Molecular Cloning and Functional Characterization of a Cell-permeable Superoxide Dismutase Targeted to Lung Adenocarcinoma Cells
Min Lu,Xingguo Gong,Yuwen Lu,Jianjun Guo,Chenhui Wang,Yuanjiang Pan
DOI: https://doi.org/10.1074/jbc.m600523200
IF: 5.485
2006-01-01
Journal of Biological Chemistry
Abstract:In clinical oncology, many trials with superoxide dismutase (SOD) have failed to demonstrate antitumor ability and in many cases even caused deleterious effects because of low tumor-targeting ability. In the current research, the Nostoc commune Fe-SOD coding sequence was amplified from genomic DNA. In addition, the single chain variable fragment (ScFv) was constructed from the cDNA of an LC-1 hybridoma cell line secreting anti-lung adenocarcinoma monoclonal antibody. After modification, the SOD and ScFv were fused and co-expressed, and the resulting fusion protein produced SOD and LC-1 antibody activity. Tracing SOD-ScFv by fluorescein isothiocyanate and superoxide anions (O2·¯) in SPC-A-1 cells showed that the fusion protein could recognize and enter SPC-A-1 cells to eliminate O2·¯. The lower oxidative stress resulting from the decrease in cellular O2·¯ delayed the cell cycle at G1 and significantly slowed SPC-A-1 cell growth in association with the dephosphorylation of the serine-threonine protein kinase Akt and expression of p27kip1. The tumor-targeting fusion protein resulting from this research overcomes two disadvantages of SODs previously used in the clinical setting, the inability to target tumor cells or permeate the cell membrane. These findings lay the groundwork for development of an efficient antitumor drug targeted by the ScFv. In clinical oncology, many trials with superoxide dismutase (SOD) have failed to demonstrate antitumor ability and in many cases even caused deleterious effects because of low tumor-targeting ability. In the current research, the Nostoc commune Fe-SOD coding sequence was amplified from genomic DNA. In addition, the single chain variable fragment (ScFv) was constructed from the cDNA of an LC-1 hybridoma cell line secreting anti-lung adenocarcinoma monoclonal antibody. After modification, the SOD and ScFv were fused and co-expressed, and the resulting fusion protein produced SOD and LC-1 antibody activity. Tracing SOD-ScFv by fluorescein isothiocyanate and superoxide anions (O2·¯) in SPC-A-1 cells showed that the fusion protein could recognize and enter SPC-A-1 cells to eliminate O2·¯. The lower oxidative stress resulting from the decrease in cellular O2·¯ delayed the cell cycle at G1 and significantly slowed SPC-A-1 cell growth in association with the dephosphorylation of the serine-threonine protein kinase Akt and expression of p27kip1. The tumor-targeting fusion protein resulting from this research overcomes two disadvantages of SODs previously used in the clinical setting, the inability to target tumor cells or permeate the cell membrane. These findings lay the groundwork for development of an efficient antitumor drug targeted by the ScFv. Superoxide dismutase (SOD), 3The abbreviations used are: SOD, superoxide dismutase; ROS, reactive oxygen species; ScFv, single chain variable fragment; mAb, monoclonal antibody; FITC, fluorescein isothiocyanate; DHE, dihydroethidium; Rh123, rhodamine 123; CM-H2TMRos, MitoTracker Orange CM-H2TMRos; DCF-DA, peroxide-sensitive fluorophore dichlorodihydrofluorescein diacetate; NDA, naphthalene-2, 3-dicarboxyaldehyde; IGF-I, insulin-like growth factor-I; and IGFBP-I, IGF binding protein-I; IPTG, isopropyl 1-thio-β-d-galactopyranoside; GSH, glutathione; LCSM, laser confocal scanning microscope. 3The abbreviations used are: SOD, superoxide dismutase; ROS, reactive oxygen species; ScFv, single chain variable fragment; mAb, monoclonal antibody; FITC, fluorescein isothiocyanate; DHE, dihydroethidium; Rh123, rhodamine 123; CM-H2TMRos, MitoTracker Orange CM-H2TMRos; DCF-DA, peroxide-sensitive fluorophore dichlorodihydrofluorescein diacetate; NDA, naphthalene-2, 3-dicarboxyaldehyde; IGF-I, insulin-like growth factor-I; and IGFBP-I, IGF binding protein-I; IPTG, isopropyl 1-thio-β-d-galactopyranoside; GSH, glutathione; LCSM, laser confocal scanning microscope. a type of metalloenzyme, is ubiquitous among oxygen-metabolizing organisms. SODs have been classified into four types based on the metal species at the enzyme active site: Cu/Zn-SOD, Mn-SOD, Fe-SOD, and Ni-SOD. The theory of free radicals and tumors held that reactive oxygen species (ROS) originating in redundant radicals attacked the organism, leading to genetic mutation and tumor formation (1Hruszkewycz A.M. Bergtold D.S. Basic Life Sci. 1988; 49: 449-456PubMed Google Scholar, 2Kodama M. Inoue F. Nakayama T. Nagata C. Arch Biochem. Biophys. 1987; 258: 574-578Crossref PubMed Scopus (2) Google Scholar, 3Schwarz M. Peres G. Kunz W. Furstenberger G. Kittstein W. Marks F. Carcinogenesis. 1984; 5: 1663-1670Crossref PubMed Scopus (28) Google Scholar). SOD is supposed to be the main cellular superoxide anion-degrading enzyme, inhibiting tumors (4Zhang Y. Zhao W. Zhang H.J. Domann F.E. Oberley L.W. Cancer Res. 2002; 62: 1205-1212PubMed Google Scholar, 5Lam E.W. Zwacka R. Engelhardt J.F. Davidson B.L. Domann F.E. Jr-Yan T. Oberley L.W. Cancer Res. 1997; 57: 5550-5556PubMed Google Scholar, 6Bravard A. Beaumatin J. Dussaulx E. Lesuffleur T. Zweibaum A. Luccioni C. Int. J. Cancer. 1994; 59: 843-847Crossref PubMed Scopus (23) Google Scholar); however, ROS are essential to the existence and development of a normal cell (7Sen C.K. Curr. Top Cell Regul. 2000; 36: 1-30Crossref PubMed Scopus (291) Google Scholar, 8Shackelford R.E. Kaufmann W.K. Paules R.S. Free Radic. Biol. Med. 2000; 28: 1387-1404Crossref PubMed Scopus (457) Google Scholar). For this reason, attempts to treat ROS-associated diseases with non-selective SOD have failed and may have caused deleterious effects in normal cells as well as in tumor cells. Using genetic techniques, Oyanagui (9Oyanagui Y. Sato S. Inoue M. Biochem. Pharmacol. 1991; 42: 991-995Crossref PubMed Scopus (15) Google Scholar) fused SOD with a polypeptide that could recognize heparin sulfates on vascular endothelial cells; the resultant HB-SOD could locate vascular endothelial cells by itself. In addition to potential non-specific cell targeting, SOD has another critical disadvantage in clinical application; its large molecular weight interferes with its ability to permeate the cell membrane, which leads to a low cellular effect. Currently, a liposome is used to enclose the SOD, and the resultant liposome-SOD can permeate the cell membrane or tissue (10Delanian S. Martin M. Bravard A. Luccioni C. Lefaix J.L. Radiother. Oncol. 2001; 58: 325-331Abstract Full Text Full Text PDF PubMed Scopus (46) Google Scholar, 11Jubeh T.T. Antler S. Haupt S. Barenholz Y. Rubinstein A. Mol. Pharm. 2005; 2: 2-11Crossref PubMed Scopus (32) Google Scholar, 12Rengel R.G. Filipovic-Grcic J. Cepelak I. Zanic-Grubisic T. Barisic K. Eur. J. Pharm. Biopharm. 2005; 60: 47-51Crossref PubMed Scopus (18) Google Scholar). But thus far, modified SODs still either cannot distinguish the target cells (tumor cells) or cannot permeate the tumor cell membrane. The single chain variable fragment (ScFv) antibody not only has a much lower immune binding than does monoclonal antibody (mAb) but also has a low molecular weight; thus, it can more easily reach intracellular targets. ScFv from the LC-1 hybridoma cell line can react at a high frequency with lung adenocarcinoma, lung squamous carcinoma, and large cell and small cell lung cancer, but not with normal and embryonic tissues (13Ge X.R. Wang J. Lin S.J. Yu N.X. Che Y.F. Gu Y. Acta Biologiae. Exprimentalis Sin. 1989; 22: 359-365Google Scholar). When this ScFv recognized and bound antigens on SPC-A-1 cells, the antigen-antibody complexes were internalized via the receptor-mediated endocytic pathway and permeated the cell membrane (14Xie W. Zhang R.X. Ge X.R. Acta Biologiae Exprimentalis Sin. 1996; 29: 207-219Google Scholar). This characteristic implies that using LC-1 ScFv to aid SOD targeting can be an effective anti-lung cancer drug because it overcomes both clinical SOD disadvantages, it will target only tumor cells and will be taken up intracellularly. In the current research, the Nostoc commune Fe-SOD and LC-1 ScFv were cloned and co-expressed, with the idea that the resultant fusion protein would possess dual activity. In theory, LC-1 ScFv would be able to introduce SOD into SPC-A-1 cells specifically and permeate the cell membrane via the receptor-mediated endocytic pathway. Once inside the cell, it would eliminate intracellular O2·¯, change the cellular redox state, and subsequently induce a pathway related to a low cellular O2·¯ level, inhibiting tumor cell proliferation. Chemicals and Antibodies—Rabbit phospho-Akt (Thr-308) antibody, p27kip1 antibody, and horseradish