17 research outputs found

    Chromo-fluorogenic probes for beta-galactosidase detection

    Full text link
    [EN] beta-Galactosidase (beta-Gal) is a widely used enzyme as a reporter gene in the field of molecular biology which hydrolyzes the beta-galactosides into monosaccharides. beta-Gal is an essential enzyme in humans and its deficiency or its overexpression results in several rare diseases. Cellular senescence is probably one of the most relevant physiological disorders that involve beta-Gal enzyme. In this review, we assess the progress made to date in the design of molecular-based probes for the detection of beta-Gal both in vitro and in vivo. Most of the reported molecular probes for the detection of beta-Gal consist of a galactopyranoside residue attached to a signalling unit through glycosidic bonds. The beta-Gal-induced hydrolysis of the glycosidic bonds released the signalling unit with remarkable changes in color and/or emission. Additional examples based on other approaches are also described. The wide applicability of these probes for the rapid and in situ detection of de-regulation beta-Gal-related diseases has boosted the research in this fertile fieldR.M laboratory members received the financial support from the Spanish Government (project RTI2018-100910-B-C41) and the Generalitat Valenciana (project PROMETEO 2018/024). B.L-T. received support from the Spanish Ministry of Economy for their PhD grants (FPU15/02707). J. F.-B received fellowship (CD19/00038)Lozano-Torres, B.; Blandez, JF.; Sancenón Galarza, F.; Martínez-Máñez, R. (2021). Chromo-fluorogenic probes for beta-galactosidase detection. Analytical and Bioanalytical Chemistry. 413(9):2361-2388. https://doi.org/10.1007/s00216-020-03111-8S236123884139Fernandes P. Enzymes in food processing: a condensed overview on strategies for better biocatalysts. Enzyme Res. 2010;2010:86253–73.Likidlilid A, Patchanans N, Peerapatdit T, Sriratanasathavorn C. Lipid peroxidation and antioxidant enzyme activities in erythrocytes of type 2 diabetic patients. J Med Assoc Thail. 2010;93(6):682–93.Pinto N, Dolan ME. Clinically relevant genetic variations in drug metabolizing enzymes. Curr Drug Metab. 2011;12(5):487–97.Giannini EG, Testa R, Savarinom V. Liver enzyme alteration: a guide for clinicians. CMAJ. 2005;172(3):367–79.Peters C, Shapiro EG, Krivit W. Hurler syndrome: past, present, and future. J Pediatr. 1998;133(1):7–9.Rodriguez M, O'Brien JS, Garrett RS, Powell HC. Canine GM1 gangliosidosis: an ultrastructural and biochemical study. J Neuropathol Exp Neurol. 1982;41(6):618–29.Cozma C, Eichler S, Wittmann G, Flores Bonet A, Kramp G, Giese AK, et al. Diagnosis of Morquio syndrome in dried blood spots based on a new MRM-MS assay. PLoS One. 2015;10(7):e0131228.Suzuki K, Suzuki Y. Globoid cell leucodystrophy (Krabbe's disease): deficiency of galactocerebroside beta-galactosidase. Proc Natl Acad Sci U S A. 1970;66(2):302–9.Holtzman D, Ulrich J. Senescent glia spell trouble in Alzheimer’s disease. Nat Neurosci. 2019;22(5):683–4.Robert L, Fulop T. Aging: facts and theories. Indian J Med Res. 2016;143(3):385–6.Dimri GP, Lee X, Basile G, Acosta M, Scott G, Roskelley C, et al. A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proc Natl Acad Sci U S A. 1995;92(20):9363–7.Biran A, Zada L, Karam PA, Vadai E, Roitman L, et al. Quantitative identification of senescent cells in aging and disease. Aging Cell. 2017;16(4):661–71.Grynkiewicz G, Poenie M, Tsien RY, Grynkiewicz G, Poenie M, Tsien RY. A new generation of Ca2+ indicators with greatly fluorescence properties. J Biol Chem. 1985;260(6):3440–50.de Silva AP, Gunaratne HQN, Gunnlaugsson T, Huxley AJ, McCoy CP, Rademacher JT, et al. Signaling recognition events with fluorescent sensors and switches. Chem Rev. 1997;97(5):1515–66.Que EL, Domaille DW, Chang CJ. Metals in neurobiology: probing their chemistry and biology with molecular imaging. Chem Rev. 2008;108(5):1517–49.Ueno T, Nagano T. Fluorescent probes for sensing and imaging. Nat Methods. 2011;8(8):642–5.Kobayashi H, Ogawa M, Alford R, Choyke PL, Urano Y. New strategies for fluorescent probe design in medical diagnostic imaging. Chem Rev. 2010;110(5):2620–40.Valeur B, Leray I. Design principles of fluorescent molecular sensors for cation recognition. Coord Chem Rev. 2000;205(1):3–40.Kim HM, Cho BR. Small-molecule two-photon probes for bioimaging applications. Chem Rev. 2015;115(11):5014–55.Huang J, Pu K. Activatable molecular probes for second near-infrared fluorescence, chemiluminescence, and photoacoustic imaging. Angew Chem Int Ed. 2020;59(29):11717–31.Miao Q, Pu K. Organic semiconducting agents for deep-tissue molecular imaging: second near-infrared fluorescence, self-luminescence, and photoacoustics. Adv Mater. 2018;30(49):e1801778.Cheng P, Miao Q, Li J, Huang J, Xie C, Pu K. Unimolecular chemo-fluoro-luminescent reporter for crosstalk-free duplex imaging of hepatotoxicity. J Am Chem Soc. 2019;141(27):10581–4.Wei H, Wu G, Tian X, Liu Z. Smart fluorescent probes for in situ imaging of enzyme activity: design strategies and applications. Future Med Chem. 2018;10(23):2729–44.Liu HW, Chen L, Xu C, Li Z, Zhang H, Zhang XB, et al. Recent progresses in small-molecule enzymatic fluorescent probes for cancer imaging. Chem Soc Rev. 2018;47(18):7140–80.Huang J, Li J, Lyu Y, Miao Q, Pu K. Molecular optical imaging probes for early diagnosis of drug-induced acute kidney injury. Nat Mater. 2019;18:1133–43.Roth ME, Green O, Gnaim S, Shabat D. Dendritic, oligomeric, and polymeric self-immolative molecular amplification. Chem Rev. 2016;116(3):1309–52.Zhang J, Cheng P, Pu K. Recent advances of molecular optical probes in imaging of β-galactosidase. Bioconjug Chem. 2019;30(8):2089–101.Rotman B. Measurement of activity of single molecules of β-D-galactosidase. Proc Natl Acad Sci U S A. 1961;47(12):1981–91.Rotman B, Zderic JA, Edelstein M. Fluorogenic substrates for beta-D-galactosidases and phosphatases derived from flurescein (3,6-dihydroxyfluoran) and its monomethylether. Proc Natl Acad Sci U S A. 1963;50(1):1–6.Mandal PK, Cattiaux L, Bensimon D, Mallet JM. Monogalactopyranosides of fluorescein and fluorescein methyl ester: synthesis, enzymatic hydrolysis by biotnylated β-galactosidase, and determination of translational diffusion coefficient. Carbohydr Res. 2012;358(40):40–6.Stracean R, Wooda J, Irschmann R. Synthesis and properties of 4-Methyl-2-oxo-1,2-benzopyran-7-yl β-D-galactoside (galactoside of 4-methylumbelliferone). J Org Chem. 1962;27(3):1074–5.Gee KR, Sun WC, Bhalgat KM, Upson RH, Klaubert DH, Latham KA, et al. Fluorogenic substrates based on fluorinated umbelliferones for continuous assays of phosphatases and beta-galactosidases. Anal Biochem. 1999;273(1):41–8.Chilvers KF, Perry JD, James AL, Reed RH. Synthesis and evaluation of novel fluorogenic substrates for the detection of bacterial beta-galactosidase. J Appl Microbiol. 2001;91(6):1118–30.Aizawa K. Studien über Carbohydrasen, I. I. Die fermentative Hydrolyse des p-nitrophenol-β-galactoside. Enzymologia. 1939;6:321–4.Na SY, Kim HJ. Fused oxazolidine-based dual optical probe for galactosidase with a dramatic chromogenic and fluorescence turn-on effect. Dyes Pigments. 