Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 669
Filter
Add more filters

Publication year range
1.
Molecules ; 29(7)2024 Mar 22.
Article in English | MEDLINE | ID: mdl-38611715

ABSTRACT

The plant-derived toxin ricin is classified as a type 2 ribosome-inactivating protein (RIP) and currently lacks effective clinical antidotes. The toxicity of ricin is mainly due to its ricin toxin A chain (RTA), which has become an important target for drug development. Previous studies have identified two essential binding pockets in the active site of RTA, but most existing inhibitors only target one of these pockets. In this study, we used computer-aided virtual screening to identify a compound called RSMI-29, which potentially interacts with both active pockets of RTA. We found that RSMI-29 can directly bind to RTA and effectively attenuate protein synthesis inhibition and rRNA depurination induced by RTA or ricin, thereby inhibiting their cytotoxic effects on cells in vitro. Moreover, RSMI-29 significantly reduced ricin-mediated damage to the liver, spleen, intestine, and lungs in mice, demonstrating its detoxification effect against ricin in vivo. RSMI-29 also exhibited excellent drug-like properties, featuring a typical structural moiety of known sulfonamides and barbiturates. These findings suggest that RSMI-29 is a novel small-molecule inhibitor that specifically targets ricin toxin A chain, providing a potential therapeutic option for ricin intoxication.


Subject(s)
Ricin , Animals , Mice , Ribosome Inactivating Proteins, Type 2 , Drug Development , Hydrolases , Liver
2.
Toxicol Appl Pharmacol ; 449: 116139, 2022 08 15.
Article in English | MEDLINE | ID: mdl-35750203

ABSTRACT

Biomacromolecules such as proteins and nucleic acids are very attractive due to their high efficiency and specificity as cancer therapeutics. In fact, the endocytosed macromolecules are often trapped in the endosomes and cannot exhibit pharmacological effects well. Many strategies have been used to address this bottleneck, and one promising approach is to exploit the endosomal escape-promoting effect of triterpenoid saponins to aid in the release of biomacromolecules. Here, Raddeanin A (RA, an oleanane-type triterpenoid saponin) was proved to significantly promote endosomal escape as it recruited Galectin-9, an endosomal escape event reporter. As expected, RA effectively enhanced the anti-tumor effect of MAP30 (a type I ribosome-inactivating protein derived from Momordica charantia). However, based on the results of fluorescent colocalization, RA did not significantly promote MAP30 release from endosomes, suggesting that RA enhances MAP30 activity not only by promoting endosomal escape. Furthermore, it was found that the inhibitors of micropinocytosis and caveolae could almost completely inhibit the cytotoxicity of MAP30 combined with RA without affecting the cytotoxicity of MAP30 alone, indicating that RA may regulate the endocytic pathway of MAP30. Meanwhile, the effect of RA is related to the intra vesicular pH and cholesterol content on cell membrane, and is also cell-type dependent. Therefore, RA enhanced the anti-tumor effect of MAP30 in multiple ways, not just by promoting endosomal escape. Our findings will help to further decipher the possible mechanisms by which triterpenoid saponins enhance drug activity, and provide a new perspective for improving the activity of endocytosed drugs.


Subject(s)
Neoplasms , Saponins , Triterpenes , Endosomes/metabolism , Humans , Neoplasms/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/pharmacology , Saponins/pharmacology , Triterpenes/pharmacology
3.
Protein Expr Purif ; 185: 105893, 2021 09.
Article in English | MEDLINE | ID: mdl-33933613