peroxide-conjugated goat antirabbit antibody were purchased from Calbiochem; anti-IGF-I antibody was kindly provided by Dr. Jianhui Fan (Boster, China) and used according to the manufacturer's recommendations. The fluorescent probes dihydroethidium (DHE), rhodamine 123 (Rh123), MitoTracker Orange CM-H2TMRos (M7511), fluo-3 AM, and peroxide-sensitive fluorophore dichlorodihydrofluorescein diacetate (DCF-DA) were purchased from Molecular Probes. Naphthalene-2,3-dicarboxyaldehyde (NDA) was obtained from Aldrich Chemicals. Cell Lines and Culture Conditions—Total RNA of the LC-1 hybridoma was kindly provided by Dr. Chen Liang of Yale University. N. commune stain CHEN, pET-22b (+) vector, pUCm-T vector, Escherichia coli (DH5α: supE44ΔlacU169; φ80 lacZΔM15, hsdR17 recA1 endA1 gurA96 thi-1 relA1), E. coli BL21 (DE3: hsdS gal; λcIts857, ind1 Sam7 nin5 lacUV5-T7), SPC-A-1 lung adenocarcinoma cell line, AGS gastric cancer cell line, and the HepG2 liver carcinoma cell line are available in our institute. N. commune was cultivated in BG11 culture medium. E. coli DH5α and E. coli BL21 (DE3) were maintained in LB culture medium. SPC-A-1 cells, AGS cells, and HepG2 cells were cultured in RPMI 1640 medium, which contained 10% fetal calf serum. Tumor cells were maintained in a CO2 cell culture incubator (Forma Scientific, 3154 CO2 Water-Jacketed Incubator) at 37 °C under 5% CO2. To prepare for every experiment, 90 μl of cells were grown in a 96-well plate until 70-80% confluent. All experiments were repeated at least three times. Data shown in the graphs for various parameters represent the mean ± S.E. Differences between means were considered significant when p < 0.05. Construction of the pET-SOD Expression Vector System—A 0.2-g N. commune sample was taken, and its genomic DNA was isolated using the method of Kim et al. (15Kim C.S. Lee C.H. Shin J.S. Chung Y.S. Hyung N.I. Nucleic Acids Res. 1997; 25: 1085-1086Crossref PubMed Scopus (171) Google Scholar). Fe-SOD was amplified from the genomic DNA, primered with degenerate primer, SOD-forward (5′-CTAGCCATGGCATTTGTACAGC-3′, including the NcoI site) and SOD-back (5′-CCGCTCGAGAGCTTT(G/A)GC(C/A)AAGTT(T/C)TC-3′, including the Xhol I site), and cloned into pET-22b (+) to be pET-SOD. Construction of the pET-ScFv Expression Vector System—The ScFv was constructed essentially following our previous approach (16Lu M. Gong X.G. Yu H. Li J.Y. J. Zhejiang Univ. Sci. B. 2005; 6: 832-837Crossref PubMed Scopus (19) Google Scholar). Briefly, Vh and Vl were amplified from the cDNA of the LC-1 hybridoma, primered with Vh-1 (5′-CATGCCATGGAACTGCAGGAGTCAGGACC-3′, including the NcoI site), Vh-2 (5′-TGAGGAGACGGTGACCGTGGTCCCTTGGCCCAG-3′), Vl-1 (5′-GACATTGAGCTCACCCAGTCTC-3′), and Vl-2 (5′-ACGCGTCGACCTTGGTCCCCCCTCCGAA-3′, including the SalI site), respectively. The Vl was modified into Vl′, primered with Vl-Mod (5′-CTGGGGCCAAGGGACCACGGTCACCGTCTCCTCAGGTGGAGGCGGTTCAGGCGGAGGTGGCTCTGGCGGTGGCGGATCGGACATTGAGCTCACCCAGTCTC-3′, including link-2: GGGGS×3) and Vl-2. Vl′ and Vh were connected by SOE-PCR, and cloned into pET-22b (+) to be pET-ScFv. Construction of the pET-SOD-ScFv Expression Vector System—The SOD fragment was amplified from the pET-SOD vector, primered with SOD-Nco (5′-CATGCCATGGCATTTGTACAGCTCCCACTACCCTTTG-3′, including the NcoI site) and SOD-Sal (5′-GCGTCGACTCCGCCTCCACCAGCTTTGGCCAAGTTTTC-3′, including the SalI site and link-1: GGGG). The ScFv fragment was amplified from the pET-ScFv vector, primered with ScFv-Sal (5′-ACGCGTCGACGTCCAACTGCAGGAGTCAGG-3′, including the SalI site) and ScFv-Not (5′-ATAAGAATGCGGCCGCTTTGATCTCGAGCTTGGTC-3′, including the NotI site). Then the SOD and ScFv fragments were cloned into pET-22b (+) to be pET-SOD-ScFv (see Fig. 1) and transformed into JM109. Expression, Purification, and Immunoblot Analyses of SOD, ScFv, and SOD-ScFv—The pET-SOD was induced in the presence of 0.5 mm IPTG and 300 μm [Fe3+] in 1 liter of LB culture medium at 37 °C for 3 h. Cells were harvested by centrifugation at 5000 × g and lysed in 50 ml of TE (20 mm Tris-HCl and 10 mm EDTA, pH 8.0). The lysate was disrupted using sonication (800 W, 20 min). The achieved sedimentation (SOD) after sonication was washed with wash buffer I (1 mm EDTA, 5% glycerol, 0.5% Triton X-100) and buffer II (wash buffer I with 2 m carbamide). Then the sedimented material was dissolved in lysis buffer (50 mm Tris-HCl, 0.3% sodium lauryl sulfate, 0.1 mm dithiothreitol) overnight. The lysate was poured into a column with 5 ml of nickel-nitrilotriacetic acid Superflow Resin, and the packed resin was washed with buffer consisting of imidazole gradient concentrations (0-500 mm). A280 was measured. The denatured protein was renatured in lysis buffer (excluding sodium lauryl sulfate) that included 100 μm [Fe3+], at 4 °C. The SOD-ScFv fusion protein was expressed and purified from pET-SOD-ScFv as described above. The ScFv was expressed and purified from pET-ScFv as described above, except that there was no [Fe3+] in the LB culture medium or the renaturing buffer. Natural Fe-SOD protein was purified from N. commune directly as follows: 500 g of N. commune was lysed in 1 liter of PBS (including 0.5 mm EDTA, pH 7.6) and disrupted using sonication (1 200 W, 60 min). Then the crude materials (Fe-SOD) were deposited in 45-85% (NH4)2SO4 for 6 h. The crude Fe-SOD was dissolved again and purified by DEAE-Sepharose fast flow and Sephadex G-100, respectively. The expression of pET-SOD and pET-SOD-ScFv was subjected to Western blot analysis (the anti-SOD antiserum was raised against natural N. commune Fe-SOD in mice). The SOD activity of purified SOD and SOD-ScFv was assayed using the pyrogallol auto-oxidation method (17Marklund S. Marklund G. Eur J Biochem. 1974; 47: 469-474Crossref PubMed Scopus (7732) Google Scholar). The antibody activity of purified ScFv and SOD-ScFv was assayed by competitive enzyme-linked immunosorbent assay as follows: 0.01 μg/μl LC-1 mAb and the purified ScFv (or SOD-ScFv) were used to bind SPC-A-1 cells competitively (AGS cells and HepG2 cells served as controls); goat anti-mouse-labeled anti-IgM secondary antibody was used to bind to the LC-1 antibody. A490 was measured. Determination of the Cell Membrane Permeation Ability of SOD-ScFv by Fluorescein Isothiocyanate (FITC) Fluorescence Tracing and Immunoblot Analyses—To trace the permeation process of SOD-ScFv through the SPC-A-1 cell membrane, SOD, ScFv, and SOD-ScFv were labeled with FITC essentially according to Lindsay et al. (18Lindsay M.P. Tamas L. Appels R. Skerritt J.H. J. Cereal Sci. 2000; 31: 321-333Crossref Scopus (30) Google Scholar). A 0.1 μm sample was dissolved in 10 ml of 100 mm carbonate buffer, pH 9.0. To each sample was added 10 μl of 1% w/v FITC in dimethyl sulfoxide, and labeling was performed at room temperature overnight. Each labeled sample was dialyzed at 4 °C for 72 h against pH 8.0 PBS. The samples were additionally separated by a Sephadex G-25 column. The fluorescence intensity of sample was measured by a fluorescence spectrophotometer (Hitachi, 850, Japan). The prepared SPC-A-1 cells were washed twice with PBS and incubated in RPMI 1640 medium containing 0.2 μm SOD-ScFv-FITC for 20, 40, and 60 min at 37 °C. Following three washes to remove unbound protein, intracellular FITC fluorescence was observed by LCSM (LSM 510, Zeiss, Germany). The intracellular stability of SOD-ScFv was estimated by Western blotting as follows. After SPC-A-1 cells were treated with 0.2 μm SOD-ScFv for 1 h, cells were washed and changed with a fresh culture medium. Then, cells were further incubated for 0, 6, 12, and 24 h followed by Western blot analysis with anti-SOD antiserum. Determination of Targeted Antitumor Ability of SOD-ScFv—A total of 90 μl of SPC-A-1 cells were grown in a 96-well plate until 30% confluent. Then, 10 μl of SOD-ScFv of the working concentrations (0.2-2 μm) were added for 1 h, and cells were washed and changed with a fresh culture medium for 48 h. Cells were counted using a hemocytometer (AGS cells and HepG2 cells served as negative controls); a cycle of cells (treated as described above) from a 6-well plate was analyzed by flow cytometry (FAC sort, Becton Dickinson) according to Mahadevan et al. (19Mahadevan B. Luch A. Seidel A. Pelling J.C. Baird W.M. Carcinogenesis. 2001; 22: 161-169Crossref PubMed Scopus (22) Google Scholar). Analysis of Cellular Superoxide Anions by LCSM—Cellular superoxide anions (O2·¯) were measured using DHE as a fluorescent dye relatively specific for O2·¯ (20Fujimura M. Morita-Fujimura Y. Kawase M. Copin J.C. Calagui B. Epstein C.J. Chan P.H. J. Neurosci. 1999; 19: C3414-C3422Crossref PubMed Google Scholar). Fluorescence intensity from O2·¯ in individual SPC-A-1 cells was traced over time essentially according to Bindokas et al. (21Bindokas V.P. Jordan J. Lee C.C. Miller R.J. J. Neurosci. 