2016;134:526–30.Corey PE, Trimmer RW, Biddlecom WG. A new chromogenic β-Galactosidase substrate: 7-β-D-galactopyranosyloxy-9,9-dimethyl-9H-acridin-2-one. Angew Chem Int Ed. 1991;30(12):1646–8.Wang P, Du J, Liu H, Bi G, Zhang G. Small quinolinium-based enzymatic probes via blue-to-red ratiometric fluorescence. Analyst. 2016;141:1483–7.Otsubo T, Minami A, Fujii H, Taguchi R, Takahashi T, Suzuki T, et al. 2-(Benzothiazol-2-yl)-phenyl-β-d-galactopyranoside derivatives as fluorescent pigment dyeing substrates and their application for the assay of β-d-galactosidase activities. Bioorg Med Chem Lett. 2013;23(7):2245–9.Sun C, Zhang X, Tanga M, Liu L, Shi L, Gao C, et al. New optical method for the determination of β-galactosidase and α-fetoprotein based on oxidase-like activity of fluorescein. Talanta. 194:164–70.Hirabayashi K, Hanaoka K, Takayanagi T, Toki Y, Egawa T, Kamiya M, et al. Analysis of chemical equilibrium of silicon-substituted fluorescein and its application to develop a scaffold for red fluorescent probes. Anal Chem. 2015;87(17):9061–9.Horwitz JP, Chua J, Curby RJ, Tomson AJ, Da Rooge MA, Fisher BE, et al. Substrates for cytochemical demonstration of enzyme activity. i. some substituted 3-Indolyl-β-D-glycopyranosides. Med Chem. 1964;7(4):574–5.Ho NH, Weissleder R, Tung CH. A self-immolative reporter for beta-galactosidase sensing. ChemBioChem. 2007;8(5):560–6.Huang Y, Feng H, Liu W, Zhang S, Tang C, Chen J, et al. Cation-driven luminescent self-assembled dots of copper nanoclusters with aggregation-induced emission for β-galactosidase activity monitoring. J Mater Chem B. 2017;5(26):5120–7.Xie X, Liana Y, Xiao L, Weia L. Facile and label-free fluorescence sensing of β-galactosidase activity by graphene quantum dots. Spectrochim Acta A Mol Biomol Spectrosc. 2020;240:118594.Hu Q, Ma K, Mei Y, He M, Kong J, Zhang X. Metal-to-ligand charge-transfer: applications to visual detection of β-galactosidase activity and sandwich immunoassay. Talanta. 2017;167:253–9.Urano Y, Kamiya M, Kanda K, Ueno T, Hirose K, Nagano T. Evolution of fluorescein as a platform for finely tunable fluorescence probes. J Am Chem Soc. 2005;127(13):4888–94.Komatsu T, Kikuchi K, Takakusa H, Hanaoka K, Ueno T, Kamiya M, et al. Design and synthesis of an enzyme activity-based labeling molecule with fluorescence spectral change. J Am Chem Soc. 2006;128(50):15946–7.Koide Y, Urano Y, Yatsushige A, Hanaoka K, Terai T, Nagano T. Design and development of enzymatically activatable photosensitizer based on unique characteristics of thiazole orange. J Am Chem Soc. 2009;131(17):6058–9.Egawa T, Koide Y, Hanaoka K, Komatsu T, Teraiab T, Nagano T. Development of a fluorescein analogue, TokyoMagenta, as a novel scaffold for fluorescence probes in red region. Chem Commun. 2011;47(14):4162–4.Kamiya M, Asanuma D, Kuranaga E, Takeishi A, Sakabe M, Miura M, et al. β-Galactosidase fluorescence probe with improved cellular accumulation based on a spirocyclized rhodol scaffold. J Am Chem Soc. 2011;133(33):12960–3.Han J, Han MS, Tung CH. A fluorogenic probe for β-galactosidase activity imaging in living cells. Mol BioSyst. 2013;9(12):3001–8.Peng L, Gao M, Cai X, Zhang R, Li K, Feng G, et al. A fluorescent light-up probe based on AIE and ESIPT processes for β-galactosidase activity detection and visualization in living cells. J Mater Chem B. 2015;3(47):9168–72.Tseng JC, Kung AL. In vivo imaging of endogenous enzyme activities using luminescent 1,2-dioxetane compounds. J Biomed Sci. 2015;22(1):45.Grimm JB, Gruber TD, Ortiz G, Brown TA, Lavis LD. Virginia Orange: a versatile, red-shifted fluorescein scaffold for single- and dual-input fluorogenic probes. Bioconjug Chem. 2016;27(2):474–80.Wei X, Hu XX, Zhang LL, Li J, Wang J. et al. Highly selective and sensitive FRET based ratiometric two-photon fluorescent probe for endogenous β-galactosidase detection in living cells and tissues Microchem. J. 2020;157:105046.Calatrava-Pérez E, Bright SA, Achermann S, Moylan C, Senge MO, Veale EB, et al. Glycosidase activated release of fluorescent 1,8-naphthalimide probes for tumor cell imaging from glycosylated pro-probes. Chem Commun. 2016;52(89):13086–9.Jiang G, Zeng G, Zhu W, Li Y, Dong X, Zhang G, et al. A selective and light-up fluorescent probe for β-galactosidase activity detection and imaging in living cells based on an AIE tetraphenylethylene derivative. Chem Commun. 2017;53(32):4505–8.Yang W, Zhao X, Zhang Y, Zhou Y, Fan S, Sheng H, et al. Hydroxyphenylquinazolinone-based turn-on fluorescent probe for β-galactosidase activity detection and application in living cells. Dyes Pigments. 2018;156:100–7.Li Y, Ning L, Yuan F, Zhang F, Zhang J, Xu Z, et al. Activatable formation of emissive excimers for highly selective detection of β-galactosidase. Anal Chem. 2020;92(8):5733–40.Huang J, Li N, Wang Q, Gu Y, Wang P. A lysosome-targetable and two-photon fluorescent probe for imaging endogenous β-galactosidase in living ovarian cancer cells. Sensor Actuat B-Chem. 2017;246:833–9.Chen X, Zhang X, Ma X, Zhang Y, Gao G, Liu J, et al. Novel fluorescent probe for rapid and ratiometric detection of β-galactosidase and live cell imaging. Talanta. 2019;192:308–13.Fu W, Yan C, Zhang Y, Ma Y, Guo Z, Zhu WH. Near-infrared aggregation-induced emission-active probe enables in situ and long-term tracking of endogenous β-galactosidase activity. Front Chem. 2019;7:291–302.Zhang X, Chen X, Zhang Y, Liu K, Shen H, et al. A near-infrared fluorescent probe for the ratiometric detection and living cell imaging of β-galactosidase. Anal Bioanal Chem. 2019;411:7957–66.Chen M, Mu L, Cao X, She G, Shi W. A novel ratiometric fluorescent probe for highly sensitive and selective detection of β-galactosidase in living cells. Chin J Chem. 2019;37(4):330–6.Kong X, Li M, Dong B, Yin Y, Song W, Lin W. An ultrasensitivity fluorescent probe based on the ict-fret dual mechanisms for imaging β-galactosidase in vitro and ex vivo. Anal Chem. 2019;91(24):15591–8.Lee HW, Lim CS, Choi H, Cho MK, Noh CH, Lee K, et al. Discrimination between human colorectal neoplasms with a dual-recognitive two-photon probe. Anal Chem. 2019;91(22):14705–11.Zhao X, Yang W, Fan S, Zhou Y, Sheng H, Cao Y, et al. A hemicyanine-based colorimetric turn-on fluorescent probe for β-galactosidase activity detection and application in living cells. J Lumin. 2019;205:310–7.Li X, Pan Y, Chen H, Duan Y, Zhou S, Wu W, et al. Specific near-infrared probe for ultrafast imaging of lysosomal β-galactosidase in ovarian cancer cells. Anal Chem. 2020;92(8):5772–9.Long R, Tang C, Yang Z, Fu Q, Xu J, Tong C, et al. A natural hyperoside based novel light-up fluorescent probe with AIE and ESIPT characteristics for on-site and long-term imaging of β-galactosidase in living cells. J Mater Chem C. 2020;8(34):11860–5.Tang C, Zhou J, Qian Z, Ma Y, Huang Y, Feng H. A universal fluorometric assay strategy for glycosidases based on functional carbon quantum dots: β-galactosidase activity detection in vitro and in living cells. J Mater Chem B. 2017;5(10):1971–9.Wang W, Vellaisamy K, Li W, Wu C, Ko CN, Leung CL, et al. Development of a long-lived luminescence probe for visualizing β-galactosidase in ovarian carcinoma cells. Anal Chem. 2017;89(21):11679–84.James AL, Perry JD, Ford M, Armstrong L, Gould FK. Evaluation of cyclohexenoesculetin-beta-D-galactoside and 8-hydroxyquinoline-beta-D-galactoside as substrates for the detection of beta-galactosidase. Appl Environ Microbiol. 