and increased survival, without damaging the gastrointestinal tract or increasing systemic changes in parameters indicative of oxidative stress. Our results demonstrate the potential clinical utility of pharmacologic ascorbate as a radiosensitizer in the treatment of pancreatic cancer.
Collapse
Affiliation(s)
- Juan Du
- Department of Surgery, University of Iowa College of Medicine, Iowa City, Iowa
| | - John A Cieslak
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | - Jessemae L Welsh
- Department of Surgery, University of Iowa College of Medicine, Iowa City, Iowa
| | - Zita A Sibenaller
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | - Bryan G Allen
- Department of Surgery, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Brett A Wagner
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | - Amanda L Kalen
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | - Claire M Doskey
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | - Robert K Strother
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa
| | | | - Sarah L Mott
- Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Brian Smith
- Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Susan Tsai
- Medical College of Wisconsin, Milwaukee, Wisconsin
| | - James Mezhir
- Department of Surgery, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Prabhat C Goswami
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Douglas R Spitz
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Garry R Buettner
- Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa
| | - Joseph J Cullen
- Department of Surgery, University of Iowa College of Medicine, Iowa City, Iowa. Department of Radiation Oncology, University of Iowa College of Medicine, Iowa City, Iowa. Holden Comprehensive Cancer Center, Iowa City, Iowa. Veterans Affairs Medical Center, Iowa City, Iowa.
| |
Collapse
|
220
|
Kagawa Y, Matsuura K, Shimizu T, Tsuneda S. Direct measurement of local dissolved oxygen concentration spatial profiles in a cell culture environment. Biotechnol Bioeng 2015; 112:1263-74. [PMID: 25565074 DOI: 10.1002/bit.25531] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2014] [Revised: 12/18/2014] [Accepted: 12/23/2014] [Indexed: 02/02/2023]
Abstract
Controlling local dissolved oxygen concentration (DO) in media is critical for cell or tissue cultures. Various biomaterials and culture methods have been developed to modulate DO. Direct measurement of local DO in cultures has not been validated as a method to test DO modulation. In the present study we developed a DO measurement system equipped with a Clark-type oxygen microelectrode manipulated with 1 μm precision in three-dimensional space to explore potential applications for tissue engineering. By determining the microelectrode tip position precisely against the bottom plane of culture dishes with rat or human cardiac cells in static monolayer culture, we successfully obtained spatial distributions of DO in the medium. Theoretical quantitative predictions fit the obtained data well. Based on analyses of the variance between samples, we found the data reflected "local" oxygen consumption in the vicinity of the microelectrode and the detection of temporal changes in oxygen consumption rates of cultured cells was limited by the diffusion rate of oxygen in the medium. This oxygen measuring system monitors local oxygen consumption and production with high spatial resolution, and can potentially be used with recently developed oxygen modulating biomaterials to design microenvironments and non-invasively monitor local DO dynamics during culture.
Collapse
Affiliation(s)
- Yuki Kagawa
- Institute for Nanoscience and Nanotechnology, Waseda University, 2-2 Wakamatsu-cho, Shinjuku, Tokyo 162-8480, Japan
| | | | | | | |
Collapse
|
221
|
Filomeni G, De Zio D, Cecconi F. Oxidative stress and autophagy: the clash between damage and metabolic needs. Cell Death Differ 2015; 22:377-88. [PMID: 25257172 PMCID: PMC4326572 DOI: 10.1038/cdd.2014.150] [Citation(s) in RCA: 1549] [Impact Index Per Article: 154.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2014] [Revised: 08/19/2014] [Accepted: 08/21/2014] [Indexed: 12/19/2022] Open
Abstract
Autophagy is a catabolic process aimed at recycling cellular components and damaged organelles in response to diverse conditions of stress, such as nutrient deprivation, viral infection and genotoxic stress. A growing amount of evidence in recent years argues for oxidative stress acting as the converging point of these stimuli, with reactive oxygen species (ROS) and reactive nitrogen species (RNS) being among the main intracellular signal transducers sustaining autophagy. This review aims at providing novel insight into the regulatory pathways of autophagy in response to glucose and amino acid deprivation, as well as their tight interconnection with metabolic networks and redox homeostasis. The role of oxidative and nitrosative stress in autophagy is also discussed in the light of its being harmful for both cellular biomolecules and signal mediator through reversible posttranslational modifications of thiol-containing proteins. The redox-independent relationship between autophagy and antioxidant response, occurring through the p62/Keap1/Nrf2 pathway, is also addressed in order to provide a wide perspective upon the interconnection between autophagy and oxidative stress. Herein, we also attempt to afford an overview of the complex crosstalk between autophagy and DNA damage response (DDR), focusing on the main pathways activated upon ROS and RNS overproduction. Along these lines, the direct and indirect role of autophagy in DDR is dissected in depth.
Collapse
Affiliation(s)
- G Filomeni
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
- IRCCS Fondazione Santa Lucia and Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
| | - D De Zio
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
- IRCCS Fondazione Santa Lucia and Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
| | - F Cecconi
- Cell Stress and Survival Unit, Danish Cancer Society Research Center, Copenhagen, Denmark
- IRCCS Fondazione Santa Lucia and Department of Biology, University of Rome ‘Tor Vergata', Rome, Italy
| |
Collapse
|
222
|
Ren X, Tapias LF, Jank BJ, Mathisen DJ, Lanuti M, Ott HC. Ex vivo non-invasive assessment of cell viability and proliferation in bio-engineered whole organ constructs. Biomaterials 2015; 52:103-12. [PMID: 25818417 DOI: 10.1016/j.biomaterials.2015.01.061] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2014] [Revised: 12/31/2014] [Accepted: 01/20/2015] [Indexed: 12/21/2022]
Abstract
Decellularized organ scaffolds allow whole organ regeneration and study of cell behavior in three-dimensional culture conditions. Cell viability within the bio-engineered organ constructs is an essential parameter reflecting the performance of participating cells during long-term ex vivo culture, and is a prerequisite for further functional performance. Resazurin-based redox metabolic assays have been used to monitor cell viability in both two- and three-dimensional cell cultures. Here we developed a method for monitoring cell viability and proliferation in bio-engineered organ constructs using a resazurin perfusion assay. This method allows non-invasive, repetitive and rapid estimation of viable cell numbers during long-term ex vivo culture. As a proof-of-principle, we assessed the performance of two different endothelial sources and the impact of different perfusion programs on endothelial viability after re-endothelialization of decellularized lung scaffolds. The resazurin-based perfusion assay revealed changes in endothelial viability and proliferation during long-term ex vivo culture, which was consistent with histological assessment at different time points. Finally, we showed that this method could be used for assessment of proliferation and cytotoxicity after pharmacological treatment on a three-dimensional non-small cell lung cancer culture model.
