Skip to main content
Log in

Imaging oxygen in neural cell and tissue models by means of anionic cell-permeable phosphorescent nanoparticles

  • Research Article
  • Published:
Cellular and Molecular Life Sciences Aims and scope Submit manuscript

Abstract

Cell-permeable phosphorescent probes enable the study of cell and tissue oxygenation, bioenergetics, metabolism, and pathological states such as stroke and hypoxia. A number of such probes have been described in recent years, the majority consisting of cationic small molecule and nanoparticle structures. While these probes continue to advance, adequate staining for the study of certain cell types using live imaging techniques remains elusive; this is particularly true for neural cells. Here we introduce novel probes for the analysis of neural cells and tissues: negatively charged poly(methyl methacrylate-co-methacrylic acid)-based nanoparticles impregnated with a phosphorescent Pt(II)-tetrakis(pentafluorophenyl)porphyrin (PtPFPP) dye (this form is referred to as PA1), and with an additional reference/antennae dye poly(9,9-diheptylfluorene-alt-9,9-di-p-tolyl-9H-fluorene) (this form is referred to as PA2). PA1 and PA2 are internalised by endocytosis, result in efficient staining in primary neurons, astrocytes, and PC12 cells and multi-cellular aggregates, and allow for the monitoring of local O2 levels on a time-resolved fluorescence plate reader and PLIM microscope. PA2 also efficiently stains rat brain slices and permits detailed O2 imaging experiments using both one and two-photon intensity-based modes and PLIM modes. Multiplexed analysis of embryonic rat brain slices reveals age-dependent staining patterns for PA2 and a highly heterogeneous distribution of O2 in tissues, which we relate to the localisation of specific progenitor cell populations. Overall, these anionic probes are useful for sensing O2 levels in various cells and tissues, particularly in neural cells, and facilitate high-resolution imaging of O2 in 3D tissue models.

This is a preview of subscription content, log in via an institution to check access.

Access this article

Price excludes VAT (USA)
Tax calculation will be finalised during checkout.

Instant access to the full article PDF.

Fig. 1
Fig. 2
Fig. 3
Fig. 4
Fig. 5

Similar content being viewed by others

Abbreviations

3D:

Three-dimensional

BLBP:

Brain lipid-binding protein

CPZ:

Chlorpromazine

CTX:

Cholera toxin, subunit B

DIV:

Days in vitro

DMF:

N,N-dimethylformamide

DMSO:

Dimethyl sulfoxide

EIPA:

5-(N-ethyl-N-isopropyl)amiloride

GFP:

Green fluorescent protein

HBSS:

Hanks balanced salt solution

HXT:

Hoechst 33342

iO2 :

Intracellular O2

MβCD:

Methyl-β-cyclodextrin

NP:

Nanoparticles

PA:

Polyacrylate NP

PBS:

Phosphate buffered saline

PDL:

Poly-d-lysine

PDT:

Photodynamic therapy

PLIM:

Phosphorescence lifetime imaging microscopy

PMMA-MA:

Poly(methyl methacrylate-co-methacrylic acid

PFO:

Poly(9,9-diheptylfluorene-alt-9,9-di-p-tolyl-9H-fluorene)

PtPFPP:

Pt(II)-tetrakis(pentafluorophenyl)porphine

ROI:

Region of interest

RT:

Room temperature

TBST:

Tris-buffered saline, tween 20

TCSPC:

Time-correlated single photon counting

TR-F:

Time-resolved fluorescence

References

  1. Kobayashi H, Ogawa M, Alford R, Choyke PL, Urano Y (2009) New strategies for fluorescent probe design in medical diagnostic imaging. Chem Rev 110(5):2620–2640

    Article  Google Scholar 

  2. Schäferling M (2012) The art of fluorescence imaging with chemical sensors. Angew Chem Int Ed 51(15):3532–3554. doi:10.1002/anie.201105459

    Article  Google Scholar 

  3. Algar WR, Prasuhn DE, Stewart MH, Jennings TL, Blanco-Canosa JB, Dawson PE, Medintz IL (2011) The controlled display of biomolecules on nanoparticles: a challenge suited to bioorthogonal chemistry. Bioconjug Chem 22(5):825–858