1996;62(10):3868–70.James AL, Perry JD, Chilvers K, Robson IS, Armstrong L, Orr KE. Alizarin-beta-D-galactoside: a new substrate for the detection of bacterial beta-galactosidase. Lett Appl Microbiol. 2000;30(4):336–40.Wei X, Wu Q, Zhang J, Zhang Y, Guo W, Chen M, et al. Synthesis of precipitating chromogenic/fluorogenic β-glucosidase/β-galactosidase substrates by a new method and their application in the visual detection of foodborne pathogenic bacteria. Chem Commun. 2017;53(1):103–6.Muñoz-Espín D, Serrano M. Cellular senescence: from physiology to pathology. Nat Rev Mol Cell Biol. 2014;15(7):482–96.Filho MS, Dao P, Gesson M, Martin AR, Benhida R. Development of highly sensitive fluorescent probes for the detection of β-galactosidase activity- application to the real-time monitoring of senescence in live cells. Analyst. 2018;143(11):2680–8.Kim EJ, Podder A, Maiti M, Lee JM, Chung BG, Bhuniya S. Selective monitoring of vascular cell senescence via β-Galactosidase detection with a fluorescent chemosensor. Sensor Actuat B-Chem. 2018;274:194–200.Jiang J, Tan Q, Zhao S, Song H, Hua L, Xie H. Late-stage difluoromethylation leading to a self-immobilizing fluorogenic probe for the visualization of enzyme activities in live cells. Chem Commun. 2019;55(99):15000–3.Qiu W, Li X, Shi D, Li X, Gao Y, Li J, et al. A rapid-response near-infrared fluorescent probe with large Stokes shift for senescence-associated β-galactosidase activity detection and imaging of senescent cells. Dyes Pigments. 2020;182(99):108657.Makau JN, Kitagawa A, Kitamura K, Yamaguchi T, Mizuta S. Design and development of an HBT-based ratiometric fluorescent probe to monitor stress-induced premature senescence. ACS Omega. 2020;5:11299–307.Senter PD, Saulnier MG, Schreiber GJ, Hirschberg DL, Brown JP, Hellström I, et al. Antitumor effect of antibody-alkaline phosphatase conjugates in combination with etoposide phosphate. Proc Natl Acad Sci U S A. 1988;85(13):4842–6.Senter PD, Springer CJ. Selective activation of anticancer prodrugs by monoclonal antibody-enzyme conjugates. Adv Drug Deliv Rev. 2001;53(3):247–64.Gu K, Xu Y, Li H, Guo Z, Zhu S, Shi P, et al. Real-time tracking and in vivo visualization of β-galactosidase activity in colorectal tumor with a ratiometric near-infrared fluorescent probe. J Am Chem Soc. 2016;138(16):5334–40.Tung CH, Zeng Q, Shah K, Kim DE, Schellingerhout D, Weissleder R. In vivo imaging of beta-galactosidase activity using far red fluorescent switch. Cancer Res. 2004;64(5):1579–83.Wehrman TS, von Degenfeld G, Krutzik PO, Nolan GP, Blau HM. Luminescent imaging of beta-galactosidase activity in living subjects using sequential reporter-enzyme luminescence. Nat Methods. 2006;3(4):295–301.Oushiki D, Kojima H, Takahashi Y, Komatsu T, Terai T, Hanaoka K, et al. Near-infrared fluorescence probes for enzymes based on binding affinity modulation of squarylium dye scaffold. Anal Chem. 2012;84(10):4404–10.Zhang XX, Wu H, Li P, Qu ZJ, Tan MQ, Han KL. A versatile two-photon fluorescent probe for ratiometric imaging E. coliβ-galactosidase in live cells and in vivo. Chem Commun. 2016;52(53):8283–6.Kim EJ, Kumar R, Sharma A, Yoon B, Kim HM, Lee H, et al. In vivo imaging of β-galactosidase stimulated activity in hepatocellular carcinoma using ligand-targeted fluorescent probe. Biomaterials. 2017;122:83–90.Shi L, Yan C, Ma Y, Wang T, Guo Z, Zhu WH. In vivo ratiometric tracking of endogenous β-galactosidase activity using an activatable near-infrared fluorescent probe. Chem Commun. 2019;55(82):12308–11.Zhen X, Zhang J, Huang J, Xie C, Miao Q, Pu K. Macrotheranostic probe with disease-activated near-infrared fluorescence, photoacoustic, and photothermal signals for imaging-guided therapy. Angew Chem Int Ed. 2018;57(26):7804–8.Li Z, Ren M, Wang L, Dai L, Lin W. Development of a red-emissive two-photon fluorescent probe for sensitive detection of beta-galactosidase in vitro and in vivo. Sensor Actuat B-Chem. 2020;307:127643.González-Gualda E, Pàez-Ribes M, Lozano-Torres B, Macias D, Wilson JR 3rd, González-López C, et al. Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity. Aging Cell. 2020;19(4):e13142.Lozano-Torres B, Galiana I, Rovira M, Garrido E, Chaib S, Bernardos A, et al. An OFF–ON two-photon fluorescent probe for tracking cell senescence in vivo. J Am Chem Soc. 2017;139(26):8808–11.Lozano-Torres B, Blandez JF, Galiana I, García-Fernández A, Alfonso M, Marcos MD, et al. Real-time in vivo detection of cellular senescence through the controlled release of the NIR fluorescent dye Nile blue. Angew Chem Int Ed. 2020;59(35):5152–6.Wang Y, Liu J, Ma X, Cui C, Deenik PR, Henderson KP, et al. Real-time imaging of senescence in tumors with DNA damage. Sci Rep. 2019;9:2102.Chen JA, Guo W, Wang Z, Sun N, Pan H, Tan J, et al. In vivo imaging of senescent vascular cells in atherosclerotic mice using a β-galactosidase-activatable nanoprobe. Anal Chem. 2020;92(18):12613–21.Liu J, Ma X, Cui C, Wang Y, Deenik PR, Cui L. A self-immobilizing NIR probe for non-invasive imaging of senescence. bioRxiv. 2020. https://doi.org/10.1101/2020.03.27.010827.Aznar E, Oroval M, Pascual L, Murguía JR, Martínez-Máñez R, Sancenón F. Gated materials for on-command release of guest molecules. Chem Rev. 2016;116(2):561–718.García-Fernández A, Aznar E, Martínez-Máñez R, Sancenón F. New advances in in vivo applications of gated mesoporous silica as drug delivery nanocarriers. Small. 2020;16(3):1902242–304.Coll C, Bernardos A, Martínez-Máñez R, Sancenón F. Gated silica mesoporous supports for controlled release and signaling applications. Acc Chem Res. 2013;46(2):339–49.Muñoz-Espín D, Rovira M, Galiana I, Giménez C, Lozano-Torres B, Paez-Ribes M. A versatile drug delivery system targeting senescent cells. EMBO Mol Med. 2018;10(9):e9355.Lozano-Torres B, Estepa-Fernández A, Rovira M, Orzáez M, Serrano M, Martínez-Máñez R, et al. The chemistry of senescence. Nat Rev Chem. 2019;3:426–41.Mazur A, Kro’l JE, Marczak M, Skorupska A. Membrane topology of PssT, the transmembrane protein component of the type I exopolysaccharide transport system in rhizobium leguminosarum bv trifolii strain TA1. J Bacteriol. 2003;85(8):2503–11.Agostini A, Mondragón L, Bernardos A, Martínez-Máñez R, Marcos MD, Sancenón F, et al. Targeted cargo delivery in senescent cells using capped mesoporous silica nanoparticles. Angew Chem Int Ed. 2012;51(42):10556–60.Asanuma D, Sakabe M, Kamiya M, Yamamoto K, Hiratake J, Ogawa M, et al. Sensitive β-galactosidase-targeting fluorescence probe for visualizing small peritoneal metastatic tumours in vivo. Nat Commun. 2015;6:6463.Sakabe M, Asanuma D, Kamiya M, Iwatate RI, Hanaoka K, Terai T, et al. Rational design of highly sensitive fluorescence probes for protease and glycosidase based on precisely controlled spirocyclization. J Am Chem Soc. 2013;135(1):409–14.Doura T, Kamiya M, Obata F, Yamaguchi Y, Hiyama TY, Matsuda T, et al. Detection of LacZ-positive cells in living tissue with single-cell resolution. Angew Chem Int Ed. 2016;55(33):9620–4.Calado RT, Young NS. Telomere diseases. N Engl J Med. 2009;361:2353–65.Chatterjee SK, Bhattacharya M, Barlow JJ. Glycosyltransferase and glycosidase activities in ovarian cancer