1996; 76: 1324-1336Crossref Google Scholar). After incubation with 0.2 μm SOD-ScFv in fresh culture medium at 37 °C for 1 h, the cells were washed with PBS and put in fresh culture medium with 20 μm DHE at 37 °C. After 5 min, the DHE fluorescence became visible on the LCSM and then cellular DHE fluorescence was scanned instantly for time-lapse images. Intracellular O2·¯ was monitored for at least 1500 s, and the temperature of the cells was maintained at 37 °C. In addition, intracellular calcium flux response to 0.2 μm SOD-ScFv (PBS and 50 μm H2O2 served as controls) was observed using fluo-3 AM for 1 h on LCSM, essentially according to Deng et al. (22Deng T.L. Yu L. Ge Y.K. Zhang L. Zheng X.X. Biochem. Biophys. Res. Commun. 2005; 338: 748-756Crossref PubMed Scopus (17) Google Scholar). Analysis of Mitochondrial Transmembrane Potential (ΔΨm) by Fluorescence Spectrophotometer—Mitochondrial membrane potential (ΔΨm) in SPC-A-1 cells was measured using two different dyes, rhodamine-123 and CM-H2TMRos. Rhodamine-123, a conventional, cationic voltage-sensitive probe that reversibly accumulates in mitochondria, was used in the present study to detect changes in ΔΨm. Cells were incubated with 1 μm rhodamine-123 at 37 °C for 30 min after 1 h of drug incubation. After three washings, the fluorescence intensity of the sample was analyzed by fluorescence spectrophotometer. CM-H2TMRos is a new fluorescent dye developed recently that is more photostable and stable in mitochondria (23Poot M. Zhang Y.Z. Kramer J.A. Wells K.S. Jones L.J. Hanzel D.K. Lugade A.G. Singer V.L. Haugland R.P. J. Histochem. Cytochem. 1996; 44: 1363-1372Crossref PubMed Scopus (394) Google Scholar). It was also used to measure the ΔΨm as described above, excepting cells that were incubated with 500 nm CM-H2TMRos. Cellular Oxidative Stress and Reducing Power Assay by Flow Cytometry—The oxidative stress of SPC-A-1 cells was determined by detection of cellular ROS with DCFH-DC (24Sawada G.A. Raub T.J. Decker D.E. Buxser S.E. Cytometry. 1996; 25: 254-262Crossref PubMed Scopus (45) Google Scholar). Briefly, after incubation with 0.2 μm SOD-ScFv in fresh culture medium for 1 h, cells were washed and incubated with 5 μm DCF-DA for 20 min at 37 °C. Cells were then washed three times with probe-free nitrate buffer. The fluorescence intensity of the sample was measured by flow cytometry. The cellular reduced glutathione (GSH) level was measured as an index to reflect the cellular reducing power in SPC-A-1 cells; GSH is oxidized to oxidized glutathione under the stress of superoxide anions or hydrogen peroxide. The GSH level was determined by staining cells with 500 μm NDA (25Orwar O. Fishman H.A. Ziv N.E. Anal. Chem. 1995; 67: 4261-4268Crossref PubMed Scopus (140) Google Scholar) as described above, then cells were washed three times, and fluorescence intensity was measured by flow cytometry. Reduced Oxidative Stress Effect on SPC-A-1 Cells through Akt/p27 kip1 Pathway—A total of 1.8 ml of SPC-A-1 cells were grown in a 6-well plate until 80% confluent, then treated with SOD-ScFv at 37 °C for 1 h (PBS served as control). After treatment, 106 cells were disrupted, and then cellular phosphorylated Akt (p-Akt) levels and insulin-like growth factor-I (IGF-I) levels were analyzed by Western blotting. To analyze the expression levels of cyclin-dependent kinase inhibitor p27kip1, 1.8 ml of SPC-A-1 cells were grown in 6-well plate until 30% confluent, 200 μl of SOD-ScFv were added for 1 h (PBS served as control), and cells were washed and changed with a fresh culture medium for 24 h. A total of 106 cells were disrupted, and the cellular p27kip1 level was analyzed by Western blotting. Based on data for other similar Fe-SODs of prokaryotic algae in GenBank™, we assumed that the Fe-SOD coding region in N. commune genome was continuous. Based on these Fe-SOD sequences, the degenerate primer SOD-forward/SOD-back was designed, and a fragment (GenBank™ accession AY830114) of 603 bp was achieved, whose deduced protein was in accordance with the molecular weight of natural Fe-SOD, which was purified from N. commune (22 kDa). These findings implied that this 603-bp coding sequence was the N. commune Fe-SOD CDS. This conclusion was supported by Western blotting analysis of expressed Fe-SOD or SOD-ScFv against natural Fe-SOD antiserum (Fig. 2A, lanes 3 and 4). BLAST search results from GenBank™ showed that this CDS had about 85% nucleotide similarity with Nostoc linckia Fe-SOD. It was found that when the cells were induced by IPTG, additional FeCl3 could improve the yield of the expected protein (26Kim Y.C. Miller C.D. Anderson A.J. Gene (Amst.). 1999; 239: 129-135Crossref PubMed Scopus (32) Google Scholar). The expressed SOD, ScFv, and SOD-ScFv proteins were composed of ∼78, 65, and 21%, respectively, of the total bacterial protein in the pET-SOD, pET-ScFv, and pET-SOD-ScFv expression vector systems, respectively (data not shown). The SOD activity assay result indicated that the specific activity of expressed SOD and SOD-ScFv was 1100 units/mg and 700 units/mg, respectively, whereas that of natural N. commune Fe-SOD was 2700 units/mg. The competitive enzyme-linked immunosorbent assay result of ScFv showed that the inhibitory rate of 0.1 μg/μl ScFv to 0.01 μg/μl LC-1 antibody was 71.0%, whereas that of ScFv-SOD was 61.2%. But neither showed the ability to bind to AGS cells or HepG2 cells (data not shown). The ability of SOD to permeate the cell membrane is a key point in clinical applications of SOD. Based on the specific spatial structure of SOD, ScFv, or SOD-ScFv, we dyed each with FITC (27Grunwaldt G. Haebel S. Spitz C. Steup M. Menzel R. J. Photochem. Photobiol. B. Biol. 2002; 67: 177-186Crossref PubMed Scopus (18) Google Scholar) to determine their ability to permeate the cell membrane and their cellular distribution after permeation. After incubation with SOD-ScFv-FITC for 20 min, the SPC-A-1 cells began to show green fluorescence (Fig. 3E). From Fig. 3I, the FITC localized inside the cell but did not reside on the cell membrane; what is more, it was distributed equally in the cells (Fig. 3, H and I). The ScFv-FITC control exhibited the same pattern (Fig. 3C, some data not shown). However, the SOD-FITC control showed no obvious fluorescence (Fig. 3B), implying that the SOD barely permeated the cell membrane by itself. The intracellular stability of SOD-ScFv in SPC-A-1 cells was shown in Fig. 2B. After permeation, the SOD-ScFv was degraded gradually; however, it could persist in cells for up to 24 h. The targeted antitumor ability of SOD-ScFv is shown in Fig. 4. Cells treated with 0.2 μm SOD-ScFv showed a 46.6% inhibition rate; this increased significantly in a SOD-ScFv dose-dependent manner (Fig. 4A). In addition, SOD-ScFv did not show the same inhibition in AGS cells or HepG2 cells (Fig. 4B), which implied the specificity of SOD-ScFv for SPC-A-1 cells. To determine how SOD-ScFv inhibited growth of SPC-A-1 cells, a cycle of treated cells was performed (Fig. 4C). It was clear that there was a change in the distribution of cells in G1 phase as a result of cellular overload of SOD. In the SOD-ScFv model, 60.4% of the cells were in G1 phase, whereas only 50.4% of the SOD-treated cells, 51.7% of the ScFv-treated cells, and 47.4% of PBS-treated cells were in G1. A considerable increase in the G1 fraction of SOD-ScFv model cells suggested that the SOD-induced inhibition in cell growth could be associated with the delay at G1. The cellular O2·¯ analysis result is shown in Fig. 5A. The oxidation rate of DHE in SPC-A-1 cells decreased significantly after pretreatment with SOD-ScFv. However, pretreatment of SOD or ScFv produced an oxidation rate not significantly different from PBS control, which showed the low permeation ability of SOD or the invalidation of ScFv in O2·¯ elimination. Interestingly, it was found that the cellular calcium level did not respond to SOD-ScFv in 1 h, whereas cellular calcium levels increased rapidly and distinctly under stimulation with 50 μm H2O2 (data not shown). It is well known that H2O2 can produce a high cellular oxidative stress. The main source of ROS in most cells is the mitochondria (28Hennet T. Richter C. Peterhans E. Biochem J. 1993; 289: 587-592Crossref PubMed Scopus (261) Google Scholar). We confirmed this in SPC-A-1 cells according to the perfect superposition of cellular DHE fluorescence and Rh123 fluorescence, which localized to the area of origin of O2·¯ and the mitochondrial area, respectively (Fig. 5B). One parameter that is altered because of ROS production is ΔΨm. Therefore, the effect of a lower O2·¯ level on ΔΨm was determined using Rh123. Fig. 6 shows an increase of Rh123 fluorescent intensity in SPC-A-1 cells (but not in AGS cells or HepG2 cells) by 1.3 times (p < 0.05) after treatment with SOD-ScFv. The CM-H2TMRos group exhibited similar results. These observations implied that lower levels of O2·¯ around the mitochondria could maintain the O2·¯. The O2·¯ impairment by cellular O2·¯ had been reported by Scanlon et al. (29Scanlon J.M. Reynolds I.J. J Neurochem. 