ABSTRACT

MAP30 (Momordica antiviral protein 30kD) is a single-chain Ⅰ-type ribosome inactivating protein with a variety of biological activities, including anti-tumor ability. It was reported that MAP30 would serve as a novel and relatively safe agent for prophylaxis and treatment of liver cancer. To determine whether adding two tumor targeting peptides could improve the antitumor activities of MAP30, we genetically modified MAP30 with an RGD motif and a EGFRi motif, which is a ligand with high affinity for αvß3 integrins and with high affinity for EGFR. The recombinant protein ELRL-MAP30 (rELRL-MAP30) containing a GST-tag was expressed in E. coli. The rELRL-MAP30 was highly expressed in the soluble fraction after induction with 0.15 mM IPTG for 20 h at 16 °C. The purified rELRL-MAP30 appeared as a band on SDS-PAGE. It was identified by western blotting. Cytotoxicity of recombinant protein to HepG2, MDA-MB-231, HUVEC and MCF-7 cells was detected by MTT analysis. Half maximal inhibitory concentration (IC50) values were 54.64 µg/mL, 70.13 µg/mL, 146 µg/mL, 466.4 µg/mL, respectively. Proliferation inhibition assays indicated that rELRL-MAP30 could inhibit the growth of Human liver cancer cell HepG2 effectively. We found that rELRL-MAP30 significantly induced apoptosis in liver cancer cells, as evidenced by nuclear staining of DAPI. In addition, rELRL-MAP30 induced apoptosis in human liver cancer HepG2 cells by up-regulation of Bax as well as down-regulation of Bcl-2. Migration of cell line were markedly inhibited by rELRL-MAP30 in a dose-dependent manner compared to the recombinant MAP30 (rMAP30). In summary, the fusion protein displaying extremely potent cytotoxicity might be highly effective for tumor therapy.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Momordica charantia/chemistry , Peptides/genetics , Recombinant Fusion Proteins/genetics , Ribosome Inactivating Proteins, Type 2/genetics , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Cloning, Molecular , ErbB Receptors/antagonists & inhibitors , ErbB Receptors/genetics , ErbB Receptors/metabolism , Escherichia coli/genetics , Escherichia coli/metabolism , Gene Expression , Genetic Vectors/chemistry , Genetic Vectors/metabolism , Glutathione Transferase/genetics , Glutathione Transferase/metabolism , Hep G2 Cells , Human Umbilical Vein Endothelial Cells , Humans , Integrin alpha5/genetics , Integrin alpha5/metabolism , Integrin beta3/genetics , Integrin beta3/metabolism , MCF-7 Cells , Peptides/metabolism , Protein Binding , Proto-Oncogene Proteins c-bcl-2/antagonists & inhibitors , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Recombinant Fusion Proteins/metabolism , Recombinant Fusion Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , bcl-2-Associated X Protein/agonists , bcl-2-Associated X Protein/genetics , bcl-2-Associated X Protein/metabolism
4.
Int J Mol Sci ; 22(15)2021 Jul 31.
Article in English | MEDLINE | ID: mdl-34361050

ABSTRACT

Sialic acid (Sia) is considered as one of the most important biomolecules of life since its derivatives and terminal orientations on cell membranes and macromolecules play a major role in many biological and pathological processes. To date, there is only a limited number of active molecules that can selectively bind to Sia and this limitation has made the study of this glycan challenging. The lectin superfamily is a well-known family of glycan binding proteins, which encompasses many strong glycan binding peptides with diverse glycan affinities. Mistletoe lectin (ML) is considered one of the most active members of lectin family which was initially classified in early studies as a galactose binding lectin; more recent studies have suggested that the peptide can also actively bind to Sia. However, the details with respect to Sia binding of ML and the domain responsible for this binding are left unanswered because no comprehensive studies have been instigated. In this study, we sought to identify the binding domain responsible for the sialic acid affinity of mistletoe lectin isoform I (MLI) in comparison to the binding activity of elderberry lectin isoform I (SNA), which has long been identified as a potent Sia binding lectin. In order to execute this, we performed computational carbohydrate-protein docking for MLB and SNA with Neu5Ac and ß-Galactose. We further analyzed the coding sequence of both lectins and identified their glycan binding domains, which were later cloned upstream and downstream to green fluorescent protein (GFP) and expressed in Escherichia coli (E. coli). Finally, the glycan affinity of the expressed fusion proteins was assessed by using different biochemical and cell-based assays and the Sia binding domains were identified.


Subject(s)
N-Acetylneuraminic Acid/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Toxins, Biological/chemistry , Binding Sites , Cell Line, Tumor , Humans , Molecular Docking Simulation , N-Acetylneuraminic Acid/chemistry , Protein Binding , Ribosome Inactivating Proteins, Type 2/metabolism , Toxins, Biological/metabolism
5.
Pharmacol Res ; 161: 105157, 2020 11.
Article in English | MEDLINE | ID: mdl-32814169

