Mitsuhiro Iyori
Kanazawa University
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Mitsuhiro Iyori.
Infection and Immunity | 2014
Masanori Mizutani; Mitsuhiro Iyori; Andrew M. Blagborough; Shinya Fukumoto; Tomohiro Funatsu; Robert E. Sinden; Shigeto Yoshida
ABSTRACT A multistage malaria vaccine targeting the pre-erythrocytic and sexual stages of Plasmodium could effectively protect individuals against infection from mosquito bites and provide transmission-blocking (TB) activity against the sexual stages of the parasite, respectively. This strategy could help prevent malaria infections in individuals and, on a larger scale, prevent malaria transmission in communities of endemicity. Here, we describe the development of a multistage Plasmodium vivax vaccine which simultaneously expresses P. vivax circumsporozoite protein (PvCSP) and P25 (Pvs25) protein of this species as a fusion protein, thereby acting as a pre-erythrocytic vaccine and a TB vaccine, respectively. A new-concept vaccine platform based on the baculovirus dual-expression system (BDES) was evaluated. The BDES-Pvs25-PvCSP vaccine displayed correct folding of the Pvs25-PvCSP fusion protein on the viral envelope and was highly expressed upon transduction of mammalian cells in vitro. This vaccine induced high levels of antibodies to Pvs25 and PvCSP and elicited protective (43%) and TB (82%) efficacies against transgenic P. berghei parasites expressing the corresponding P. vivax antigens in mice. Our data indicate that our BDES, which functions as both a subunit and DNA vaccine, can offer a promising multistage vaccine capable of delivering a potent antimalarial pre-erythrocytic and TB response via a single immunization regimen.
Vaccine | 2015
Katarzyna A. Sala; Hidesato Nishiura; Leanna M. Upton; Sara E. Zakutansky; Michael J. Delves; Mitsuhiro Iyori; Masanori Mizutani; Robert E. Sinden; Shigeto Yoshida; Andrew M. Blagborough
Anti-malarial transmission-blocking vaccines (TBVs) aim to inhibit the transmission of Plasmodium from humans to mosquitoes by targeting the sexual/ookinete stages of the parasite. Successful use of such interventions will subsequently result in reduced cases of malarial infection within a human population, leading to local elimination. There are currently only five lead TBV candidates under examination. There is a consequent need to identify novel antigens to allow the formulation of new potent TBVs. Here we describe the design and evaluation of a potential TBV (BDES-PbPSOP12) targeting Plasmodium berghei PSOP12 based on the baculovirus dual expression system (BDES), enabling expression of antigens on the surface of viral particles and within infected mammalian cells. In silico studies have previously suggested that PSOP12 (Putative Secreted Ookinete Protein 12) is expressed within the sexual stages of the parasite (gametocytes, gametes and ookinetes), and is a member of the previously characterized 6-Cys family of plasmodial proteins. We demonstrate that PSOP12 is expressed within the sexual/ookinete forms of the parasite, and that sera obtained from mice immunized with BDES-PbPSOP12 can recognize the surface of the male and female gametes, and the ookinete stages of the parasite. Immunization of mice with BDES-PbPSOP12 confers modest but significant transmission-blocking activity in vivo by active immunization (53.1% reduction in oocyst intensity, 10.9% reduction in oocyst prevalence). Further assessment of transmission-blocking potency ex vivo shows a dose-dependent response, with up to a 76.4% reduction in intensity and a 47.2% reduction in prevalence observed. Our data indicates that PSOP12 in Plasmodium spp. could be a potential new TBV target candidate, and that further experimentation to examine the protein within human malaria parasites would be logical.