1998; 71: 2392-2400Crossref PubMed Scopus (68) Google Scholar) and Kimura et al. (30Kimura S. Zhang G.X. Nishiyama A. Shokoji T. Yao L. Fan Y.Y. Rahman M. Abe Y. Hypertension. 2005; 45: 438-444Crossref PubMed Scopus (179) Google Scholar). Based on the fact that SOD-ScFv can eliminate O2·¯ in SPC-A-1 cells, we determined the cellular redox state aroused by SOD-ScFv after 1 h by detecting cellular ROS and GSH levels. As Fig. 7 shows, it is notable that SOD-ScFv resulted in significant decreases in cellular ROS fluorescence intensity (0.54-fold, p < 0.05) and increases in GSH fluorescence intensity (5.4-fold, p < 0.05), implying that cellular oxidative stress can be regulated by O2·¯ levels in SPC-A-1 cells. Increases in GSH with decreased oxidative stress have been demonstrated in previous studies (31Paz-Miguel J.E. Flores R. Sanchez-Velasco P. Ocejo-Vinyals G. Escribano de Diego J. Lopez de Rego J. Leyva-Cobian F. J. Immunol. 1999; 163: 5399-5410PubMed Google Scholar, 32Wefers H. Sies H. Eur. J. Biochem. 1983; 137: 29-36Crossref PubMed Scopus (190) Google Scholar). To understand the mechanism of how SOD-ScFv retarded the cell cycle, we examined the effects of reduced levels of intracellular ROS on some signal-transduction pathways. As shown in Fig. 8, it was found that SOD-ScFv dose dependently inhibited Akt phosphorylation (Thr-308) in a short time (1 h); in the subsequent 24 h, the expression of cyclin-dependent kinase inhibitors p27kip1 (a downstream protein of Akt) was up-regulated in SPC-A-1 cells. This outcome indicated that the Akt/p27kip1 pathway was activated under the reduced level of intracellular ROS in SPC-A-1 cells. However, the mechanism of Akt/p27kip1 pathway activation by ROS remains unclear. We predicted the tertiary structures of N. commune Fe-SOD by ExPASy and found that the four Fe3+-bound sites in the SOD active center, His-28, His-80, and His-166 and Asp-162 were close (a water molecule or a hydroxyl ion is the fifth bound site of Fe3+) (33Lim J.H. Yu Y.G. Choi I.G. Ryu J.R. Ahn B.Y. Kim S.H. Han Y.S. FEBS Lett. 1997; 406: 142-146Crossref PubMed Scopus (35) Google Scholar). Also, the C terminus amino acid residues of SOD were farther from the enzymatic active center than the N terminus; thus, the ScFv was added to the C terminus of SOD with link-1 to weaken the spatial interference between SOD and ScFv. The dual activity of SOD-ScFv indicated that SOD and ScFv were well separated by link-1. High intracellular ROS levels and low SOD activity were found in many tumor cells. Based on these findings in M14 melanoma cells, Pervaiz et al. (34Pervaiz S. Ramalingam J.K. Hirpara J.L. Clement M.V. FEBS Lett. 1999; 459: 343-348Crossref PubMed Scopus (86) Google Scholar) suggested that an intracellular environment with high ROS concentrations provided tumor cells with a survival advantage over their normal counterparts. It was suggested that ROS acts as an essential intracellular secondary messenger in the regulation of protein kinase activity and relative gene expression and in modulating cell growth, apoptosis, differentiation, and transformation (35Undaresan M. Yu Z. Ferrons V.J. Irani K. Finkel T. Science. 1995; 270: 296-299Crossref PubMed Scopus (2285) Google Scholar, 36Suh Y.A. Arnold R.S. Lassegue B. Shi J. Xu X. Sorescu D. Chung A.B. Griendling K.K. Lambeth J.D. Nature. 1999; 401: 79-82Crossref PubMed Scopus (1263) Google Scholar). Shi et al. (37Shi D.Y. Deng Y.R. Liu S.L. Zha
What problem does this paper attempt to address?
-
Construction and functional characterization of lung adenocarcinoma targeting SOD
Min Lu,Xingguo Gong,ChenHui Wang,Le Zheng,Jianjun Guo,ShenFeng Zhang
DOI: https://doi.org/10.3321/j.issn:1000-3282.2006.01.008
2006-01-01
PROGRESS IN BIOCHEMISTRY AND BIOPHYSICS
Abstract:In clinic, the low tumour-targeted ability of SOD is a critical shortage in its application, which is a difficulty for scientist at all time. For solving this problem, Nostoc commune CHEN iron-superoxide dismutase (Fe-SOD) and anti-lung adenocarcinoma LC-1 single chain Fv (ScFv) were fused, and the fusion protein named SOD-ScFv was expressed. After purification, fusion protein demonstrated both SOD and LC-1 antibody activities. The result of tracing of SOD-ScFv by FITC dyeing and quantification of ROS(reactive oxygan species) in SPC-A-1 (lung adenocarcinoma) cells showed that the fusion protein possessed the ability to recognize SPC-A-1 cells and eliminate the cellular ROS. The tumour-targeted theory put up in this research will overcome two applied disadvantages of SOD in clinic: it neither target the tumour cell nor permeates through the cell membrane. Also, the research provides a feasible idea that ScFv can be used to target the anticancer drug to tumour.
-
A novel molecular target, superoxide dismutase 1, in ALK inhibitor-resistant lung cancer cells, detected through proteomic analysis
Noriko Miyake,Nobuaki Ochi,Masami Takeyama,Hideko Isozaki,Eiki Ichihara,Hiromichi Yamane,Takuya Fukazawa,Yasunari Nagasaki,Tatsuyuki Kawahara,Hidekazu Nakanishi,Akio Hiraki,Katsuyuki Kiura,Nagio Takigawa
DOI: https://doi.org/10.1016/j.yexcr.2024.114266
2024-10-01
Abstract:Backgrounds: To the best of our knowledge, there are no reports of proteomic analysis for the identification of unknown proteins involved in resistance to anaplastic lymphoma kinase (ALK) inhibitors. In this study, we investigated the proteins involved in resistance to alectinib, a representative ALK inhibitor, through proteomic analysis and the possibility of overcoming resistance. Methods: An ALK-positive lung adenocarcinoma cell line (ABC-11) and the corresponding alectinib-resistant cell line (ABC-11/CHR2) were used. Two-dimensional difference gel electrophoresis (2D DIGE) was performed; the stained gel was scanned and the spots were analyzed using DeCyder TM2D 7.0. Mass spectrometry (MS) with the UltrafleXtreme matrix-assisted laser desorption ionization-tandem time-of-flight (MALDI-TOF/TOF) MS system was performed. For the MS/MS analysis, the samples were spotted on an AnchorChipTM 600 TF plate. The peptide masses obtained in the reflector positive mode were acquired at m/z of 400-6000. MS/MS data were searched against the NCBI protein databases. Growth inhibition was measured using an MTT assay. The isobologram and combination index were calculated based on the median-effect analysis. Western blotting was performed using antibodies, including superoxide dismutase (SOD) 1, MET, ERK, PARP, AKT, and BRCA1. Results: The 2D DIGE for ABC-11 and ABC-11/CHR2 showed different expression levels in about 2000 spots. SOD was identified from spots highly expressed in resistant strains. Western blotting also confirmed SOD1 overexpression in ABC-11/CHR2. siSOD1 enhanced the growth inhibitory effects of alectinib, increased cleaved PARP levels, and decreased pERK, pAKT, and BRCA1 levels with a combination of alectinib. In addition, the combination of LCS-1, an SOD1 inhibitor, and alectinib synergistically suppressed the growth in ABC-11/CHR2, but not in ABC-11. Conclusions: SOD1 overexpression is thought to be a mechanism for alectinib resistance, suggesting the possibility of overcoming resistance using SOD1 inhibitors.
-
Anti-proliferative Effects of Recombinant Iron Superoxide Dismutase on HepG2 Cells Via a Redox-Dependent PI3k/Akt Pathway.
Min Lu,Chong-shan Bi,Xing-guo Gong,Han-min Chen,Xie-huang Sheng,Tong-le Deng,Ke-di Xu
DOI: https://doi.org/10.1007/s00253-007-0939-3
IF: 5
2007-01-01
Applied Microbiology and Biotechnology
Abstract:The coding sequence for an iron superoxide dismutase (fe-sod) was amplified from the Nostoc commune genome. Recombinant Fe-SOD was overexpressed in Escherichia coli, accounting for ∼76% of total bacterial protein. Fe-SOD was purified from bacterial lysate by Ni-NTA column chromatography and used to generate an anti-SOD antibody. The purified Fe-SOD was encapsulated in liposomes and delivered to HepG2 liver tumor cells to eliminate cellular superoxide anions. The SOD-loaded cells exhibited lower reactive oxygen species (ROS) levels and higher reduced glutathione (GSH) levels. In Fe-SOD-treated cells, the cell cycle was delayed in the G1 phase, and HepG2 cell growth slowed in association with dephosphorylation of the serine–threonine kinase Akt. Low-dose H2O2 stimulated Akt phosphorylation, implying that Akt activation in HepG2 cells is redox-sensitive. Akt phosphorylation was abrogated by phosphatidylinositol 3-kinase (PI3K) inhibitors, suggesting that PI3K is an upstream mediator of Akt activation in HepG2 cells. This study provides insight into recombinant Fe-SOD-induced signaling mechanisms in liver tumor cells and suggests the feasibility of using Fe-SOD as an antitumor agent.
