- -

A translational approach to assess the metabolomic impact of stabilized gold nanoparticles by NMR spectroscopy

RiuNet: Repositorio Institucional de la Universidad Politécnica de Valencia

Compartir/Enviar a

Citas

Estadísticas

  • Estadisticas de Uso

A translational approach to assess the metabolomic impact of stabilized gold nanoparticles by NMR spectroscopy

Mostrar el registro completo del ítem

Herance, JR.; García Gómez, H.; Guitierrez Carcedo, P.; Navalón Oltra, S.; Pineda-Lucena, A.; Palomino-Schätzlein, M. (2019). A translational approach to assess the metabolomic impact of stabilized gold nanoparticles by NMR spectroscopy. The Analyst. 144(4):1265-1274. https://doi.org/10.1039/c8an01827h

Por favor, use este identificador para citar o enlazar este ítem: http://hdl.handle.net/10251/154119

Ficheros en el ítem

Metadatos del ítem

Título: A translational approach to assess the metabolomic impact of stabilized gold nanoparticles by NMR spectroscopy
Autor: Herance, Jose Raul García Gómez, Hermenegildo Guitierrez Carcedo, Patricia Navalón Oltra, Sergio Pineda-Lucena, Antonio Palomino-Schätzlein, M.
Entidad UPV: Universitat Politècnica de València. Departamento de Química - Departament de Química
Fecha difusión:
Resumen:
[EN] Gold nanoparticles have high potential in the biomedical area, especially in disease diagnosis and treatment. The application of these nanoparticles requires the presence of stabilizers to avoid their agglomeration. ...[+]
Palabras clave: Cells , Cytotoxicity , Perturbations , Proteomics , Reveals , Assay , Ceria
Derechos de uso: Reserva de todos los derechos
Fuente:
The Analyst. (issn: 0003-2654 )
DOI: 10.1039/c8an01827h
Editorial:
The Royal Society of Chemistry
Versión del editor: https://doi.org/10.1039/c8an01827h
Código del Proyecto:
info:eu-repo/grantAgreement/MINECO//SAF2014-53977-R/ES/HERRAMIENTAS DE RMN PARA EL DESARROLLO DE UNA PLATAFORMA PARA LA IDENTIFIC. DE DIANAS, LA EVAL. DE FARMACOS Y LA PERSONAL. DE TRATAMIENTOS BASADA EN APROX. METABOLOMICAS/
info:eu-repo/grantAgreement/MINECO//CP13%2F00252/ES/CP13%2F00252/
info:eu-repo/grantAgreement/AEI/Plan Estatal de Investigación Científica y Técnica y de Innovación 2013-2016/SAF2017-89229-R/ES/CONTRIBUCIONES DE LA METABOLOMICA POR RMN A LA CLASIFICACION CLINICA DE PACIENTES: UN PASO ADELANTE HACIA LA MEDICINA DE PRECISION/
info:eu-repo/grantAgreement/MINECO//RD12%2F0036%2F0025/ES/Cáncer/
nfo:eu-repo/grantAgreement/MINECO//PI16%2F02064/ES/Resistencia insulínica en miocardio como factor de riesgo cardiovascular en Diabetes Mellitus tipo 2/
Agradecimientos:
This work was supported by the Carlos III Health Institute, the European Regional Development Fund (PI16/02064 and CP13/00252) and the Spanish Ministerio de Economia y Competitividad (SAF2014-53977-R, SAF2017-89229-R and ...[+]
Tipo: Artículo

References

Shi, J., Kantoff, P. W., Wooster, R., & Farokhzad, O. C. (2016). Cancer nanomedicine: progress, challenges and opportunities. Nature Reviews Cancer, 17(1), 20-37. doi:10.1038/nrc.2016.108

Gioria, S., Lobo Vicente, J., Barboro, P., La Spina, R., Tomasi, G., Urbán, P., … Chassaigne, H. (2016). A combined proteomics and metabolomics approach to assess the effects of gold nanoparticlesin vitro. Nanotoxicology, 10(6), 736-748. doi:10.3109/17435390.2015.1121412

Beik, J., Khademi, S., Attaran, N., Sarkar, S., Shakeri-Zadeh, A., Ghaznavi, H., & Ghadiri, H. (2017). A Nanotechnology-based Strategy to Increase the Efficiency of Cancer Diagnosis and Therapy: Folate-conjugated Gold Nanoparticles. Current Medicinal Chemistry, 24(39). doi:10.2174/0929867324666170810154917 [+]
Shi, J., Kantoff, P. W., Wooster, R., & Farokhzad, O. C. (2016). Cancer nanomedicine: progress, challenges and opportunities. Nature Reviews Cancer, 17(1), 20-37. doi:10.1038/nrc.2016.108