PLOS ONE | 2013
Mitsuhiro Iyori; Hiroki Nakaya; Katsuya Inagaki; Sathit Pichyangkul; Daisuke S. Yamamoto; Masanori Kawasaki; Kyungtak Kwak; Yoshihiro Goto; Hiroyuki Matsuoka; Makoto Matsumoto; Shigeto Yoshida
We have previously developed a new malaria vaccine delivery system based on the baculovirus dual expression system (BDES). In this system, expression of malaria antigens is driven by a dual promoter consisting of the baculovirus-derived polyhedrin and mammal-derived cytomegalovirus promoters. To test this system for its potential as a vaccine against human malaria parasites, we investigated immune responses against the newly developed BDES-based Plasmodium falciparum circumsporozoite protein vaccines (BDES-PfCSP) in mice and Rhesus monkeys. Immunization of mice with BDES-PfCSP induced Th1/Th2-mixed type immune responses with high PfCSP-specific antibody (Ab) titers, and provided significant protection against challenge from the bites of mosquitoes infected with a transgenic P. berghei line expressing PfCSP. Next, we evaluated the immunogenicity of the BDES-PfCSP vaccine in a rhesus monkey model. Immunization of BDES-PfCSP elicited high levels of anti-PfCSP Ab responses in individual monkeys. Moreover, the sera from the immunized monkeys remarkably blocked sporozoite invasion of HepG2 cells. Taken together with two animal models, our results indicate that this novel vaccine platform (BDES) has potential clinical application as a vaccine against malaria.
Journal of Biological Chemistry | 2014
Kanako Sugiyama; Mitsuhiro Iyori; Asuka Sawaguchi; Satoko Akashi; Jeremy R. H. Tame; Sam-Yong Park; Shigeto Yoshida
Background: Naturally occurring anticoagulant proteins provide models for new medications with highly desirable properties. Results: The crystal structure of the active region of mosquito protein AAPP has been solved. Conclusion: The mosquito protein AAPP uses a small turn region to block coagulation extremely effectively by binding collagen. Significance: New small molecule anti-coagulants may be developed with completely new mechanisms and none of the drawbacks of current treatments. Blood clotting is a vitally important process that must be carefully regulated to prevent blood loss on one hand and thrombosis on the other. Severe injury and hemophilia may be treated with pro-coagulants, whereas risk of obstructive clotting or embolism may be reduced with anti-coagulants. Anti-coagulants are an extremely important class of drug, one of the most widely used types of medication, but there remains a pressing need for novel treatments, however, as present drugs such as warfarin have significant drawbacks. Nature provides a number of examples of anti-coagulant proteins produced by blood-sucking animals, which may provide templates for the development of new small molecules with similar physiological effects. We have, therefore, studied an Anopheles anti-platelet protein from a malaria vector mosquito and report its crystal structure in complex with an antibody. Overall the protein is extremely sensitive to proteolysis, but the crystal structure reveals a stable domain built from two helices and a turn, which corresponds to the functional region. The antibody raised against Anopheles anti-platelet protein prevents it from binding collagen. Our work, therefore, opens new avenues to the development of both novel small molecule anti-clotting agents and anti-malarials.
Malaria Journal | 2016
Masanori Mizutani; Shinya Fukumoto; Adam Patrice Soubeiga; Akira Soga; Mitsuhiro Iyori; Shigeto Yoshida
BackgroundThe approach of using transgenic rodent malaria parasites to assess the immune system’s response to antigenic targets from a human malaria parasite has been shown to be useful for preclinical evaluation of new vaccine formulations. The transgenic Plasmodium berghei parasite line [PvCSP(VK210)/Pb] generated previously expresses the full-length circumsporozoite protein (CSP) VK210 from Plasmodium vivax. The transgenic parasite expresses one of the two most common alleles of CSP, defined by nine amino acids at the central repeat region of this protein. In the present study, a transgenic P. berghei parasite line [PvCSP(VK247)/Pb] expressing the full-length PvCSP(VK247), which is the alternative common allele, was generated and characterized.MethodsThe P. berghei expressing full-length PvCSP(VK247) was generated and examined its applicability to CSP-based vaccine research by examining its biological characteristics in mosquitoes and mice.ResultsSimilar to PvCSP(VK210)/Pb, PvCSP(VK247)/Pb developed normally in mosquitoes and produced infectious sporozoites equipped to generate patent infections in mice. Invasion of HepG2 cells by PvCSP(VK247)/Pb sporozoites was inhibited by an anti-PvCSP(VK247) repeat monoclonal antibody (mAb), but not by an anti-PvCSP(VK210) repeat mAb.ConclusionsThese two transgenic parasites thus far can be used to evaluate the potential efficacy of PvCSP-based vaccine candidates encompassing the two major genetic variants in preclinical trials.