ABSTRACT

Increasing evidence shows that Traditional Chinese Medicine (TCM) has an obvious appeal for cancer treatment, but there is still a lack of scientific investigation of its underlying molecular mechanisms. Bitter melon or bitter gourd (Momordica charantia) is an edible fruit that is commonly consumed, and it is used to cure different diseases in various ancient folk medical practices. We report that a bioactive protein, MAP30, isolated from bitter melon seeds exhibited potent anticancer and anti-chemoresistant effects on ovarian cancer cells. Functional studies revealed that MAP30 inhibited cancer cell migration, cell invasion, and cell proliferation in various ovarian cancer cells but not normal immortalized ovarian epithelial cells. When administered with cisplatin, MAP30 produced a synergistic effect on cisplatin-induced cell cytotoxicity in ovarian cancer cells. When low doses of cisplatin and MAP30 were co-injected intraperitoneally, a remarkable reduction of tumor dissemination and tumor growth was observed in an ovarian cancer ascites mouse model. Notably, blood tests confirmed that MAP30 did not cause any adverse effects on liver and kidney functions in the treated mice. MAP30 activated AMP-activated protein kinase (AMPK) signaling via CaMKKß and induced cell cycle arrest in the S-phase. MAP30 modulated cell metabolism of ovarian cancer cells via suppression of GLUT-1/-3-mediated glucose uptake, adipogenesis, and lipid droplet formation in tumor development and progression. MAP30 also induced an increase in intracellular Ca2+ ion concentration, which triggered ROS-mediated cancer cell death via apoptosis and ferroptosis. Collectively, these findings suggest that natural MAP30 is a non-toxic supplement that may enhance chemotherapeutic outcomes and benefit ovarian cancer patients with peritoneal metastases.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Cisplatin/pharmacology , Energy Metabolism/drug effects , Ferroptosis/drug effects , Momordica charantia , Ovarian Neoplasms/drug therapy , Ribosome Inactivating Proteins, Type 2/pharmacology , AMP-Activated Protein Kinases/metabolism , Animals , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Cycle Checkpoints/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Drug Synergism , Female , Glycolysis/drug effects , Humans , Lipogenesis/drug effects , Mice, Inbred BALB C , Mice, Nude , Momordica charantia/chemistry , Neoplasm Invasiveness , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Ribosome Inactivating Proteins, Type 2/isolation & purification , Xenograft Model Antitumor Assays
6.
Cell Mol Biol (Noisy-le-grand) ; 66(1): 42-48, 2020 Apr 20.
Article in English | MEDLINE | ID: mdl-32359382

ABSTRACT

To investigate the antitumor mechanism of MAP30 in human bladder cell line (T24) and its potential toxic effects in mice.  In this study, the biological behavior of MAP30's influence on bladder cell was investigated to reveal the antitumor mechanism and role of MAP30 in bladder cancer. MAP30 gene sequence optimized by gene synthesis codon was inserted into the prokaryotic expression vector pET-28a to produce a large amount of target protein in Escherichia coli. The protein product was obtained after purification. Membrane hydration method was used to prepare MAP30 liposome in order to enhance its membrane permeability. The effects of MAP30 on the viability, apoptosis and migration of T24 cell were assessed using 3­(4,5­dimethyl­thiazol­2­yl)­2,5­diphenyl­2H­tetrazolium bromide (MTT), flow cytometric and TUNEL assays, respectively. Mice were transfected with bladder cancer cells for 48 h. The expressions of apoptotic and non-apoptotic proteins were determined using Western blotting. Changes in tumor volume and occurrence of metastasis were assessed using luciferase assay. After 7 days, liver and kidney were excised for histological examination. The levels of reactive oxygen species (ROS), malondialdehyde (MDA), and reduced glutathione (GSH), and activities of catalase and glutathione peroxidase (GPx) were determined in serum or homogenate using enzyme-linked immunosorbent assay (ELISA). The yield of MAP30 after purification was significantly increased. The results of MTT assay showed that MAP30 significantly and concentration-dependently inhibited the proliferation and migration of T24 cells (p < 0.05). The prepared liposomes had uniform hydrated particle size of 132.6 nm, with encapsulation efficiency of 78 %. The inhibitory effect of MAP30 liposome on T24 cells was significantly higher than that of MAP30, and MAP30 significantly increased the number of apoptotic cells (p < 0.05). Western blotting showed that MAP30 significantly promoted the expression of caspase 3 (p < 0.05), but did not significantly affect the expressions of bcl-2 and bax (p > 0.05). It also significantly down-regulated the expressions of NF-kB, JNK and MMP2 (p < 0.05). Tumor formation was significantly inhibited, and tumor volume reduced in bladder cancer-bearing mice after treatment with MAP30 (p < 0.05). Histological examination showed that MAP30 induced mild histological changes in the liver and kidney of mice, and significantly increased the level of MDA at day 1 (p < 0.05). It also significantly and time-dependently increased ROS, but reduced GSH levels and activities of catalase and GPx (p < 0.05). However, MAP30 had no significant effect on DNA (p > 0.05). The apoptotic effect of MAP30 in T24 cells is mediated via activation of caspase-3 signaling pathway. The protein produces mild histological changes in the liver and kidney of mice, but has no significant effect on DNA.