-
677. Polycationic Liposome-Mediated Extracellular Superoxide Dismutase Gene Delivery Leads to High Levels of the Transgene Expression and Prevents Acute Liver Injury in Mice
Jian Wu,Li Liu,Roy D. Yen,Andrea Catana,Mark A. Zern,Michael H. Nantz,Stefan L. Marklund
DOI: https://doi.org/10.1016/j.ymthe.2004.06.562
IF: 12.91
2004-01-01
Molecular Therapy
Abstract:BACKGROUND: We have developed a formulation of polycationic liposomes that has been shown to be non-toxic, highly stable in the bloodsteam, and very effective in liver gene transfer in mice (Gene Therapy 2003; 10:180–187). Extracellular superoxide dismutase (EC-SOD) inhibits the generation of superoxide anions in the interstitial space of tissues, and may play a role in minimizing oxidative stress in liver injury. The aim of the present study is to investigate whether our polycationic liposome-mediated human EC-SOD gene delivery will protect against lipopolysaccharide (LPS)-induced liver toxicity in D-galactosamine (GalN)-sensitized mice. METHODS: Polycationic liposomes were generated from polycationic lipid (PCL) and cholesterol (Chol). Lipoplexes were formed by complexing liposomes with control plasmid (pEGFP-C1) or EC-SOD plasmid (pEGFP-C1-ECSOD) before use. Mice were injected with thyroid hormone (T3, 4 mg/kg, s.c.) two days before lipoplexes were injected via the portal vein. One day following lipoplex injection, mice were treated with GalN (500 mg/kg, i.p.) plus LPS (25 mg/kg, i.p.). Serum alanine aminotransferase (ALT), SOD activity, liver histology, glutathione (GSH) content and lipid peroxidation were evaluated one day after GalN/LPS exposure. Human EC-SOD gene expression in mouse liver tissue was determined by quantitative RT-PCR two days after lipoplex injection. RESULTS: somes and the control lipoplexes were stable in their size for three months at 4°C. Injection of T3, PCL-Chol liposomes or control lipoplexes did not cause a significant change in serum ALT levels. Real time quantitative RT-PCR analysis showed that human EC-SOD mRNA levels in mouse liver tissue in EC-SOD lipoplex-injected group were 55-fold higher than saline, liposome or control lipoplex controls when mouse -actin was employed as a house-keeping control gene. Serum ALT levels in animals receiving portal vein injections of EC-SOD lipoplexes plus GalN/LPS exposure were much lower than in those receiving normal saline, liposomes alone, or control lipoplexes plus GalN/LPS exposure (62583 vs. 150865, 153194, 1880520 units/ml, p<0.01). Liver histology showed much less hepatocyte death in EC-SOD lipoplex-treated group than liposome- or control lipoplex-treated groups. Serum SOD activity in animals receiving injection of EC-SOD lipoplexes were higher than in those treated with liposomes only, or control lipoplexes (28.8 3.3 vs. 15.2 0.9, 17.4 3.4 units/ml, p<0.05). Liver GSH content in animals receiving EC-SOD lipoplex injection was preserved with less lipid peroxidation, as indicated by a lower level of malondialdehyde content. CONCLUSIONS: liposome-mediated EC-SOD gene delivery leads to the marked transgene expression, and attenuates acute liver injury due to reduced oxidative stress in GalN/LPS-exposed mice. To our knowledge, this is a first report which demonstrates that polycationic liposome-mediated EC-SOD gene delivery to the liver represents a potential therapy for hepatotoxin-induced liver injury.
-
Advanced biomimetic nanoreactor for specifically killing tumor cells through multi-enzyme cascade
Wen Liu,Jinpei Wu,Xin Ji,Yandong Ma,Lamei Liu,Xiaoqing Zong,Haiyuan Yang,Jian Dai,Xiaoyuan Chen,Wei Xue
DOI: https://doi.org/10.7150/thno.45456
IF: 11.6
2020-01-01
Theranostics
Abstract:<p>Although the enzyme catalytic nanoreactors reported so far have achieved excellent therapeutic efficacy, how to accurately exert enzyme activity in the tumor microenvironment to specifically kill tumor cells and avoid systemic oxidative damage would be an inevitable challenge for catalytic nanomedicine. At the present study, we fabricate an advanced biomimetic nanoreactor, SOD-Fe<sup>0</sup>@Lapa-ZRF for tumor multi-enzyme cascade delivery that combined specifically killing tumor cells and protect cells from oxidative stress.</p><p><b>Methods</b>: We first synthesized the FeNP-embedded SOD (SOD-Fe<sup>0</sup>) by reduction reaction using sodium borohydride. Next, SOD-Fe<sup>0</sup> and Lapa cargo were encapsulated in ZIF-8 by self-assembly. In order to protect the cargo enzyme from digestion by protease and prolong blood circulating time, SOD-Fe<sup>0</sup>@Lapa-Z was further cloaked with RBC membrane and functionalized with folate targeting, resulting in the final advanced biomimetic nanoreactor SOD-Fe<sup>0</sup>@Lapa-ZRF.</p><p><b>Results</b>: Once internalized, ZIF-8 achieves pH-triggered disassembly in weakly acidic tumor microenvironment. The released SOD-Fe<sup>0</sup> and Lapa were further endocytosed by tumor cells and the Lapa produces superoxide anion (O<sub>2</sub><sup>-•</sup>) through the catalysis of NQO1 that is overexpressed in tumor cells, while O<sub>2</sub><sup>-•</sup> is converted to H<sub>2</sub>O<sub>2</sub> via SOD. At this time, the released ferrous ions from SOD-Fe<sup>0</sup> and H<sub>2</sub>O<sub>2</sub> are further transformed to highly toxic hydroxyl radicals (•OH) for specifically killing tumor cells, and there was no obvious toxicological response during long-term treatment. Importantly, SOD-Fe<sup>0</sup>@Lapa-ZRF enhanced the normal cell's anti-oxidation ability, and thus had little effect on the secretion of TNF-α, IL-6 and IL-1β pro-inflammatory cytokines, while effectively reversed the decreased activity of T-SOD and GSH-Px and remained stable MDA content after tumor treatment. <i>In vitro</i> and <i>in vivo</i> results indicate that the tumor microenvironment-responsive release multi-enzyme cascade have high tumor specificity and effective anti-tumor efficacy, and can protect cells from oxidative stress damage.</p><p><b>Conclusion</b>: The biomimetic nanoreactor will have a great potential in cancer nanomedicine and provide a novel strategy to regulate oxidative stress.</p>
medicine, research & experimental
-
A new recombinant MS-superoxide dismutase alleviates 5-fluorouracil-induced intestinal mucositis in mice
Xiao-Xia Yan,Hai-Long Li,Yi-Ting Zhang,Shou-Yan Wu,Heng-Lei Lu,Xiao-Lu Yu,Fan-Guo Meng,Jian-Hua Sun,Li-Kun Gong
DOI: https://doi.org/10.1038/s41401-019-0295-8
Abstract:Intestinal mucositis is a common side effect of anticancer regimens that exerts a negative impact on chemotherapy. Superoxide dismutase (SOD) is a potential therapy for mucositis but efficient product is not available because the enzyme is degraded following oral administration or induces an immune reaction after intravascular infusion. Multi-modified Stable Anti-Oxidant Enzymes® (MS-AOE®) is a new recombinant SOD with better resistance to pepsin and trypsin. We referred it as MS-SOD to distinguish from other SODs. In this study we investigated its potential to alleviate 5-FU-induced intestinal injury and the mechanisms. An intestinal mucositis model was established in C57/BL6 mice by 5-day administration of 5-FU (50 mg/kg every day, ip). MS-SOD (800 IU/10 g, ig) was given once daily for 9 days. 5-FU caused severe mucositis with intestinal morphological damage, bodyweight loss and diarrhea; MS-SOD significantly decreased the severity. 5-FU markedly increased reactive oxygen species (ROS) and inflammatory cytokines in the intestine which were ameliorated by MS-SOD. Furthermore, MS-SOD modified intestinal microbes, particularly reduced Verrucomicrobia, compared with the 5-FU group. In Caco2 cells, MS-SOD (250-1000 U/mL) dose-dependently decreased tBHP-induced ROS generation. In RAW264.7 cells, MS-SOD (500 U/mL) had no effect on LPS-induced inflammatory cytokines, but inhibited iNOS expression. These results demonstrate that MS-SOD can scavenge ROS at the initial stage of injury, thus play an indirect role in anti-inflammatory and barrier protein protection. In conclusion, MS-SOD attenuates 5-FU-induced intestinal mucositis by suppressing oxidative stress and inflammation, and influencing microbes. MS-SOD may exert beneficial effect in prevention of intestinal mucositis during chemotherapy in clinic.
-
GST-TAT-SOD: Cell Permeable Bifunctional Antioxidant Enzyme-A Potential Selective Radioprotector
Jianru Pan,Huocong He,Ying Su,Guangjin Zheng,Junxin Wu,Shutao Liu,Pingfan Rao
DOI: https://doi.org/10.1155/2016/5935080
Abstract:Superoxide dismutase (SOD) fusion of TAT was proved to be radioprotective in our previous work. On that basis, a bifunctional recombinant protein which was the fusion of glutathione S-transferase (GST), SOD, and TAT was constructed and named GST-TAT-SOD. Herein we report the investigation of the cytotoxicity, cell-penetrating activity, and in vitro radioprotective effect of GST-TAT-SOD compared with wild SOD, single-function recombinant protein SOD-TAT, and amifostine. We demonstrated that wild SOD had little radioprotective effect on irradiated L-02 and Hep G2 cells while amifostine was protective to both cell lines. SOD-TAT or GST-TAT-SOD pretreatment 3 h prior to radiation protects irradiated normal liver cells against radiation damage by eliminating intracellular excrescent superoxide, reducing cellular MDA level, enhancing cellular antioxidant ability and colony formation ability, and reducing apoptosis rate. Compared with SOD-TAT, GST-TAT-SOD was proved to have better protective effect on irradiated normal liver cells and minimal effect on irradiated hepatoma cells. Besides, GST-TAT-SOD was safe for normal cells and effectively transduced into different organs in mice, including the brain. The characteristics of this protein suggest that it may be a potential radioprotective agent in cancer therapy better than amifostine. Fusion of two antioxidant enzymes and cell-penetrating peptides is potentially valuable in the development of radioprotective agent.