Platelets | 2013
Hideki Hayashi; Hiroyuki Kyushiki; Keisuke Nagano; Toshiki Sudo; Mitsuhiro Iyori; Hiroyuki Matsuoka; Shigeto Yoshida
We previously identified an anti-platelet protein, anopheline anti-platelet protein (AAPP), from the salivary gland of female Anopheles stephensi (a mosquito vector of human malaria). AAPP specifically blocks platelet adhesion to collagen by binding directly to collagen and subsequently causing platelet aggregation. The aim of this study was to identify the active region of AAPP responsible for the anti-thrombotic activity because we hypothesized that AAPP could be used as a candidate anti-platelet drug. Various truncated forms of AAPP were produced using an Escherichia coli expression system. Each protein was examined for binding activities to soluble/fibrillar collagen and anti-thrombotic activity using a plate assay and platelet/whole blood aggregation study, respectively. Among the truncated forms examined, only a protein encoded by exon 3–4 (rAAPPex3–4) effectively bound to soluble/fibrillar collagen in a concentration-dependent and saturable manner. The EC50 values of full-length AAPP and rAAPPex3–4 for soluble collagen binding were 35 nM and 36 nM, respectively. In contrast to soluble collagen, there was a difference in binding affinity to fibrillar collagen between full-length AAPP and rAAPPex3–4, with EC50 values of 31 nM and 51 nM, respectively. rAAPPex3–4 also inhibited aggregation of platelets/whole blood, and the IC50 values of full-length AAPP and rAAPPex3–4 for platelet aggregation were 35 nM and 93 nM, respectively. These results indicated that the essential moiety of AAPP for collagen binding and anti-thrombotic activity was in the region encoded by exon 3–4, which is highly conserved among the counterpart regions of other mosquito species.
Malaria Journal | 2017
Mitsuhiro Iyori; Daisuke S. Yamamoto; Miako Sakaguchi; Masanori Mizutani; Sota Ogata; Hidesato Nishiura; Takahiko Tamura; Hiroyuki Matsuoka; Shigeto Yoshida
BackgroundPrevious studies have shown that the baculovirus-vectored vaccine based on the “baculovirus dual expression system (BDES)” is an effective vaccine delivery platform for malaria. However, a point of weakness remaining for use of this vaccine platform in vivo concerns viral inactivation by serum complement. In an effort to achieve complement resistance, the gene encoding the human decay-accelerating factor (hDAF) was incorporated into the BDES malaria vaccine expressing the Plasmodium falciparum circumsporozoite protein (PfCSP).ResultsThe newly-developed BDES vaccine, designated BDES-sPfCSP2-Spider, effectively displayed hDAF and PfCSP on the surface of the viral envelope, resulting in complement resistance both in vitro and in vivo. Importantly, upon intramuscular inoculation into mice, the BDES-sPfCSP2-Spider vaccine had a higher protective efficacy (60%) than that of the control vaccine BDES-sPfCSP2-Spier (30%) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP.ConclusionDAF-shielded BDES-vaccines offer great potential for development as a new malaria vaccine platform against the sporozoite challenge.