    Article  CAS  PubMed  Google Scholar 

  4. Canton I, Battaglia G (2012) Endocytosis at the nanoscale. Chem Soc Rev 41(7):2718–2739

    Article  CAS  PubMed  Google Scholar 

  5. Soenen SJ, Himmelreich U, Nuytten N, Pisanic TR, Ferrari A, De Cuyper M (2010) Intracellular nanoparticle coating stability determines nanoparticle diagnostics efficacy and cell functionality. Small 6(19):2136–2145

    Article  CAS  PubMed  Google Scholar 

  6. Karamchand L, Kim G, Wang S, Hah HJ, Ray A, Jiddou R, Koo Lee Y-E, Philbert MA, Kopelman R (2013) Modulation of hydrogel nanoparticle intracellular trafficking by multivalent surface engineering with tumor targeting peptide. Nanoscale 5(21):10327–10344. doi:10.1039/c3nr00908d

    Article  CAS  PubMed  Google Scholar 

  7. Koo Lee Y-E, Ulbrich EE, Kim G, Hah H, Strollo C, Fan W, Gurjar R, Koo S, Kopelman R (2010) Near infrared luminescent oxygen nanosensors with nanoparticle matrix tailored sensitivity. Anal Chem 82(20):8446–8455. doi:10.1021/ac1015358

    Article  CAS  Google Scholar 

  8. Napp J, Behnke T, Fischer L, Würth C, Wottawa M, Katschinski DM, Alves F, Resch-Genger U, Schäferling M (2011) Targeted luminescent near-infrared polymer-nanoprobes for in vivo imaging of tumor hypoxia. Anal Chem 83(23):9039–9046. doi:10.1021/ac201870b

    Article  CAS  PubMed  Google Scholar 

  9. Xiang H, Cheng J, Ma X, Zhou X, Chruma JJ (2013) Near-infrared phosphorescence: materials and applications. Chem Soc Rev. doi:10.1039/c3cs60029g

    PubMed  Google Scholar 

  10. Dmitriev RI, Papkovsky DB (2012) Optical probes and techniques for O2 measurement in live cells and tissue. Cell Mol Life Sci 69(12):2025–2039

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  11. Quaranta M, Borisov SM, Klimant I (2012) Indicators for optical oxygen sensors. Bioanal Rev 4(2–4):115–157

    Article  PubMed Central  PubMed  Google Scholar 

  12. Ruggi A, van Leeuwen FW, Velders AH (2011) Interaction of dioxygen with the electronic excited state of Ir(III) and Ru (II) complexes: principles and biomedical applications. Coord Chem Rev 255(21):2542–2554

    Article  CAS  Google Scholar 

  13. Lee Y-EK, Kopelman R, Smith R (2009) Nanoparticle PEBBLE sensors in live cells and in vivo. Annu Rev Anal Chem (Palo Alto, Calif) 2:57

    Article  Google Scholar 

  14. Devor A, Sakadžić S, Yaseen MA, Roussakis E, Tian P, Slovin H, Vanzetta I, Teng I, Saisan PA, Sinks LE (2014) Functional imaging of cerebral oxygenation with intrinsic optical contrast and phosphorescent probes. Optical imaging of neocortical dynamics. Springer, Berlin, pp 225–253

    Chapter  Google Scholar 

  15. Papkovsky DB, Dmitriev RI (2013) Biological detection by optical oxygen sensing. Chem Soc Rev 42(22):8700–8732

    Article  CAS  PubMed  Google Scholar 

  16. Tsytsarev V, Arakawa H, Borisov S, Pumbo E, Erzurumlu RS, Papkovsky DB (2013) In vivo imaging of brain metabolism activity using a phosphorescent oxygen-sensitive probe. J Neurosci Methods 216(2):146–151

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  17. Dmitriev RI, Zhdanov AV, Nolan YM, Papkovsky DB (2013) Imaging of neurosphere oxygenation with phosphorescent probes. Biomaterials 34(37):9307–9317

    Article  CAS  PubMed  Google Scholar 

  18. Lambrechts D, Roeffaers M, Kerckhofs G, Roberts SJ, Hofkens J, Van de Putte T, Van Oosterwyck H, Schrooten J (2013) Fluorescent oxygen sensitive microbead incorporation for measuring oxygen tension in cell aggregates. Biomaterials 34(4):922–929

    Article  CAS  PubMed  Google Scholar 

  19. Dmitriev RI, Kondrashina AV, Koren K, Klimant I, Zhdanov AV, Pakan JMP, McDermott KW, Papkovsky DB (2014) Small molecule phosphorescent probes for O2 imaging in 3D tissue models. Biomater Sci 2:853–866. doi:10.1039/c3bm60272a