Collapse
Affiliation(s)
- Xi Ren
- Center for Regenerative Medicine, Massachusetts General Hospital, United States; Harvard Medical School, United States
| | - Luis F Tapias
- Harvard Medical School, United States; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States
| | - Bernhard J Jank
- Center for Regenerative Medicine, Massachusetts General Hospital, United States; Harvard Medical School, United States
| | - Douglas J Mathisen
- Harvard Medical School, United States; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States
| | - Michael Lanuti
- Harvard Medical School, United States; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States
| | - Harald C Ott
- Center for Regenerative Medicine, Massachusetts General Hospital, United States; Harvard Medical School, United States; Division of Thoracic Surgery, Department of Surgery, Massachusetts General Hospital, United States; Harvard Stem Cell Institute, United States.
| |
Collapse
|
223
|
Buettner GR. Moving free radical and redox biology ahead in the next decade(s). Free Radic Biol Med 2015; 78:236-8. [PMID: 25450329 PMCID: PMC4331124 DOI: 10.1016/j.freeradbiomed.2014.10.578] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Accepted: 10/27/2014] [Indexed: 10/24/2022]
Affiliation(s)
- Garry R Buettner
- Free Radical and Radiation Biology, Department of Radiation Oncology and ESR Facility, College of Medicine, The University of Iowa, Iowa City, IA52242-1181, USA.
| |
Collapse
|
224
|
Son JH, Cho YC, Sung IY, Kim IR, Park BS, Kim YD. Melatonin promotes osteoblast differentiation and mineralization of MC3T3-E1 cells under hypoxic conditions through activation of PKD/p38 pathways. J Pineal Res 2014; 57:385-92. [PMID: 25250639 DOI: 10.1111/jpi.12177] [Citation(s) in RCA: 55] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/20/2014] [Accepted: 09/12/2014] [Indexed: 11/30/2022]
Abstract
Osteoblastic differentiation and bone-forming capacity are known to be suppressed under hypoxic conditions. Melatonin has been shown to influence cell differentiation. A number of in vitro and in vivo studies have suggested that melatonin also has an anabolic effect on bone, by promoting osteoblastic differentiation. However, the precise mechanisms and the signaling pathways involved in this process, particularly under hypoxic conditions, are unknown. This study investigated whether melatonin could promote osteoblastic differentiation and mineralization of preosteoblastic MC3T3-E1 cells under hypoxic conditions. Additionally, we examined the molecular signaling pathways by which melatonin mediates this process. We found that melatonin is capable of promoting differentiation and mineralization of MC3T3-E1 cells cultured under hypoxic conditions. Melatonin upregulated ALP activity and mRNA levels of Alp, Osx, Col1, and Ocn in a time- and concentration-dependent manner. Alizarin red S staining showed that the mineralized matrix in hypoxic MC3T3-E1 cells formed in a manner that was dependent on melatonin concentration. Moreover, melatonin stimulated phosphorylation of p38 Mapk and Prkd1 in these MC3T3-E1 cells. We concluded that melatonin promotes osteoblastic differentiation of MC3T3-E1 cells under hypoxic conditions via the p38 Mapk and Prkd1 signaling pathways.
Collapse
Affiliation(s)
- Jang-Ho Son
- Department of Oral and Maxillofacial Surgery, Ulsan University Hospital, College of Medicine, Ulsan University, Ulsan, South Korea
| | | | | | | | | | | |
Collapse
|
225
|
Ozden O, Park SH, Wagner BA, Song HY, Zhu Y, Vassilopoulos A, Jung B, Buettner GR, Gius D. SIRT3 deacetylates and increases pyruvate dehydrogenase activity in cancer cells. Free Radic Biol Med 2014; 76:163-172. [PMID: 25152236 PMCID: PMC4364304 DOI: 10.1016/j.freeradbiomed.2014.08.001] [Citation(s) in RCA: 148] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/13/2013] [Revised: 07/10/2014] [Accepted: 08/05/2014] [Indexed: 12/15/2022]
Abstract
Pyruvate dehydrogenase E1α (PDHA1) is the first component enzyme of the pyruvate dehydrogenase (PDH) complex that transforms pyruvate, via pyruvate decarboxylation, into acetyl-CoA that is subsequently used by both the citric acid cycle and oxidative phosphorylation to generate ATP. As such, PDH links glycolysis and oxidative phosphorylation in normal as well as cancer cells. Herein we report that SIRT3 interacts with PDHA1 and directs its enzymatic activity via changes in protein acetylation. SIRT3 deacetylates PDHA1 lysine 321 (K321), and a PDHA1 mutant mimicking a deacetylated lysine (PDHA1(K321R)) increases PDH activity, compared to the K321 acetylation mimic (PDHA1(K321Q)) or wild-type PDHA1. Finally, PDHA1(K321Q) exhibited a more transformed in vitro cellular phenotype compared to PDHA1(K321R). These results suggest that the acetylation of PDHA1 provides another layer of enzymatic regulation, in addition to phosphorylation, involving a reversible acetyllysine, suggesting that the acetylome, as well as the kinome, links glycolysis to respiration.
