ribosome inactivating protein
Recently Published Documents


TOTAL DOCUMENTS

415
(FIVE YEARS 23)

H-INDEX

38
(FIVE YEARS 1)

2021 ◽  
Vol 157 ◽  
pp. 112590
Author(s):  
Simin Chen ◽  
Cláudia Figueiredo Lóssio ◽  
Isabel Verbeke ◽  
Joost Verduijn ◽  
Bogdan Parakhonskiy ◽  
...  

Toxins ◽  
2021 ◽  
Vol 13 (10) ◽  
pp. 684
Author(s):  
Rossella Rotondo ◽  
Sara Ragucci ◽  
Salvatore Castaldo ◽  
Maria Antonietta Oliva ◽  
Nicola Landi ◽  
...  

Ribosome-inactivating proteins (RIPs) are found in several edible plants and are well characterized. Many studies highlight their use in cancer therapy, alone or as immunoconjugates, linked to monoclonal antibodies directed against target cancer cells. In this context, we investigate the cytotoxicity of quinoin, a novel type 1 RIP from quinoa seeds, on human continuous and primary glioblastoma cell lines. The cytotoxic effect of quinoin was assayed on human continuous glioblastoma U87Mg cells. Moreover, considering that common conventional glioblastoma multiforme (GBM) cell lines are genetically different from the tumors from which they derive, the cytotoxicity of quinoin was subsequently tested towards primary cells NULU and ZAR (two cell lines established from patients’ gliomas), also in combination with the chemotherapeutic agent temozolomide (TMZ), currently used in glioblastoma treatment. The present study demonstrated that quinoin (2.5 and 5.0 nM) strongly reduced glioblastoma cells’ growth. The mechanisms responsible for the inhibitory action of quinoin are different in the tested primary cell lines, reproducing the heterogeneous response of glioblastoma cells. Interestingly, primary cells treated with quinoin in combination with TMZ were more sensitive to the treatment. Overall, our data highlight that quinoin could represent a novel tool for glioblastoma therapy and a possible adjuvant for the treatment of the disease in combination with TMZ, alone or as possible immunoconjugates/nanoconstructs.


2021 ◽  
Vol 22 (16) ◽  
pp. 8964
Author(s):  
Sara Ragucci ◽  
Daniela Bulgari ◽  
Nicola Landi ◽  
Rosita Russo ◽  
Angela Clemente ◽  
...  

Quinoin is a type 1 ribosome-inactivating protein (RIP) we previously isolated from the seeds of pseudocereal quinoa (Chenopodium quinoa) and is known as a functional food for its beneficial effects on human health. As the presence of RIPs in edible plants could be potentially risky, here we further characterised biochemically the protein (complete amino acid sequence, homologies/differences with other RIPs and three-dimensional homology modeling) and explored its possible defensive role against pathogens. Quinoin consists of 254 amino acid residues, without cysteinyl residues. As demonstrated by similarities and homology modeling, quinoin preserves the amino acid residues of the active site (Tyr75, Tyr122, Glu177, Arg180, Phe181 and Trp206; quinoin numbering) and the RIP-fold characteristic of RIPs. The polypeptide chain of quinoin contains two N-glycosylation sites at Asn115 and Asp231, the second of which appears to be linked to sugars. Moreover, by comparative MALDI-TOF tryptic peptide mapping, two differently glycosylated forms of quinoin, named pre-quinoin-1 and pre-quinoin-2 (~0.11 mg/100 g and ~0.85 mg/100 g of seeds, respectively) were characterised. Finally, quinoin possesses: (i) strong antiviral activity, both in vitro and in vivo towards Tobacco Necrosis Virus (TNV); (ii) a growth inhibition effect on the bacterial pathogens of plants; and (iii) a slight antifungal effect against two Cryphonectria parasitica strains.


2021 ◽  
Vol 12 (3) ◽  
pp. 165-169
Author(s):  
T. Ghosh ◽  
◽  
Y. Vashi ◽  
K. Barman ◽  
L. I. Singha ◽  
...  

