Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria

Malaria and babesiosis, the two primary intraerythrocytic protozoan diseases of humans, have been reported in multiple cases of co-infection in endemic regions. As the geographic range and incidence of arthropod-borne infectious diseases is being affected by climate change, co-infection cases with P...

Full description

Bibliographic Details
Main Authors: Artemis Efstratiou, Eloiza May S. Galon, Guanbo Wang, Kousuke Umeda, Daisuke Kondoh, Mohamad Alaa Terkawi, Aiko Kume, Mingming Liu, Aaron Edmond Ringo, Huanping Guo, Yang Gao, Seung-Hun Lee, Jixu Li, Paul Franck Adjou Moumouni, Yoshifumi Nishikawa, Hiroshi Suzuki, Ikuo Igarashi, Xuenan Xuan
Format: Article
Language:English
Published: Frontiers Media S.A. 2020-04-01
Series:Frontiers in Cellular and Infection Microbiology
Subjects:
Online Access:https://www.frontiersin.org/article/10.3389/fcimb.2020.00193/full
id doaj-6a03d58795f0417e9ef8c613e3f9bd34
record_format Article
collection DOAJ
language English
format Article
sources DOAJ
author Artemis Efstratiou
Eloiza May S. Galon
Guanbo Wang
Kousuke Umeda
Daisuke Kondoh
Mohamad Alaa Terkawi
Mohamad Alaa Terkawi
Aiko Kume
Mingming Liu
Aaron Edmond Ringo
Huanping Guo
Yang Gao
Seung-Hun Lee
Jixu Li
Paul Franck Adjou Moumouni
Yoshifumi Nishikawa
Hiroshi Suzuki
Ikuo Igarashi
Xuenan Xuan
spellingShingle Artemis Efstratiou
Eloiza May S. Galon
Guanbo Wang
Kousuke Umeda
Daisuke Kondoh
Mohamad Alaa Terkawi
Mohamad Alaa Terkawi
Aiko Kume
Mingming Liu
Aaron Edmond Ringo
Huanping Guo
Yang Gao
Seung-Hun Lee
Jixu Li
Paul Franck Adjou Moumouni
Yoshifumi Nishikawa
Hiroshi Suzuki
Ikuo Igarashi
Xuenan Xuan
Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
Frontiers in Cellular and Infection Microbiology
Plasmodium chabaudi
malaria
Babesia microti
babesiosis
infection
macrophages
author_facet Artemis Efstratiou
Eloiza May S. Galon
Guanbo Wang
Kousuke Umeda
Daisuke Kondoh
Mohamad Alaa Terkawi
Mohamad Alaa Terkawi
Aiko Kume
Mingming Liu
Aaron Edmond Ringo
Huanping Guo
Yang Gao
Seung-Hun Lee
Jixu Li
Paul Franck Adjou Moumouni
Yoshifumi Nishikawa
Hiroshi Suzuki
Ikuo Igarashi
Xuenan Xuan
author_sort Artemis Efstratiou
title Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
title_short Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
title_full Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
title_fullStr Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
title_full_unstemmed Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine Malaria
title_sort babesia microti confers macrophage-based cross-protective immunity against murine malaria
publisher Frontiers Media S.A.
series Frontiers in Cellular and Infection Microbiology
issn 2235-2988
publishDate 2020-04-01
description Malaria and babesiosis, the two primary intraerythrocytic protozoan diseases of humans, have been reported in multiple cases of co-infection in endemic regions. As the geographic range and incidence of arthropod-borne infectious diseases is being affected by climate change, co-infection cases with Plasmodium and Babesia are likely to increase. The two parasites have been used in experimental settings, where prior infection with Babesia microti has been shown to protect against fatal malarial infections in mice and primates. However, the immunological mechanisms behind such phenomena of cross-protection remain unknown. Here, we investigated the effect of a primary B. microti infection on the outcome of a lethal P. chabaudi challenge infection using a murine model. Simultaneous infection with both pathogens led to high mortality rates in immunocompetent BALB/c mice, similar to control mice infected with P. chabaudi alone. On the other hand, mice with various stages of B. microti primary infection were thoroughly immune to a subsequent P. chabaudi challenge. Protected mice exhibited decreased levels of serum antibodies and pro-inflammatory cytokines during early stages of challenge infection. Mice repeatedly immunized with dead B. microti quickly succumbed to P. chabaudi infection, despite induction of high antibody responses. Notably, cross-protection was observed in mice lacking functional B and T lymphocytes. When the role of other innate immune effector cells was examined, NK cell-depleted mice with chronic B. microti infection were also found to be protected against P. chabaudi. Conversely, in vivo macrophage depletion rendered the mice vulnerable to P. chabaudi. The above results show that the mechanism of cross-protection conferred by B. microti against P. chabaudi is innate immunity-based, and suggest that it relies predominantly upon the function of macrophages. Further research is needed for elucidating the malaria-suppressing effects of babesiosis, with a vision toward development of novel tools to control malaria.