    Influence of the organic linker substituent on the catalytic activity of MIL-101(Cr) for the oxidative coupling of benzylamines to imines

    Full text link
    [EN] MIL-101(Cr) having substituents at the terephthalate linker (X = H, NO2, SO3H, Cl, CH3 and NH2) promotes the aerobic oxidation of benzylamines to the corresponding N-benzylidene benzylamines at different rates. MIL-101(Cr)¿NO2 was the most active catalyst, about 6-fold more active than the parent MIL-101(Cr). MIL-101(Cr)¿NO2 does not deactivate significantly upon five consecutive reuses, does not leach the metal to the solution and maintains its crystallinity. MIL-101(Cr)¿NO2 is active for a wide range of benzylamines including para-substituted, heterocyclic benzylamines and di- and tribenzylamines.Financial support by the Spanish Ministry of Economy and Competitiveness (CTQ 2015-69153-CO2-1, CTQ2014-53292-R, Severo Ochoa) and Generalitat Valenciana (Prometeo 2013014) is gratefully acknowledged.Santiago-Portillo, A.; Blandez, JF.; Navalón Oltra, S.; Alvaro Rodríguez, MM.; García Gómez, H. (2017). Influence of the organic linker substituent on the catalytic activity of MIL-101(Cr) for the oxidative coupling of benzylamines to imines. Catalysis Science & Technology. 7(6):1351-1362. https://doi.org/10.1039/c6cy02577cS135113627

    Real-Time In Vivo Detection of Cellular Senescence through the Controlled Release of the NIR Fluorescent Dye Nile Blue

    Full text link
    This is the peer reviewed version of the following article: B. Lozano-Torres, J. F. Blandez, I. Galiana, A. García-Fernández, M. Alfonso, M. D. Marcos, M. Orzáez, F. Sancenón, R. Martínez-Máñez, Angew. Chem. Int. Ed. 2020, 59, 15152., which has been published in final form at https://doi.org/10.1002/anie.202004142. This article may be used for non-commercial purposes in accordance with Wiley Terms and Conditions for Self-Archiving.[EN] In vivo detection of cellular senescence is accomplished by using mesoporous silica nanoparticles loaded with the NIR-FDA approved Nile blue (NB) dye and capped with a galactohexasaccharide (S3). NB emission at 672 nm is highly quenched inside S3, yet a remarkable emission enhancement is observed upon cap hydrolysis in the presence of beta-galactosidase and dye release. The efficacy of the probe to detect cellular senescence is tested in vitro in melanoma SK-Mel-103 and breast cancer 4T1 cells and in vivo in palbociclib-treated BALB/cByJ mice bearing breast cancer tumor.R.M. thanks financial support from the Spanish Government (RTI2018-100910-B-C41 and RTI2018-101599-B-C22 (MCUI/AEI/FEDER, UE)) and the Generalitat Valenciana (PROMETEO 2018/024). M.O. thanks the financial support from SAF2017-84689-R project and MINECO/AEI/FEDER, UE and the Generalitat Valenciana (PROMETEO/2019/065). B.L.-T. is grateful to the Spanish Ministry of Economy for her PhD grant. I.G. thanks her contract from IDM. J.F.-B. and M.A. thank the UPV for their postdoctoral fellowship.Lozano-Torres, B.; Blandez, JF.; Galiana, I.; García-Fernández, A.; Alfonso-Navarro, M.; Marcos Martínez, MD.; Orzáez, M.... (2020). Real-Time In Vivo Detection of Cellular Senescence through the Controlled Release of the NIR Fluorescent Dye Nile Blue. Angewandte Chemie International Edition. 59(35):15152-15156. https://doi.org/10.1002/anie.202004142S15152151565935He, S., & Sharpless, N. E. (2017). Senescence in Health and Disease. Cell, 169(6), 1000-1011. doi:10.1016/j.cell.2017.05.015Lozano-Torres, B., Estepa-Fernández, A., Rovira, M., Orzáez, M., Serrano, M., Martínez-Máñez, R., & Sancenón, F. (2019). The chemistry of senescence. Nature Reviews Chemistry, 3(7), 426-441. doi:10.1038/s41570-019-0108-0Muñoz-Espín, D., & Serrano, M. (2014). Cellular senescence: from physiology to pathology. Nature Reviews Molecular Cell Biology, 15(7), 482-496. doi:10.1038/nrm3823Hernandez-Segura, A., Nehme, J., & Demaria, M. (2018). Hallmarks of Cellular Senescence. Trends in Cell Biology, 28(6), 436-453. doi:10.1016/j.tcb.2018.02.001Baker, D. J., Childs, B. G., Durik, M., Wijers, M. E., Sieben, C. J., Zhong, J., … van Deursen, J. M. (2016). Naturally occurring p16Ink4a-positive cells shorten healthy lifespan. Nature, 530(7589), 184-189. doi:10.1038/nature16932Soto-Gamez, A., & Demaria, M. (2017). Therapeutic interventions for aging: the case of cellular senescence. Drug Discovery Today, 22(5), 786-795. doi:10.1016/j.drudis.2017.01.004Kirkland, J. L., Tchkonia, T., Zhu, Y., Niedernhofer, L. J., & Robbins, P. D. (2017). The Clinical Potential of Senolytic Drugs. Journal of the American Geriatrics Society, 65(10), 2297-2301. doi:10.1111/jgs.14969Niedernhofer, L. J., & Robbins, P. D. (2018). Senotherapeutics for healthy ageing. Nature Reviews Drug Discovery, 17(5), 377-377. doi:10.1038/nrd.2018.44Hayflick, L., & Moorhead, P. S. (1961). The serial cultivation of human diploid cell strains. Experimental Cell Research, 25(3), 585-621. doi:10.1016/0014-4827(61)90192-6Zhang, R., & Adams, P. D. (2007). Heterochromatin and its Relationship to Cell Senescence and Cancer Therapy. Cell Cycle, 6(7), 784-789. doi:10.4161/cc.6.7.4079Campisi, J. (2005). Senescent Cells, Tumor Suppression, and Organismal Aging: Good Citizens, Bad Neighbors. Cell, 120(4), 513-522. doi:10.1016/j.cell.2005.02.003Dimri, G. P., Lee, X., Basile, G., Acosta, M., Scott, G., Roskelley, C., … Pereira-Smith, O. (1995). A biomarker that identifies senescent human cells in culture and in aging skin in vivo. Proceedings of the National Academy of Sciences, 92(20), 9363-9367. doi:10.1073/pnas.92.20.9363Lozano-Torres, B., Galiana, I., Rovira, M., Garrido, E., Chaib, S., Bernardos, A., … Sancenón, F. (2017). An OFF–ON Two-Photon Fluorescent Probe for Tracking Cell Senescence in Vivo. Journal of the American Chemical Society, 139(26), 8808-8811. doi:10.1021/jacs.7b04985Asanuma, D., Sakabe, M., Kamiya, M., Yamamoto, K., Hiratake, J., Ogawa, M., … Urano, Y. (2015). Sensitive β-galactosidase-targeting fluorescence probe for visualizing small peritoneal metastatic tumours in vivo. Nature Communications, 6(1). doi:10.1038/ncomms7463Muñoz-Espín, D. (2019). Nanocarriers targeting senescent cells. Translational Medicine of Aging, 3, 1-5. doi:10.1016/j.tma.2019.01.001Ekpenyong-Akiba, A. E., Canfarotta, F., Abd H., B., Poblocka, M., Casulleras, M., Castilla-Vallmanya, L., … Macip, S. (2019). Detecting and targeting senescent cells using molecularly imprinted nanoparticles. Nanoscale Horizons, 4(3), 757-768. doi:10.1039/c8nh00473kAlberti, S., Soler-Illia, G. J. A. A., & Azzaroni, O. (2015). Gated supramolecular chemistry in hybrid mesoporous silica nanoarchitectures: controlled delivery and molecular transport in response to chemical, physical and biological stimuli. Chemical Communications, 51(28), 6050-6075. doi:10.1039/c4cc10414eDe la Torre, C., Casanova, I., Acosta, G., Coll, C., Moreno, M. J., Albericio, F., … Martínez-Máñez, R. (2014). Gated Mesoporous Silica Nanoparticles Using a Double-Role Circular Peptide for the Controlled and Target-Preferential Release of Doxorubicin in CXCR4-Expresing Lymphoma Cells. Advanced Functional Materials, 25(5), 687-695. doi:10.1002/adfm.201403822Bernardos, A., Aznar, E., Marcos, M. D., Martínez-Máñez, R., Sancenón, F., Soto, J., … Amorós, P. (2009). Enzyme-Responsive Controlled Release Using Mesoporous Silica Supports Capped with Lactose. Angewandte Chemie International Edition, 48(32), 5884-5887. doi:10.1002/anie.200900880Bernardos, A., Aznar, E., Marcos, M. D., Martínez-Máñez, R., Sancenón, F., Soto, J., … Amorós, P. (2009). Enzyme-Responsive Controlled Release Using Mesoporous Silica Supports Capped with Lactose. Angewandte Chemie, 121(32), 5998-6001. doi:10.1002/ange.200900880Agostini, A., Mondragón, L., Bernardos, A., Martínez-Máñez, R., Marcos, M. D., Sancenón, F., … Murguía, J. R. (2012). Targeted Cargo Delivery in Senescent Cells Using Capped Mesoporous Silica Nanoparticles. Angewandte Chemie International Edition, 51(42), 10556-10560. doi:10.1002/anie.201204663Agostini, A., Mondragón, L., Bernardos, A., Martínez-Máñez, R., Marcos, M. D., Sancenón, F., … Murguía, J. R. (2012). Targeted Cargo Delivery in Senescent Cells Using Capped Mesoporous Silica Nanoparticles. Angewandte Chemie, 124(42), 10708-10712. doi:10.1002/ange.201204663Muñoz‐Espín, D., Rovira, M., Galiana, I., Giménez, C., Lozano‐Torres, B., Paez‐Ribes, M., … Serrano, M. (2018). A versatile drug delivery system targeting senescent cells. EMBO Molecular Medicine, 10(9). doi:10.15252/emmm.201809355Mérian, J., Gravier, J., Navarro, F., & Texier, I. (2012). Fluorescent Nanoprobes Dedicated to in Vivo Imaging: From Preclinical Validations to Clinical Translation. Molecules, 17(5), 5564-5591. doi:10.3390/molecules17055564Fu, W., Yan, C., Guo, Z., Zhang, J., Zhang, H., Tian, H., & Zhu, W.-H. (2019). Rational Design of Near-Infrared Aggregation-Induced-Emission-Active Probes: In Situ Mapping of Amyloid-β Plaques with Ultrasensitivity and High-Fidelity. Journal of the American Chemical Society, 141(7), 3171-3177. doi:10.1021/jacs.8b12820Ovchinnikov, O. V., Evtukhova, A. V., Kondratenko, T. S., Smirnov, M. S., Khokhlov, V. Y., & Erina, O. V. (2016). Manifestation of intermolecular interactions in FTIR spectra of methylene blue molecules. Vibrational Spectroscopy, 86, 181-189. doi:10.1016/j.vibspec.2016.06.016Aznar, E., Oroval, M., Pascual, L., Murguía, J. R., Martínez-Máñez, R., & Sancenón, F. (2016). Gated Materials for On-Command Release of Guest Molecules. Chemical Reviews, 116(2), 561-718. doi:10.1021/acs.chemrev.5b00456Llopis-Lorente, A., Lozano-Torres, B., Bernardos, A., Martínez-Máñez, R., & Sancenón, F. (2017). Mesoporous silica materials for controlled delivery based on enzymes. Journal of Materials Chemistry B, 5(17), 3069-3083. doi:10.1039/c7tb00348jGarcía‐Fernández, A., Aznar, E., Martínez‐Máñez, R., & Sancenón, F. (2019). New Advances in In Vivo Applications of Gated Mesoporous Silica as Drug Delivery Nanocarriers. Small, 16(3), 1902242. doi:10.1002/smll.201902242Kozlovskaya, V., Xue, B., & Kharlampieva, E. (2016). Shape-Adaptable Polymeric Particles for Controlled Delivery. Macromolecules, 49(22), 8373-8386. doi:10.1021/acs.macromol.6b01740Mishra, D. K., Shandilya, R., & Mishra, P. K. (2018). Lipid based nanocarriers: a translational perspective. Nanomedicine: Nanotechnology, Biology and Medicine, 14(7), 2023-2050. doi:10.1016/j.nano.2018.05.021Seidi, F., Jenjob, R., Phakkeeree, T., & Crespy, D. (2018). Saccharides, oligosaccharides, and polysaccharides nanoparticles for biomedical applications. Journal of Controlled Release, 284, 188-212. doi:10.1016/j.jconrel.2018.06.026Chen, W., Zhou, S., Ge, L., Wu, W., & Jiang, X. (2018). Translatable High Drug Loading Drug Delivery Systems Based on Biocompatible Polymer Nanocarriers. Biomacromolecules, 19(6), 1732-1745. doi:10.1021/acs.biomac.8b00218Vázquez-González, M., & Willner, I. (2018). DNA-Responsive SiO2 Nanoparticles, Metal–Organic Frameworks, and Microcapsules for Controlled Drug Release. Langmuir, 34(49), 14692-14710. doi:10.1021/acs.langmuir.8b00478Farid, R. M., Youssef, N. A. H. A., & Kassem, A. A. (2018). Platform for Lipid Based Nanocarriers’ Formulation Components and their Potential Effects: A Literature Review. Current Pharmaceutical Design, 23(43), 6613-6629. doi:10.2174/1381612824666171128104814Lombardo, D., Calandra, P., Barreca, D., Magazù, S., & Kiselev, M. (2016). Soft Interaction in Liposome Nanocarriers for Therapeutic Drug Delivery. Nanomaterials, 6(7), 125. doi:10.3390/nano6070125Bansal, A., & Zhang, Y. (2014). Photocontrolled Nanoparticle Delivery Systems for Biomedical Applications. Accounts of Chemical Research, 47(10), 3052-3060. doi:10.1021/ar500217wKamaly, N., Yameen, B., Wu, J., & Farokhzad, O. C. (2016). Degradable Controlled-Release Polymers and Polymeric Nanoparticles: Mechanisms of Controlling Drug Release. Chemical Reviews, 116(4), 2602-2663. doi:10.1021/acs.chemrev.5b00346Trewyn, B. G., Slowing, I. I., Giri, S., Chen, H.-T., & Lin, V. S.-Y. (2007). Synthesis and Functionalization of a Mesoporous Silica Nanoparticle Based on the Sol–Gel Process and Applications in Controlled Release. Accounts of Chemical Research, 40(9), 846-853. doi:10.1021/ar600032