Collapse
Affiliation(s)
- Ozkan Ozden
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Seong-Hoon Park
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Brett A. Wagner
- Department of Radiation Oncology, Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA 52242
| | - Ha Yong Song
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Yueming Zhu
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Athanassios Vassilopoulos
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
| | - Barbara Jung
- Department of Medicine, Division of Gastroenterology and Hepatology, University of Illinois at Chicago, Chicago, IL 60612
| | - Garry R. Buettner
- Department of Radiation Oncology, Free Radical and Radiation Biology, The University of Iowa, Iowa City, IA 52242
| | - David Gius
- Department of Radiation Oncology, Robert Lurie Cancer Center, Northwestern University Feinberg School of Medicine, Chicago, IL 60611
- Corresponding Author: David Gius, M.D., Ph.D. Professor Department of Radiation Oncology Robert Lurie Cancer Center Northwestern University 303 East Superior, Rm 3-119 Chicago, IL 60611
| |
Collapse
|
226
|
Marsch E, Theelen TL, Demandt JAF, Jeurissen M, van Gink M, Verjans R, Janssen A, Cleutjens JP, Meex SJR, Donners MM, Haenen GR, Schalkwijk CG, Dubois LJ, Lambin P, Mallat Z, Gijbels MJ, Heemskerk JWM, Fisher EA, Biessen EAL, Janssen BJ, Daemen MJAP, Sluimer JC. Reversal of hypoxia in murine atherosclerosis prevents necrotic core expansion by enhancing efferocytosis. Arterioscler Thromb Vasc Biol 2014; 34:2545-53. [PMID: 25256233 DOI: 10.1161/atvbaha.114.304023] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
Abstract
OBJECTIVE Advanced murine and human plaques are hypoxic, but it remains unclear whether plaque hypoxia is causally related to atherogenesis. Here, we test the hypothesis that reversal of hypoxia in atherosclerotic plaques by breathing hyperoxic carbogen gas will prevent atherosclerosis. APPROACH AND RESULTS Low-density lipoprotein receptor-deficient mice (LDLR(-/-)) were fed a Western-type diet, exposed to carbogen (95% O2, 5% CO2) or air, and the effect on plaque hypoxia, size, and phenotype was studied. First, the hypoxic marker pimonidazole was detected in murine LDLR(-/-) plaque macrophages from plaque initiation onwards. Second, the efficacy of breathing carbogen (90 minutes, single exposure) was studied. Compared with air, carbogen increased arterial blood pO2 5-fold in LDLR(-/-) mice and reduced plaque hypoxia in advanced plaques of the aortic root (-32%) and arch (-84%). Finally, the effect of repeated carbogen exposure on progression of atherosclerosis was studied in LDLR(-/-) mice fed a Western-type diet for an initial 4 weeks, followed by 4 weeks of diet and carbogen or air (both 90 min/d). Carbogen reduced plaque hypoxia (-40%), necrotic core size (-37%), and TUNEL(+) (terminal uridine nick-end labeling positive) apoptotic cell content (-50%) and increased efferocytosis of apoptotic cells by cluster of differentiation 107b(+) (CD107b, MAC3) macrophages (+36%) in advanced plaques of the aortic root. Plaque size, plasma cholesterol, hematopoiesis, and systemic inflammation were unchanged. In vitro, hypoxia hampered efferocytosis by bone marrow-derived macrophages, which was dependent on the receptor Mer tyrosine kinase. CONCLUSIONS Carbogen restored murine plaque oxygenation and prevented necrotic core expansion by enhancing efferocytosis, likely via Mer tyrosine kinase. Thus, plaque hypoxia is causally related to necrotic core expansion.
Collapse
Affiliation(s)
- Elke Marsch
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Thomas L Theelen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Jasper A F Demandt
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mike Jeurissen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mathijs van Gink
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Robin Verjans
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Anique Janssen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Jack P Cleutjens
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Steven J R Meex
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Marjo M Donners
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Guido R Haenen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Casper G Schalkwijk
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ludwig J Dubois
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Philippe Lambin
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ziad Mallat
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Marion J Gijbels
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Johan W M Heemskerk
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Edward A Fisher
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Erik A L Biessen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Ben J Janssen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Mat J A P Daemen
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.)
| | - Judith C Sluimer
- From the Department of Pathology, Cardiovascular Research Institute Maastricht (CARIM) (E.M., T.L.T., J.A.F.D., M.J., M.v.G., R.V., A.J., J.P.C., M.M.D., M.J.G., E.A.L.B., J.C.S.), Department of Clinical Chemistry (S.J.R.M.), Department of Toxicology (G.R.H.), Department of Internal Medicine, CARIM (C.G.S.), Department of Radiation Oncology (Maastro Lab), GROW (L.J.D., P.L.), Department of Molecular Genetics, CARIM (M.J.G.), Department of Biochemistry, CARIM (J.W.M.H.), Department of Pharmacology, CARIM (B.J.J.), Maastricht University Medical Centre, Maastricht, The Netherlands; Paris Centre de Recherche Cardiovasculaire (PARCC) Inserm-UMR 970, Paris, France (Z.M.); Department of Medicine, University of Cambridge, Cambridge, United Kingdom (Z.M.); Department of Medical Biochemistry (M.J.G.) and Department of Pathology (M.J.A.P.D.), AMC, Amsterdam, The Netherlands; and Department of Medicine (Cardiology), New York University School of Medicine, New York (E.A.F.).
| |
Collapse
|
227
|
Abeille F, Mittler F, Obeid P, Huet M, Kermarrec F, Dolega ME, Navarro F, Pouteau P, Icard B, Gidrol X, Agache V, Picollet-D'hahan N. Continuous microcarrier-based cell culture in a benchtop microfluidic bioreactor. LAB ON A CHIP 2014; 14:3510-8. [PMID: 25012393 DOI: 10.1039/c4lc00570h] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/04/2023]
Abstract
Microfluidic bioreactors are expected to impact cell therapy and biopharmaceutical production due to their ability to control cellular microenvironments. This work presents a novel approach for continuous cell culture in a microfluidic system. Microcarriers (i.e., microbeads) are used as growth support for anchorage-dependent mammalian cells. This approach eases the manipulation of cells within the system and enables harmless extraction of cells. Moreover, the microbioreactor uses a perfusion function based on the biocompatible integration of a porous membrane to continuously feed the cells. The perfusion rate is optimized through simulations to provide a stable biochemical environment. Thermal management is also addressed to ensure a homogeneous bioreactor temperature. Eventually, incubator-free cell cultures of Drosophila S2 and PC3 cells are achieved over the course of a week using this bioreactor. In future applications, a more efficient alternative to harvesting cells from microcarriers is also anticipated as suggested by our positive results from the microcarrier digestion experiments.
