Cargando…

Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses

D-mannose can be transported into a variety of cells via glucose transporter (GLUT), and supraphysiological levels of D-mannose impairs tumor growth and modulates immune cell function through mechanisms such as interference with glycolysis and induction of oxidative stress. Blood-stage Plasmodium ma...

Descripción completa

Detalles Bibliográficos
Autores principales: Lv, Li, Xu, Zihao, Zhao, Meichen, Gao, Jian, Jiang, Rumeng, Wang, Qian, Shi, Xiaoyu
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9546034/
https://www.ncbi.nlm.nih.gov/pubmed/36211381
http://dx.doi.org/10.3389/fimmu.2022.859228
_version_ 1784804952211718144
author Lv, Li
Xu, Zihao
Zhao, Meichen
Gao, Jian
Jiang, Rumeng
Wang, Qian
Shi, Xiaoyu
author_facet Lv, Li
Xu, Zihao
Zhao, Meichen
Gao, Jian
Jiang, Rumeng
Wang, Qian
Shi, Xiaoyu
author_sort Lv, Li
collection PubMed
description D-mannose can be transported into a variety of cells via glucose transporter (GLUT), and supraphysiological levels of D-mannose impairs tumor growth and modulates immune cell function through mechanisms such as interference with glycolysis and induction of oxidative stress. Blood-stage Plasmodium mainly depends on glycolysis for energy supply and pathological immune response plays a vital role in cerebral malaria. However, it is not clear whether mannose affects malaria blood-stage infection. Here, we fed D-mannose to Plasmodium berghei-infected mice and found weight loss and reduced parasitemia without apparent side effects. Compromised parasitemia in C57BL/6 mice was accompanied by an increase in splenic macrophages compared to an untreated group. When mannose was applied to a rodent experimental cerebral malaria (ECM) model, the incidence of ECM decreased. Expression of activation marker CD69 on T cells in peripheral blood and the brain were reduced, and cerebral migration of activated T cells was prevented by decreased expression of CXCR3. These findings suggest that mannose inhibits Plasmodium infection by regulating multiple host immune responses and could serve as a potential strategy for facilitating malaria treatment.
format Online
Article
Text
id pubmed-9546034
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-95460342022-10-08 Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses Lv, Li Xu, Zihao Zhao, Meichen Gao, Jian Jiang, Rumeng Wang, Qian Shi, Xiaoyu Front Immunol Immunology D-mannose can be transported into a variety of cells via glucose transporter (GLUT), and supraphysiological levels of D-mannose impairs tumor growth and modulates immune cell function through mechanisms such as interference with glycolysis and induction of oxidative stress. Blood-stage Plasmodium mainly depends on glycolysis for energy supply and pathological immune response plays a vital role in cerebral malaria. However, it is not clear whether mannose affects malaria blood-stage infection. Here, we fed D-mannose to Plasmodium berghei-infected mice and found weight loss and reduced parasitemia without apparent side effects. Compromised parasitemia in C57BL/6 mice was accompanied by an increase in splenic macrophages compared to an untreated group. When mannose was applied to a rodent experimental cerebral malaria (ECM) model, the incidence of ECM decreased. Expression of activation marker CD69 on T cells in peripheral blood and the brain were reduced, and cerebral migration of activated T cells was prevented by decreased expression of CXCR3. These findings suggest that mannose inhibits Plasmodium infection by regulating multiple host immune responses and could serve as a potential strategy for facilitating malaria treatment. Frontiers Media S.A. 2022-09-23 /pmc/articles/PMC9546034/ /pubmed/36211381 http://dx.doi.org/10.3389/fimmu.2022.859228 Text en Copyright © 2022 Lv, Xu, Zhao, Gao, Jiang, Wang and Shi 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
Lv, Li
Xu, Zihao
Zhao, Meichen
Gao, Jian
Jiang, Rumeng
Wang, Qian
Shi, Xiaoyu
Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title_full Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title_fullStr Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title_full_unstemmed Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title_short Mannose inhibits Plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
title_sort mannose inhibits plasmodium parasite growth and cerebral malaria development via regulation of host immune responses
topic Immunology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9546034/
https://www.ncbi.nlm.nih.gov/pubmed/36211381
http://dx.doi.org/10.3389/fimmu.2022.859228
work_keys_str_mv AT lvli mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT xuzihao mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT zhaomeichen mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT gaojian mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT jiangrumeng mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT wangqian mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses
AT shixiaoyu mannoseinhibitsplasmodiumparasitegrowthandcerebralmalariadevelopmentviaregulationofhostimmuneresponses