-
Novel Oxygen-Dependent Degradable Immunotoxin Regulated by the Ubiquitin-Proteasome System Reduces Nonspecific Cytotoxicity
Min Wei,Anxiang Chen,Jian Zhang,Yuhong Ren
DOI: https://doi.org/10.1021/acs.molpharmaceut.2c00503
2022-01-01
Molecular Pharmaceutics
Abstract:The use of bacterial toxins as antitumor agents has received considerable attention. Immunotoxins based on antigen recognition of single-chain antibodies have been widely explored for cancer therapy. Despite their impressive killing effect on tumor cells, immunotoxins still display unspecific toxicity with undesired side effects. High levels of hypoxia-inducible factor 1α (HIF-1α) are well-known indicators of hypoxia in cancer cells. In this study, different linkers were employed to fuse the immunotoxin DAB389-4D5 scFv (DS) with the oxygen-dependent degradation domain (ODDD) of HIF-1α, a domain selectively facilitating the accumulation of HIF-1α under hypoxia, to construct the oxygen-dependent degradable immunotoxin DS-ODDD (DSO). The engineered fusion protein DSO-2 containing a linker (G4S)3 possesses the best killing effect on cancer cells under hypoxia and displayed considerably reduced nonspecific toxicity to normal cells under normoxic conditions. Flow cytometry, immunofluorescence, and immunoblot analyses demonstrated that DSO-2 was degraded via the ubiquitin-proteasome pathway regulated by the oxygen-sensitive mechanism. Western blot analysis indicated that the degradation of DSO-2 significantly decreased the activation of apoptosis-related molecules in normal cells. The engineered immunotoxin with oxygen-sensing properties developed herein is a potential therapeutic agent for cancer treatment.
-
The Effect of Extracellular Superoxide Dismutase (SOD3) Gene in Lung Cancer
Yundi Zhang,Xuan Lu,Yueyang Zhang,Dongbo Zhao,Haoming Gong,Yuxin Du,Hua Sun
DOI: https://doi.org/10.3389/fonc.2022.722646
IF: 4.7
2022-03-08
Frontiers in Oncology
Abstract:Background The recognition of new diagnostic and prognostic biological markers for lung cancer, the most severe malignant tumor, is an essential and eager study. In a microenvironment, superoxide dismutase 3 (SOD3) can adjust active oxygen, and it refers to a secreted antioxidant enzyme. It was also found to be cancer-related, and in lung cancer, it was remarkably down-regulated. More and more new cancer research focuses on the function of SOD3. Despite this, there is no good description of SOD3 function in the LC progression. Methods Through bioinformatics analysis, we found that SOD3 was a possible novel lung cancer gene in this study. We analyzed data sets from Gene Expression Comprehensive Database (GEO) and the Cancer Genome Atlas (TCGA), and SOD3 expression was studied in lung cancer. This study estimated the SOD3 diagnosis and prognosis through gene expression differential display, gene set enrichment analysis (GSEA), enrichment and genomes (KEGG) analysis, and gene ontology (GO). Then in order to investigate the SOD3 presentation in lung cancer cells, we used Western Blot and also applied Flow cytometry to detect the impact of anti-tumor medicine on tumor cell apoptosis. Results We found that the expression level of SOD3 in lung cancer was low (P = 4.218E-29), while the survival of lung cancer patients with high SOD3 expression was shorter (LUSC p =0.00086, LUAD p=0.00038). According to the result of western blot, the expression of SOD3 in tumor cells was higher than that in normal cells. The ratio of early apoptosis induced by anti-cancer drugs was 10.5% in normal cells, 35.1% in squamous cell carcinoma and 36.9% in adenocarcinoma.The SOD3 high expression was associated with poor survival probability by multivariate analysis (HR: 1.006, 95% CI 1.002–1.011, p=0.006). Moreover, SOD3 high expression group had higher ESTIMATE scores, and larger amount of immune infiltrating cells. SOD3 expression is correlated with PDCD1 and CTLA4 expression. Conclusions SOD3 gene can be used as a prognostic gene in lung cancer patients, and lung cancer patients with high expression of this gene can reap worse prognostic outcome. It can be used as a new clinical method and prognosticator for lung cancer patients.
oncology
-
Liposome-mediated Extracellular Superoxide Dismutase Gene Delivery Protects Against Acute Liver Injury in Mice.
J Wu,L Liu,RD Yen,A Catana,MH Nantz,MA Zern
DOI: https://doi.org/10.1002/hep.20288
IF: 17.298
2004-01-01
Hepatology
Abstract:Our previous study demonstrated that polycationic liposomes are highly stable in the bloodstream and represent an effective agent for liver gene delivery. We report here that liposome-mediated extracellular superoxide dismutase (EC-SOD) gene delivery successfully prevented acute liver injury in mice. The therapeutic efficacy of EC-SOD gene delivery by polycationic liposomes was determined against the toxicity of superoxide anions and hydroxyethyl radicals in HepG2 cells and in a mouse model of acute liver injury caused by D-galactosamine and lipopolysaccharide intoxication. Transfection of HepG2 cells with an EC-SOD plasmid led to a striking increase in superoxide dismutase activity in the medium. The transfected cells had much less cell death after reactive oxygen species exposure compared with untransfected or control plasmid-transfected cells. In a model of acute liver injury, serum alanine aminotransferase levels in mice receiving portal vein injections of EC-SOD lipoplexes were much lower than in those receiving normal saline, liposomes alone, or control lipoplexes. Liver histology confirmed that there was less cell death in the EC-SOD lipoplex-treated group. Quantitative reverse transcriptase polymerase chain reaction showed a 55-fold increase in human EC-SOD gene expression in the liver of mice injected with EC-SOD lipoplexes. Serum superoxide dismutase activity in EC-SOD lipoplex-treated mice was higher than in the control groups; this was associated with higher liver glutathione levels and reduced lipid peroxidation. In conclusion, polycationic liposome-mediated EC-SOD gene delivery protects against reactive oxygen species toxicity in vitro and against lipopolysaccharide-induced acute liver injury in D-galactosamine-sensitized mice.
-
Preparation of Recombinant Its Biological Hybrid Human SOD2/3 and Activity Assay
程安阳,范立强,赵健,李小灵,王刚
DOI: https://doi.org/10.14135/j.cnki.1006-3080.2011.06.005
2011-01-01
Abstract:To obtain recombinant hybrid human superoxide dismutase (rhSOD2/3) and investigate whether SOD2/3 fusion protein could be transduced into and inhibit the growth of human cervical carcinoma cell, the recombinant plasmid pET-SOD2/3 was constructed and transformed into E. coli BL21 (DE3) to express rhSOD2/3, rhSOD2/3 was purified by Ni-NTA affinity chromatography with the property of its N-terminal (His)6 affinity tag. Heparin affinity chromatography was used to test the heparin-binding ability of rhSOD2/3. The purified SOD2 and SOD2/3 were incubated with Hela cells. FITC and MTT assay was used to test their anti-tumor growth activities. The hybrid SOD2/3 was successfully expressed in E. coli, accounted for about 30% of the total bacterial protein and had normal SOD activity. After one-step Ni-NTA affinity chromatography, SOD2/3 of 90% pure was obtained. rhSOD2/3 could bind on a heparin affinity column, indicating that it was successfully targeted to heparin. Cell experiment showed that rhSOD2/3 inhibited the growth of Hela cells, while SOD2 did not. Relative pure hybrid rhSOD2/3 was obtained. Compared to SOD2, rhSOD2/3 fusion protein with natural and active form could be transduced more effectively into Hela ceils and inhibit more strongly cell growth. The hybrid rhSOD2/3 can bind to cell surfaces and may aid in anti-tumor activity of SOD2 and may function as a rational therapeutic agent for treating free-radical-mediated diseases.
-
Fe-SOD Cooperates with Nutlin3 to Selectively Inhibit Cancer Cells in Vitro and in Vivo
Yong Qin,Wei Dai,Yu Wang,Xing-Guo Gong,Min Lu
DOI: https://doi.org/10.1016/j.bbrc.2013.01.001
IF: 3.1
2013-01-01
Biochemical and Biophysical Research Communications
Abstract:Nutlin3, a non-genotoxic agonist of p53, is currently in phase II clinical trials for cancer treatment. However, its effects on normal tissues and cell types remain largely to be determined. Drugs that can selectively target cancer cells as well as cooperate with the p53 pathway are thus greatly needed. Iron-superoxide dismutase (Fe-SOD) is a potential candidate as it selectively targets cancer cells by eliminating the abnormally high levels of reactive oxygen species (ROS) in cancer cells; it also inhibits cancer cell growth by induction of p27. Here, we show evidence that modulating redox and ROS homeostasis cooperates with Nutlin3 to selectively inhibit cancer cells in vitro and in vivo. Co-treatment of Fe-SOD and Nutlin3 showed synergistic inhibition on cancer cells in vitro, and the induction of p27 appeared to be involved. No effects were observed on normal cells. In addition, such co-treatment further exhibited synergistic inhibition on tumor growth in vivo in a murine B16 xenograft model, while the individual treatments only achieved very limited inhibition. Thus, Fe-SOD cooperated with Nutlin3 to selectively inhibit cancer cells in vitro and in vivo.
