10 research outputs found

    PNC-27, a Chimeric p53-Penetratin Peptide Binds to HDM-2 in a p53 Peptide-like Structure, Induces Selective Membrane-Pore Formation and Leads to Cancer Cell Lysis

    Get PDF
    PNC-27, a 32-residue peptide that contains an HDM-2 binding domain and a cell-penetrating peptide (CPP) leader sequence kills cancer, but not normal, cells by binding to HDM-2 associated with the plasma membrane and induces the formation of pores causing tumor cell lysis and necrosis. Conformational energy calculations on the structure of PNC-27 bound to HDM-2 suggest that 1:1 complexes form between PNC-27 and HDM-2 with the leader sequence pointing away from the complex. Immuno-scanning electron microscopy was carried out with cancer cells treated with PNC-27 and decorated with an anti-PNC-27 antibody coupled to 6 nm gold particles and an anti-HDM-2 antibody linked to 15 nm gold particles. We found multiple 6 nm- and 15 nm-labeled gold particles in approximately 1:1 ratios in layered ring-shaped structures in the pores near the cell surface suggesting that these complexes are important to the pore structure. No pores formed in the control, PNC-27-treated untransformed fibroblasts. Based on the theoretical and immuno-EM studies, we propose that the pores are lined by PNC-27 bound to HDM-2 at the membrane surface with the PNC-27 leader sequence lining the pores or by PNC-27 bound to HDM-2

    Plant Gel-Mediated Synthesis of Gold-Coated Nanoceria Using Ferula gummosa: Characterization and Estimation of Its Cellular Toxicity toward Breast Cancer Cell Lines

    Get PDF
    In this study, a novel method using Ferula gummosa gums as a capping agent was used to synthesize the nanoceria for the first time. The method was economical and performed at room temperature. Furthermore, it was coated with gold (Au/nanoceria) and fully characterized using X-ray powder diffraction (XRD), field emission scanning electron microscopy with energy-dispersive X-ray spectroscopy (FESEM-EDX), Fourier-transform infrared spectroscopy (FTIR), dynamic light scattering (DLS), and zeta potential (& zeta; potential). The crystallite size obtained from the results was 28.09 nm for Au/nanoceria. The energy-dispersive X-ray spectroscopy (EDX) analysis of Au/nanoceria revealed the compositional constituents of the product, which display the purity of the Au/nanoceria. The cell toxicity properties of the non-doped and Au-coated nanoceria were identified by a MTT analysis on a breast cancer cell line (MCF7). Additionally, human foreskin fibroblast cells (HFF) were used as a normal cell line. The cytotoxicity results indicated that the toxicological effect of Au/nanoceria on cancer cells was significant while having little toxic effect on normal cells. The toxicity effect of nanoceria clearly shows the dependence on dose and time, so, with increasing the dose of Au/nanoceria, the death of cancer cells also increases

    Effect of Organic Fertilizer on the Growth and Physiological Parameters of a Traditional Medicinal Plant under Salinity Stress Conditions

    No full text
    Foeniculum vulgare (fennel) is a medicinal and aromatic plant species from Apiaceae (Umbelliferae) and has been extensively used to treat digestive and pulmonary diseases. This plant is relatively sensitive to salinity. To investigate the effect of salinity stress at levels of 0, 40, and 80 mM NaCl in combination with 0 and 5% v/v vermicompost mixed with soil on the growth as well as the physiological and biochemical traits of two fennel landraces planted in Urmia and Shiraz areas, a factorial experiment was conducted as a randomized complete block design in three replications under greenhouse conditions. The plants were sampled in the flowering stage eleven weeks after cultivation. As the results showed, vermicompost treatment together with salinity stress could enhance the growth traits of the plants, such as the length and dry weight of shoots; leaf area and dry weight of roots; photosynthetic pigments, i.e., chlorophylls and carotenoids; membrane stability index; relative water content, soluble sugar, soluble protein, proline, total phenol, and anthocyanin in the shoots; mineral elements, i.e., phosphate, nitrate, zinc, molybdenum, magnesium, and iron in the shoots; and potassium and calcium in the shoots and roots. The interaction of vermicompost and salinity also decreased the aldehydes, total flavonoids, activity of catalase enzyme and shoot starch, soluble sugar and root proline, and sodium content of both shoots and roots. In a comparison of the two studied fennel landraces, the Shiraz landrace emerged to be less affected by salinity stress. In saline conditions, vermicompost caused a change in the physiological and biochemical parameters of both fennel landraces and improved their growth. The improvement in the growth conditions in the Urmia landrace was more obvious due to the use of vermicompost. Using vermicompost plus 40 mM NaCl salinity, the dry weight of the shoot and leaf surface of the Urmia landrace increased by about 3 and 2.5 times, respectively, and under 80 mM NaCl, the dry weight of the shoot and leaf surface increased by 2.7 and 1.2 times compared to the control. According to the experiments, it seems that vermicompost can limit the harmful effects of salinity on fennel plants by affecting photosynthetic pigments, osmolytes, phenolic compounds, antioxidants, the stability of membranes, and the availability of water and essential minerals

    Ketone Bodies Induce Unique Inhibition of Tumor Cell Proliferation and Enhance the Efficacy of Anti-Cancer Agents