topic Plasmodium chabaudi
malaria
Babesia microti
babesiosis
infection
macrophages
url https://www.frontiersin.org/article/10.3389/fcimb.2020.00193/full
work_keys_str_mv AT artemisefstratiou babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT eloizamaysgalon babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT guanbowang babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT kousukeumeda babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT daisukekondoh babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT mohamadalaaterkawi babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT mohamadalaaterkawi babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT aikokume babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT mingmingliu babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT aaronedmondringo babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT huanpingguo babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT yanggao babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT seunghunlee babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT jixuli babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT paulfranckadjoumoumouni babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT yoshifuminishikawa babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT hiroshisuzuki babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT ikuoigarashi babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
AT xuenanxuan babesiamicroticonfersmacrophagebasedcrossprotectiveimmunityagainstmurinemalaria
_version_ 1724654424117215232
spelling doaj-6a03d58795f0417e9ef8c613e3f9bd342020-11-25T03:11:23ZengFrontiers Media S.A.Frontiers in Cellular and Infection Microbiology2235-29882020-04-011010.3389/fcimb.2020.00193515440Babesia microti Confers Macrophage-Based Cross-Protective Immunity Against Murine MalariaArtemis Efstratiou0Eloiza May S. Galon1Guanbo Wang2Kousuke Umeda3Daisuke Kondoh4Mohamad Alaa Terkawi5Mohamad Alaa Terkawi6Aiko Kume7Mingming Liu8Aaron Edmond Ringo9Huanping Guo10Yang Gao11Seung-Hun Lee12Jixu Li13Paul Franck Adjou Moumouni14Yoshifumi Nishikawa15Hiroshi Suzuki16Ikuo Igarashi17Xuenan Xuan18National Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanDepartment of Basic Veterinary Medicine, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanDepartment of Orthopedic Surgery, Hokkaido University, Sapporo, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanNational Research Center for Protozoan Diseases, Obihiro University of Agriculture and Veterinary Medicine, Obihiro, JapanMalaria and babesiosis, the two primary intraerythrocytic protozoan diseases of humans, have been reported in multiple cases of co-infection in endemic regions. As the geographic range and incidence of arthropod-borne infectious diseases is being affected by climate change, co-infection cases with Plasmodium and Babesia are likely to increase. The two parasites have been used in experimental settings, where prior infection with Babesia microti has been shown to protect against fatal malarial infections in mice and primates. However, the immunological mechanisms behind such phenomena of cross-protection remain unknown. Here, we investigated the effect of a primary B. microti infection on the outcome of a lethal P. chabaudi challenge infection using a murine model. Simultaneous infection with both pathogens led to high mortality rates in immunocompetent BALB/c mice, similar to control mice infected with P. chabaudi alone. On the other hand, mice with various stages of B. microti primary infection were thoroughly immune to a subsequent P. chabaudi challenge. Protected mice exhibited decreased levels of serum antibodies and pro-inflammatory cytokines during early stages of challenge infection. Mice repeatedly immunized with dead B. microti quickly succumbed to P. chabaudi infection, despite induction of high antibody responses. Notably, cross-protection was observed in mice lacking functional B and T lymphocytes. When the role of other innate immune effector cells was examined, NK cell-depleted mice with chronic B. microti infection were also found to be protected against P. chabaudi. Conversely, in vivo macrophage depletion rendered the mice vulnerable to P. chabaudi. The above results show that the mechanism of cross-protection conferred by B. microti against P. chabaudi is innate immunity-based, and suggest that it relies predominantly upon the function of macrophages. Further research is needed for elucidating the malaria-suppressing effects of babesiosis, with a vision toward development of novel tools to control malaria.https://www.frontiersin.org/article/10.3389/fcimb.2020.00193/fullPlasmodium chabaudimalariaBabesia microtibabesiosisinfectionmacrophages