    Electro-responsive films containing voltage responsive gated mesoporous silica nanoparticles grafted onto PEDOT-based conducting polymer

    Full text link
    [EN] The characteristics and electromechanical properties of conductive polymers together to their biocompatibility have boosted their application as a suitable tool in regenerative medicine and tissue engineering. However, conducting polymers as drug release materials are far from being ideal. A possibility to overcome this drawback is to combine conducting polymers with on-command delivery particles with inherent high-loading capacity. In this scenario, we report here the preparation of conduction polymers containing gated mesoporous silica nanoparticles (MSN) loaded with a cargo that is delivered on command by electro-chemical stimuli increasing the potential use of conducting polymers as controlled delivery systems. MSNs are loaded with Rhodamine B (Rh B), anchored to the conductive polymer poly(3,4-ethylenedioxythiophene) (PEDOT) doped with poly[(4-styrenesulfonic acid)-co-(maleic acid)], functionalized with a bipyridinium derivative and pores are capped with heparin (P3) by electrostatic interactions. P3 releases the entrapped cargo after the application of ¿640 mV voltage versus the saturated calomel electrode (SCE). Pore opening in the nanoparticles and dye delivery is ascribed to both (i) the reduction of the grafted bipyridinium derivative and (ii) the polarization of the conducting polymer electrode to negative potentials that induce detachment of positively charged heparin from the surface of the nanoparticles. Biocompatibility and cargo release studies were carried out in HeLa cells cultures.Alba Garcia-Fernandez, Beatriz Lozano-Torres contributed equally to this work. A. Garcia-Fernandez and B. Lozano-Torres are grateful to the "Ministerio de Economia y Competitividad" of the Spanish Government for her PhD fellowships. J. F. Blandez thanks the "Universitat Politecnica de Valencia" for his postdoctoral fellowship (PAID-10-17). The authors thank to the Spanish Government (Projects RTI2018-100910-B-C41 and RTI2018-101599-B-C22 (MCUI/AEI/FEDER, EU)) and the Generalitat Valencia (Project PROMETEO2018-024) for support.García-Fernández, A.; Lozano-Torres, B.; Blandez, JF.; Monreal-Trigo, J.; Soto Camino, J.; Collazos-Castro, JE.; Alcañiz Fillol, M.... (2020). Electro-responsive films containing voltage responsive gated mesoporous silica nanoparticles grafted onto PEDOT-based conducting polymer. Journal of Controlled Release. 323:421-430. https://doi.org/10.1016/j.jconrel.2020.04.048S421430323Aznar, E., Oroval, M., Pascual, L., Murguía, J. R., Martínez-Máñez, R., & Sancenón, F. (2016). Gated Materials for On-Command Release of Guest Molecules. Chemical Reviews, 116(2), 561-718. doi:10.1021/acs.chemrev.5b00456Mura, S., Nicolas, J., & Couvreur, P. (2013). Stimuli-responsive nanocarriers for drug delivery. Nature Materials, 12(11), 991-1003. doi:10.1038/nmat3776Llopis-Lorente, A., Lozano-Torres, B., Bernardos, A., Martínez-Máñez, R., & Sancenón, F. (2017). Mesoporous silica materials for controlled delivery based on enzymes. Journal of Materials Chemistry B, 5(17), 3069-3083. doi:10.1039/c7tb00348jTarn, D., Ashley, C. E., Xue, M., Carnes, E. C., Zink, J. I., & Brinker, C. J. (2013). Mesoporous Silica Nanoparticle Nanocarriers: Biofunctionality and Biocompatibility. Accounts of Chemical Research, 46(3), 792-801. doi:10.1021/ar3000986Mauriello Jimenez, C., Aggad, D., Croissant, J. G., Tresfield, K., Laurencin, D., Berthomieu, D., … Durand, J.-O. (2018). Porous Porphyrin-Based Organosilica Nanoparticles for NIR Two-Photon Photodynamic Therapy and Gene Delivery in Zebrafish. Advanced Functional Materials, 28(21), 1800235. doi:10.1002/adfm.201800235Alberti, S., Soler-Illia, G. J. A. A., & Azzaroni, O. (2015). Gated supramolecular chemistry in hybrid mesoporous silica nanoarchitectures: controlled delivery and molecular transport in response to chemical, physical and biological stimuli. Chemical Communications, 51(28), 6050-6075. doi:10.1039/c4cc10414eLlopis-Lorente, A., de Luis, B., García-Fernández, A., Jimenez-Falcao, S., Orzáez, M., Sancenón, F., … Martínez-Máñez, R. (2018). Hybrid Mesoporous Nanocarriers Act by Processing Logic Tasks: Toward the Design of Nanobots Capable of Reading Information from the Environment. ACS Applied Materials & Interfaces, 10(31), 26494-26500. doi:10.1021/acsami.8b05920Yang, P., Gai, S., & Lin, J. (2012). Functionalized mesoporous silica materials for controlled drug delivery. Chemical Society Reviews, 41(9), 3679. doi:10.1039/c2cs15308dSong, N., & Yang, Y.-W. (2015). Molecular and supramolecular switches on mesoporous silica nanoparticles. Chemical Society Reviews, 44(11), 3474-3504. doi:10.1039/c5cs00243eOroval, M., Díez, P., Aznar, E., Coll, C., Marcos, M. D., Sancenón, F., … Martínez-Máñez, R. (2016). Self-Regulated Glucose-Sensitive Neoglycoenzyme-Capped Mesoporous Silica Nanoparticles for Insulin Delivery. Chemistry - A European Journal, 23(6), 1353-1360. doi:10.1002/chem.201604104De la Torre, C., Domínguez-Berrocal, L., Murguía, J. R., Marcos, M. D., Martínez-Máñez, R., Bravo, J., & Sancenón, F. (2018). ϵ -Polylysine-Capped Mesoporous Silica Nanoparticles as Carrier of the C 9h Peptide to Induce Apoptosis in Cancer Cells. Chemistry - A European Journal, 24(8), 1890-1897. doi:10.1002/chem.201704161Llopis-Lorente, A., Díez, P., Sánchez, A., Marcos, M. D., Sancenón, F., Martínez-Ruiz, P., … Martínez-Máñez, R. (2017). Interactive models of communication at the nanoscale using nanoparticles that talk to one another. Nature Communications, 8(1). doi:10.1038/ncomms15511Pascual, L., Baroja, I., Aznar, E., Sancenón, F., Marcos, M. D., Murguía, J. R., … Martínez-Máñez, R. (2015). Oligonucleotide-capped mesoporous silica nanoparticles as DNA-responsive dye delivery systems for genomic DNA detection. Chemical Communications, 51(8), 1414-1416. doi:10.1039/c4cc08306gArgyo, C., Weiss, V., Bräuchle, C., & Bein, T. (2013). Multifunctional Mesoporous Silica Nanoparticles as a Universal Platform for Drug Delivery. Chemistry of Materials, 26(1), 435-451. doi:10.1021/cm402592tLi, Z., Barnes, J. C., Bosoy, A., Stoddart, J. F., & Zink, J. I. (2012). Mesoporous silica nanoparticles in biomedical applications. Chemical Society Reviews, 41(7), 2590. doi:10.1039/c1cs15246gKumar, P., Tambe, P., Paknikar, K. M., & Gajbhiye, V. (2018). Mesoporous silica nanoparticles as cutting-edge theranostics: Advancement from merely a carrier to tailor-made smart delivery platform. Journal of Controlled Release, 287, 35-57. doi:10.1016/j.jconrel.2018.08.024Lai, C.-Y., Trewyn, B. G., Jeftinija, D. M., Jeftinija, K., Xu, S., Jeftinija, S., & Lin, V. S.-Y. (2003). A Mesoporous Silica Nanosphere-Based Carrier System with Chemically Removable CdS Nanoparticle Caps for Stimuli-Responsive Controlled Release of Neurotransmitters and Drug Molecules. Journal of the American Chemical Society, 125(15), 4451-4459. doi:10.1021/ja028650lLiu, R., Zhao, X., Wu, T., & Feng, P. (2008). Tunable Redox-Responsive Hybrid Nanogated Ensembles. Journal of the American Chemical Society, 130(44), 14418-14419. doi:10.1021/ja8060886Qu, H., Yang, L., Yu, J., Dong, T., Rong, M., Zhang, J., … Liu, H. (2017). A redox responsive controlled release system using mesoporous silica nanoparticles capped with Au nanoparticles. RSC Advances, 7(57), 35704-35710. doi:10.1039/c7ra04444eGiménez, C., de la Torre, C., Gorbe, M., Aznar, E., Sancenón, F., Murguía, J. R., … Amorós, P. (2015). Gated Mesoporous Silica Nanoparticles for the Controlled Delivery of Drugs in Cancer Cells. Langmuir, 31(12), 3753-3762. doi:10.1021/acs.langmuir.5b00139Luo, Z., Hu, Y., Cai, K., Ding, X., Zhang, Q., Li, M., … Zhao, Y. (2014). Intracellular redox-activated anticancer drug delivery by functionalized hollow mesoporous silica nanoreservoirs with tumor specificity. Biomaterials, 35(27), 7951-7962. doi:10.1016/j.biomaterials.2014.05.058Du, X., Xiong, L., Dai, S., Kleitz, F., & Qiao, S. Z. (2014). Intracellular Microenvironment-Responsive Dendrimer-Like Mesoporous Nanohybrids for Traceable, Effective, and Safe Gene Delivery. Advanced Functional Materials, 24(48), 7627-7637. doi:10.1002/adfm.201402408Raza, A., Hayat, U., Rasheed, T., Bilal, M., & Iqbal, H. M. N. (2018). Redox-responsive nano-carriers as tumor-targeted drug delivery systems. European Journal