-
GSH/Ph Dual Activatable Crosslinked And Fluorinated Pei for Cancer Gene Therapy Through Endogenous Iron De‐Hijacking And in Situ Ros Amplification
Suleixin Yang,Yi Wu,Wenzhao Zhong,Ruie Chen,Meilin Wang,Meiwan Chen
DOI: https://doi.org/10.1002/adma.202304098
IF: 29.4
2023-09-11
Advanced Materials
Abstract:Ferroptosis‐related cancer therapy is limited by insufficient Fe2+/Fe3+ redox pair and hydrogen peroxide (H2O2) for producing lethal hydroxyl radicals (•OH). Although exogenous iron or ROS‐producing drugs can enhance ferroptosis, exploiting endogenous iron (labile iron pool, LIP) stored in ferritin and promoting ROS generation may be safer. Herein, a metal/drug‐free nanomedicine was developed for responsive LIP release and H2O2 generation on the mitochondria membranes, amplifying hydroxyl radical production to enhance ferroptosis‐mediated anti‐tumor effects. A GSH/pH dual activatable fluorinated and crosslinked polyethyleneimine with dialdehyde polyethylene glycol layer nanocomplex loaded with MTS‐KR‐SOD (Mitochondria‐targeting‐sequence‐KillerRed‐Superoxide Dismutase) and CRISPR/Cas9‐CA IX (Carbonic anhydrase IX) plasmids (FP@MC) were developed for enhanced ferroptosis through endogenous iron de‐hijacking and in situ ROS amplification. Two plasmids were constructed to knock down carbonic anhydrase IX and translate KillerRed‐SuperOxide Dismutase recombinant protein specifically on mitochondria membranes, respectively. The CA IX knockdown acidified the intracellular environment, leading the release of LIP from ferritin as a "flare" to initiate endogenous chemodynamic therapy. Meanwhile, MTS‐KR‐SOD generated H2O2 when irradiated by a 590 nm laser to assist chemodynamic therapy, leading to ROS amplification for mitochondria damage and lipid peroxide accumulation. The combined therapeutic effects aggravated cancer ferroptosis and suppressed tumor growth, providing a new paradigm for amplifying ROS and iron ions to promote ferroptosis‐related cancer therapy. This article is protected by copyright. All rights reserved
materials science, multidisciplinary,chemistry, physical,physics, applied, condensed matter,nanoscience & nanotechnology
-
A Compartmental Silica Nanoreactor for Multienzyme‐Regulated Superactive Catalytic Therapy
Chu Wu,Limei Qin,Dechao Niu,Xia Wang,Qi Zhang,Yinghui Shang,Min Hu,Xiaoke Han,Saiji Shen,Xing Qin,Songyan Xi,Yongsheng Li,Qigang Wang
DOI: https://doi.org/10.1002/adfm.202103531
IF: 19
2021-07-31
Advanced Functional Materials
Abstract:Many tumor therapies take advantage of upsetting the redox balance in tumor cells, but to do so requires excessive biochemical or physical attacks. The high-throughput simulation using multi-pathway techniques described herein can yield an increased efficacy in bio-oxidation. In this study, compartmental hierarchical nanoreactors are developed as an efficient multi-pathway singlet oxygen (1O2) generation system for superactive biocatalytic tumor therapy. The penetrated super cavity and connected dual-mesopore channels of the compartmental multienzyme nanoreactors are designed using the proposed heterogeneous template assembly for multi-enzyme complex (superoxide dismutase (SOD)-lactoperoxidase (LPO)) and photosensitizer molecule (indocyanine green (ICG)) encapsulation. Benefiting by the enhanced direct substrate diffusion between the interacting SOD–LPO complex and decrease in external diffusion, the parallel catalysis combined by the superactive cascade biocatalysis and enzyme-promoted photosensitization effect is verified by this compartmental silica nanoreactor system. The parallel pathways not only make full use of the products of SOD (H2O2 and O2), but also exhibit outstanding capability for 1O2 production, at ≈2.15 and 1.70 times augmented 1O2, respectively. Both in vitro and in vivo studies demonstrate the synergetic 1O2-mediated inhibition of tumor proliferation, lending this strategy great potential for the treatment of hypoxic tumors.
materials science, multidisciplinary,chemistry, physical,physics, applied, condensed matter,nanoscience & nanotechnology
-
Targeting Human Lung Adenocarcinoma with a Suppressor of Mitochondrial Superoxide Production
Nivea Dias Amoedo,Laetitia Dard,Saharnaz Sarlak,Walid Mahfouf,Wendy Blanchard,Benoît Rousseau,Julien Izotte,Stéphane Claverol,Didier Lacombe,Hamid Reza Rezvani,Ciro Leonardo Pierri,Rodrigue Rossignol
DOI: https://doi.org/10.1089/ars.2019.7892
2020-11-01
Abstract:Aims: REDOX signaling from reactive oxygen species (ROS) generated by the mitochondria (mitochondrial reactive oxygen species [mtROS]) has been implicated in cancer growth and survival. Here, we investigated the effect of 5-(4-methoxyphenyl)-3H-1,2-dithiole-3-thione (AOL), a recently characterized member of the new class of mtROS suppressors (S1QELs), on human lung adenocarcinoma proteome reprogramming, bioenergetics, and growth. Results: AOL reduced steady-state cellular ROS levels in human lung cancer cells without altering the catalytic activity of complex I. AOL treatment induced dose-dependent inhibition of lung cancer cell proliferation and triggered a reduction in tumor growth in vivo. Molecular investigations demonstrated that AOL reprogrammed the proteome of human lung cancer cells. In particular, AOL suppressed the determinants of the Warburg effect and increased the expression of the complex I subunit NDUFV1 which was also identified as AOL binding site using molecular modeling computer simulations. Comparison of the molecular changes induced by AOL and MitoTEMPO, an mtROS scavenger that is not an S1QEL, identified a core component of 217 proteins commonly altered by the two treatments, as well as drug-specific targets. Innovation: This study provides proof-of-concept data on the anticancer effect of AOL on mouse orthotopic human lung tumors. A unique dataset on proteomic reprogramming by AOL and MitoTEMPO is also provided. Lastly, our study revealed the repression of NDUFV1 by S1QEL AOL. Conclusion: Our findings demonstrate the preclinical anticancer properties of S1QEL AOL and delineate its mode of action on REDOX and cancer signaling.
-
Heme Sequestration Effectively Suppresses the Development and Progression of Both Lung Adenocarcinoma and Squamous Cell Carcinoma
Sanchareeka Dey,Adnin Ashrafi,Chantal Vidal,Nivesh Jain,Sarada Preeta Kalainayakan,Poorva Ghosh,Parinaz Sadat Alemi,Narges Salamat,Purna Chaitanya Konduri,Jung-Whan Kim,Li Zhang
DOI: https://doi.org/10.1158/1541-7786.MCR-21-0385
Abstract:Lung adenocarcinoma (ADC) and squamous cell carcinoma (SCC) are two most common subtypes of lung cancer. Here, to identify new, targetable molecular properties of both subtypes, we monitored changes in the levels of heme- and oxidative phosphorylation (OXPHOS)-related proteins during lung tumorigenesis. Heme is a central molecule for oxidative metabolism and ATP generation via OXPHOS. Notably, both lung ADC and SCC tumors can be induced in the genetically engineered KLLuc mouse model harboring the G12D Kras mutation and a conditional Lkb1 knockout. We found that the levels of the rate-limiting heme synthesis enzyme ALAS1 and uptake protein SLC48A1, along with OXPHOS complex subunits, progressively increased as lung tumorigenesis advanced. Our data demonstrated that elevated levels of heme- and OXPHOS-related proteins were associated with both ADC and SCC. Importantly, treatment of KLLuc mice with a heme-sequestering protein, HeSP2, that inhibits heme uptake in tumor cells effectively arrested lung tumor progression, and both ADC and SCC tumors were strongly suppressed. Additionally, HeSP2 effectively suppressed the growth of both SCC and ADC tumor xenografts in NOD/SCID mice. Further analyses indicated that HeSP2 effectively diminished OXPHOS in both ADC and SCC, reduced angiogenesis, alleviated tumor hypoxia, and suppressed cell proliferation. These results show that the advancing of lung tumorigenesis requires progressive increase in cellular heme synthesis and uptake, leading to intensified OXPHOS activity and ATP generation and promoting aggressive tumorigenic functions. IMPLICATIONS: Heme sequestration is an effective strategy for the suppression of both ADC and SCC tumor initiation and development.
