Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Hiromi Tanji is active.

Publication


Featured researches published by Hiromi Tanji.


Science | 2013

Structural Reorganization of the Toll-Like Receptor 8 Dimer Induced by Agonistic Ligands

Hiromi Tanji; Umeharu Ohto; Takuma Shibata; Kensuke Miyake; Toshiyuki Shimizu

Dissecting TLR8 Interactions Toll-like receptors (TLRs) activate the innate immune system in response to invading pathogens. TLR7 and TLR8 recognize single-stranded RNA from viruses and also contribute to the pathogenesis of autoimmune diseases. Tanji et al. (p. 1426) now report the crystal structure of the unliganded TLR8 ectodomain and the TLR8 ectodomain bound to three different small-molecule agonists. Ligand binding to preformed TLR8 dimers induced conformational changes that brought the C-terminal domains closer together, presumably initiating downstream signaling. The crystal structure of unbound and ligand-bound Toll-like receptor 8 reveals ligand-induced conformational changes. Toll-like receptor 7 (TLR7) and TLR8 recognize single-stranded RNA and initiate innate immune responses. Several synthetic agonists of TLR7-TLR8 display novel therapeutic potential; however, the molecular basis for ligand recognition and activation of signaling by TLR7 or TLR8 is largely unknown. In this study, the crystal structures of unliganded and ligand-induced activated human TLR8 dimers were elucidated. Ligand recognition was mediated by a dimerization interface formed by two protomers. Upon ligand stimulation, the TLR8 dimer was reorganized such that the two C termini were brought into proximity. The loop between leucine-rich repeat 14 (LRR14) and LRR15 was cleaved; however, the N- and C-terminal halves remained associated and contributed to ligand recognition and dimerization. Thus, ligand binding induces reorganization of the TLR8 dimer, which enables downstream signaling processes.


Nature | 2015

Structural basis of CpG and inhibitory DNA recognition by Toll-like receptor 9

Umeharu Ohto; Takuma Shibata; Hiromi Tanji; Hanako Ishida; Elena Krayukhina; Susumu Uchiyama; Kensuke Miyake; Toshiyuki Shimizu

Innate immunity serves as the first line of defence against invading pathogens such as bacteria and viruses. Toll-like receptors (TLRs) are examples of innate immune receptors, which sense specific molecular patterns from pathogens and activate immune responses. TLR9 recognizes bacterial and viral DNA containing the cytosine–phosphate–guanine (CpG) dideoxynucleotide motif. The molecular basis by which CpG-containing DNA (CpG-DNA) elicits immunostimulatory activity via TLR9 remains to be elucidated. Here we show the crystal structures of three forms of TLR9: unliganded, bound to agonistic CpG-DNA, and bound to inhibitory DNA (iDNA). Agonistic-CpG-DNA-bound TLR9 formed a symmetric TLR9–CpG-DNA complex with 2:2 stoichiometry, whereas iDNA-bound TLR9 was a monomer. CpG-DNA was recognized by both protomers in the dimer, in particular by the amino-terminal fragment (LRRNT–LRR10) from one protomer and the carboxy-terminal fragment (LRR20–LRR22) from the other. The iDNA, which formed a stem-loop structure suitable for binding by intramolecular base pairing, bound to the concave surface from LRR2–LRR10. This structure serves as an important basis for improving our understanding of the functional mechanisms of TLR9.


Nature Structural & Molecular Biology | 2015

Toll-like receptor 8 senses degradation products of single-stranded RNA

Hiromi Tanji; Umeharu Ohto; Takuma Shibata; Masato Taoka; Yoshio Yamauchi; Toshiaki Isobe; Kensuke Miyake; Toshiyuki Shimizu

Toll-like receptor 8 (TLR8) recognizes viral or bacterial single-stranded RNA (ssRNA) and activates innate immune systems. TLR8 is activated by uridine- and guanosine-rich ssRNA as well as by certain synthetic chemicals; however, the molecular basis for ssRNA recognition has remained unknown. In this study, to elucidate the recognition mechanism of ssRNA, we determined the crystal structures of human TLR8 in complex with ssRNA. TLR8 recognized two degradation products of ssRNA—uridine and a short oligonucleotide—at two distinct sites: uridine bound the site on the dimerization interface where small chemical ligands are recognized, whereas short oligonucleotides bound a newly identified site on the concave surface of the TLR8 horseshoe structure. Site-directed mutagenesis revealed that both binding sites were essential for activation of TLR8 by ssRNA. These results demonstrate that TLR8 is a sensor for both uridine and a short oligonucleotide derived from RNA.