of Medicinal Chemistry, 157, 705-715. doi:10.1016/j.ejmech.2018.08.034Xiao, Y., Wang, T., Cao, Y., Wang, X., Zhang, Y., Liu, Y., & Huo, Q. (2015). Enzyme and voltage stimuli-responsive controlled release system based on β-cyclodextrin-capped mesoporous silica nanoparticles. Dalton Transactions, 44(9), 4355-4361. doi:10.1039/c4dt03758hWang, T., Sun, G., Wang, M., Zhou, B., & Fu, J. (2015). Voltage/pH-Driven Mechanized Silica Nanoparticles for the Multimodal Controlled Release of Drugs. ACS Applied Materials & Interfaces, 7(38), 21295-21304. doi:10.1021/acsami.5b05619Jiao, X., Sun, R., Cheng, Y., Li, F., Du, X., Wen, Y., … Zhang, X. (2017). A Voltage-Responsive Free-Blockage Controlled-Release System Based on Hydrophobicity Switching. ChemPhysChem, 18(10), 1317-1323. doi:10.1002/cphc.201700117Khashab, N. M., Trabolsi, A., Lau, Y. A., Ambrogio, M. W., Friedman, D. C., Khatib, H. A., … Stoddart, J. F. (2009). Redox- and pH-Controlled Mechanized Nanoparticles. European Journal of Organic Chemistry, 2009(11), 1669-1673. doi:10.1002/ejoc.200801300Guimard, N. K., Gomez, N., & Schmidt, C. E. (2007). Conducting polymers in biomedical engineering. Progress in Polymer Science, 32(8-9), 876-921. doi:10.1016/j.progpolymsci.2007.05.012Wang, X., Zhi, L., & Müllen, K. (2007). Transparent, Conductive Graphene Electrodes for Dye-Sensitized Solar Cells. Nano Letters, 8(1), 323-327. doi:10.1021/nl072838rLeigh, S. J., Bradley, R. J., Purssell, C. P., Billson, D. R., & Hutchins, D. A. (2012). A Simple, Low-Cost Conductive Composite Material for 3D Printing of Electronic Sensors. PLoS ONE, 7(11), e49365. doi:10.1371/journal.pone.0049365Zhang, D., Ryu, K., Liu, X., Polikarpov, E., Ly, J., Tompson, M. E., & Zhou, C. (2006). Transparent, Conductive, and Flexible Carbon Nanotube Films and Their Application in Organic Light-Emitting Diodes. Nano Letters, 6(9), 1880-1886. doi:10.1021/nl0608543Kenry, & Liu, B. (2018). Recent Advances in Biodegradable Conducting Polymers and Their Biomedical Applications. Biomacromolecules, 19(6), 1783-1803. doi:10.1021/acs.biomac.8b00275Palza, H., Zapata, P., & Angulo-Pineda, C. (2019). Electroactive Smart Polymers for Biomedical Applications. Materials, 12(2), 277. doi:10.3390/ma12020277Naseri, M., Fotouhi, L., & Ehsani, A. (2018). Recent Progress in the Development of Conducting Polymer-Based Nanocomposites for Electrochemical Biosensors Applications: A Mini-Review. The Chemical Record, 18(6), 599-618. doi:10.1002/tcr.201700101Inal, S., Rivnay, J., Suiu, A.-O., Malliaras, G. G., & McCulloch, I. (2018). Conjugated Polymers in Bioelectronics. Accounts of Chemical Research, 51(6), 1368-1376. doi:10.1021/acs.accounts.7b00624Aqrawe, Z., Montgomery, J., Travas-Sejdic, J., & Svirskis, D. (2017). Conducting Polymers as Electrode Coatings for Neuronal Multi-electrode Arrays. Trends in Biotechnology, 35(2), 93-95. doi:10.1016/j.tibtech.2016.06.007Vara, H., & Collazos-Castro, J. E. (2015). Biofunctionalized Conducting Polymer/Carbon Microfiber Electrodes for Ultrasensitive Neural Recordings. ACS Applied Materials & Interfaces, 7(48), 27016-27026. doi:10.1021/acsami.5b09594Green, R., & Abidian, M. R. (2015). Conducting Polymers for Neural Prosthetic and Neural Interface Applications. Advanced Materials, 27(46), 7620-7637. doi:10.1002/adma.201501810Svirskis, D., Travas-Sejdic, J., Rodgers, A., & Garg, S. (2010). Electrochemically controlled drug delivery based on intrinsically conducting polymers. Journal of Controlled Release, 146(1), 6-15. doi:10.1016/j.jconrel.2010.03.023Uppalapati, D., Boyd, B. J., Garg, S., Travas-Sejdic, J., & Svirskis, D. (2016). Conducting polymers with defined micro- or nanostructures for drug delivery. Biomaterials, 111, 149-162. doi:10.1016/j.biomaterials.2016.09.021Alves-Sampaio, A., García-Rama, C., & Collazos-Castro, J. E. (2016). Biofunctionalized PEDOT-coated microfibers for the treatment of spinal cord injury. Biomaterials, 89, 98-113. doi:10.1016/j.biomaterials.2016.02.037Guo, B., & Ma, P. X. (2018). Conducting Polymers for Tissue Engineering. Biomacromolecules, 19(6), 1764-1782. doi:10.1021/acs.biomac.8b00276Wadhwa, R., Lagenaur, C. F., & Cui, X. T. (2006). Electrochemically controlled release of dexamethasone from conducting polymer polypyrrole coated electrode. Journal of Controlled Release, 110(3), 531-541. doi:10.1016/j.jconrel.2005.10.027Shamaeli, E., & Alizadeh, N. (2014). Nanostructured biocompatible thermal/electrical stimuli-responsive biopolymer-doped polypyrrole for controlled release of chlorpromazine: Kinetics studies. International Journal of Pharmaceutics, 472(1-2), 327-338. doi:10.1016/j.ijpharm.2014.06.036Esrafilzadeh, D., Razal, J. M., Moulton, S. E., Stewart, E. M., & Wallace, G. G. (2013). Multifunctional conducting fibres with electrically controlled release of ciprofloxacin. Journal of Controlled Release, 169(3), 313-320. doi:10.1016/j.jconrel.2013.01.022Richardson, R. T., Wise, A. K., Thompson, B. C., Flynn, B. O., Atkinson, P. J., Fretwell, N. J., … Clark, G. M. (2009). Polypyrrole-coated electrodes for the delivery of charge and neurotrophins to cochlear neurons. Biomaterials, 30(13), 2614-2624. doi:10.1016/j.biomaterials.2009.01.015Boehler, C., Kleber, C., Martini, N., Xie, Y., Dryg, I., Stieglitz, T., … Asplund, M. (2017). Actively controlled release of Dexamethasone from neural microelectrodes in a chronic in vivo study. Biomaterials, 129, 176-187. doi:10.1016/j.biomaterials.2017.03.019Collazos-Castro, J. E., Polo, J. L., Hernández-Labrado, G. R., Padial-Cañete, V., & García-Rama, C. (2010). Bioelectrochemical control of neural cell development on conducting polymers. Biomaterials, 31(35), 9244-9255. doi:10.1016/j.biomaterials.2010.08.057Cho, Y., Shi, R., Ivanisevic, A., & Ben Borgens, R. (2009). A mesoporous silica nanosphere-based drug delivery system using an electrically conducting polymer. Nanotechnology, 20(27), 275102. doi:10.1088/0957-4484/20/27/275102García-Fernández, A., García-Laínez, G., Ferrándiz, M. L., Aznar, E., Sancenón, F., Alcaraz, M. J., … Orzáez, M. (2017). Targeting inflammasome by the inhibition of caspase-1 activity using capped mesoporous silica nanoparticles. Journal of Controlled Release, 248, 60-70. doi:10.1016/j.jconrel.2017.01.002Lozano-Torres, B., Pascual, L., Bernardos, A., Marcos, M. D., Jeppesen, J. O., Salinas, Y., … Sancenón, F. (2017). Pseudorotaxane capped mesoporous silica nanoparticles for 3,4-methylenedioxymethamphetamine (MDMA) detection in water. Chemical Communications, 53(25), 3559-3562. doi:10.1039/c7cc00186jCollazos-Castro, J. E., Hernández-Labrado, G. R., Polo, J. L., & García-Rama, C. (2013). N-Cadherin- and L1-functionalised conducting polymers for synergistic stimulation and guidance of neural cell growth. Biomaterials, 34(14), 3603-3617. doi:10.1016/j.biomaterials.2013.01.097Garcia-Breijo, E., Peris, R. M., Pinatti, C. O., Fillol, M. A., Civera, J. I., & Prats, R. B. (2013). Low-Cost Electronic Tongue System and Its Application to Explosive Detection. IEEE Transactions on Instrumentation and Measurement, 62(2), 424-431. doi:10.1109/tim.2012.2215156Orgill, J. J., Chen, C., Schirmer, C. R., Anderson, J. L., & Lewis, R. S. (2015). Prediction of methyl viologen redox states for biological applications. Biochemical Engineering Journal, 94, 15-21. doi:10.1016/j.bej.2014.11.005Zhang, L. (2010). Glycosaminoglycan (GAG) Biosynthesis and GAG-Binding Proteins. Glycosaminoglycans in Development, Health and Disease, 1-17. doi:10.1016/s1877-1173(10)93001-9Collazos-Castro, J. E., García-Rama, C., & Alves-Sampaio, A. (2016). Glial progenitor cell migration promotes CNS axon growth on functionalized electroconducting microfibers. Acta Biomaterialia, 35, 42-56. doi:10.1016/j.actbio.2016.02.023Ito, M., & Kuwana, T. (1971). Spectroelectrochemical study of indirect reduction of triphosphopyridine nucleotide. Journal of Electroanalytical Chemistry and Interfacial Electrochemistry, 32(3), 415-425. doi:10.1016/s0022-0728(71)80144-4Nezakati, T., Seifalian, A., Tan, A., & Seifalian, A. M. (2018). Conductive Polymers: Opportunities and Challenges in Biomedical Applications. Chemical Reviews, 118(14), 6766-6843. doi:10.1021/acs.chemrev.6b00275Vitale, F., Summerson, S. R., Aazhang, B., Kemere, C., & Pasquali, M. (2015). Neural Stimulation and Recording with Bidirectional, Soft Carbon Nanotube Fiber Microelectrodes. ACS Nano, 9(4), 4465-4474. doi:10.1021/acsnano.5b0106