Collapse
Affiliation(s)
- F Abeille
- Univ. Grenoble Alpes, F-38000 Grenoble, France
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
228
|
Browne SM, Daud H, Murphy WG, Al-Rubeai M. Measuring dissolved oxygen to track erythroid differentiation of hematopoietic progenitor cells in culture. J Biotechnol 2014; 187:135-8. [PMID: 25107508 DOI: 10.1016/j.jbiotec.2014.07.433] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Revised: 07/16/2014] [Accepted: 07/18/2014] [Indexed: 10/24/2022]
Abstract
As stem cell technologies move from the developmental to the commercial stage strategies must be developed to monitor culture operations. These will ensure consistency of differentiation programs and maintenance of optimum cell viability during production runs. Due to the sensitivity of stem cells to their environment, and their variability in response to external stimuli, accurate monitoring of in vitro conditions will be crucial for effective large-scale culturing of therapeutic stem cells. Here we describe a simple method to monitor the expansion and maturation of adult human haematopoietic stem/progenitor cells into red blood cells in vitro by measuring the oxygen consumption rate of cultures. Cell cultures followed a characteristic pattern of oxygen consumption that is reflective of in vivo erythroid maturation. This method could be easily developed as an online system to map erythroid differentiation and maturation of cultured cells as effectively as the more time consuming process of flow cytometric analysis of surface marker expression patterns.
Collapse
Affiliation(s)
- Susan M Browne
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - Hasbullah Daud
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland
| | - William G Murphy
- School of Medicine and Medical Science, University College Dublin, Belfield, Dublin 4, Ireland; Health Service Executive, Dublin, Ireland
| | - Mohamed Al-Rubeai
- School of Chemical and Bioprocess Engineering, University College Dublin, Belfield, Dublin 4, Ireland.
| |
Collapse
|
229
|
Speranza G, Della Volpe C, Catapano G. Surface wettability of model microporous membranes enhances rat liver cell functions in sub-confluent adherent culture in a continuous-flow recycle bioreactor depending on the ammonia concentration challenge. J Memb Sci 2014. [DOI: 10.1016/j.memsci.2014.04.007] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023]
|
230
|
Abstract
SIGNIFICANCE Most solid tumors contain regions of low oxygenation or hypoxia. Tumor hypoxia has been associated with a poor clinical outcome and plays a critical role in tumor radioresistance. RECENT ADVANCES Two main types of hypoxia exist in the tumor microenvironment: chronic and cycling hypoxia. Chronic hypoxia results from the limited diffusion distance of oxygen, and cycling hypoxia primarily results from the variation in microvessel red blood cell flux and temporary disturbances in perfusion. Chronic hypoxia may cause either tumor progression or regressive effects depending on the tumor model. However, there is a general trend toward the development of a more aggressive phenotype after cycling hypoxia. With advanced hypoxia imaging techniques, spatiotemporal characteristics of tumor hypoxia and the changes to the tumor microenvironment can be analyzed. CRITICAL ISSUES In this review, we focus on the biological and clinical consequences of chronic and cycling hypoxia on radiation treatment. We also discuss the advanced non-invasive imaging techniques that have been developed to detect and monitor tumor hypoxia in preclinical and clinical studies. FUTURE DIRECTIONS A better understanding of the mechanisms of tumor hypoxia with non-invasive imaging will provide a basis for improved radiation therapeutic practices.
Collapse
Affiliation(s)
- Chen-Ting Lee
- 1 Department of Radiation Oncology, Duke University Medical Center , Durham, North Carolina
| | | | | |
Collapse
|
231
|
Quantitative redox biology: an approach to understand the role of reactive species in defining the cellular redox environment. Cell Biochem Biophys 2014; 67:477-83. [PMID: 22161621 DOI: 10.1007/s12013-011-9320-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Systems biology is now recognized as a needed approach to understand the dynamics of inter- and intra-cellular processes. Redox processes are at the foundation of nearly all aspects of biology. Free radicals, related oxidants, and antioxidants are central to the basic functioning of cells and tissues. They set the cellular redox environment and, therefore, are the key to regulation of biochemical pathways and networks, thereby influencing organism health. To understand how short-lived, quasi-stable species, such as superoxide, hydrogen peroxide, and nitric oxide, connect to the metabolome, proteome, lipidome, and genome we need absolute quantitative information on all redox active compounds as well as thermodynamic and kinetic information on their reactions, i.e., knowledge of the complete redoxome. Central to the state of the redoxome are the interactive details of the superoxide/peroxide formation and removal systems. Quantitative information is essential to establish the dynamic mathematical models needed to reveal the temporal evolution of biochemical pathways and networks. This new field of Quantitative Redox Biology will allow researchers to identify new targets for intervention to advance our efforts to achieve optimal human health.
Collapse
|
232
|
Ebrahimkhani MR, Neiman JAS, Raredon MSB, Hughes DJ, Griffith LG. Bioreactor technologies to support liver function in vitro. Adv Drug Deliv Rev 2014; 69-70:132-57. [PMID: 24607703 PMCID: PMC4144187 DOI: 10.1016/j.addr.2014.02.011] [Citation(s) in RCA: 83] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2014] [Revised: 02/18/2014] [Accepted: 02/24/2014] [Indexed: 02/08/2023]
Abstract
Liver is a central nexus integrating metabolic and immunologic homeostasis in the human body, and the direct or indirect target of most molecular therapeutics. A wide spectrum of therapeutic and technological needs drives efforts to capture liver physiology and pathophysiology in vitro, ranging from prediction of metabolism and toxicity of small molecule drugs, to understanding off-target effects of proteins, nucleic acid therapies, and targeted therapeutics, to serving as disease models for drug development. Here we provide perspective on the evolving landscape of bioreactor-based models to meet old and new challenges in drug discovery and development, emphasizing design challenges in maintaining long-term liver-specific function and how emerging technologies in biomaterials and microdevices are providing new experimental models.