    Article  CAS  Google Scholar 

  20. Dmitriev RI, Zhdanov AV, Jasionek G, Papkovsky DB (2012) Assessment of cellular oxygen gradients with a panel of phosphorescent oxygen-sensitive probes. Anal Chem 84(6):2930–2938

    Article  CAS  PubMed  Google Scholar 

  21. Zhdanov AV, Dmitriev RI, Golubeva AV, Gavrilova SA, Papkovsky DB (1830) Chronic hypoxia leads to a glycolytic phenotype and suppressed HIF-2 signaling in PC12 cells. Biochim Biophys Acta Gen Subj 6:3553–3569

    Google Scholar 

  22. Zhdanov AV, Waters AH, Golubeva AV, Dmitriev RI, Papkovsky DB (1837) Availability of the key metabolic substrates dictates the respiratory response of cancer cells to the mitochondrial uncoupling. Biochim Biophys Acta Bioenerg 1:51–62

    Google Scholar 

  23. Apreleva SV, Wilson DF, Vinogradov SA (2006) Tomographic imaging of oxygen by phosphorescence lifetime. Appl Opt 45(33):8547–8559

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  24. Zhdanov AV, Ogurtsov VI, Taylor CT, Papkovsky DB (2010) Monitoring of cell oxygenation and responses to metabolic stimulation by intracellular oxygen sensing technique. Integr Biol 2(9):443–451

    Article  CAS  Google Scholar 

  25. Takahashi E, Asano K (2002) Mitochondrial respiratory control can compensate for intracellular O2 gradients in cardiomyocytes at low Po 2. Am J Physiol Heart Circ Physiol 283(3):H871–H878

    CAS  PubMed  Google Scholar 

  26. Finikova OS, Lebedev AY, Aprelev A, Troxler T, Gao F, Garnacho C, Muro S, Hochstrasser RM, Vinogradov SA (2008) Oxygen microscopy by two-photon-excited phosphorescence. ChemPhysChem 9(12):1673–1679

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  27. Mik EG, Stap J, Sinaasappel M, Beek JF, Aten JA, van Leeuwen TG, Ince C (2006) Mitochondrial PO2 measured by delayed fluorescence of endogenous protoporphyrin IX. Nat Methods 3(11):939–945

    Article  CAS  PubMed  Google Scholar 

  28. Clanton T, Hogan M, Gladden L (2013) Regulation of cellular gas exchange, oxygen sensing, and metabolic control. Compr Physiol. doi:10.1002/cphy.c120030

    PubMed  Google Scholar 

  29. Erecińska M, Silver IA (2001) Tissue oxygen tension and brain sensitivity to hypoxia. Respir Physiol 128(3):263–276

    Article  PubMed  Google Scholar 

  30. Mohyeldin A, Garzón-Muvdi T, Quiñones-Hinojosa A (2010) Oxygen in stem cell biology: a critical component of the stem cell niche. Cell Stem Cell 7(2):150–161

    Article  CAS  PubMed  Google Scholar 

  31. Toussaint O, Weemaels G, Debacq-Chainiaux F, Scharffetter-Kochanek K, Wlaschek M (2011) Artefactual effects of oxygen on cell culture models of cellular senescence and stem cell biology. J Cell Physiol 226(2):315–321

    Article  CAS  PubMed  Google Scholar 

  32. Semenza GL (2009) Regulation of oxygen homeostasis by hypoxia-inducible factor 1. Physiology 24(2):97–106

    Article  CAS  PubMed  Google Scholar 

  33. McHugh SB, Fillenz M, Lowry JP, Rawlins JNP, Bannerman DM (2011) Brain tissue oxygen amperometry in behaving rats demonstrates functional dissociation of dorsal and ventral hippocampus during spatial processing and anxiety. Eur J Neurosci 33(2):322–337

    Article  PubMed Central  PubMed  Google Scholar 

  34. Choi SW, Gerencser AA, Ng R, Flynn JM, Melov S, Danielson SR, Gibson BW, Nicholls DG, Bredesen DE, Brand MD (2012) No consistent bioenergetic defects in presynaptic nerve terminals isolated from mouse models of Alzheimer’s disease. J Neurosci 32(47):16775–16784