Gioria, S., Lobo Vicente, J., Barboro, P., La Spina, R., Tomasi, G., Urbán, P., … Chassaigne, H. (2016). A combined proteomics and metabolomics approach to assess the effects of gold nanoparticlesin vitro. Nanotoxicology, 10(6), 736-748. doi:10.3109/17435390.2015.1121412

Beik, J., Khademi, S., Attaran, N., Sarkar, S., Shakeri-Zadeh, A., Ghaznavi, H., & Ghadiri, H. (2017). A Nanotechnology-based Strategy to Increase the Efficiency of Cancer Diagnosis and Therapy: Folate-conjugated Gold Nanoparticles. Current Medicinal Chemistry, 24(39). doi:10.2174/0929867324666170810154917

Nagi, N. M. S., Khair, Y. A. M., & Abdalla, A. M. E. (2017). Capacity of gold nanoparticles in cancer radiotherapy. Japanese Journal of Radiology, 35(10), 555-561. doi:10.1007/s11604-017-0671-6

Gharatape, A., & Salehi, R. (2017). Recent progress in theranostic applications of hybrid gold nanoparticles. European Journal of Medicinal Chemistry, 138, 221-233. doi:10.1016/j.ejmech.2017.06.034

Fang, J., Nakamura, H., & Maeda, H. (2011). The EPR effect: Unique features of tumor blood vessels for drug delivery, factors involved, and limitations and augmentation of the effect. Advanced Drug Delivery Reviews, 63(3), 136-151. doi:10.1016/j.addr.2010.04.009

Haume, K., Rosa, S., Grellet, S., Śmiałek, M. A., Butterworth, K. T., Solov’yov, A. V., … Mason, N. J. (2016). Gold nanoparticles for cancer radiotherapy: a review. Cancer Nanotechnology, 7(1). doi:10.1186/s12645-016-0021-x

Azevedo, R. S. S., de Sousa, J. R., Araujo, M. T. F., Martins Filho, A. J., de Alcantara, B. N., Araujo, F. M. C., … Vasconcelos, P. F. C. (2018). In situ immune response and mechanisms of cell damage in central nervous system of fatal cases microcephaly by Zika virus. Scientific Reports, 8(1). doi:10.1038/s41598-017-17765-5

Lin, Y., Wu, Z., Wen, J., Ding, K., Yang, X., Poeppelmeier, K. R., & Marks, L. D. (2015). Adhesion and Atomic Structures of Gold on Ceria Nanostructures: The Role of Surface Structure and Oxidation State of Ceria Supports. Nano Letters, 15(8), 5375-5381. doi:10.1021/acs.nanolett.5b02694

Menchón, C., Martín, R., Apostolova, N., Victor, V. M., Álvaro, M., Herance, J. R., & García, H. (2012). Gold Nanoparticles Supported on Nanoparticulate Ceria as a Powerful Agent against Intracellular Oxidative Stress. Small, 8(12), 1895-1903. doi:10.1002/smll.201102255

Tiwari, P., Vig, K., Dennis, V., & Singh, S. (2011). Functionalized Gold Nanoparticles and Their Biomedical Applications. Nanomaterials, 1(1), 31-63. doi:10.3390/nano1010031

Fathy, M. M., Mohamed, F. S., Elbialy, N., & Elshemey, W. M. (2018). Multifunctional Chitosan-Capped Gold Nanoparticles for enhanced cancer chemo-radiotherapy: An invitro study. Physica Medica, 48, 76-83. doi:10.1016/j.ejmp.2018.04.002

Guo, X., Zhuang, Q., Ji, T., Zhang, Y., Li, C., Wang, Y., … Du, L. (2018). Multi-functionalized chitosan nanoparticles for enhanced chemotherapy in lung cancer. Carbohydrate Polymers, 195, 311-320. doi:10.1016/j.carbpol.2018.04.087

Lee, Y.-H., Kim, J.-S., Kim, J.-E., Lee, M.-H., Jeon, J.-G., Park, I.-S., & Yi, H.-K. (2017). Nanoparticle mediated PPARγ gene delivery on dental implants improves osseointegration via mitochondrial biogenesis in diabetes mellitus rat model. Nanomedicine: Nanotechnology, Biology and Medicine, 13(5), 1821-1832. doi:10.1016/j.nano.2017.02.020