Subject(s)
Antineoplastic Agents/therapeutic use , Ribosome Inactivating Proteins, Type 2/toxicity , Ribosome Inactivating Proteins, Type 2/therapeutic use , Urinary Bladder Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation/drug effects , Humans , Liposomes , Male , Mice , Ribosome Inactivating Proteins, Type 2/isolation & purification , Tumor Burden/drug effects
7.
Molecules ; 25(11)2020 May 31.
Article in English | MEDLINE | ID: mdl-32486427

ABSTRACT

Mistletoe (Viscum album) extracts have been used as alternative and complementary therapeutic preparations in multiple cancers for decades. Mistletoe lectins (ML-I, ML-II, and ML-III) are considered to be the main anticancer components of such preparations. In the present study, ML-II was transiently expressed in Nicotiana benthamiana using the pEAQ-HT expression system. Expression levels of up to 60 mg/kg of the infiltrated plant tissue were obtained, and a three-fold increase was achieved by adding the endoplasmic reticulum (ER) retention signal KDEL to the native ML-II sequence. The native protein containing His-tag and KDEL was purified by immobilized metal affinity chromatography (IMAC) and gel filtration. We found that the recombinant ML-II lectin was glycosylated and retained its carbohydrate-binding activity. In addition, we demonstrated that plant produced ML-II displayed anticancer activity in vitro, inhibiting non-small cell lung cancer H460 and A549 cells with EC50 values of 4 and 3.5 µg/mL, respectively. Annexin V-448A and PI double staining revealed that cell cytotoxicity occurred via apoptosis induction. These results indicate that ML-II transiently expressed in N. benthamiana plants is a promising candidate as an anticancer agent, although further optimization of production and purification methods is required to enable further in vitro testing, as well as in vivo assays.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Mistletoe/chemistry , Nicotiana/chemistry , Ribosome Inactivating Proteins, Type 2/chemistry , Toxins, Biological/chemistry , A549 Cells , Apoptosis , Carbohydrates/chemistry , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Line, Tumor , Cell Survival , Chromatography, Gel , Cloning, Molecular , Drug Screening Assays, Antitumor , Endoplasmic Reticulum/metabolism , Endotoxins/metabolism , Humans , Lectins/chemistry , Lung Neoplasms/drug therapy , Plants, Genetically Modified/chemistry , Nicotiana/genetics
8.
Dokl Biochem Biophys ; 494(1): 219-221, 2020 Sep.
Article in English | MEDLINE | ID: mdl-33119820

ABSTRACT

In the present study, we assessed the role of annexin 13 membrane-binding protein (ANXA13) in the intracellular transport of vesicles containing type II ribosome-inactivating proteins (RIP-IIs). A modified human intestinal epithelial cell line HT29 was used, in which the expression of ANXA13 was significantly reduced. The cytotoxic effect of ricin and viscumin was evaluated by modification of 28S ribosome RNA. The observed differences in the activity of toxins on the parental and modified HT29 lines indicate that ANXA13 plays a different role in the intracellular transport of vesicles containing the RIP-IIs.


Subject(s)
Annexins/metabolism , Chemical Warfare Agents/pharmacology , Colonic Neoplasms/pathology , Ribosome Inactivating Proteins, Type 2/pharmacology , Ribosome Inactivating Proteins/metabolism , Ribosomes/drug effects , Ricin/pharmacology , Toxins, Biological/pharmacology , Biological Transport , Cell Survival/drug effects , Colonic Neoplasms/drug therapy , Colonic Neoplasms/metabolism , HT29 Cells , Humans
9.
Dokl Biochem Biophys ; 493(1): 198-200, 2020 Jul.
Article in English | MEDLINE | ID: mdl-32894464

ABSTRACT

The role of proteasome proteins and proteins of the ERAD system in the cytotoxicity of type II ribosome-inactivating proteins ricin and viscumin was investigated. For this, the cell line of colorectal adenocarcinoma HT29, as well as the HT29-sh002 line obtained on its basis, were used. On the basis on the proteome analysis of these lines and the estimation of the proportion of inactivated ribosomes, it was shown that the contribution of the proteasome to the degradation of the catalytic subunits of toxins is different. The role of the Cdc37 co-chaperone in maintaining the stability of A subunit of viscumin in the cytoplasm is shown.


Subject(s)
Cell Cycle Proteins/metabolism , Chaperonins/metabolism , Colorectal Neoplasms/drug therapy , Proteasome Endopeptidase Complex/biosynthesis , Ribosome Inactivating Proteins, Type 2/pharmacology , Ricin/pharmacology , Toxins, Biological/pharmacology , Adenocarcinoma/drug therapy , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Cell Cycle Proteins/genetics , Chaperonins/genetics , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Cytoplasm/metabolism , Humans , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Ribosomes/metabolism , Tumor Cells, Cultured
10.
Biotechnol Bioeng ; 116(9): 2236-2249, 2019 09.
Article in English | MEDLINE | ID: mdl-31140580

