Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.

The synthesis of inactive enzyme precursors, also known as "zymogens," serves as a mechanism for regulating the execution of selected catalytic activities in a desirable time and/or site. Zymogens are usually activated by proteolytic cleavage. Many viruses encode proteases that execute key...

Descripción completa

Guardado en:
Detalles Bibliográficos
Autores principales: Assaf Shapira, Meital Gal-Tanamy, Limor Nahary, Dana Litvak-Greenfeld, Romy Zemel, Ran Tur-Kaspa, Itai Benhar
Formato: article
Lenguaje:EN
Publicado: Public Library of Science (PLoS) 2011
Materias:
R
Q
Acceso en línea:https://doaj.org/article/52f1c18b1d9a4efdb713441d923e9515
Etiquetas: Agregar Etiqueta
Sin Etiquetas, Sea el primero en etiquetar este registro!
id oai:doaj.org-article:52f1c18b1d9a4efdb713441d923e9515
record_format dspace
spelling oai:doaj.org-article:52f1c18b1d9a4efdb713441d923e95152021-11-18T07:00:29ZEngineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.1932-620310.1371/journal.pone.0015916https://doaj.org/article/52f1c18b1d9a4efdb713441d923e95152011-01-01T00:00:00Zhttps://www.ncbi.nlm.nih.gov/pmc/articles/pmid/21264238/pdf/?tool=EBIhttps://doaj.org/toc/1932-6203The synthesis of inactive enzyme precursors, also known as "zymogens," serves as a mechanism for regulating the execution of selected catalytic activities in a desirable time and/or site. Zymogens are usually activated by proteolytic cleavage. Many viruses encode proteases that execute key proteolytic steps of the viral life cycle. Here, we describe a proof of concept for a therapeutic approach to fighting viral infections through eradication of virally infected cells exclusively, thus limiting virus production and spread. Using the hepatitis C virus (HCV) as a model, we designed two HCV NS3 protease-activated "zymogenized" chimeric toxins (which we denote "zymoxins"). In these recombinant constructs, the bacterial and plant toxins diphtheria toxin A (DTA) and Ricin A chain (RTA), respectively, were fused to rationally designed inhibitor peptides/domains via an HCV NS3 protease-cleavable linker. The above toxins were then fused to the binding and translocation domains of Pseudomonas exotoxin A in order to enable translocation into the mammalian cells cytoplasm. We show that these toxins exhibit NS3 cleavage dependent increase in enzymatic activity upon NS3 protease cleavage in vitro. Moreover, a higher level of cytotoxicity was observed when zymoxins were applied to NS3 expressing cells or to HCV infected cells, demonstrating a potential therapeutic window. The increase in toxin activity correlated with NS3 protease activity in the treated cells, thus the therapeutic window was larger in cells expressing recombinant NS3 than in HCV infected cells. This suggests that the "zymoxin" approach may be most appropriate for application to life-threatening acute infections where much higher levels of the activating protease would be expected.Assaf ShapiraMeital Gal-TanamyLimor NaharyDana Litvak-GreenfeldRomy ZemelRan Tur-KaspaItai BenharPublic Library of Science (PLoS)articleMedicineRScienceQENPLoS ONE, Vol 6, Iss 1, p e15916 (2011)
institution DOAJ
collection DOAJ
language EN
topic Medicine
R
Science
Q
spellingShingle Medicine
R
Science
Q
Assaf Shapira
Meital Gal-Tanamy
Limor Nahary
Dana Litvak-Greenfeld
Romy Zemel
Ran Tur-Kaspa
Itai Benhar
Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
description The synthesis of inactive enzyme precursors, also known as "zymogens," serves as a mechanism for regulating the execution of selected catalytic activities in a desirable time and/or site. Zymogens are usually activated by proteolytic cleavage. Many viruses encode proteases that execute key proteolytic steps of the viral life cycle. Here, we describe a proof of concept for a therapeutic approach to fighting viral infections through eradication of virally infected cells exclusively, thus limiting virus production and spread. Using the hepatitis C virus (HCV) as a model, we designed two HCV NS3 protease-activated "zymogenized" chimeric toxins (which we denote "zymoxins"). In these recombinant constructs, the bacterial and plant toxins diphtheria toxin A (DTA) and Ricin A chain (RTA), respectively, were fused to rationally designed inhibitor peptides/domains via an HCV NS3 protease-cleavable linker. The above toxins were then fused to the binding and translocation domains of Pseudomonas exotoxin A in order to enable translocation into the mammalian cells cytoplasm. We show that these toxins exhibit NS3 cleavage dependent increase in enzymatic activity upon NS3 protease cleavage in vitro. Moreover, a higher level of cytotoxicity was observed when zymoxins were applied to NS3 expressing cells or to HCV infected cells, demonstrating a potential therapeutic window. The increase in toxin activity correlated with NS3 protease activity in the treated cells, thus the therapeutic window was larger in cells expressing recombinant NS3 than in HCV infected cells. This suggests that the "zymoxin" approach may be most appropriate for application to life-threatening acute infections where much higher levels of the activating protease would be expected.
format article
author Assaf Shapira
Meital Gal-Tanamy
Limor Nahary
Dana Litvak-Greenfeld
Romy Zemel
Ran Tur-Kaspa
Itai Benhar
author_facet Assaf Shapira
Meital Gal-Tanamy
Limor Nahary
Dana Litvak-Greenfeld
Romy Zemel
Ran Tur-Kaspa
Itai Benhar
author_sort Assaf Shapira
title Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
title_short Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
title_full Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
title_fullStr Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
title_full_unstemmed Engineered toxins "zymoxins" are activated by the HCV NS3 protease by removal of an inhibitory protein domain.
title_sort engineered toxins "zymoxins" are activated by the hcv ns3 protease by removal of an inhibitory protein domain.
publisher Public Library of Science (PLoS)
publishDate 2011
url https://doaj.org/article/52f1c18b1d9a4efdb713441d923e9515
work_keys_str_mv AT assafshapira engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT meitalgaltanamy engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT limornahary engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT danalitvakgreenfeld engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT romyzemel engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT ranturkaspa engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
AT itaibenhar engineeredtoxinszymoxinsareactivatedbythehcvns3proteasebyremovalofaninhibitoryproteindomain
_version_ 1718424038891585536