Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis

Detalhes bibliográficos
Autor(a) principal: Pires, David
Data de Publicação: 2023
Outros Autores: Mandal, Manoj, Matos, Ana I., Peres, Carina, Catalão, Maria João, Azevedo-Pereira, José Miguel, Satchi-Fainaro, Ronit, Florindo, Helena F., Anes, Elsa
Tipo de documento: Artigo
Idioma: eng
Título da fonte: Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
Texto Completo: http://hdl.handle.net/10400.14/41077
Resumo: The golden age of antibiotics for tuberculosis (TB) is marked by its success in the 1950s of the last century. However, TB is not under control, and the rise in antibiotic resistance worldwide is a major threat to global health care. Understanding the complex interactions between TB bacilli and their host can inform the rational design of better TB therapeutics, including vaccines, new antibiotics, and host-directed therapies. We recently demonstrated that the modulation of cystatin C in human macrophages via RNA silencing improved the anti-mycobacterial immune responses to Mycobacterium tuberculosis infection. Available in vitro transfection methods are not suitable for the clinical translation of host-cell RNA silencing. To overcome this limitation, we developed different RNA delivery systems (DSs) that target human macrophages. Human peripheral blood-derived macrophages and THP1 cells are difficult to transfect using available methods. In this work, a new potential nanomedicine based on chitosan (CS-DS) was efficiently developed to carry a siRNA-targeting cystatin C to the infected macrophage models. Consequently, an effective impact on the intracellular survival/replication of TB bacilli, including drug-resistant clinical strains, was observed. Altogether, these results suggest the potential use of CS-DS in adjunctive therapy for TB in combination or not with antibiotics.
id RCAP_78293ef416061f46692fdc9f64f5c175
oai_identifier_str oai:repositorio.ucp.pt:10400.14/41077
network_acronym_str RCAP
network_name_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository_id_str 7160
spelling Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosisAntibiotic resistanceCathepsin inhibitorsChitosanCystatinsHost-directed therapiesNanomedicinesTuberculosisThe golden age of antibiotics for tuberculosis (TB) is marked by its success in the 1950s of the last century. However, TB is not under control, and the rise in antibiotic resistance worldwide is a major threat to global health care. Understanding the complex interactions between TB bacilli and their host can inform the rational design of better TB therapeutics, including vaccines, new antibiotics, and host-directed therapies. We recently demonstrated that the modulation of cystatin C in human macrophages via RNA silencing improved the anti-mycobacterial immune responses to Mycobacterium tuberculosis infection. Available in vitro transfection methods are not suitable for the clinical translation of host-cell RNA silencing. To overcome this limitation, we developed different RNA delivery systems (DSs) that target human macrophages. Human peripheral blood-derived macrophages and THP1 cells are difficult to transfect using available methods. In this work, a new potential nanomedicine based on chitosan (CS-DS) was efficiently developed to carry a siRNA-targeting cystatin C to the infected macrophage models. Consequently, an effective impact on the intracellular survival/replication of TB bacilli, including drug-resistant clinical strains, was observed. Altogether, these results suggest the potential use of CS-DS in adjunctive therapy for TB in combination or not with antibiotics.Veritati - Repositório Institucional da Universidade Católica PortuguesaPires, DavidMandal, ManojMatos, Ana I.Peres, CarinaCatalão, Maria JoãoAzevedo-Pereira, José MiguelSatchi-Fainaro, RonitFlorindo, Helena F.Anes, Elsa2023-05-10T09:10:34Z2023-04-082023-04-08T00:00:00Zinfo:eu-repo/semantics/publishedVersioninfo:eu-repo/semantics/articleapplication/pdfhttp://hdl.handle.net/10400.14/41077eng2079-638210.3390/antibiotics1204072985153749019PMC1013532037107091000977007200001info:eu-repo/semantics/openAccessreponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãoinstacron:RCAAP2023-07-12T17:46:39Zoai:repositorio.ucp.pt:10400.14/41077Portal AgregadorONGhttps://www.rcaap.pt/oai/openaireopendoar:71602024-03-19T18:33:45.462344Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informaçãofalse
dc.title.none.fl_str_mv Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
title Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
spellingShingle Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
Pires, David
Antibiotic resistance
Cathepsin inhibitors
Chitosan
Cystatins
Host-directed therapies
Nanomedicines
Tuberculosis
title_short Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
title_full Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
title_fullStr Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
title_full_unstemmed Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
title_sort Development of chitosan particles loaded with siRNA for cystatin C to control intracellular drug-resistant mycobacterium tuberculosis
author Pires, David
author_facet Pires, David
Mandal, Manoj
Matos, Ana I.