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  35. Nicholls DG, Johnson-Cadwell L, Vesce S, Jekabsons M, Yadava N (2007) Bioenergetics of mitochondria in cultured neurons and their role in glutamate excitotoxicity. J Neurosci Res 85(15):3206–3212. doi:10.1002/jnr.21290

    Article  CAS  PubMed  Google Scholar 

  36. Lancaster MA, Renner M, Martin C-A, Wenzel D, Bicknell LS, Hurles ME, Homfray T, Penninger JM, Jackson AP, Knoblich JA (2013) Cerebral organoids model human brain development and microcephaly. Nature 501:373–379. doi:10.1038/nature12517

    Article  CAS  PubMed  Google Scholar 

  37. Noraberg J, Kristensen BW, Zimmer J (1999) Markers for neuronal degeneration in organotypic slice cultures. Brain Res Protoc 3(3):278–290. doi:10.1016/S1385-299X(98)00050-6

    Article  CAS  Google Scholar 

  38. Zimmer J, Kristensen BW, Jakobsen B, Noraberg J (2000) Excitatory amino acid neurotoxicity and modulation of glutamate receptor expression in organotypic brain slice cultures. Amino Acids 19(1):7–21

    Article  CAS  PubMed  Google Scholar 

  39. Kazmi S, Salvaggio AJ, Estrada AD, Hemati MA, Shaydyuk NK, Roussakis E, Jones TA, Vinogradov SA, Dunn AK (2013) Three-dimensional mapping of oxygen tension in cortical arterioles before and after occlusion. Biomed Optics Express 4(7):1061–1073

    Article  Google Scholar 

  40. Lecoq J, Parpaleix A, Roussakis E, Ducros M, Houssen YG, Vinogradov SA, Charpak S (2011) Simultaneous two-photon imaging of oxygen and blood flow in deep cerebral vessels. Nat Med 17(7):893–898

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  41. Wolfbeis OS (2013) Editorial: probes, sensors, and labels: Why is real progress slow? Angew Chem Int Ed 52(38):9864–9865. doi:10.1002/anie.201305915

    Article  CAS  Google Scholar 

  42. Kondrashina AV, Dmitriev RI, Borisov SM, Klimant I, O’Brien I, Nolan YM, Zhdanov AV, Papkovsky DB (2012) A phosphorescent nanoparticle-based probe for sensing and imaging of (Intra) cellular oxygen in multiple detection modalities. Adv Funct Mater 22(23):4931–4939. doi:10.1002/adfm.201201387

    Article  CAS  Google Scholar 

  43. Liu H, Yang H, Hao X, Xu H, Lv Y, Xiao D, Wang H, Tian Z (2013) Development of polymeric nanoprobes with improved lifetime dynamic range and stability for intracellular oxygen sensing. Small 9(15):2639–2648

    Article  CAS  PubMed  Google Scholar 

  44. X-d Wang, Gorris HH, Stolwijk JA, Meier RJ, Groegel DB, Wegener J, Wolfbeis OS (2011) Self-referenced RGB colour imaging of intracellular oxygen. Chem Sci 2(5):901–906

    Article  Google Scholar 

  45. X-d Wang, Stolwijk JA, Lang T, Sperber M, Meier RJ, Wegener J, Wolfbeis OS (2012) Ultra-small, highly stable, and sensitive dual nanosensors for imaging intracellular oxygen and pH in cytosol. J Am Chem Soc 134(41):17011–17014

    Article  Google Scholar 

  46. Wu C, Bull B, Christensen K, McNeill J (2009) Ratiometric single-nanoparticle oxygen sensors for biological imaging. Angew Chem Int Ed 48(15):2741–2745

    Article  CAS  Google Scholar 

  47. Yim H, S-j Park, Bae YH, Na K (2013) Biodegradable cationic nanoparticles loaded with an anticancer drug for deep penetration of heterogeneous tumours. Biomaterials 34(31):7674–7682

    Article  CAS  PubMed  Google Scholar 

  48. Owens Iii DE, Peppas NA (2006) Opsonization, biodistribution, and pharmacokinetics of polymeric nanoparticles. Int J Pharm 307(1):93–102. doi:10.1016/j.ijpharm.2005.10.010

    Article  Google Scholar 

  49. Ingram J, Zhang C, Cressman JR, Hazra A, WEI Y, Koo Y-E, Ziburkus J, Kopelman R, Xu J, Schiff SJ (2014) Oxygen and seizure dynamics: I. Experiments. J Neurophysiol 00540:02013