ABSTRACT

Cancer is the leading cause of death in industrialized countries. Cancer therapy often involves monoclonal antibodies or small-molecule drugs, but carbohydrate-binding lectins such as mistletoe (Viscum album) viscumin offer a potential alternative treatment strategy. Viscumin is toxic in mammalian cells, ruling them out as an efficient production system, and it forms inclusion bodies in Escherichia coli such that purification requires complex and lengthy refolding steps. We therefore investigated the transient expression of viscumin in intact Nicotiana benthamiana plants and Nicotiana tabacum Bright Yellow 2 plant-cell packs (PCPs), comparing a full-length viscumin gene construct to separate constructs for the A and B chains. As determined by capillary electrophoresis the maximum yield of purified heterodimeric viscumin in N. benthamiana was ~7 mg/kg fresh biomass with the full-length construct. The yield was about 50% higher in PCPs but reduced 10-fold when coexpressing A and B chains as individual polypeptides. Using a single-step lactosyl-Sepharose affinity resin, we purified viscumin to ~54%. The absence of refolding steps resulted in estimated cost savings of more than 80% when transient expression in tobacco was compared with E. coli. Furthermore, the plant-derived product was ~3-fold more toxic than the bacterially produced counterpart. We conclude that plants offer a suitable alternative for the production of complex biopharmaceutical proteins that are toxic to mammalian cells and that form inclusion bodies in bacteria.


Subject(s)
Antineoplastic Agents, Phytogenic , Escherichia coli , Gene Expression , Nicotiana , Plant Cells/metabolism , Plant Proteins , Plants, Genetically Modified , Ribosome Inactivating Proteins, Type 2 , Toxins, Biological , Antineoplastic Agents, Phytogenic/biosynthesis , Antineoplastic Agents, Phytogenic/isolation & purification , Escherichia coli/genetics , Escherichia coli/metabolism , Plant Proteins/biosynthesis , Plant Proteins/genetics , Plant Proteins/isolation & purification , Plants, Genetically Modified/genetics , Plants, Genetically Modified/metabolism , Recombinant Proteins/biosynthesis , Recombinant Proteins/genetics , Recombinant Proteins/isolation & purification , Ribosome Inactivating Proteins, Type 2/biosynthesis , Ribosome Inactivating Proteins, Type 2/genetics , Ribosome Inactivating Proteins, Type 2/isolation & purification , Nicotiana/genetics , Nicotiana/metabolism , Toxins, Biological/biosynthesis , Toxins, Biological/genetics , Toxins, Biological/isolation & purification
11.
Anticancer Drugs ; 29(8): 736-747, 2018 09.
Article in English | MEDLINE | ID: mdl-29912010

ABSTRACT

Cyclosporin A (CsA) is a calcium antagonist and can enhance the efficacy of some protein drugs, but its mechanism remains unknown. In this study, MAP30, a ribosome-inactivating protein reported to have apoptotic effects on cancer cells, was fused with S3, an epidermal growth factor receptor (EGFR)-targeting peptide. In addition, CsA was used to investigate whether it can further promote the apoptotic effects of S3 fused MAP30 (MAP30-S3). Our result showed that the internalization of FITC-labeled MAP30-S3 was increased significantly by S3 in HeLa cells. Unexpectedly, MAP30-S3 only showed a minor decrease in the viability of EGFR-overexpressing cancer cells, including HeLa, SMMC-7721, and MGC803 (IC50>5 µmol/l). However, 2 µmol/l CsA significantly increased the cytotoxicity of MAP30-S3, especially for HeLa cells (IC50=40.3 nmol/l). In comparison, CsA did not further decrease the cytotoxicity of MAP30-S3 on MRC-5, an EGFR low-expressing cell line from normal lung tissue, indicating that CsA did not affect the cancer-targeting specificity of MAP30-S3. Our results also showed that CsA further increased the apoptotic activity of MAP30-S3 in HeLa cells. CsA could promote the endosomal escape of FITC-MAP30-S3 with a diffused pattern in the cytoplasm. Five endocytic inhibitors were used to investigate the cellular uptake mechanism of MAP30-S3, and the results showed that the endosomal escape-enhancing effect of CsA on MAP30-S3 may be associated with the clathrin-dependent endocytic pathways. Our study suggested that CsA could be a novel endosomal escape enhancer to potentiate the intracellular release of anticancer protein drugs, resulting in their improved therapeutic efficacy.