Peres, Carina
Catalão, Maria João
Azevedo-Pereira, José Miguel
Satchi-Fainaro, Ronit
Florindo, Helena F.
Anes, Elsa
author_role author
author2 Mandal, Manoj
Matos, Ana I.
Peres, Carina
Catalão, Maria João
Azevedo-Pereira, José Miguel
Satchi-Fainaro, Ronit
Florindo, Helena F.
Anes, Elsa
author2_role author
author
author
author
author
author
author
author
dc.contributor.none.fl_str_mv Veritati - Repositório Institucional da Universidade Católica Portuguesa
dc.contributor.author.fl_str_mv Pires, David
Mandal, Manoj
Matos, Ana I.
Peres, Carina
Catalão, Maria João
Azevedo-Pereira, José Miguel
Satchi-Fainaro, Ronit
Florindo, Helena F.
Anes, Elsa
dc.subject.por.fl_str_mv Antibiotic resistance
Cathepsin inhibitors
Chitosan
Cystatins
Host-directed therapies
Nanomedicines
Tuberculosis
topic Antibiotic resistance
Cathepsin inhibitors
Chitosan
Cystatins
Host-directed therapies
Nanomedicines
Tuberculosis
description The golden age of antibiotics for tuberculosis (TB) is marked by its success in the 1950s of the last century. However, TB is not under control, and the rise in antibiotic resistance worldwide is a major threat to global health care. Understanding the complex interactions between TB bacilli and their host can inform the rational design of better TB therapeutics, including vaccines, new antibiotics, and host-directed therapies. We recently demonstrated that the modulation of cystatin C in human macrophages via RNA silencing improved the anti-mycobacterial immune responses to Mycobacterium tuberculosis infection. Available in vitro transfection methods are not suitable for the clinical translation of host-cell RNA silencing. To overcome this limitation, we developed different RNA delivery systems (DSs) that target human macrophages. Human peripheral blood-derived macrophages and THP1 cells are difficult to transfect using available methods. In this work, a new potential nanomedicine based on chitosan (CS-DS) was efficiently developed to carry a siRNA-targeting cystatin C to the infected macrophage models. Consequently, an effective impact on the intracellular survival/replication of TB bacilli, including drug-resistant clinical strains, was observed. Altogether, these results suggest the potential use of CS-DS in adjunctive therapy for TB in combination or not with antibiotics.
publishDate 2023
dc.date.none.fl_str_mv 2023-05-10T09:10:34Z
2023-04-08
2023-04-08T00:00:00Z
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/10400.14/41077
url http://hdl.handle.net/10400.14/41077
dc.language.iso.fl_str_mv eng
language eng
dc.relation.none.fl_str_mv 2079-6382
10.3390/antibiotics12040729
85153749019
PMC10135320
37107091
000977007200001
dc.rights.driver.fl_str_mv info:eu-repo/semantics/openAccess
eu_rights_str_mv openAccess
dc.format.none.fl_str_mv application/pdf
dc.source.none.fl_str_mv reponame:Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
instname:Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron:RCAAP
instname_str Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
instacron_str RCAAP
institution RCAAP
reponame_str Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
collection Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos)
repository.name.fl_str_mv Repositório Científico de Acesso Aberto de Portugal (Repositórios Cientìficos) - Agência para a Sociedade do Conhecimento (UMIC) - FCT - Sociedade da Informação
repository.mail.fl_str_mv
_version_ 1799132064372490240