    Google Scholar 

  50. Ingram JM, Zhang C, Xu J, Schiff SJ (2013) FRET excited ratiometric oxygen sensing in living tissue. J Neurosci Methods 214(1):45–51. doi:10.1016/j.jneumeth.2013.01.002

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  51. Koren K, Dmitriev RI, Borisov SM, Papkovsky DB, Klimant I (2012) Complexes of IrIII-Octaethylporphyrin with Peptides as Probes for Sensing Cellular O2. Chembiochem 13(8):1184–1190. doi:10.1002/cbic.201200083

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  52. Borisov SM, Mayr T, Mistlberger G, Waich K, Koren K, Chojnacki P, Klimant I (2009) Precipitation as a simple and versatile method for preparation of optical nanochemosensors. Talanta 79(5):1322–1330

    Article  CAS  PubMed  Google Scholar 

  53. Albanese A, Tang PS, Chan WC (2012) The effect of nanoparticle size, shape, and surface chemistry on biological systems. Annu Rev Biomed Eng 14:1–16

    Article  CAS  PubMed  Google Scholar 

  54. Yang W-H, Smolen VF, Peppas NA (1981) Oxygen permeability coefficients of polymers for hard and soft contact lens applications. J Membr Sci 9(1):53–67

    Article  CAS  Google Scholar 

  55. Fercher A, Borisov SM, Zhdanov AV, Klimant I, Papkovsky DB (2011) Intracellular O2 sensing probe based on cell-penetrating phosphorescent nanoparticles. ACS Nano 5(7):5499–5508

    Article  CAS  PubMed  Google Scholar 

  56. Mendes AN, Hubber I, Siqueira M, Barbosa GM, de Lima Moreira D, Holandino C, Pinto JC, Nele M (2012) Preparation and Cytotoxicity of Poly (Methyl Methacrylate) Nanoparticles for Drug Encapsulation. In: Macromolecular Symposia, Wiley Online Library, pp 34–40

  57. Araujo L, Sheppard M, Löbenberg R, Kreuter J (1999) Uptake of PMMA nanoparticles from the gastrointestinal tract after oral administration to rats: modification of the body distribution after suspension in surfactant solutions and in oil vehicles. Int J Pharm 176(2):209–224

    Article  CAS  Google Scholar 

  58. Ivanov AI (2008) Pharmacological inhibition of endocytic pathways: is it specific enough to be useful? In: Exocytosis and Endocytosis. Springer, pp 15–33

  59. Carraway E, Demas J, DeGraff B, Bacon J (1991) Photophysics and photochemistry of oxygen sensors based on luminescent transition-metal complexes. Anal Chem 63(4):337–342

    Article  CAS  Google Scholar 

  60. DeGraff B, Demas J (2005) Luminescence-based oxygen sensors. Reviews in Fluorescence 2005:125–151

    Google Scholar 

  61. Foster KA, Galeffi F, Gerich FJ, Turner DA, Müller M (2006) Optical and pharmacological tools to investigate the role of mitochondria during oxidative stress and neurodegeneration. Prog Neurobiol 79(3):136–171

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  62. Pakan JM, McDermott KW (2014) A method to investigate radial glia cell behavior using two-photon time-lapse microscopy in an ex vivo model of spinal cord development. Front Neuroanat 8:22. doi:10.3389/fnana.2014.00022

    Article  PubMed Central  PubMed  Google Scholar 

  63. Schmid RS, Yokota Y, Anton E (2006) Generation and characterization of brain lipid-binding protein promoter-based transgenic mouse models for the study of radial glia. Glia 53(4):345–351

    Article  PubMed  Google Scholar 

  64. Becker W (2012) Fluorescence lifetime imaging—techniques and applications. J Microsc 247(2):119–136. doi:10.1111/j.1365-2818.2012.03618.x

    Article  CAS  PubMed  Google Scholar 

  65. Ingaramo M, York AG, Wawrzusin P, Milberg O, Hong A, Weigert R, Shroff H, Patterson GH (2014) Two-photon excitation improves multifocal structured illumination microscopy in thick scattering tissue. Proc Natl Acad Sci 111(14):5254–5259

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  66. Pampaloni F, Ansari N, Stelzer EH (2013) High-resolution deep imaging of live cellular spheroids with light-sheet-based fluorescence microscopy. Cell Tissue Res 352:161–177

