Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy

Detalhes bibliográficos
Autor(a) principal: Carolina Angelica da Silva Parada
Data de Publicação: 2022
Outros Autores: William Tadeu Lara Festuccia, Luciano Borges Censoni, Ivarne Luis dos Santos Tersariol, Emer Suavinho Ferro, Ivan Pires de Oliveira, Mayara Calegaro Ferrari Gewehr, João Agostinho Machado Neto, Keli Cristina de Lima, Rosangela Aparecida dos Santos Eichler, Lucia Rossetti Lopes, Luiz Roberto Grassmann Bechara, Julio Cesar Batista Ferreira
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Institucional da UFMG
Texto Completo: http://hdl.handle.net/1843/62125
Resumo: Intracellular peptides (InPeps) generated by proteasomes were previously suggested as putative natural regulators of protein–protein interactions (PPI). Here, the main aim was to investigate the intracellular effects of intracellular peptide VFDVELL (VFD7) and related peptides on PPI. The internalization of the peptides was achieved using a C-terminus covalently bound cell-penetrating peptide (cpp; YGRKKRRQRRR). The possible inhibition of PPI was investigated using a NanoBiT® luciferase structural complementation reporter system, with a pair of plasmids vectors each encoding, simultaneously, either FK506-binding protein (FKBP) or FKBP-binding domain (FRB) of mechanistic target of rapamycin complex 1 (mTORC1). The interaction of FKBP–FRB within cells occurs under rapamycin induction. Results shown that rapamycin-induced interaction between FKBP–FRB within human embryonic kidney 293 (HEK293) cells was inhibited by VFD7-cpp (10–500 nM) and FDVEL- LYGRKKRRQRRR (VFD6-cpp; 1–500 nM); additional VFD7-cpp derivatives were either less or not effective in inhibiting FKBP–FRB interaction induced by rapamycin. Molecular dynamics simula- tions suggested that selected peptides, such as VFD7-cpp, VFD6-cpp, VFAVELLYGRKKKRRQRRR (VFA7-cpp), and VFEVELLYGRKKKRRQRRR (VFA7-cpp), bind to FKBP and to FRB protein surfaces. However, only VFD7-cpp and VFD6-cpp induced changes on FKBP structure, which could help with understanding their mechanism of PPI inhibition. InPeps extracted from HEK293 cells were found mainly associated with macromolecular components (i.e., proteins and/or nucleic acids), contributing to understanding InPeps’ intracellular proteolytic stability and mechanism of action-inhibiting PPI within cells. In a model of cell death induced by hypoxia-reoxygenation, VFD6-cpp (1 μM) increased the viability of mouse embryonic fibroblasts cells (MEF) expressing mTORC1-regulated autophagy-related gene 5 (Atg5), but not in autophagy-deficient MEF cells lacking the expression of Atg5. These data suggest that VFD6-cpp could have therapeutic applications reducing undesired side effects of rapamycin long-term treatments. In summary, the present report provides further evidence that In Peps have biological significance and could be valuable tools for the rational design of therapeutic molecules targeting intracellular PPI.
id UFMG_30d46d3e293a2733e6cc82fd64b18772
oai_identifier_str oai:repositorio.ufmg.br:1843/62125
network_acronym_str UFMG
network_name_str Repositório Institucional da UFMG
repository_id_str
spelling 2023-12-21T20:01:40Z2023-12-21T20:01:40Z20221132073-4409http://hdl.handle.net/1843/62125Intracellular peptides (InPeps) generated by proteasomes were previously suggested as putative natural regulators of protein–protein interactions (PPI). Here, the main aim was to investigate the intracellular effects of intracellular peptide VFDVELL (VFD7) and related peptides on PPI. The internalization of the peptides was achieved using a C-terminus covalently bound cell-penetrating peptide (cpp; YGRKKRRQRRR). The possible inhibition of PPI was investigated using a NanoBiT® luciferase structural complementation reporter system, with a pair of plasmids vectors each encoding, simultaneously, either FK506-binding protein (FKBP) or FKBP-binding domain (FRB) of mechanistic target of rapamycin complex 1 (mTORC1). The interaction of FKBP–FRB within cells occurs under rapamycin induction. Results shown that rapamycin-induced interaction between FKBP–FRB within human embryonic kidney 293 (HEK293) cells was inhibited by VFD7-cpp (10–500 nM) and FDVEL- LYGRKKRRQRRR (VFD6-cpp; 1–500 nM); additional VFD7-cpp derivatives were either less or not effective in inhibiting FKBP–FRB interaction induced by rapamycin. Molecular dynamics simula- tions suggested that selected peptides, such as VFD7-cpp, VFD6-cpp, VFAVELLYGRKKKRRQRRR (VFA7-cpp), and VFEVELLYGRKKKRRQRRR (VFA7-cpp), bind to FKBP and to FRB protein