    A Two-Photon Probe Based on Naphthalimide-Styrene Fluorophore for the In Vivo Tracking of Cellular Senescence

    Get PDF
    Cellular senescence is a state of stable cell cycle arrest that can negatively affect the regenerative capacities of tissues and can contribute to inflammation and the progression of various aging-related diseases. Advances in the in vivo detection of cellular senescence are still crucial to monitor the action of senolytic drugs and to assess the early onset or accumulation of senescent cells. Here, we describe a naphthalimide-styrene-based probe (HeckGal) for the detection of cellular senescence both in vitro and in vivo. HeckGal is hydrolyzed by the increased lysosomal β-galactosidase activity of senescent cells, resulting in fluorescence emission. The probe was validated in vitro using normal human fibroblasts and various cancer cell lines undergoing senescence induced by different stress stimuli. Remarkably, HeckGal was also validated in vivo in an orthotopic breast cancer mouse model treated with senescence-inducing chemotherapy and in a renal fibrosis mouse model. In all cases, HeckGal allowed the unambiguous detection of senescence in vitro as well as in tissues and tumors in vivo. This work is expected to provide a potential technology for senescence detection in aged or damaged tissues

    Galacto-conjugation of Navitoclax as an efficient strategy to increase senolytic specificity and reduce platelet toxicity.