Subject(s)
Cyclosporine/pharmacology , Endosomes/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Recombinant Fusion Proteins/pharmacology , Ribosomal Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/pharmacology , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Synergism , HeLa Cells , Humans , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics , Ribosomal Proteins/chemistry , Ribosomal Proteins/genetics , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/genetics
12.
Biochim Biophys Acta Gen Subj ; 1862(3): 460-473, 2018 Mar.
Article in English | MEDLINE | ID: mdl-29154940

ABSTRACT

BACKGROUND: Sambucus ebulus is a rich source of ribosome-inactivating proteins (RIPs) and RIP-related lectins generated from multiple genes. These proteins differ in their structure, enzymatic activity and sugar binding specificity. METHODS: We have purified and characterized ebulin-RP from S. ebulus leaves and determined the amino acid sequence by cDNA cloning. Cytotoxicity was studied in a variety of cancer cells and a comparative study of the ability of ebulin-RP to bind sugars using "in vitro" and "in silico" approaches was performed. RESULTS: Ebulin-RP is a novel heterodimeric type 2 RIP present in S. ebulus leaves together with the type 2 RIP ebulin l, which displayed rRNA N-glycosidase activity but unlike ebulin l, lacked functional sugar binding domains. As a consequence of changes in its B-chain, ebulin-RP displayed lower cytotoxicity than ebulin l towards cancer cells and induced apoptosis as the predominant pattern of cell death. CONCLUSIONS: Ebulin-RP is a novel member of the ebulin gene family with low cytotoxicity as a result of deficient sugar binding domains. Type 2 RIP genes from Sambucus have evolved to render proteins with different sugar affinities that may be related to different biological activities and could result in an advantage for the plant. GENERAL SIGNIFICANCE: The ebulin family of RIPs and lectins can serve as a good model for studying the evolutionary process which may have occurred in RIPs. The lack of cytotoxicity of ebulin-RP makes it a good candidate as a toxic moiety in the construction of immunotoxins and conjugates directed against specific targets.


Subject(s)
Cytotoxins/isolation & purification , Ribosome Inactivating Proteins, Type 2/isolation & purification , Sambucus/enzymology , Sugars/metabolism , Amino Acid Sequence , Animals , Apoptosis/drug effects , Binding Sites , Cell Line , Cell Line, Tumor , Cell-Free System , Cytotoxins/chemistry , Cytotoxins/metabolism , Cytotoxins/pharmacology , Drug Screening Assays, Antitumor , Evolution, Molecular , Humans , Mesenchymal Stem Cells/drug effects , Mice , Models, Molecular , Molecular Docking Simulation , Nucleic Acids/drug effects , Phylogeny , Plant Leaves/enzymology , Protein Conformation , Protein Domains , Recombinant Proteins/metabolism , Recombinant Proteins/pharmacology , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Sequence Alignment , Sequence Homology, Amino Acid , Substrate Specificity
13.
Mol Biol (Mosk) ; 52(4): 675-682, 2018.
Article in Russian | MEDLINE | ID: mdl-30113033

ABSTRACT

The mistletoe lectin viscumin (MLI) is a ribosome-inactivating protein from Viscum album widely used in cancer therapy. Its antitumor properties are due to its immunomodulating action, previously demonstrated in experiments involving intravenous, subcutaneous, and oral administration of viscumin. To investigate whether viscumin has a cytotoxic effect on the intestinal epithelium, its safety was assessed using (i) impedance spectroscopy to measure the integrity of the colorectal adenocarcinoma Caco-2 cell monolayer after exposure to viscumin and (ii) a novel technique of determining the portion of viscumin-inactivated ribosomes. It was shown that inactivation of at least 20% of the ribosomes within 6 h did not lead to disruption of the Caco-2 cell monolayer or alter the physicochemical parameters of enterocyte membranes.


Subject(s)
Colorectal Neoplasms/drug therapy , Intestinal Mucosa/drug effects , Ribosome Inactivating Proteins, Type 2/pharmacology , Ribosomes/drug effects , Toxins, Biological/pharmacology , Caco-2 Cells , Cell Membrane/drug effects , Cell Survival/drug effects , Colorectal Neoplasms/pathology , Electric Impedance , Enterocytes/drug effects , Humans , Ribosomes/genetics
14.
Mol Carcinog ; 56(2): 389-401, 2017 02.
Article in English | MEDLINE | ID: mdl-27182794

ABSTRACT

Abrus agglutinin (AGG), a type II ribosome-inactivating protein has been found to induce mitochondrial apoptosis. In the present study, we documented that AGG-mediated Akt dephosphorylation led to ER stress resulting the induction of autophagy-dependent cell death through the canonical pathway in cervical cancer cells. Inhibition of autophagic death with 3-methyladenine (3-MA) and siRNA of Beclin-1 and ATG5 increased AGG-induced apoptosis. Further, inhibiting apoptosis by Z-DEVD-FMK and N-acetyl cysteine (NAC) increased autophagic cell death after AGG treatment, suggesting that AGG simultaneously induced autophagic and apoptotic death in HeLa cells. Additionally, it observed that AGG-induced autophagic cell death in Bax knock down (Bax-KD) and 5-FU resistant HeLa cells, confirming as an alternate cell killing pathway to apoptosis. At the molecular level, AGG-induced ER stress in PERK dependent pathway and inhibition of ER stress by salubrinal, eIF2α phosphatase inhibitor as well as siPERK reduced autophagic death in the presence of AGG. Further, our in silico and colocalization study showed that AGG interacted with pleckstrin homology (PH) domain of Akt to suppress its phosphorylation and consequent downstream mTOR dephosphorylation in HeLa cells. We showed that Akt overexpression could not augment GRP78 expression and reduced autophagic cell death by AGG as compared to pcDNA control, indicating Akt modulation was the upstream signal during AGG's ER stress mediated autophagic cell death. In conclusion, we established that AGG stimulated cell death by autophagy might be used as an alternative tumor suppressor mechanism in human cervical cancer. © 2016 Wiley Periodicals, Inc.