Sun, I.-C., Na, J. H., Jeong, S. Y., Kim, D.-E., Kwon, I. C., Choi, K., … Kim, K. (2013). Biocompatible Glycol Chitosan-Coated Gold Nanoparticles for Tumor-Targeting CT Imaging. Pharmaceutical Research, 31(6), 1418-1425. doi:10.1007/s11095-013-1142-0

Costa, P. M., & Fadeel, B. (2016). Emerging systems biology approaches in nanotoxicology: Towards a mechanism-based understanding of nanomaterial hazard and risk. Toxicology and Applied Pharmacology, 299, 101-111. doi:10.1016/j.taap.2015.12.014

Lv, M., Huang, W., Chen, Z., Jiang, H., Chen, J., Tian, Y., … Xu, F. (2015). Metabolomics techniques for nanotoxicity investigations. Bioanalysis, 7(12), 1527-1544. doi:10.4155/bio.15.83

Nagana Gowda, G. A., Barding, G. A., Dai, J., Gu, H., Margineantu, D. H., Hockenbery, D. M., & Raftery, D. (2018). A Metabolomics Study of BPTES Altered Metabolism in Human Breast Cancer Cell Lines. Frontiers in Molecular Biosciences, 5. doi:10.3389/fmolb.2018.00049

Ali, M. R. K., Wu, Y., Han, T., Zang, X., Xiao, H., Tang, Y., … El-Sayed, M. A. (2016). Simultaneous Time-Dependent Surface-Enhanced Raman Spectroscopy, Metabolomics, and Proteomics Reveal Cancer Cell Death Mechanisms Associated with Gold Nanorod Photothermal Therapy. Journal of the American Chemical Society, 138(47), 15434-15442. doi:10.1021/jacs.6b08787

Esumi, K., Takei, N., & Yoshimura, T. (2003). Antioxidant-potentiality of gold–chitosan nanocomposites. Colloids and Surfaces B: Biointerfaces, 32(2), 117-123. doi:10.1016/s0927-7765(03)00151-6

Du, S., Kendall, K., Toloueinia, P., Mehrabadi, Y., Gupta, G., & Newton, J. (2012). Aggregation and adhesion of gold nanoparticles in phosphate buffered saline. Journal of Nanoparticle Research, 14(3). doi:10.1007/s11051-012-0758-z

Ulrich, E. L., Akutsu, H., Doreleijers, J. F., Harano, Y., Ioannidis, Y. E., Lin, J., … Markley, J. L. (2007). BioMagResBank. Nucleic Acids Research, 36(Database), D402-D408. doi:10.1093/nar/gkm957

Wishart, D. S., Feunang, Y. D., Marcu, A., Guo, A. C., Liang, K., Vázquez-Fresno, R., … Scalbert, A. (2017). HMDB 4.0: the human metabolome database for 2018. Nucleic Acids Research, 46(D1), D608-D617. doi:10.1093/nar/gkx1089

Mosmann, T. (1983). Rapid colorimetric assay for cellular growth and survival: Application to proliferation and cytotoxicity assays. Journal of Immunological Methods, 65(1-2), 55-63. doi:10.1016/0022-1759(83)90303-4

Yang, L., Shang, L., & Nienhaus, G. U. (2013). Mechanistic aspects of fluorescent gold nanocluster internalization by live HeLa cells. Nanoscale, 5(4), 1537. doi:10.1039/c2nr33147k

Zhitomirsky, B., Farber, H., & Assaraf, Y. G. (2018). LysoTracker and MitoTracker Red are transport substrates of P‐glycoprotein: implications for anticancer drug design evading multidrug resistance. Journal of Cellular and Molecular Medicine, 22(4), 2131-2141. doi:10.1111/jcmm.13485

Chong, J., Soufan, O., Li, C., Caraus, I., Li, S., Bourque, G., … Xia, J. (2018). MetaboAnalyst 4.0: towards more transparent and integrative metabolomics analysis. Nucleic Acids Research, 46(W1), W486-W494. doi:10.1093/nar/gky310

Stepanenko, A. A., & Dmitrenko, V. V. (2015). Pitfalls of the MTT assay: Direct and off-target effects of inhibitors can result in over/underestimation of cell viability. Gene, 574(2), 193-203. doi:10.1016/j.gene.2015.08.009