    No full text
    The ketone bodies, sodium and lithium salts of acetoacetate (AcAc) and sodium 3-hydroxybutyrate (3-HB; commonly called beta-hydroxybutyrate) have been found to inhibit the proliferation of cancer cells. Previous studies have suggested that lithium itself may be an inhibiting agent but may be additive or synergistic with the effect of AcAc. We previously found that sodium acetoacetate (NaAcAc) inhibits the growth of human colon cancer cell line SW480. We report here similar results for several other cancer cell lines including ovarian, cervical and breast cancers. We found that NaAcAc does not kill cancer cells but rather blocks their proliferation. Similar inhibition of growth was seen in the effect of lithium ion alone (as LiCl). The effect of LiAcAc appears to be due to the combined effects of acetoacetate and the lithium ion. The ketone bodies, when given together with chemotherapeutic agents, rapamycin, methotrexate and the new peptide anti-cancer agent, PNC-27, substantially lowers their IC50 values for cancer cell, killing suggesting that ketone bodies and ketogenic diets may be powerful adjunct agents in treating human cancers

    PNC-27, a Chimeric p53-Penetratin Peptide Binds to HDM-2 in a p53 Peptide-like Structure, Induces Selective Membrane-Pore Formation and Leads to Cancer Cell Lysis

    Get PDF
    PNC-27, a 32-residue peptide that contains an HDM-2 binding domain and a cell-penetrating peptide (CPP) leader sequence kills cancer, but not normal, cells by binding to HDM-2 associated with the plasma membrane and induces the formation of pores causing tumor cell lysis and necrosis. Conformational energy calculations on the structure of PNC-27 bound to HDM-2 suggest that 1:1 complexes form between PNC-27 and HDM-2 with the leader sequence pointing away from the complex. Immuno-scanning electron microscopy was carried out with cancer cells treated with PNC-27 and decorated with an anti-PNC-27 antibody coupled to 6 nm gold particles and an anti-HDM-2 antibody linked to 15 nm gold particles. We found multiple 6 nm- and 15 nm-labeled gold particles in approximately 1:1 ratios in layered ring-shaped structures in the pores near the cell surface suggesting that these complexes are important to the pore structure. No pores formed in the control, PNC-27-treated untransformed fibroblasts. Based on the theoretical and immuno-EM studies, we propose that the pores are lined by PNC-27 bound to HDM-2 at the membrane surface with the PNC-27 leader sequence lining the pores or by PNC-27 bound to HDM-2

    Anticancer peptide PNC-27 adopts an HDM-2-binding conformation and kills cancer cells by binding to HDM-2 in their membranes

    No full text
    The anticancer peptide PNC-27, which contains an HDM-2-binding domain corresponding to residues 12-26 of p53 and a transmembrane-penetrating domain, has been found to kill cancer cells (but not normal cells) by inducing membranolysis. We find that our previously determined 3D structure of the p53 residues of PNC-27 is directly superimposable on the structure for the same residues bound to HDM-2, suggesting that the peptide may target HDM-2 in the membranes of cancer cells. We now find significant levels of HDM-2 in the membranes of a variety of cancer cells but not in the membranes of several untransformed cell lines. In colocalization experiments, we find that PNC-27 binds to cell membrane-bound HDM-2. We further transfected a plasmid expressing full-length HDM-2 with a membrane-localization signal into untransformed MCF-10-2A cells not susceptible to PNC-27 and found that these cells expressing full-length HDM-2 on their cell surface became susceptible to PNC-27. We conclude that PNC-27 targets HDM-2 in the membranes of cancer cells, allowing it to induce membranolysis of these cells selectively

    The penetratin sequence in the anticancer PNC-28 peptide causes tumor cell necrosis rather than apoptosis of human pancreatic cancer cells

    No full text
    Background PNC-27 and PNC-28 are p53-derived peptides from the human double minute (hdm-2) binding domain attached to penetratin. These peptides induce tumor cell necrosis of cancer cells, but not normal cells. The anticancer activity and mechanism of PNC-28 (p53 aa17–26-penetratin) was specifically studied against human pancreatic cancer. Methods MiaPaCa-2 cells were treated with PNC-28. Necrosis was determined by measuring lactate dehydrogenase (LDH) and apoptosis as assayed for measuring elevation of proapoptotic proteins. PNC-29, an unrelated peptide, and hdm-2-binding domain p53 aa12-26 without penetratin (PNC-26) were used as controls. Since there is evidence that penetratin is required for tumor cell necrosis, we tested “naked” p53 peptide without penetratin by transfecting a plasmid that encodes p53 aa17–26 segment of PNC-28 into MiaPaCa-2 and an untransformed rat pancreatic acinar cell line, BMRPA1. Time-lapse electron microscopy was employed to further elucidate anticancer mechanism. Results Treatment with PNC-28 does not result in the elevation of proapoptotic proteins found in p53-induced apoptosis, but elicits rapid release of LDH, indicative of tumor cell necrosis. Accordingly, we observed membrane pore formation and dose-dependent killing. In direct contrast, transfected MiaPaCa-2 cells underwent apoptosis, and not necrosis, as evidenced by expression of high levels of caspases-3 and 7 and annexin V with background levels of LDH. Conclusion These results suggest that PNC-28 may be effective in treating human pancreatic cancer. The penetratin sequence appears to be responsible for the fundamental change in the mechanism of action, inducing rapid necrosis initiated by membrane pore formation. Cancer cell death by apoptosis was observed in the absence of penetratin
    corecore