-
Augment of Ferroptosis with Photothermal Enhanced Fenton Reaction and Glutathione Inhibition for Tumor Synergistic Nano-Catalytic Therapy
Qingcheng Song,Yiran Zhang,Hongzhi Hu,Xuemei Yang,Xin Xing,Jianhua Wu,Yanbin Zhu,Yingze Zhang
DOI: https://doi.org/10.2147/ijn.s480586
IF: 7.033
2024-11-20
International Journal of Nanomedicine
Abstract:Qingcheng Song, 1, 2, &ast Yiran Zhang, 3, &ast Hongzhi Hu, 4, &ast Xuemei Yang, 5 Xin Xing, 1, 2 Jianhua Wu, 6 Yanbin Zhu, 1, 2 Yingze Zhang 1, 2 1 Department of Orthopaedic Surgery, Third Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China; 2 Orthopaedic Institution of Hebei Province, Shijiazhuang, Hebei, People's Republic of China; 3 School of Medicine, Nankai University, Tianjin, People's Republic of China; 4 Department of Orthopedics, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, Hubei, People's Republic of China; 5 The Fourth Hospital of Shijiazhuang, Shijiazhuang, Hebei, People's Republic of China; 6 The Fourth Hospital of Hebei Medical University, Shijiazhuang, Hebei, People's Republic of China &astThese authors contributed equally to this work Correspondence: Yingze Zhang; Yanbin Zhu, Email ; Introduction: Ferroptosis-driven tumor ablation strategies based on nanotechnology could be achieved by elevating intracellular iron levels or inhibiting glutathione peroxidase 4 (GPX4) activity. However, the intracellular antioxidative defense mechanisms endow tumor cells with ferroptosis resistance capacity. The purpose of this study was to develop a synergistic therapeutic platform to enhance the efficacy of ferroptosis-based tumor therapy. Methods: In this study, a multifunctional nano-catalytic therapeutic platform (mFeB@PDA-FA) based on chemodynamic therapy (CDT) and photothermal therapy (PTT) was developed to effectively trigger ferroptosis in tumor. In our work, iron-based mesoporous Fe 3 O 4 nanoparticles (mFe 3 O 4 NPs) were employed for the encapsulation of L-buthionine sulfoximine (BSO), followed by the modification of folic acid-functionalized polydopamine (PDA) coating on the periphery. Then, the antitumor effect of mFeB@PDA-FA NPs was evaluated using Human OS cells (MNNG/HOS) and a subcutaneous xenograft model of osteosarcoma. Results: mFe 3 O 4 harboring multivalent elements (Fe 2+/3+ ) could catalyze hydrogen peroxide (H 2 O 2 ) into highly cytotoxic &dotOH, while the tumor microenvironment (TME)-responsive released BSO molecules inhibit the biosynthesis of GSH, thus achieving the deactivation of GPX4 and the enhancement of ferroptosis. Moreover, thanks to the remarkable photothermal conversion performance of mFe 3 O 4 and PDA shell, PTT further synergistically enhanced the efficacy of CDT and facilitated ferroptosis. Both in vivo and in vitro experiments confirmed that this synergistic therapy could achieve excellent tumor inhibition effects. Conclusion: The nanotherapeutic platform mFeB@PDA-FA could effectively disrupted the redox homeostasis in tumor cells for boosting ferroptosis through the combination of CDT, PTT and GSH elimination, which provided a new perspective for the treatment of ferroptosis sensitive tumors. Keywords: ferroptosis, chemodynamic therapy, photothermal therapy, Fenton reaction, BSO Graphical Osteosarcoma (OS) is the most common primary malignant bone tumor and the leading cause of cancer-related death in children and adolescents. 1,2 At present, the common treatment methods of OS are surgical resection, systemic chemotherapy and radiotherapy, but the therapeutic approaches are far from satisfactory due to chemotherapy resistance and postoperative distant metastasis. 3–5 Hence, the development of new strategies that combine biomedicine with emerging nanotechnology to find alternative ways to stimulate cell death may overcome the current therapeutic dilemma. 6 Ferroptosis, as a non-apoptotic form of programmed cell death, has received considerable attention due to its unique properties associated with various diseases, including cancer. 7,8 Different from traditional apoptosis and necrosis, ferroptosis is mainly characterized by iron-dependent ROS generation and lipid peroxidation (LPO), which is of great significance for killing tumor cells resistant to necrosis and apoptosis. 9,10 Therefore, the intracellular redox active iron (Fe 2+ ) level is the key elements in the process of inducing ferroptosis, which could catalyze the decomposition of hydrogen peroxide (H 2 O 2 ) in the tumor microenvironment (TME) to generate highly toxic hydroxyl radicals ( ̇OH) based on Fenton reaction and promote the accumulation of lipid peroxides, thus causing damage to the structure and inte -Abstract Truncated-
pharmacology & pharmacy,nanoscience & nanotechnology
-
Radiation-induced SOD2 Overexpression Sensitizes Colorectal Cancer to Radiation While Protecting Normal Tissue
Zhiqiang Zhang,Jinyi Lang,Zhi Cao,Rong Li,Xingyong Wang,Weidong Wang
DOI: https://doi.org/10.18632/oncotarget.13954
2016-01-01
Oncotarget
Abstract:This study investigated whether radiation-induced overexpression of superoxide dismutase 2 (SOD2) exerts radio-sensitizing effects on tumor cells while having radio-protective effects on normal cells during radio-activated gene therapy for human colorectal cancer. A chimeric promoter, C9BC, was generated by directly linking nine tandem CArG boxes to a CMV basic promoter, after which lentiviral vectors containing GFP and SOD2 gene driven by the C9BC promoter were constructed. Stably transfected HT-29 colorectal cancer cells and CCD 841 CoN normal colorectal cells were irradiated to a dose of 6-Gy, and cell proliferation and apoptosis were observed. Tumor xenografts and peritumoral skin tissue in BALB/c mice were infected with the therapeutic lentivirus and subsequently irradiated with a total dose of 6 Gy. In vitro experiments revealed that radiation-induced SOD2 overexpression inhibited tumor cell proliferation (61.89% vs. 40.17%, P < 0.01) and decreased apoptosis among normal cells (14.8% vs. 9.6%, P = 0.02) as compared to untransfected cells. Similar effects were observed in vivo. Thus radiation-induced SOD2 overexpression via the chimeric C9BC promoter increased the radiosensitivity of HT-29 human colorectal cancer cells and concurrently protected normal CCD 841 CoN colorectal cells from radiation damage.
-
The anti-tumor and renoprotection study of E-[c(RGDfK)2]/folic acid co-modified nanostructured lipid carrier loaded with doxorubicin hydrochloride/salvianolic acid A
Bing Zhang,Ying Zhang,Wenli Dang,Bin Xing,Changxiang Yu,Pan Guo,Jiaxin Pi,Xiuping Deng,Dongli Qi,Zhidong Liu
DOI: https://doi.org/10.1186/s12951-022-01628-x
2022-09-24
Abstract:Background: Poor in vivo targeting of tumors by chemotherapeutic drugs reduces their anti-cancer efficacy in the clinic. The discovery of over-expressed components on the tumor cell surface and their specific ligands provide a basis for targeting tumor cells. However, the differences in the expression levels of these receptors on the tumor cell surface limit the clinical application of anti-tumor preparations modified by a single ligand. Meanwhile, toxicity of chemotherapeutic drugs leads to poor tolerance to anti-tumor therapy. The discovery of natural active products antagonizing these toxic side effects offers an avenue for relieving cancer patients' pain during the treatment process. Since the advent of nanotechnology, interventions, such as loading appropriate drug combinations into nano-sized carriers and multiple tumor-targeting functional modifications on the carrier surface to enhance the anti-tumor effect and reduce toxic and side effects, have been widely used for treating tumors. Results: Nanocarriers containing doxorubicin hydrochloride (DOX) and salvianolic acid A (Sal A) are spherical with a diameter of about 18 nm; the encapsulation efficiency of both DOX and salvianolic acid A is greater than 80%. E-[c(RGDfK)2]/folic acid (FA) co-modification enabled nanostructured lipid carriers (NLC) to efficiently target a variety of tumor cells, including 4T1, MDA-MB-231, MCF-7, and A549 cells in vitro. Compared with other preparations (Sal A solution, NLC-Sal A, DOX solution, DOX injection, Sal A/DOX solution, NLC-DOX, NLC-Sal A/DOX, and E-[c(RGDfK)2]/FA-NLC-Sal A/DOX) in this experiment, the prepared E-[c(RGDfK)2]/FA-NLC-Sal A/DOX had the best anti-tumor effect. Compared with the normal saline group, it had the highest tumor volume inhibition rate (90.72%), the highest tumor weight inhibition rate (83.94%), led to the highest proportion of apoptosis among the tumor cells (61.30%) and the lowest fluorescence intensity of proliferation among the tumor cells (0.0083 ± 0.0011). Moreover, E-[c(RGDfK)2]/FA-NLC-Sal A/DOX had a low level of nephrotoxicity, with a low creatinine (Cre) concentration of 52.58 μmoL/L in the blood of mice, and no abnormalities were seen on pathological examination of the isolated kidneys at the end of the study. Sal A can antagonize the nephrotoxic effect of DOX. Free Sal A reduced the Cre concentration of the free DOX group by 61.64%. In NLC groups, Sal A reduced the Cre concentration of the DOX group by 42.47%. The E-[c(RGDfK)2]/FA modification reduced the side effects of the drug on the kidney, and the Cre concentration was reduced by 46.35% compared with the NLC-Sal A/DOX group. These interventions can potentially improve the tolerance of cancer patients to chemotherapy. Conclusion: The E-[c(RGDfK)2]/FA co-modified DOX/Sal A multifunctional nano-drug delivery system has a good therapeutic effect on tumors and low nephrotoxicity and is a promising anti-cancer strategy.
-
Asymmetric Coordination of Iron Single‐Atom Nanozymes with Efficient Self‐Cascade Catalysis for Ferroptosis Therapy
Wendong Liu,Qian Chen,Jiaxin Wu,Fanghua Zhang,Lu Han,Jia Liu,Hongyan Zhang,Zhe Hao,Enyu Shi,Yuzhu Sun,Ruizhong Zhang,Yinsong Wang,Libing Zhang
DOI: https://doi.org/10.1002/adfm.202312308
IF: 19
2023-12-27
Advanced Functional Materials
Abstract:Asymmetrically coordinated Fe‐N3S1 single‐atom nanozymes (Fe‐S/N‐C SAzymes) exhibit significantly stronger peroxidase (POD)‐ and glutathione oxidase (GSHOx)‐like activities than those of their symmetric Fe‐N4 counterparts (Fe‐N‐C) SAzymes and the classical Fe3O4 nanozyme. By camouflaging with macrophage membranes, the tumor‐targeting nanocatalytic agents (M@Fe‐S/N‐C) trigger enhanced self‐cascade catalysis in the tumor microenvironment for ferroptosis‐based tumor‐specific therapy. Single‐atom nanozymes (SAzymes) hold great promise in tumor therapy due to their maximized atomic utilization and well‐defined electronic structures. However, they still face challenges of activity, specificity, and targeting that impede therapeutic efficacy. Herein, a practical strategy is reported to construct asymmetric N, S‐coordinated Fe SAzymes (Fe‐S/N‐C). Benefiting from the regulatory influence of S atoms on the disruption of local charge symmetry of center Fe atoms, the Fe‐S/N‐C SAzymes exhibit significantly enhanced peroxidase (POD)‐ and glutathione oxidase (GSHOx)‐like activities, with catalytic efficiencies being 6.33 and 47.88 times higher than their symmetric Fe‐N4 counterparts, respectively. Theoretical calculations demonstrate that the asymmetric atomic interface configuration increases electron localization around center Fe sites, thus facilitating the adsorption and activation of H2O2 and O2. By camouflaging with macrophage membranes, the tumor‐targeting nanocatalytic agents (M@Fe‐S/N‐C) trigger enhanced self‐cascade catalysis in the tumor microenvironment for ferroptosis‐based tumor‐specific therapy. These results open up a promising avenue for addressing the limitations associated with current SAzymes‐based tumor therapies.
materials science, multidisciplinary,chemistry, physical,physics, applied, condensed matter,nanoscience & nanotechnology