Subject(s)
Antineoplastic Agents/pharmacology , Autophagy/drug effects , Endoplasmic Reticulum Stress/drug effects , Plant Lectins/pharmacology , Pleckstrin Homology Domains/drug effects , Proto-Oncogene Proteins c-akt/metabolism , Ribosome Inactivating Proteins, Type 2/pharmacology , Abrus/chemistry , Antineoplastic Agents/isolation & purification , Endoplasmic Reticulum Chaperone BiP , Female , HeLa Cells , Humans , Models, Molecular , Plant Lectins/isolation & purification , Proto-Oncogene Proteins c-akt/chemistry , Ribosome Inactivating Proteins, Type 2/isolation & purification , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/metabolism , Uterine Cervical Neoplasms/pathology , eIF-2 Kinase/metabolism
15.
J Membr Biol ; 250(1): 77-87, 2017 02.
Article in English | MEDLINE | ID: mdl-27837242

ABSTRACT

Fluorescence correlation spectroscopy (FCS) is a sensitive analytical tool for investigation of processes accompanied by changes in the mobility of molecules and complexes. In the present work, peak intensity analysis (PIA) in combination with the solution stirring using FCS setup was applied to explore the interaction between fluorescently labeled protein ligands and corresponding receptors located on membranes. In the system composed of biotinylated liposomes and fluorescently labeled streptavidin as a ligand, PIA allowed us to determine the optimum receptor concentration and demonstrate the essential dependence of the binding efficacy on the length of the linker between the biotin group and the polar head group of the lipid. The binding was dependent on the size of liposomes which was varied by lipid extrusion through filters of different pore diameters. The sensitivity of the method was higher with the liposomes of larger sizes. The PIA approach can be applied not only to liposomes but also to relatively large objects, e.g., erythrocytes or Sepharose beads derivatized with lactose as a receptor for the binding of viscumin and ricin.


Subject(s)
Erythrocytes/metabolism , Lipids/chemistry , Proteins/chemistry , Spectrometry, Fluorescence , Animals , Biotin , Cattle , Liposomes/chemistry , Particle Size , Protein Binding , Proteins/metabolism , Ribosome Inactivating Proteins, Type 2/chemistry , Ribosome Inactivating Proteins, Type 2/metabolism , Ricin/chemistry , Ricin/metabolism , Staining and Labeling , Toxins, Biological/chemistry , Toxins, Biological/metabolism
16.
Mol Cell Biochem ; 426(1-2): 87-99, 2017 Feb.
Article in English | MEDLINE | ID: mdl-27868169

ABSTRACT

Leukemia is among the most aggressive and prevalent human malignant carcinoma. Chemotherapy is the preferred therapeutic strategy; however, recurrence of cancer and non-selective cytotoxicity are the major concerns. Unlike synthetic chemotherapeutic agents, mistletoe ribosome-inactivating protein (RIP) displays anti-tumor function in various types of cancers. However, its effect on leukemia cells is little explored. In this study, we assessed the impact of Viscum articulatum RIP (Articulatin-D) on the survival of acute T-cell leukemia cells and the involved molecular and cellular mechanisms. Cell proliferation assay showed that Articulatin-D suppressed the viability of leukemia cells selectively. We further confirmed that the elevation of mitochondrial membrane potential and exposure of phosphatidylserine are the early events of apoptosis induction in Articulatin-D-treated Jurkat cells. Subsequently, we found that Articulatin-D treatment induces apoptosis in Jurkat cells in a time- and concentration-dependent manner. In conclusion, we provided evidence that Articulatin-D efficiently activates caspase-8 involved in extrinsic pathway of apoptosis induction, which ultimately results in caspase-3-dependent DNA fragmentation of Jurkat cells. Further evaluation of Articulatin-D in cell culture and animal models may provide novel information on selective cytotoxicity to acute T-cell leukemia and its involvement in targeting tumor cell survival pathways.