Collapse
Affiliation(s)
- Mohammad R Ebrahimkhani
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Jaclyn A Shepard Neiman
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | - Micha Sam B Raredon
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Department of Materials Science and Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA
| | | | - Linda G Griffith
- Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA; Center for Gynepathology Research, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| |
Collapse
|
233
|
Sanders PN, Koval OM, Jaffer OA, Prasad AM, Businga TR, Scott JA, Hayden PJ, Luczak ED, Dickey DD, Allamargot C, Olivier AK, Meyerholz DK, Robison AJ, Winder DG, Blackwell TS, Dworski R, Sammut D, Wagner BA, Buettner GR, Pope RM, Miller FJ, Dibbern ME, Haitchi HM, Mohler PJ, Howarth PH, Zabner J, Kline JN, Grumbach IM, Anderson ME. CaMKII is essential for the proasthmatic effects of oxidation. Sci Transl Med 2014; 5:195ra97. [PMID: 23884469 DOI: 10.1126/scitranslmed.3006135] [Citation(s) in RCA: 51] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Increased reactive oxygen species (ROS) contribute to asthma, but little is known about the molecular mechanisms connecting increased ROS with characteristic features of asthma. We show that enhanced oxidative activation of the Ca(2+)/calmodulin-dependent protein kinase (ox-CaMKII) in bronchial epithelium positively correlates with asthma severity and that epithelial ox-CaMKII increases in response to inhaled allergens in patients. We used mouse models of allergic airway disease induced by ovalbumin (OVA) or Aspergillus fumigatus (Asp) and found that bronchial epithelial ox-CaMKII was required to increase a ROS- and picrotoxin-sensitive Cl(-) current (ICl) and MUC5AC expression, upstream events in asthma progression. Allergen challenge increased epithelial ROS by activating NADPH oxidases. Mice lacking functional NADPH oxidases due to knockout of p47 and mice with epithelial-targeted transgenic expression of a CaMKII inhibitory peptide or wild-type mice treated with inhaled KN-93, an experimental small-molecule CaMKII antagonist, were protected against increases in ICl, MUC5AC expression, and airway hyperreactivity to inhaled methacholine. Our findings support the view that CaMKII is a ROS-responsive, pluripotent proasthmatic signal and provide proof-of-concept evidence that CaMKII is a therapeutic target in asthma.
Collapse
Affiliation(s)
- Philip N Sanders
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
234
|
Moreno-Sánchez R, Marín-Hernández A, Saavedra E, Pardo JP, Ralph SJ, Rodríguez-Enríquez S. Who controls the ATP supply in cancer cells? Biochemistry lessons to understand cancer energy metabolism. Int J Biochem Cell Biol 2014; 50:10-23. [PMID: 24513530 DOI: 10.1016/j.biocel.2014.01.025] [Citation(s) in RCA: 150] [Impact Index Per Article: 13.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 01/21/2014] [Accepted: 01/26/2014] [Indexed: 11/17/2022]
Abstract
Applying basic biochemical principles, this review analyzes data that contrasts with the Warburg hypothesis that glycolysis is the exclusive ATP provider in cancer cells. Although disregarded for many years, there is increasing experimental evidence demonstrating that oxidative phosphorylation (OxPhos) makes a significant contribution to ATP supply in many cancer cell types and under a variety of conditions. Substrates oxidized by normal mitochondria such as amino acids and fatty acids are also avidly consumed by cancer cells. In this regard, the proposal that cancer cells metabolize glutamine for anabolic purposes without the need for a functional respiratory chain and OxPhos is analyzed considering thermodynamic and kinetic aspects for the reductive carboxylation of 2-oxoglutarate catalyzed by isocitrate dehydrogenase. In addition, metabolic control analysis (MCA) studies applied to energy metabolism of cancer cells are reevaluated. Regardless of the experimental/environmental conditions and the rate of lactate production, the flux-control of cancer glycolysis is robust in the sense that it involves the same steps: glucose transport, hexokinase, hexosephosphate isomerase and glycogen degradation, all at the beginning of the pathway; these steps together with phosphofructokinase 1 also control glycolysis in normal cells. The respiratory chain complexes exert significantly higher flux-control on OxPhos in cancer cells than in normal cells. Thus, determination of the contribution of each pathway to ATP supply and/or the flux-control distribution of both pathways in cancer cells is necessary in order to identify differences from normal cells which may lead to the design of rational alternative therapies that selectively target cancer energy metabolism.
Collapse
Affiliation(s)
- Rafael Moreno-Sánchez
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Tlalpan, México D.F., Mexico.
| | - Alvaro Marín-Hernández
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Tlalpan, México D.F., Mexico
| | - Emma Saavedra
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Tlalpan, México D.F., Mexico
| | - Juan P Pardo
- Universidad Nacional Autónoma de México, Facultad de Medicina, Departamento de Bioquímica, México D.F., Mexico
| | - Stephen J Ralph
- School of Medical Sciences, Griffith University, Gold Coast Campus, Qld, Australia
| | - Sara Rodríguez-Enríquez
- Instituto Nacional de Cardiología, Departamento de Bioquímica, Tlalpan, México D.F., Mexico; Instituto Nacional de Cancerología, Laboratorio de Medicina Translacional, Tlalpan, México D.F., Mexico
| |
Collapse
|
235
|
Vozzi F, Bianchi F, Ahluwalia A, Domenici C. Hydrostatic pressure and shear stress affect endothelin-1 and nitric oxide release by endothelial cells in bioreactors. Biotechnol J 2013; 9:146-54. [PMID: 23959971 DOI: 10.1002/biot.201300016] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2013] [Revised: 06/27/2013] [Accepted: 08/16/2013] [Indexed: 02/04/2023]
Abstract
Abundant experimental evidence demonstrates that endothelial cells are sensitive to flow; however, the effect of fluid pressure or pressure gradients that are used to drive viscous flow is not well understood. There are two principal physical forces exerted on the blood vessel wall by the passage of intra-luminal blood: pressure and shear. To analyze the effects of pressure and shear independently, these two stresses were applied to cultured cells in two different types of bioreactors: a pressure-controlled bioreactor and a laminar flow bioreactor, in which controlled levels of pressure or shear stress, respectively, can be generated. Using these bioreactor systems, endothelin-1 (ET-1) and nitric oxide (NO) release from human umbilical vein endothelial cells were measured under various shear stress and pressure conditions. Compared to the controls, a decrease of ET-1 production by the cells cultured in both bioreactors was observed, whereas NO synthesis was up-regulated in cells under shear stress, but was not modulated by hydrostatic pressure. These results show that the two hemodynamic forces acting on blood vessels affect endothelial cell function in different ways, and that both should be considered when planning in vitro experiments in the presence of flow. Understanding the individual and synergic effects of the two forces could provide important insights into physiological and pathological processes involved in vascular remodeling and adaptation.
