Cargando…

Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections

INTRODUCTION: Recurrent urinary tract infections (RUTIs) and recurrent vulvovaginal candidiasis (RVVCs) represent major healthcare problems all over the world. Antibiotics and antifungals are widely used for such infectious diseases, which is linked with microbial resistances and microbiota deleteri...

Descripción completa

Detalles Bibliográficos
Autores principales: Martín-Cruz, Leticia, Angelina, Alba, Baydemir, Ilayda, Bulut, Özlem, Subiza, José Luis, Netea, Mihai G., Domínguez-Andrés, Jorge, Palomares, Oscar
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9729253/
https://www.ncbi.nlm.nih.gov/pubmed/36505433
http://dx.doi.org/10.3389/fimmu.2022.1066383
_version_ 1784845448092057600
author Martín-Cruz, Leticia
Angelina, Alba
Baydemir, Ilayda
Bulut, Özlem
Subiza, José Luis
Netea, Mihai G.
Domínguez-Andrés, Jorge
Palomares, Oscar
author_facet Martín-Cruz, Leticia
Angelina, Alba
Baydemir, Ilayda
Bulut, Özlem
Subiza, José Luis
Netea, Mihai G.
Domínguez-Andrés, Jorge
Palomares, Oscar
author_sort Martín-Cruz, Leticia
collection PubMed
description INTRODUCTION: Recurrent urinary tract infections (RUTIs) and recurrent vulvovaginal candidiasis (RVVCs) represent major healthcare problems all over the world. Antibiotics and antifungals are widely used for such infectious diseases, which is linked with microbial resistances and microbiota deleterious effects. The development of novel approaches for genitourinary tract infections (GUTIs) such as trained immunity-based vaccines (TIbV) is therefore highly required. MV140 is a sublingual whole-cell heat-inactivated polybacterial preparation with demonstrated clinical efficacy for RUTIs. The sublingual heat-inactivated Candida albicans vaccine V132 has been developed for RVVCs. We previously showed that the combination of MV140 and V132 promotes potent Th1/Th17 and regulatory T-cell responses against antigens contained in the formulation and unrelated antigens. The specific contribution of each preparation to such effects and the underlying molecular mechanisms remain incompletely understood. METHODS: PBMC and monocytes were isolated from healthy donors and in vitro stimulated with V132, MV140 or MV140/V132. After 6 days of resting, cells were reestimulated with LPS and MV140. Analysis of cytokine production by ELISA, Seahorse assays for functional metabolic experiments and chromatin immunoprecipitation assays were performed. BALB/c mice were intraperitoneally and sublingually immunized with V132. RESULTS: We uncover that V132 induces trained immunity in human PBMCs and purified monocytes, significantly increasing the responses triggered by subsequent stimulation with MV140. Mechanistically, V132 drives metabolic rewiring towards increased glycolysis and oxidative phosphorylation and induces epigenetic reprogramming that enhances the transcription of the pro-inflammatory genes IL6 and TNFA. Splenocytes and peritoneal cells from V132-immunize mice show increased responses upon in vitro stimulation with MV140. Remarkably, splenocytes from sublingually V132-immunized and MV140 in vivo treatment mice show stronger Th17 responses than mice exposed to excipients upon in vitro stimulation with MV140. CONCLUSION: Overall, we provide novel mechanistic insights into how V132-induced trained immunity enhances both innate and adaptive immune responses triggered by MV140, which might open the door for new interventions for GUTIs with important clinical implications.
format Online
Article
Text
id pubmed-9729253
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-97292532022-12-09 Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections Martín-Cruz, Leticia Angelina, Alba Baydemir, Ilayda Bulut, Özlem Subiza, José Luis Netea, Mihai G. Domínguez-Andrés, Jorge Palomares, Oscar Front Immunol Immunology INTRODUCTION: Recurrent urinary tract infections (RUTIs) and recurrent vulvovaginal candidiasis (RVVCs) represent major healthcare problems all over the world. Antibiotics and antifungals are widely used for such infectious diseases, which is linked with microbial resistances and microbiota deleterious effects. The development of novel approaches for genitourinary tract infections (GUTIs) such as trained immunity-based vaccines (TIbV) is therefore highly required. MV140 is a sublingual whole-cell heat-inactivated polybacterial preparation with demonstrated clinical efficacy for RUTIs. The sublingual heat-inactivated Candida albicans vaccine V132 has been developed for RVVCs. We previously showed that the combination of MV140 and V132 promotes potent Th1/Th17 and regulatory T-cell responses against antigens contained in the formulation and unrelated antigens. The specific contribution of each preparation to such effects and the underlying molecular mechanisms remain incompletely understood. METHODS: PBMC and monocytes were isolated from healthy donors and in vitro stimulated with V132, MV140 or MV140/V132. After 6 days of resting, cells were reestimulated with LPS and MV140. Analysis of cytokine production by ELISA, Seahorse assays for functional metabolic experiments and chromatin immunoprecipitation assays were performed. BALB/c mice were intraperitoneally and sublingually immunized with V132. RESULTS: We uncover that V132 induces trained immunity in human PBMCs and purified monocytes, significantly increasing the responses triggered by subsequent stimulation with MV140. Mechanistically, V132 drives metabolic rewiring towards increased glycolysis and oxidative phosphorylation and induces epigenetic reprogramming that enhances the transcription of the pro-inflammatory genes IL6 and TNFA. Splenocytes and peritoneal cells from V132-immunize mice show increased responses upon in vitro stimulation with MV140. Remarkably, splenocytes from sublingually V132-immunized and MV140 in vivo treatment mice show stronger Th17 responses than mice exposed to excipients upon in vitro stimulation with MV140. CONCLUSION: Overall, we provide novel mechanistic insights into how V132-induced trained immunity enhances both innate and adaptive immune responses triggered by MV140, which might open the door for new interventions for GUTIs with important clinical implications. Frontiers Media S.A. 2022-11-24 /pmc/articles/PMC9729253/ /pubmed/36505433 http://dx.doi.org/10.3389/fimmu.2022.1066383 Text en Copyright © 2022 Martín-Cruz, Angelina, Baydemir, Bulut, Subiza, Netea, Domínguez-Andrés and Palomares https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Immunology
Martín-Cruz, Leticia
Angelina, Alba
Baydemir, Ilayda
Bulut, Özlem
Subiza, José Luis
Netea, Mihai G.
Domínguez-Andrés, Jorge
Palomares, Oscar
Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title_full Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title_fullStr Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title_full_unstemmed Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title_short Candida albicans V132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine MV140 for genitourinary tract infections
title_sort candida albicans v132 induces trained immunity and enhances the responses triggered by the polybacterial vaccine mv140 for genitourinary tract infections
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9729253/
https://www.ncbi.nlm.nih.gov/pubmed/36505433
http://dx.doi.org/10.3389/fimmu.2022.1066383
work_keys_str_mv AT martincruzleticia candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT angelinaalba candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT baydemirilayda candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT bulutozlem candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT subizajoseluis candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT neteamihaig candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT dominguezandresjorge candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections
AT palomaresoscar candidaalbicansv132inducestrainedimmunityandenhancestheresponsestriggeredbythepolybacterialvaccinemv140forgenitourinarytractinfections