surfaces. However, only VFD7-cpp and VFD6-cpp induced changes on FKBP structure, which could help with understanding their mechanism of PPI inhibition. InPeps extracted from HEK293 cells were found mainly associated with macromolecular components (i.e., proteins and/or nucleic acids), contributing to understanding InPeps’ intracellular proteolytic stability and mechanism of action-inhibiting PPI within cells. In a model of cell death induced by hypoxia-reoxygenation, VFD6-cpp (1 μM) increased the viability of mouse embryonic fibroblasts cells (MEF) expressing mTORC1-regulated autophagy-related gene 5 (Atg5), but not in autophagy-deficient MEF cells lacking the expression of Atg5. These data suggest that VFD6-cpp could have therapeutic applications reducing undesired side effects of rapamycin long-term treatments. In summary, the present report provides further evidence that In Peps have biological significance and could be valuable tools for the rational design of therapeutic molecules targeting intracellular PPI.engUniversidade Federal de Minas GeraisUFMGBrasilICA - INSTITUTO DE CIÊNCIAS AGRÁRIASCellsPeptídeosProteinasProteina de ligaçãoIntracellular peptidesProtein–protein interactionmTORC1EdgotypeEffect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagyinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articlehttps://doi.org/10.3390/cells11030385Carolina Angelica da Silva ParadaWilliam Tadeu Lara FestucciaLuciano Borges CensoniIvarne Luis dos Santos TersariolEmer Suavinho FerroIvan Pires de OliveiraMayara Calegaro Ferrari GewehrJoão Agostinho Machado NetoKeli Cristina de LimaRosangela Aparecida dos Santos EichlerLucia Rossetti LopesLuiz Roberto Grassmann BecharaJulio Cesar Batista Ferreirainfo:eu-repo/semantics/openAccessreponame:Repositório Institucional da UFMGinstname:Universidade Federal de Minas Gerais (UFMG)instacron:UFMGLICENSELicense.txtLicense.txttext/plain; charset=utf-82042https://repositorio.ufmg.br/bitstream/1843/62125/1/License.txtfa505098d172de0bc8864fc1287ffe22MD51ORIGINALEffect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy_Ivan Pires de Oliveira.pdfEffect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy_Ivan Pires de Oliveira.pdfapplication/pdf5101258https://repositorio.ufmg.br/bitstream/1843/62125/2/Effect%20of%20fkbp12-derived%20intracellular%20peptides%20on%20rapamycin-induced%20fkbp-frb%20interaction%20and%20autophagy_Ivan%20Pires%20de%20Oliveira.pdf90e8d22624459a0efa57a795e3aa0b36MD521843/621252024-01-02 13:50:00.115oai:repositorio.ufmg.br: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Repositório de PublicaçõesPUBhttps://repositorio.ufmg.br/oaiopendoar:2024-01-02T16:50Repositório Institucional da UFMG - Universidade Federal de Minas Gerais (UFMG)false
dc.title.pt_BR.fl_str_mv Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
title Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
spellingShingle Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
Carolina Angelica da Silva Parada
Intracellular peptides
Protein–protein interaction
mTORC1
Edgotype
Peptídeos
Proteinas
Proteina de ligação
title_short Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
title_full Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
title_fullStr Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
title_full_unstemmed Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
title_sort Effect of fkbp12-derived intracellular peptides on rapamycin-induced fkbp-frb interaction and autophagy
author Carolina Angelica da Silva Parada
author_facet Carolina Angelica da Silva Parada
William Tadeu Lara Festuccia
Luciano Borges Censoni
Ivarne Luis dos Santos Tersariol
Emer Suavinho Ferro
Ivan Pires de Oliveira
Mayara Calegaro Ferrari Gewehr
João Agostinho Machado Neto
Keli Cristina de Lima
Rosangela Aparecida dos Santos Eichler
Lucia Rossetti Lopes
Luiz Roberto Grassmann Bechara
Julio Cesar Batista Ferreira
author_role author
author2 William Tadeu Lara Festuccia
Luciano Borges Censoni
Ivarne Luis dos Santos Tersariol
Emer Suavinho Ferro
Ivan Pires de Oliveira
Mayara Calegaro Ferrari Gewehr
João Agostinho Machado Neto
Keli Cristina de Lima
Rosangela Aparecida dos Santos Eichler
Lucia Rossetti Lopes
Luiz Roberto Grassmann Bechara
Julio Cesar Batista Ferreira
author2_role author
author
author
author
author
author
author
author
author
author
author
author
dc.contributor.author.fl_str_mv Carolina Angelica da Silva Parada
William Tadeu Lara Festuccia
Luciano Borges Censoni
Ivarne Luis dos Santos Tersariol
Emer Suavinho Ferro
Ivan Pires de Oliveira
Mayara Calegaro Ferrari Gewehr
João Agostinho Machado Neto