Collapse
Affiliation(s)
- Federico Vozzi
- Biomimetic Materials and Tissue Engineering Laboratory, C.N.R. Institute of Clinical Physiology, Pisa, Italy
| | | | | | | |
Collapse
|
236
|
Sobotta MC, Barata AG, Schmidt U, Mueller S, Millonig G, Dick TP. Exposing cells to H2O2: a quantitative comparison between continuous low-dose and one-time high-dose treatments. Free Radic Biol Med 2013; 60:325-35. [PMID: 23485584 DOI: 10.1016/j.freeradbiomed.2013.02.017] [Citation(s) in RCA: 81] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2012] [Revised: 01/07/2013] [Accepted: 02/15/2013] [Indexed: 01/01/2023]
Abstract
Most studies investigating the influence of H2O2 on cells in culture apply nonphysiological concentrations over nonphysiological time periods (i.e., a one-time bolus that is metabolized in minutes). As an alternative, the glucose oxidase/catalase (GOX/CAT) system allows application of physiologically relevant H2O2 concentrations (300nM-10µM) over physiologically relevant time periods (up to 24h). Recent findings suggest that bolus and GOX/CAT treatments can lead to opposing cellular responses, thus warranting a quantitative comparison between the two approaches. First, we established a reaction-diffusion model that can predict the behavior of the GOX/CAT system with spatiotemporal resolution, thus aiding selection of optimal experimental conditions for its application. Measurements of H2O2 concentration in the cellular supernatant with the luminol/hypochlorite system were consistent with the predictions of the model. Second, we compared the impact of bolus and GOX/CAT treatments on cytosolic H2O2 levels over time. Intracellular H2O2 was monitored by the response of the thiol peroxidase Prx2 and the H2O2 sensor roGFP2-Orp1. We found that Prx2 rapidly and reversibly responds to submicromolar H2O2 levels and accurately reflects kinetic competition with cellular catalase. Our measurements reveal fundamental differences in the dynamic response of cellular H2O2 concentrations following either bolus or GOX/CAT treatments. Thus, different, or even opposing, biological outcomes from differing means of H2O2 delivery may be expected. Cellular responses induced by bolus treatment may not occur under GOX/CAT conditions, and vice versa.
Collapse
Affiliation(s)
- Mirko C Sobotta
- Division of Redox Regulation, German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, 69120 Heidelberg, Germany
| | | | | | | | | | | |
Collapse
|
237
|
Howard MA, Asmis R, Evans KK, Mustoe TA. Oxygen and wound care: a review of current therapeutic modalities and future direction. Wound Repair Regen 2013; 21:503-11. [PMID: 23756299 DOI: 10.1111/wrr.12069] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/25/2012] [Accepted: 03/18/2013] [Indexed: 11/29/2022]
Abstract
While the importance of oxygen to the wound healing process is well accepted, research and technological advances continue in this field and efforts are ongoing to further utilize oxygen as a therapeutic modality. In this paper, the authors briefly review the role of oxygen in wound healing and discuss the distinct mechanism of action as well as the advantages and disadvantages of the three major oxygen-based therapies currently in clinical use (Hyperbaric Oxygen and Topical Oxygen and Continuous Diffusion of Oxygen), as well as review the existing literature regarding these distinct therapeutic modalities.
Collapse
Affiliation(s)
- Michael A Howard
- Division of Plastic Surgery, North Shore University HealthSystem, University of Chicago Pritzker School of Medicine, Chicago, IL 60062, USA.
| | | | | | | |
Collapse
|
238
|
Meyer W, Kacza J, Hornickel IN, Schoennagel B. Immunolocalization of succinate dehydrogenase in the esophagus epithelium of domesticated mammals. Eur J Histochem 2013; 57:e18. [PMID: 23807297 PMCID: PMC3794344 DOI: 10.4081/ejh.2013.e18] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2013] [Revised: 04/30/2013] [Accepted: 04/15/2013] [Indexed: 11/23/2022] Open
Abstract
Using immunohistochemistry and transmission electron microscopy (TEM), the esophagus epithelia of seven domesticated mammals (horse, cattle, goat, pig, dog, laboratory rat, cat) of three nutrition groups (herbivorous, omnivorous, carnivorous) were studied to get first information about energy generation, as demonstrated by succinate dehydrogenase (SDH) activities. Distinct reaction intensities could be observed in all esophageal cell layers of the different species studied reflecting moderate to strong metabolic activities. The generally strong staining in the stratum basale indicated that new cells are continuously produced. The latter feature was confirmed by a thick, and in the horse generally highly active stratum spinosum. Only in the pig, reaction intensity variations occurred, obviously related to differences in physical feed quality or restricted feed allocation. The immunohistochemical results were corroborated by the presence of intact mitochondria in the esophageal cells of all species and nutrition types studied, except for the horse. Possible relationships between SDH reaction intensities and feed structure, mass or consistency are discussed.