    Get PDF
    Pharmacologically active compounds with preferential cytotoxic activity for senescent cells, known as senolytics, can ameliorate or even revert pathological manifestations of senescence in numerous preclinical mouse disease models, including cancer models. However, translation of senolytic therapies to human disease is hampered by their suboptimal specificity for senescent cells and important toxicities that narrow their therapeutic windows. We have previously shown that the high levels of senescence-associated lysosomal β-galactosidase (SA-β-gal) found within senescent cells can be exploited to specifically release tracers and cytotoxic cargoes from galactose-encapsulated nanoparticles within these cells. Here, we show that galacto-conjugation of the BCL-2 family inhibitor Navitoclax results in a potent senolytic prodrug (Nav-Gal), that can be preferentially activated by SA-β-gal activity in a wide range of cell types. Nav-Gal selectively induces senescent cell apoptosis and has a higher senolytic index than Navitoclax (through reduced activation in nonsenescent cells). Nav-Gal enhances the cytotoxicity of standard senescence-inducing chemotherapy (cisplatin) in human A549 lung cancer cells. Concomitant treatment with cisplatin and Nav-Gal in vivo results in the eradication of senescent lung cancer cells and significantly reduces tumour growth. Importantly, galacto-conjugation reduces Navitoclax-induced platelet apoptosis in human and murine blood samples treated ex vivo, and thrombocytopenia at therapeutically effective concentrations in murine lung cancer models. Taken together, we provide a potentially versatile strategy for generating effective senolytic prodrugs with reduced toxicities

    Influence of functionalization of terephthalate linker on the catalytic activity of UiO-66 for epoxide ring opening

    Full text link
    [EN] A series of five isostructural zirconium terephthalate UiO-66 metal organic frameworks bearing different functional groups on the terephthalate linker (UiO-66-X; X = H, NH2, NO2, Br, Cl,) have been successfully prepared and characterized. UiO-66-X materials were evaluated as heterogeneous catalysts for the epoxide ring opening of styrene oxide by methanol, observing an increase in the initial reaction rate from UiO-66-H to UiO-66-Br, over one order of magnitude. The reactivity order, however, does not follow a linear relationship between the Hammett constant value of the substituent and the initial reaction rate. UiO-66-Br exhibits a wide scope, its activity depending on the structure of epoxide and nucleophile. The absence of Zr leaching to the solution together with the preservation of the UiO-66-X crystallinity confirms the stability of the framework under the reaction conditions. Nevertheless, UiO-66 undergoes a progressive deactivation upon reuse that was attributed to a strong adsorption of the reaction product.Financial support by the Spanish Ministry of Economy and Competitiveness (Severo Ochoa and CTQ2015-69153 and CTQ2014-53292-R and Generalitat Valenciana (Prometeo 2013/14) and is grateful acknowledged. We also thank EU under the Being Energy contract for partial funding. J.F.B. thanks the Universitat Politecnica de Valencia for a postgraduate scholarship. S.N. thanks the Spanish Ministerio de Educacion, Cultura y Deporte for Jose Castillejo mobility programme (CAS14/00067).Blandez, JF.; Santiago-Portillo, A.; Navalón Oltra, S.; Gimenez Marques, M.; Alvaro Rodríguez, MM.; Horcajada, P.; García Gómez, H. (2016). Influence of functionalization of terephthalate linker on the catalytic activity of UiO-66 for epoxide ring opening. Journal of Molecular Catalysis A Chemical. 425:332-339. https://doi.org/10.1016/j.molcata.2016.10.022S33233942

    Lipofuscin labeling through biorthogonal strain-promoted azide-alkyne cycloaddition for the detection of senescent cells

    Full text link
    [EN] A new method for senescent cell detection is described, which is based on lipofuscin labeling with a fluorescent reporter through a biorthogonal strain-promoted azide-alkyne cycloaddition. The sensing protocol involves a first step where the interaction of lipofuscin with a Sudan Black B derivative containing an azide moiety (SBB-N3 ) is carried out. In the final step, the azide moiety reacts with a fluorophore containing a cyclooctene ring (BODIPY). The efficacy of this two-step protocol is assessed in senescent melanoma SK-MEL-103 cells, senescent triple-negative breast cancer MDA-MB-231 cells and senescent WI-38 fibroblasts. In all cases, a clear fluorescence pattern was observed in senescent cells, compared to proliferative cells, only when the SBB-N3 -BODIPY probe was formed. Our results provide an alternative tool for the detection of senescent cells, based on an in situ bio-orthogonal reaction for lipofuscin labelingRM laboratory members are grateful for financial support from the FEDER foundation of European Union (IDIFEDER/2021/044), the Spanish Government (projects RTI2018-100910-B-C41 and RTI2018-101599-B-C22) and the Generalitat Valenciana (project PROMETEO 2018/024). BL-T and AG-F acknowledge their current Margarita Salas postdoctoral fellowship from UPV-MIU and 'Next Generation EU' program. JF-B is grateful for his Sara Borrell postdoctoral fellowship (CD19/00038).Lozano-Torres, B.; Blandez, JF.; García-Fernández, A.; Sancenón Galarza, F.; Martínez-Máñez, R. (2023). Lipofuscin labeling through biorthogonal strain-promoted azide-alkyne cycloaddition for the detection of senescent cells. FEBS Journal. 290(5):1314-1325. https://doi.org/10.1111/febs.1647713141325290

    Nickel nanoparticles supported on graphene as catalysts for aldehyde hydrosilylation

    Full text link
    [EN] Nickel nanoparticles (NPs) supported on different undoped or doped with N or B graphenes (Gs) have been tested as catalyst for the hydrosilylation of aldehydes to obtain the corresponding siloxanes with high conversion and good selectivity in short reaction time. The different Gs employed were obtained by pyrolysis under inert atmosphere of alginate or chitosan, modified or not with boric acid. Then the metal NPs obtained by polyol reduction method using ethylene glycol were adsorbed on Gs. The Ni-containing G catalysts were characterized by electron microscopy, XPS and Raman spectroscopy. The scope of the Ni/G catalyst includes aliphatic and aromatic aldehydes as well as a variety of hydrosilanes. (C) 2015 Elsevier B.V. All rights reserved.Financial support by the Spanish Ministry of Economy and Competitiveness (Severo Ochoa and CTQ2012-32315) and Generalitat Valenciana (Prometeo 2013-019) in gratefully acknowledged. J.F.B. and I.E.A. thanks to the Technical University of Valencia and the Spanish Ministry of Economy and Competitiveness for postgraduate scholarships, respectively.Blandez, JF.; Esteve-Adell, I.; Primo Arnau, AM.; Alvaro Rodríguez, MM.; García Gómez, H. (2016). Nickel nanoparticles supported on graphene as catalysts for aldehyde hydrosilylation. Journal of Molecular Catalysis A Chemical. 412:13-19. https://doi.org/10.1016/j.molcata.2015.11.011S131941

    Synthesis, Structural Characterization, and Catalytic Activity of IPrNi(styrene)2 in the Amination of Aryl Tosylates

    Full text link
    [EN] A novel bis-styrene IPrNi0 derivative has been synthesized from the reaction of Ni(COD)2 and free 1,3-bis(2,6-diisopropylphenyl)imidazolidene (IPr) ligand in the presence of styrene. The complex has been characterized by spectroscopic data as well as by X-ray crystallography. Its catalytic performance in the amination reaction of aryl tosylates is also reported. The catalytic reactions proceed in a very selective manner, affording moderate to high yields of cross-coupling products in short reaction times at 110 °C.This research was financially supported by the MINECO (Proyecto CTQ2011-24502 and Consolider Ingenio 2010, Grant No. CSD2006-003) and the Junta de Andalucia (Proyecto P07-FQM-02745). M.J.I. thanks the Junta de Andalucia for a research fellowship.Iglesia, MJ.; Blandez, JF.; Fructos, MR.; Prieto, A.; Alvarez, E.; Belderrain, TR.; Nicasio, MC. (2012). Synthesis, Structural Characterization, and Catalytic Activity of IPrNi(styrene)2 in the Amination of Aryl Tosylates. Organometallics. 31(17):6312-6316. https://doi.org/10.1021/om300566mS63126316311
    corecore