Choi, S. Y., Jang, S. H., Park, J., Jeong, S., Park, J. H., Ock, K. S., … Lee, S. Y. (2012). Cellular uptake and cytotoxicity of positively charged chitosan gold nanoparticles in human lung adenocarcinoma cells. Journal of Nanoparticle Research, 14(12). doi:10.1007/s11051-012-1234-5

De Carvalho, T. G., Garcia, V. B., de Araújo, A. A., da Silva Gasparotto, L. H., Silva, H., Guerra, G. C. B., … de Araújo Júnior, R. F. (2018). Spherical neutral gold nanoparticles improve anti-inflammatory response, oxidative stress and fibrosis in alcohol-methamphetamine-induced liver injury in rats. International Journal of Pharmaceutics, 548(1), 1-14. doi:10.1016/j.ijpharm.2018.06.008

Carrola, J., Bastos, V., Ferreira de Oliveira, J. M. P., Oliveira, H., Santos, C., Gil, A. M., & Duarte, I. F. (2016). Insights into the impact of silver nanoparticles on human keratinocytes metabolism through NMR metabolomics. Archives of Biochemistry and Biophysics, 589, 53-61. doi:10.1016/j.abb.2015.08.022

Fröhlich, E. (2012). The role of surface charge in cellular uptake and cytotoxicity of medical nanoparticles. International Journal of Nanomedicine, 5577. doi:10.2147/ijn.s36111

Gürer, H., Özgünes, H., Saygin, E., & Ercal, N. (2001). Antioxidant Effect of Taurine Against Lead-Induced Oxidative Stress. Archives of Environmental Contamination and Toxicology, 41(4), 397-402. doi:10.1007/s002440010265

Lee, S.-H., Wang, T.-Y., Hong, J.-H., Cheng, T.-J., & Lin, C.-Y. (2016). NMR-based metabolomics to determine acute inhalation effects of nano- and fine-sized ZnO particles in the rat lung. Nanotoxicology, 10(7), 924-934. doi:10.3109/17435390.2016.1144825

Saborano, R., Wongpinyochit, T., Totten, J. D., Johnston, B. F., Seib, F. P., & Duarte, I. F. (2017). Metabolic Reprogramming of Macrophages Exposed to Silk, Poly(lactic-co-glycolic acid), and Silica Nanoparticles. Advanced Healthcare Materials, 6(14), 1601240. doi:10.1002/adhm.201601240

Bo, Y., Jin, C., Liu, Y., Yu, W., & Kang, H. (2014). Metabolomic analysis on the toxicological effects of TiO2nanoparticles in mouse fibroblast cells: from the perspective of perturbations in amino acid metabolism. Toxicology Mechanisms and Methods, 24(7), 461-469. doi:10.3109/15376516.2014.939321

Shea, T. B., Ekinci, F. J., Ortiz, D., Dawn-Linsley, M., Wilson, T. O., & Nicolosi, R. J. (2002). Efficacy of vitamin E, phosphatidyl choline, and pyruvate on buffering neuronal degeneration and oxidative stress in cultured cortical neurons and in central nervous tissue of apolipoprotein E-deficient mice. Free Radical Biology and Medicine, 33(2), 276-282. doi:10.1016/s0891-5849(02)00872-9

Schätzlein, M. P., Becker, J., Schulze-Sünninghausen, D., Pineda-Lucena, A., Herance, J. R., & Luy, B. (2018). Rapid two-dimensional ALSOFAST-HSQC experiment for metabolomics and fluxomics studies: application to a 13C-enriched cancer cell model treated with gold nanoparticles. Analytical and Bioanalytical Chemistry, 410(11), 2793-2804. doi:10.1007/s00216-018-0961-6

HUNTER, A. (2006). Molecular hurdles in polyfectin design and mechanistic background to polycation induced cytotoxicity☆. Advanced Drug Delivery Reviews, 58(14), 1523-1531. doi:10.1016/j.addr.2006.09.008

Ratnasekhar, C., Sonane, M., Satish, A., & Mudiam, M. K. R. (2015). Metabolomics reveals the perturbations in the metabolome ofCaenorhabditis elegansexposed to titanium dioxide nanoparticles. Nanotoxicology, 9(8), 994-1004. doi:10.3109/17435390.2014.993345

[-]

recommendations

 

Este ítem aparece en la(s) siguiente(s) colección(ones)

Mostrar el registro completo del ítem