Real-time tracking of delayed-onset cellular apoptosis induced by intracellular magnetic hyperthermia.

AIM To assess cell death pathways in response to magnetic hyperthermia. MATERIALS & METHODS Human melanoma cells were loaded with citric acid-coated iron-oxide nanoparticles, and subjected to a time-varying magnetic field. Pathways were monitored in vitro in suspensions and in situ in monolayers using fluorophores to report on early-stage apoptosis and late-stage apoptosis and/or necrosis. RESULTS Delayed-onset effects were observed, with a rate and extent proportional to the thermal-load-per-cell. At moderate loads, membranal internal-to-external lipid exchange preceded rupture and death by a few hours (the timeline varying cell-to-cell), without any measurable change in the local environment temperature. CONCLUSION Our observations support the proposition that intracellular heating may be a viable, controllable and nonaggressive in vivo treatment for human pathological conditions.

[1]  S. Bossmann,et al.  Attenuation of mouse melanoma by A/C magnetic field after delivery of bi-magnetic nanoparticles by neural progenitor cells. , 2010, ACS nano.

[2]  S. Dutz,et al.  Magnetic nanoparticle heating and heat transfer on a microscale: Basic principles, realities and physical limitations of hyperthermia for tumour therapy , 2013, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[3]  M. Horton,et al.  Integrin expression in human melanoma cell lines: Heterogeneity of vitronectin receptor composition and function , 1991, International journal of cancer.

[4]  R. Bajpai,et al.  Efficient propagation of single cells accutase‐dissociated human embryonic stem cells , 2008, Molecular reproduction and development.

[5]  V. Connord,et al.  Real-Time Analysis of Magnetic Hyperthermia Experiments on Living Cells under a Confocal Microscope. , 2015, Small.

[6]  F. Lin,et al.  The characterization and evaluation of cisplatin-loaded magnetite–hydroxyapatite nanoparticles (mHAp/CDDP) as dual treatment of hyperthermia and chemotherapy for lung cancer therapy , 2015 .

[7]  M. Ibarra,et al.  Controlled Cell Death by Magnetic Hyperthermia: Effects of Exposure Time, Field Amplitude, and Nanoparticle Concentration , 2012, Pharmaceutical Research.

[8]  M. Morales,et al.  Synthesis of high intrinsic loss power aqueous ferrofluids of iron oxide nanoparticles by citric acid-assisted hydrothermal-reduction route , 2012 .

[9]  C. Innocenti,et al.  A smart platform for hyperthermia application in cancer treatment: cobalt-doped ferrite nanoparticles mineralized in human ferritin cages. , 2014, ACS nano.

[10]  Viktor Chikan,et al.  A/C magnetic hyperthermia of melanoma mediated by iron(0)/iron oxide core/shell magnetic nanoparticles: a mouse study , 2010, BMC Cancer.

[11]  K. Paknikar,et al.  Temperature-dependent and time-dependent effects of hyperthermia mediated by dextran-coated La0.7Sr0.3MnO3: in vitro studies , 2015, International journal of nanomedicine.

[12]  J. Gabriel,et al.  Highly focalised thermotherapy using a ferrimagnetic cement in the treatment of a melanoma mouse model by low temperature hyperthermia , 2013, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[13]  I. Hilger In vivo applications of magnetic nanoparticle hyperthermia , 2013, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[14]  C. Reutelingsperger,et al.  A novel assay to measure loss of plasma membrane asymmetry during apoptosis of adherent cells in culture. , 1996, Cytometry.

[15]  B. Alberts,et al.  Molecular Biology of the Cell 4th edition , 2007 .

[16]  H. Honda,et al.  4‐S‐Cysteaminylphenol‐loaded magnetite cationic liposomes for combination therapy of hyperthermia with chemotherapy against malignant melanoma , 2007, Cancer science.

[17]  Q. Pankhurst,et al.  High performance multi-core iron oxide nanoparticles for magnetic hyperthermia: microwave synthesis, and the role of core-to-core interactions. , 2015, Nanoscale.

[18]  W. Bubb,et al.  Singlet Oxygen–mediated Protein Oxidation: Evidence for the Formation of Reactive Side Chain Peroxides on Tyrosine Residues ¶ , 2002, Photochemistry and photobiology.

[19]  J. K. Sugden,et al.  Photochemistry of dyes and fluorochromes used in biology and medicine: some physicochemical background and current applications , 2004, Biotechnic & histochemistry : official publication of the Biological Stain Commission.

[20]  M. Lens,et al.  Global perspectives of contemporary epidemiological trends of cutaneous malignant melanoma , 2004, The British journal of dermatology.

[21]  E. Falcieri,et al.  Analysis of cell death by electron microscopy. , 2013, Methods in molecular biology.

[22]  T. Schmitz-Rode,et al.  Synthesis, physicochemical characterization and MR relaxometry of aqueous ferrofluids. , 2008, Journal of nanoscience and nanotechnology.

[23]  Carlos Rinaldi,et al.  EGFR-targeted magnetic nanoparticle heaters kill cancer cells without a perceptible temperature rise. , 2011, ACS nano.

[24]  Donald Wlodkowic,et al.  Real‐time cell viability assays using a new anthracycline derivative DRAQ7® , 2013, Cytometry. Part A : the journal of the International Society for Analytical Cytology.

[25]  M. Dewhirst,et al.  Thresholds for thermal damage to normal tissues: An update , 2011, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[26]  J. Hickman Programmed Cell Death in Tumours and Tissues , 1991, British Journal of Cancer.

[27]  A. Telser Molecular Biology of the Cell, 4th Edition , 2002 .

[28]  I. Andreu,et al.  Accuracy of available methods for quantifying the heat power generation of nanoparticles for magnetic hyperthermia , 2013, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[29]  P. Decuzzi,et al.  Heat-generating iron oxide nanocubes: subtle "destructurators" of the tumoral microenvironment. , 2014, ACS nano.

[30]  Q. Pankhurst,et al.  On the reliable measurement of specific absorption rates and intrinsic loss parameters in magnetic hyperthermia materials , 2014 .

[31]  Rakesh K. Gupta,et al.  Innovative synthesis of citrate-coated superparamagnetic Fe3O4 nanoparticles and its preliminary applications. , 2011, Journal of colloid and interface science.

[32]  Y Rabin,et al.  Is intracellular hyperthermia superior to extracellular hyperthermia in the thermal sense? , 2002, International journal of hyperthermia : the official journal of European Society for Hyperthermic Oncology, North American Hyperthermia Group.

[33]  H. Honda,et al.  N-propionyl-cysteaminylphenol-magnetite conjugate (NPrCAP/M) is a nanoparticle for the targeted growth suppression of melanoma cells. , 2009, The Journal of investigative dermatology.

[34]  E. Sahai,et al.  Membrane blebbing during apoptosis results from caspase-mediated activation of ROCK I , 2001, Nature Cell Biology.

[35]  K. Dawson,et al.  Surface coatings shape the protein corona of SPIONs with relevance to their application in vivo. , 2012, Langmuir : the ACS journal of surfaces and colloids.

[36]  Liberato Manna,et al.  Water-soluble iron oxide nanocubes with high values of specific absorption rate for cancer cell hyperthermia treatment. , 2012, ACS nano.

[37]  A. Friedman,et al.  Biodegradable chitosan nanoparticles in drug delivery for infectious disease. , 2015, Nanomedicine.

[38]  Timothy O'Brien,et al.  Superparamagnetic iron oxide nanoparticle targeting of MSCs in vascular injury. , 2013, Biomaterials.