Collapse
Affiliation(s)
- W Meyer
- Institute for Anatomy, University of Veterinary Medicine Hannover Foundation, 30173 Hannover, Germany.
| | | | | | | |
Collapse
|
239
|
Danhier P, Copetti T, De Preter G, Leveque P, Feron O, Jordan BF, Sonveaux P, Gallez B. Influence of cell detachment on the respiration rate of tumor and endothelial cells. PLoS One 2013; 8:e53324. [PMID: 23382841 PMCID: PMC3559693 DOI: 10.1371/journal.pone.0053324] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2012] [Accepted: 11/30/2012] [Indexed: 01/30/2023] Open
Abstract
Cell detachment is a procedure routinely performed in cell culture and a necessary step in many biochemical assays including the determination of oxygen consumption rates (OCR) in vitro. In vivo, cell detachment has been shown to exert profound metabolic influences notably in cancer but also in other pathologies, such as retinal detachment for example. In the present study, we developed and validated a new technique combining electron paramagnetic resonance (EPR) oximetry and the use of cytodex 1 and collagen-coated cytodex 3 dextran microbeads, which allowed the unprecedented comparison of the OCR of adherent and detached cells with high sensitivity. Hence, we demonstrated that both B16F10 melanoma cells and human umbilical vein endothelial cells (HUVEC) experience strong OCR decrease upon trypsin or collagenase treatments. The reduction of cell oxygen consumption was more pronounced with a trypsin compared to a collagenase treatment. Cells remaining in suspension also encounter a marked intracellular ATP depletion and an increase in the lactate production/glucose uptake ratio. These findings highlight the important influence exerted by cell adhesion/detachment on cell respiration, which can be probed with the unprecedented experimental assay that was developed and validated in this study.
Collapse
Affiliation(s)
- Pierre Danhier
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Tamara Copetti
- Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Géraldine De Preter
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Philippe Leveque
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Olivier Feron
- Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Bénédicte F. Jordan
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université catholique de Louvain (UCL), Brussels, Belgium
| | - Pierre Sonveaux
- Institut de Recherche Expérimentale et Clinique (IREC), Université catholique de Louvain (UCL), Brussels, Belgium
| | - Bernard Gallez
- Louvain Drug Research Institute, Biomedical Magnetic Resonance Research Group, Université catholique de Louvain (UCL), Brussels, Belgium
- * E-mail:
| |
Collapse
|
240
|
Olney KE, Du J, van 't Erve TJ, Witmer JR, Sibenaller ZA, Wagner BA, Buettner GR, Cullen JJ. Inhibitors of hydroperoxide metabolism enhance ascorbate-induced cytotoxicity. Free Radic Res 2013. [PMID: 23205739 DOI: 10.3109/10715762.2012.755263] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Pharmacological ascorbate, via its oxidation, has been proposed as a pro-drug for the delivery of H(2)O(2) to tumors. Pharmacological ascorbate decreases clonogenic survival of pancreatic cancer cells, which can be reversed by treatment with scavengers of H(2)O(2). The goal of this study was to determine if inhibitors of intracellular hydroperoxide detoxification could enhance the cytotoxic effects of ascorbate. Human pancreatic cancer cells were treated with ascorbate alone or in combination with inhibitors of hydroperoxide removal including the glutathione disulfide reductase inhibitor 1,3 bis (2-chloroethyl)-1-nitrosurea (BCNU), siRNA targeted to glutathione disulfide reductase (siGR), and 2-deoxy-D-glucose (2DG), which inhibits glucose metabolism. Changes in the intracellular concentration of H(2)O(2) were determined by analysis of the rate of aminotriazole-mediated inactivation of endogenous catalase activity. Pharmacological ascorbate increased intracellular H(2)O(2) and depleted intracellular glutathione. When inhibitors of H(2)O(2) metabolism were combined with pharmacological ascorbate the increase in intracellular H(2)O(2) was amplified and cytotoxicity was enhanced. We conclude that inclusion of agents that inhibit cellular peroxide removal produced by pharmacological ascorbate leads to changes in the intracellular redox state resulting in enhanced cytotoxicity.
Collapse
Affiliation(s)
- K E Olney
- Department of Radiation Oncology, University of Iowa , Iowa City, IA 52242, USA
| | | | | | | | | | | | | | | |
Collapse
|
241
|
Wagner BA, Witmer JR, van 't Erve TJ, Buettner GR. An Assay for the Rate of Removal of Extracellular Hydrogen Peroxide by Cells. Redox Biol 2013; 1:210-217. [PMID: 23936757 PMCID: PMC3736862 DOI: 10.1016/j.redox.2013.01.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Cells have a wide range of capacities to remove extracellular hydrogen peroxide. At higher concentrations of extracellular H2O2 (micromolar) the rate of removal can be approximated by a rate equation that is first-order in the concentration of H2O2 and cell density. Here we present a method to determine the observed rate constant for the removal of extracellular H2O2 on a per cell basis. In the cells examined, when exposed to 20 μM H2O2, these rate constants (kcell) range from 0.46×10−12 s−1 cell−1 L for Mia-PaCa-2 cells (human pancreatic carcinoma) to 10.4×10−12 s−1 cell−1 L for U937 cells (human histiocytic lymphoma). For the relatively small red blood cell kcell=2.9×10−12 s−1 cell−1 L. These rate constants, kcell, can be used to compare the capacity of cells to remove higher levels of extracellular H2O2, as often presented in cell culture experiments. They also provide a means to estimate the rate of removal of extracellular H2O2, rate=−kcell [H2O2] (cells L−1), and the half-life of a bolus of H2O2. This information is essential to optimize experimental design and interpret data from experiments that expose cells to extracellular H2O2.