    Article  PubMed  Google Scholar 

  67. Golub AS, Pittman RN (2008) PO2 measurements in the microcirculation using phosphorescence quenching microscopy at high magnification. Am J Physiol Heart Circ Physiol 63(6):H2905

    Article  Google Scholar 

  68. Devor A, Sakadzic S, Srinivasan VJ, Yaseen MA, Nizar K, Saisan PA, Tian P, Dale AM, Vinogradov SA, Franceschini MA, Boas DA (2012) Frontiers in optical imaging of cerebral blood flow and metabolism. J Cereb Blood Flow Metab 32(7):1259–1276

    Article  CAS  PubMed Central  PubMed  Google Scholar 

  69. Spencer JA, Ferraro F, Roussakis E, Klein A, Wu J, Runnels JM, Zaher W, Mortensen LJ, Alt C, Turcotte R, Yusuf R, Cote D, Vinogradov SA, Scadden DT, Lin CP (2014) Direct measurement of local oxygen concentration in the bone marrow of live animals. Nature advance online publication. doi:10.1038/nature13034

    Google Scholar 

  70. Verma A, Stellacci F (2010) Effect of surface properties on nanoparticle-cell interactions. Small 6(1):12–21. doi:10.1002/smll.200901158

    Article  CAS  PubMed  Google Scholar 

  71. Nichols AJ, Roussakis E, Klein OJ, Evans CL (2014) Click-Assembled, Oxygen-Sensing Nanoconjugates for Depth-Resolved, Near-Infrared Imaging in a 3 D Cancer Model. Angew Chem Int Ed n/a-n/a. doi:10.1002/anie.201311303

    Google Scholar 

  72. Dmitriev RI, Ropiak HM, Ponomarev GV, Yashunsky DV, Papkovsky DB (2011) Cell-penetrating conjugates of coproporphyrins with oligoarginine peptides: rational design and application for sensing intracellular O2. Bioconjugate Chem 22(12):2507–2518

    Article  CAS  Google Scholar 

  73. Anilkumar U, Weisová P, Düssmann H, Concannon CG, König H-G, Prehn JHM (2013) AMP-activated protein kinase (AMPK)–induced preconditioning in primary cortical neurons involves activation of MCL-1. J Neurochem 124(5):721–734. doi:10.1111/jnc.12108

    Article  CAS  PubMed  Google Scholar 

  74. Stoppini L, Buchs P-A, Muller D (1991) A simple method for organotypic cultures of nervous tissue. J Neurosci Methods 37(2):173–182

    Article  CAS  PubMed  Google Scholar 

  75. Murphy RC, Messer A (2001) Gene transfer methods for CNS organotypic cultures: a comparison of three nonviral methods. Mol Ther 3(1):113–121

    Article  CAS  PubMed  Google Scholar 

Download references

Acknowledgments

This work was supported by the Science Foundation Ireland, Grant 12/RC/2276, the European Commission FP7 Program, grant FP7-HEALTH-2012-INNOVATION-304842-2, the Irish Research Council for Science, Engineering and Technology, the Health Research Board, the Programme for Research at Third Level Institutions Cycle 4 and Marie Curie IAPP Oxy-Sense (No. 230641). We thank T. Foley and Dr. Y. Nolan (Department of Anatomy and Neuroscience, UCC) for the help with primary neuronal cultures and Dr. Heiko Dussmann (Royal College of Surgeons in Ireland, Dublin) for help with microscopy imaging.

Author information

Authors and Affiliations

Authors

Corresponding author

Correspondence to Ruslan I. Dmitriev.

Electronic supplementary material

Below is the link to the electronic supplementary material.

Supplementary material 1 (PDF 492 kb)

Rights and permissions

Reprints and permissions

About this article

Check for updates. Verify currency and authenticity via CrossMark

Cite this article

Dmitriev, R.I., Borisov, S.M., Kondrashina, A.V. et al. Imaging oxygen in neural cell and tissue models by means of anionic cell-permeable phosphorescent nanoparticles. Cell. Mol. Life Sci. 72, 367–381 (2015). https://doi.org/10.1007/s00018-014-1673-5

Download citation

  • Received:

  • Revised:

  • Accepted:

  • Published:

  • Issue Date:

  • DOI: https://doi.org/10.1007/s00018-014-1673-5

Keywords

Navigation