Keli Cristina de Lima
Rosangela Aparecida dos Santos Eichler
Lucia Rossetti Lopes
Luiz Roberto Grassmann Bechara
Julio Cesar Batista Ferreira
dc.subject.por.fl_str_mv Intracellular peptides
Protein–protein interaction
mTORC1
Edgotype
topic Intracellular peptides
Protein–protein interaction
mTORC1
Edgotype
Peptídeos
Proteinas
Proteina de ligação
dc.subject.other.pt_BR.fl_str_mv Peptídeos
Proteinas
Proteina de ligação
description Intracellular peptides (InPeps) generated by proteasomes were previously suggested as putative natural regulators of protein–protein interactions (PPI). Here, the main aim was to investigate the intracellular effects of intracellular peptide VFDVELL (VFD7) and related peptides on PPI. The internalization of the peptides was achieved using a C-terminus covalently bound cell-penetrating peptide (cpp; YGRKKRRQRRR). The possible inhibition of PPI was investigated using a NanoBiT® luciferase structural complementation reporter system, with a pair of plasmids vectors each encoding, simultaneously, either FK506-binding protein (FKBP) or FKBP-binding domain (FRB) of mechanistic target of rapamycin complex 1 (mTORC1). The interaction of FKBP–FRB within cells occurs under rapamycin induction. Results shown that rapamycin-induced interaction between FKBP–FRB within human embryonic kidney 293 (HEK293) cells was inhibited by VFD7-cpp (10–500 nM) and FDVEL- LYGRKKRRQRRR (VFD6-cpp; 1–500 nM); additional VFD7-cpp derivatives were either less or not effective in inhibiting FKBP–FRB interaction induced by rapamycin. Molecular dynamics simula- tions suggested that selected peptides, such as VFD7-cpp, VFD6-cpp, VFAVELLYGRKKKRRQRRR (VFA7-cpp), and VFEVELLYGRKKKRRQRRR (VFA7-cpp), bind to FKBP and to FRB protein surfaces. However, only VFD7-cpp and VFD6-cpp induced changes on FKBP structure, which could help with understanding their mechanism of PPI inhibition. InPeps extracted from HEK293 cells were found mainly associated with macromolecular components (i.e., proteins and/or nucleic acids), contributing to understanding InPeps’ intracellular proteolytic stability and mechanism of action-inhibiting PPI within cells. In a model of cell death induced by hypoxia-reoxygenation, VFD6-cpp (1 μM) increased the viability of mouse embryonic fibroblasts cells (MEF) expressing mTORC1-regulated autophagy-related gene 5 (Atg5), but not in autophagy-deficient MEF cells lacking the expression of Atg5. These data suggest that VFD6-cpp could have therapeutic applications reducing undesired side effects of rapamycin long-term treatments. In summary, the present report provides further evidence that In Peps have biological significance and could be valuable tools for the rational design of therapeutic molecules targeting intracellular PPI.
publishDate 2022
dc.date.issued.fl_str_mv 2022
dc.date.accessioned.fl_str_mv 2023-12-21T20:01:40Z
dc.date.available.fl_str_mv 2023-12-21T20:01:40Z
dc.type.status.fl_str_mv info:eu-repo/semantics/publishedVersion
dc.type.driver.fl_str_mv info:eu-repo/semantics/article
format article
status_str publishedVersion
dc.identifier.uri.fl_str_mv http://hdl.handle.net/1843/62125
dc.identifier.issn.pt_BR.fl_str_mv 2073-4409
identifier_str_mv 2073-4409
url http://hdl.handle.net/1843/62125
dc.language.iso.fl_str_mv eng
language eng
dc.relation.ispartof.none.fl_str_mv Cells
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.publisher.none.fl_str_mv Universidade Federal de Minas Gerais
dc.publisher.initials.fl_str_mv UFMG
dc.publisher.country.fl_str_mv Brasil
dc.publisher.department.fl_str_mv ICA - INSTITUTO DE CIÊNCIAS AGRÁRIAS
publisher.none.fl_str_mv Universidade Federal de Minas Gerais
dc.source.none.fl_str_mv reponame:Repositório Institucional da UFMG
instname:Universidade Federal de Minas Gerais (UFMG)
instacron:UFMG
instname_str Universidade Federal de Minas Gerais (UFMG)
instacron_str UFMG
institution UFMG
reponame_str Repositório Institucional da UFMG
collection Repositório Institucional da UFMG
bitstream.url.fl_str_mv https://repositorio.ufmg.br/bitstream/1843/62125/1/License.txt
https://repositorio.ufmg.br/bitstream/1843/62125/2/Effect%20of%20fkbp12-derived%20intracellular%20peptides%20on%20rapamycin-induced%20fkbp-frb%20interaction%20and%20autophagy_Ivan%20Pires%20de%20Oliveira.pdf
bitstream.checksum.fl_str_mv fa505098d172de0bc8864fc1287ffe22
90e8d22624459a0efa57a795e3aa0b36
bitstream.checksumAlgorithm.fl_str_mv MD5
MD5
repository.name.fl_str_mv Repositório Institucional da UFMG - Universidade Federal de Minas Gerais (UFMG)
repository.mail.fl_str_mv
_version_ 1803589198893023232