Collapse
Affiliation(s)
- Brett A Wagner
- The University of Iowa, Free Radical and Radiation Biology Program & ESR Facility, Iowa City, IA 52242-1181, USA
| | | | | | | |
Collapse
|
242
|
Sarsour EH, Kalen AL, Xiao Z, Veenstra TD, Chaudhuri L, Venkataraman S, Reigan P, Buettner GR, Goswami PC. Manganese superoxide dismutase regulates a metabolic switch during the mammalian cell cycle. Cancer Res 2012; 72:3807-16. [PMID: 22710435 PMCID: PMC3429130 DOI: 10.1158/0008-5472.can-11-1063] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Proliferating cells consume more glucose to cope with the bioenergetics and biosynthetic demands of rapidly dividing cells as well as to counter a shift in cellular redox environment. This study investigates the hypothesis that manganese superoxide dismutase (MnSOD) regulates cellular redox flux and glucose consumption during the cell cycle. A direct correlation was observed between glucose consumption and percentage of S-phase cells in MnSOD wild-type fibroblasts, which was absent in MnSOD homozygous knockout fibroblasts. Results from electron paramagnetic resonance spectroscopy and flow cytometric assays showed a significant increase in cellular superoxide levels in S-phase cells, which was associated with an increase in glucose and oxygen consumption, and a decrease in MnSOD activity. Mass spectrometry results showed a complex pattern of MnSOD-methylation at both lysine (68, 89, 122, and 202) and arginine (197 and 216) residues. MnSOD protein carrying a K89A mutation had significantly lower activity compared with wild-type MnSOD. Computational-based simulations indicate that lysine and arginine methylation of MnSOD during quiescence would allow greater accessibility to the enzyme active site as well as increase the positive electrostatic potential around and within the active site. Methylation-dependent changes in the MnSOD conformation and subsequent changes in the electrostatic potential around the active site during quiescence versus proliferation could increase the accessibility of superoxide, a negatively charged substrate. These results support the hypothesis that MnSOD regulates a "metabolic switch" during progression from quiescent through the proliferative cycle. We propose MnSOD as a new molecular player contributing to the Warburg effect.
Collapse
Affiliation(s)
- Ehab H. Sarsour
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Amanda L. Kalen
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Zhen Xiao
- Laboratory of Proteomics and Analytical Technologies, National Cancer Institute, Frederick, Maryland, USA
| | - Timothy D. Veenstra
- Laboratory of Proteomics and Analytical Technologies, National Cancer Institute, Frederick, Maryland, USA
| | - Leena Chaudhuri
- Division of Hematology and Oncology, Mayo Clinic, Scottsdale, Arizona, USA
| | | | - Philip Reigan
- School of Pharmacy, University of Colorado, Denver, USA
| | - Garry R. Buettner
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| | - Prabhat C. Goswami
- Free Radical and Radiation Biology Division, Department of Radiation Oncology, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
243
|
van de Linde S, Heilemann M, Sauer M. Live-cell super-resolution imaging with synthetic fluorophores. Annu Rev Phys Chem 2012; 63:519-40. [PMID: 22404589 DOI: 10.1146/annurev-physchem-032811-112012] [Citation(s) in RCA: 208] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Super-resolution imaging methods now can provide spatial resolution that is well below the diffraction limit approaching virtually molecular resolution. They can be applied to biological samples and provide new and exciting views on the structural organization of cells and the dynamics of biomolecular assemblies on wide timescales. These revolutionary developments come with novel requirements for fluorescent probes, labeling techniques, and data interpretation strategies. Synthetic fluorophores have a small size, are available in many colors spanning the whole spectrum, and can easily be chemically modified and used for stoichiometric labeling of proteins in live cells. Because of their brightness, their photostability, and their ability to be operated as photoswitchable fluorophores even in living cells under physiological conditions, synthetic fluorophores have the potential to substantially accelerate the broad application of live-cell super-resolution imaging methods.
Collapse
Affiliation(s)
- Sebastian van de Linde
- Department of Biotechnology and Biophysics, Julius-Maximilians-University Würzburg, Germany
| | | | | |
Collapse
|
244
|
Huang SH, Hsu YH, Wu CW, Wu CJ. Light-addressable measurements of cellular oxygen consumption rates in microwell arrays based on phase-based phosphorescence lifetime detection. BIOMICROFLUIDICS 2012; 6:44118. [PMID: 24348889 PMCID: PMC3555697 DOI: 10.1063/1.4772604] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Accepted: 12/04/2012] [Indexed: 05/02/2023]
Abstract
A digital light modulation system that utilizes a modified commercial digital micromirror device (DMD) projector, which is equipped with a UV light-emitting diode as a light modulation source, has been developed to spatially direct excited light toward a microwell array device to detect the oxygen consumption rate (OCR) of single cells via phase-based phosphorescence lifetime detection. The microwell array device is composed of a combination of two components: an array of glass microwells containing Pt(II) octaethylporphine (PtOEP) as the oxygen-sensitive luminescent layer and a microfluidic module with pneumatically actuated glass lids set above the microwells to controllably seal the microwells of interest. By controlling the illumination pattern on the DMD, the modulated excitation light can be spatially projected to only excite the sealed microwell for cellular OCR measurements. The OCR of baby hamster kidney-21 fibroblast cells cultivated on the PtOEP layer within a sealed microwell has been successfully measured at 104 ± 2.96 amol s(-1) cell(-1). Repeatable and consistent measurements indicate that the oxygen measurements did not adversely affect the physiological state of the measured cells. The OCR of the cells exhibited a good linear relationship with the diameter of the microwells, ranging from 400 to 1000 μm and containing approximately 480 to 1200 cells within a microwell. In addition, the OCR variation of single cells in situ infected by Dengue virus with a different multiplicity of infection was also successfully measured in real-time. This proposed platform provides the potential for a wide range of biological applications in cell-based biosensing, toxicology, and drug discovery.
Collapse
Affiliation(s)
- Shih-Hao Huang
- Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, No. 2, Beining Rd., Keelung 202-24, Taiwan ; Center for Marine Bioenvironment and Biotechnology (CMBB), National Taiwan Ocean University, Keelung 202-24, Taiwan
| | - Yu-Hsuan Hsu
- Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, No. 2, Beining Rd., Keelung 202-24, Taiwan
| | - Chih-Wei Wu
- Department of Mechanical and Mechatronic Engineering, National Taiwan Ocean University, No. 2, Beining Rd., Keelung 202-24, Taiwan
| | - Chang-Jer Wu
- Department of Food Science, National Taiwan Ocean University, Keelung 202-24, Taiwan
| |
Collapse
|