Subject(s)
Apoptosis/drug effects , Caspase 8/metabolism , Cell Proliferation/drug effects , Plant Preparations/pharmacology , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/drug therapy , Ribosome Inactivating Proteins, Type 2/pharmacology , Toxins, Biological/pharmacology , Viscum/chemistry , DNA Fragmentation/drug effects , Enzyme Activation/drug effects , Humans , Jurkat Cells , Plant Preparations/chemistry , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/metabolism , Precursor T-Cell Lymphoblastic Leukemia-Lymphoma/pathology , Ribosome Inactivating Proteins, Type 2/chemistry , Toxins, Biological/chemistry
17.
Molecules ; 22(1)2017 Jan 06.
Article in English | MEDLINE | ID: mdl-28067841

ABSTRACT

Elderberry contains healthy low molecular weight nutraceuticals and lectins which are sequence-related to the elderberry allergen Sam n1. Some of these lectins are type II ribosome-inactivating proteins. The sensitivity of native lectins present in elderberry fruits and bark to the proteolysis triggered by in vitro simulated gastric and duodenal fluids has been investigated. It was found that these lectins are refractory to proteolysis. Nonetheless, incubation for 5-10 min in a boiling water bath completely sensitized them to the hydrolytic enzymes in vitro. Under these conditions neither total Folin-Ciocalteau's reagent reactive compounds, total anthocyanins and the mixture of cyanidin-3-glucoside plus cyanidin-3-sambubioside, nor antioxidant and free-radical scavenging activities were affected by more than 10% for incubations of up to 20 min. Therefore, short-time heat treatment reduces potential allergy-related risks deriving from elderberry consumption without seriously affecting its properties as an antioxidant and free-radical scavenging food.


Subject(s)
Allergens/chemistry , Antioxidants/chemistry , Fruit/chemistry , Plant Lectins/chemistry , Ribosome Inactivating Proteins, Type 2/chemistry , Sambucus nigra/chemistry , Allergens/isolation & purification , Antioxidants/isolation & purification , Hot Temperature , Pepsin A/chemistry , Plant Bark/chemistry , Plant Extracts/chemistry , Plant Lectins/isolation & purification , Plants, Medicinal , Protein Stability , Proteolysis , Ribosome Inactivating Proteins, Type 2/isolation & purification , Spain
19.
Bull Exp Biol Med ; 163(4): 482-485, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28853065

ABSTRACT

We studied the possibility of using viscumin lectin (MLI) for targeted delivery of antitumor drugs. Affinity of MLI for more than 600 oligosaccharide structures was determined and the glycosylation profiles of cell surface in various mouse tissues were analyzed. It was found that biodistribution of MLI was determined by not only expression of oligosaccharides specifically recognized by the lectin in tissue cells, but also by the structure of glycan in general.


Subject(s)
Plant Lectins/metabolism , Ribosome Inactivating Proteins, Type 2/metabolism , Toxins, Biological/metabolism , Animals , Female , Glycosylation , Male , Mice , Mice, Inbred BALB C , Oligosaccharides/metabolism , Polysaccharides/metabolism
20.
Bull Exp Biol Med ; 163(6): 745-748, 2017 Oct.
Article in English | MEDLINE | ID: mdl-29063321

ABSTRACT

External magnetic field is characterized by low toxicity and existence of magnetic properties, which contributes to an interest in the development of products from ferromagnetic nanoparticles (FNP) for antitumor therapy. Previously we synthesized a conjugate of ferromagnetic magnetite nanoparticles and viscumin (mistletoe lectin I, MLI), which exhibits the antitumor activity. Studying the pharmacological properties of this conjugate (FNP-MLI) was directed to the evaluation of FNP-MLI elimination after intratumor injection in mice. The elimination rate of FNP-MLI was much lower than that of native plant MLI. The presence of FNP-MLI was not accompanied by undesired changes in the tumor tissue. The use of a FNP-MLI conjugate allowed us to prolong the time of MLI presence in tissues without increasing the dose of exogenous lectin. These features contribute to the prolongation of an immunomodulatory effect of MLI.


Subject(s)
Adenocarcinoma/drug therapy , Antineoplastic Agents, Phytogenic/pharmacokinetics , Breast Neoplasms/drug therapy , Drug Carriers/administration & dosage , Magnetite Nanoparticles/administration & dosage , Ribosome Inactivating Proteins, Type 2/pharmacokinetics , Toxins, Biological/pharmacokinetics , Adenocarcinoma/diagnostic imaging , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Breast Neoplasms/diagnostic imaging , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Drug Carriers/pharmacokinetics , Female , Humans , Injections, Intralesional , Injections, Subcutaneous , Magnetic Resonance Imaging , Mice , Mice, SCID , Ribosome Inactivating Proteins, Type 2/pharmacology , Toxins, Biological/pharmacology , Xenograft Model Antitumor Assays
SELECTION OF CITATIONS
SEARCH DETAIL