Parasite Immunology | 2017
Mitsuhiro Iyori; Andrew M. Blagborough; Katarzyna A. Sala; Hidesato Nishiura; Kentaro Takagi; Shigeto Yoshida
Interleukin‐12 (IL‐12) plays an important role in antigen‐specific adaptive immunity against Plasmodium sporozoites, and this requirement allows for a new approach to developing an effective malaria vaccine. In this study, we examined whether IL‐12 could enhance protective efficacy of a baculovirus‐based malaria vaccine. For this aim, a baculoviral vector expressing murine IL‐12 (mIL‐12) under the control of CMV promoter (BES‐mIL‐12‐Spider) and a baculoviral vector expressing Plasmodium falciparum circumsporozoite protein (PfCSP) with post‐transcriptional regulatory element of woodchuck hepatitis virus (BDES‐sPfCSP2‐WPRE‐Spider) were generated. BES‐mIL‐12‐Spider produced bioactive IL‐12 which activates splenocytes, resulting in induction of IFN‐γ. When co‐immunized with BES‐mIL‐12‐Spider and BDES‐sPfCSP2‐WPRE‐Spider, the mouse number for high IgG2a/IgG1 ratios and the geometric mean in this group were both increased as compared with those of the other groups, indicating a shift towards a Th1‐type response following immunization with BES‐mIL‐12‐Spider. Finally, immunization with BDES‐sPfCSP2‐WPRE‐Spider plus BES‐mIL‐12‐Spider had a higher protective efficacy (73%) than immunization with BDES‐sPfCSP2‐WPRE‐Spider alone (30%) against challenge with transgenic Plasmodium berghei sporozoites expressing PfCSP. These results suggest that co‐administration of IL‐12 expressing baculoviral vector, instead of IL‐12 cDNA, with viral‐vectored vaccines provides a new feasible vaccine platform to enhance Th1‐type cellular immune responses against Plasmodium parasites.
Scientific Reports | 2018
Kunitaka Yoshida; Mitsuhiro Iyori; Andrew M. Blagborough; Ahmed M. Salman; Pawan Dulal; Katarzyna A. Sala; Daisuke S. Yamamoto; Shahid M. Khan; Chris J. Janse; Sumi Biswas; Tatsuya Yoshii; Yenni Yusuf; Masaharu Tokoro; Adrian V. S. Hill; Shigeto Yoshida
With the increasing prevalence of artemisinin-resistant malaria parasites, a highly efficacious and durable vaccine for malaria is urgently required. We have developed an experimental virus-vectored vaccine platform based on an envelope-modified baculovirus dual-expression system (emBDES). Here, we show a conceptually new vaccine platform based on an adenovirus-prime/emBDES-boost heterologous immunization regimen expressing the Plasmodium falciparum circumsporozoite protein (PfCSP). A human adenovirus 5-prime/emBDES-boost heterologous immunization regimen consistently achieved higher sterile protection against transgenic P. berghei sporozoites expressing PfCSP after a mosquito-bite challenge than reverse-ordered or homologous immunization. This high protective efficacy was also achieved with a chimpanzee adenovirus 63-prime/emBDES-boost heterologous immunization regimen against an intravenous sporozoite challenge. Thus, we show that the adenovirus-prime/emBDES-boost heterologous immunization regimen confers sterile protection against sporozoite challenge by two individual routes, providing a promising new malaria vaccine platform for future clinical use.
bioRxiv | 2018
Talha Bin Emran; Mitsuhiro Iyori; Yuki Ono; Fitri Amelia; Yenni Yusuf; Ashekul Islam; Asrar Alam; Ryohei Ogawa; Hiroyuki Matsuoka; Daisuke S. Yamamoto; Shigeto Yoshida
Baculovirus (BV), an enveloped insect virus with a circular double-stranded DNA genome, possesses unique characteristics that induce strong innate immune responses in mammalian cells. Here, we show that BV administration not only sterilely protects BALB/c mice for at least 7 days from subsequent Plasmodium berghei sporozoite infection but also eliminates existing liver-stage parasites completely, effects superior to those of primaquine, and does so in a TLR9-independent manner. Six hours post-BV administration, IFN-α and IFN-γ were robustly produced in serum, and RNA transcripts of interferon-stimulated genes were drastically upregulated in the liver. The in vivo passive transfer of post-BV administration serum effectively eliminated liver-stage parasites, and IFN-α neutralization abolished this effect, indicating that the BV liver-stage parasite killing mechanism is downstream of the type I IFN signaling pathway. Our results demonstrate that BV is a potent IFN-inducing prophylactic and therapeutic agent with great potential for further development as a new malaria vaccine and/or anti-hypnozoite drug.
Collaboration
Dive into the Mitsuhiro Iyori's collaboration.
Obihiro University of Agriculture and Veterinary Medicine
View shared research outputs