Ribosome-inactivating proteins (RIPs) are toxic N-glycosidases that depurinate eukaryotic and prokaryotic rRNAs and thus interrupt protein synthesis during translation. In the present study, a protein of around 32 kDa, supposedly a RIP isolated from Trichosanthes dioica, was assessed for its potential to induce apoptosis in HeLa cells. Cell viability assay was done to measure cell proliferation and survivability. It was observed that cells viability decreased with the increase of decrease in dilution, i.e. when the sample was an undiluted one, the viability decreased drastically and almost came to less than 10%. To further check whether the isolated RIP could induce apoptosis, HeLa cells were treated with the test RIP. Immunoblotting was carried out using PARP poly (ADP-ribose) polymerase (PARP-1), a 113 kDa nuclear enzyme, which is considered a hallmark of cells undergoing apoptosis. HeLa cells were further analyzed for loss of mitochondrial membrane potential with JC-1 dye, which is an early event during apoptosis. Increased PARP breakdown in the RIP treated cells indicates that cells undergoing apoptosis and progressive loss of red J-aggregate fluorescence indicate that the isolated RIP from Trichosanthes dioica induces apoptosis in HeLa cells. The ability of apoptosis induction is comparable to another known RIP from Momordica charantia, which was used as a positive control. Promising results from the present study warrants the isolated RIP to be further explored for anticancer activities.


Cells ◽  
2021 ◽  
Vol 10 (6) ◽  
pp. 1476
Author(s):  
Aurora K. Vikan ◽  
Michal Kostas ◽  
Ellen Margrethe Haugsten ◽  
Pål K. Selbo ◽  
Jørgen Wesche

Fibroblast growth factor receptors (FGFRs) have become an attractive target in cancer research and therapy due to their implication in several cancers. Limitations of current treatment options require a need for additional, more specific and potent strategies to overcome cancers driven by FGFRs. Photochemical internalization (PCI) is a light-controlled method for cytosolic delivery of drugs that are entrapped in endosomes and lysosomes. We here evaluated the efficacy and selectivity of PCI of FGF2-saporin (FGF-SAP) in cells overexpressing FGFR1. FGF-SAP is a conjugate of FGF2 and the highly cytotoxic ribosome-inactivating protein (RIP) saporin, which is used as payload to eliminate cancer cells. Evaluation of the targeting effect of PCI of FGF-SAP was done by comparing the cytotoxic response in osteosarcoma cells with very low levels of FGFR1 (U2OS) to cells overexpressing FGFR1 (U2OS-R1). We demonstrate that PCI greatly enhances cytotoxicity of the drug showing efficient cell killing at pM concentrations of the drug in U2OS-R1 cells. However, U2OS cells were also sensitive to the toxin after PCI. Binding experiments using confocal microscopy and Western blotting techniques indicate that FGF-SAP is taken up by cells through heparan sulfate proteoglycans (HSPGs) in U2OS cells. We further show that the cytotoxicity of FGF-SAP in U2OS cells was reduced when cells were co-treated with heparin to compete out binding to HSPG, demonstrating that the cytotoxic effect was due to internalization by HSPGs. We conclude that to prevent off-target effects of FGF-based toxins, it will be necessary to circumvent binding to HSPGs, for example by mutating the binding site of FGF2 to HSPGs.


2021 ◽  
Vol 12 ◽  
Author(s):  
Luana di Leandro ◽  
Francesco Giansanti ◽  
Sabrina Mei ◽  
Sara Ponziani ◽  
Martina Colasante ◽  
...  

A novel suicide gene therapy approach was tested in U87 MG glioblastoma multiforme cells. A 26nt G-rich double-stranded DNA aptamer (AS1411) was integrated into a vector at the 5′ of a mammalian codon-optimized saporin gene, under CMV promoter. With this plasmid termed “APTSAP”, the gene encoding ribosome-inactivating protein saporin is driven intracellularly by the glioma-specific aptamer that binds to cell surface-exposed nucleolin and efficiently kills target cells, more effectively as a polyethyleneimine (PEI)-polyplex. Cells that do not expose nucleolin at the cell surface such as 3T3 cells, used as a control, remain unaffected. Suicide gene-induced cell killing was not observed when the inactive saporin mutant SAPKQ DNA was used in the (PEI)-polyplex, indicating that saporin catalytic activity mediates the cytotoxic effect. Rather than apoptosis, cell death has features resembling autophagic or methuosis-like mechanisms. These main findings support the proof-of-concept of using PEI-polyplexed APTSAP for local delivery in rat glioblastoma models.


Author(s):  
Wallady da Silva Barroso ◽  
Talita Abrante Leite ◽  
Vanessa de Abreu Feitosa ◽  
Samara Sena da Penha ◽  
Elionai Gomes Freire ◽  
...  

Sign in / Sign up

Export Citation Format

Share Document