Immunity | 2016

Structural Analysis Reveals that Toll-like Receptor 7 Is a Dual Receptor for Guanosine and Single-Stranded RNA

Zhikuan Zhang; Umeharu Ohto; Takuma Shibata; Elena Krayukhina; Masato Taoka; Yoshio Yamauchi; Hiromi Tanji; Toshiaki Isobe; Susumu Uchiyama; Kensuke Miyake; Toshiyuki Shimizu

Toll-like receptor 7 (TLR7) is a single-stranded RNA (ssRNA) sensor in innate immunity and also responds to guanosine and chemical ligands, such as imidazoquinoline compounds. However, TLR7 activation mechanism by these ligands remain largely unknown. Here, we generated crystal structures of three TLR7 complexes, and found that all formed an activated m-shaped dimer with two ligand-binding sites. The first site conserved in TLR7 and TLR8 was used for small ligand-binding essential for its activation. The second site spatially distinct from that of TLR8 was used for a ssRNA-binding that enhanced the affinity of the first-site ligands. The first site preferentially recognized guanosine and the second site specifically bound to uridine moieties in ssRNA. Our structural, biochemical, and mutagenesis studies indicated that TLR7 is a dual receptor for guanosine and uridine-containing ssRNA. Our findings have important implications for understanding of TLR7 function, as well as for therapeutic manipulation of TLR7 activation.


ChemMedChem | 2014

Structure‐Based Design of Novel Human Toll‐like Receptor 8 Agonists

Hari Prasad Kokatla; Diptesh Sil; Hiromi Tanji; Umeharu Ohto; Subbalakshmi S. Malladi; Lauren M. Fox; Toshiyoki Shimizu; Sunil A. David

Toll‐like receptor (TLR)‐8 agonists activate adaptive immune responses by inducing robust production of T helper 1‐polarizing cytokines, suggesting that TLR8‐active compounds might be promising candidate vaccine adjuvants. Recently, a C2‐butyl furo[2,3‐c]quinoline was reported with purely TLR8 agonistic activity. This compound was successfully co‐crystallized with the human TLR8 ectodomain, and the co‐crystal structure revealed ligand‐induced reorganization of the binding pocket of TLR8. The loss of a key hydrogen bond between the oxygen atom of the furanyl ring of the agonist and Thr 574 in TLR8 suggested that the furan ring is dispensable. Employing a disconnection strategy, 3‐ and 4‐substituted aminoquinolines were investigated. Focused structure‐based ligand design studies led to the identification of 3‐pentyl‐quinoline‐2‐amine as a novel, structurally simple, and highly potent human TLR8‐specific agonist (EC50=0.2 μM). Preliminary evaluation of this compound in ex vivo human blood assay systems revealed that it retains prominent cytokine‐inducing activity. Together, these results indicate the suitability of this compound as a novel vaccine adjuvant, warranting further investigation.


Journal of Medicinal Chemistry | 2014

Determinants of Activity at Human Toll-like Receptors 7 and 8: Quantitative Structure-Activity Relationship (QSAR) of Diverse Heterocyclic Scaffolds

Euna Yoo; Deepak B. Salunke; Diptesh Sil; Xiaoqiang Guo; Alex C. D. Salyer; Alec R. Hermanson; Manoj Kumar; Subbalakshmi S. Malladi; Rajalakshmi Balakrishna; Ward H. Thompson; Hiromi Tanji; Umeharu Ohto; Toshiyuki Shimizu; Sunil A. David

Toll-like receptor (TLR) 7 and 8 agonists are potential vaccine adjuvants, since they directly activate APCs and enhance Th1-driven immune responses. Previous SAR investigations in several scaffolds of small molecule TLR7/8 activators pointed to the strict dependence of the selectivity for TLR7 vis-à-vis TLR8 on the electronic configurations of the heterocyclic systems, which we sought to examine quantitatively with the goal of developing “heuristics” to define structural requisites governing activity at TLR7 and/or TLR8. We undertook a scaffold-hopping approach, entailing the syntheses and biological evaluations of 13 different chemotypes. Crystal structures of TLR8 in complex with the two most active compounds confirmed important binding interactions playing a key role in ligand occupancy and biological activity. Density functional theory based quantum chemical calculations on these compounds followed by linear discriminant analyses permitted the classification of inactive, TLR8-active, and TLR7/8 dual-active compounds, confirming the critical role of partial charges in determining biological activity.


Microbes and Infection | 2014

Structure and function of toll-like receptor 8

Umeharu Ohto; Hiromi Tanji; Toshiyuki Shimizu

Toll-like receptors (TLRs) sense pathogen-associated molecular patterns originating from invading microorganisms and initiate innate immune responses. Recent structural studies of TLR-ligand complexes have revealed the detailed molecular mechanisms by which each TLR specifically recognizes its own ligands. This review focuses on the structure of TLR8 and discusses the similarities and diversities of TLR-ligand interactions and signaling mechanisms.


Journal of Medicinal Chemistry | 2015

Structure-Based Design of Human TLR8-Specific Agonists with Augmented Potency and Adjuvanticity.

Mallesh Beesu; Giuseppe Caruso; Alex C. D. Salyer; Karishma K. Khetani; Diptesh Sil; Mihiri Weerasinghe; Hiromi Tanji; Umeharu Ohto; Toshiyuki Shimizu; Sunil A. David

Human Toll-like receptor 8 (hTLR8) is expressed in myeloid dendritic cells, monocytes, and monocyte-derived dendritic cells. Engagement by TLR8 agonists evokes a distinct cytokine profile which favors the development of type 1 helper T cells. Crystal structures of the ectodomain of hTLR8 cocrystallized with two regioisomers of a dual TLR7/8-agonistic N1-substituted imidazoquinolines showed subtle differences in their interactions in the binding site of hTLR8. We hypothesized that the potency of a previously reported best-in-class pure TLR8 agonist, 3-pentylquinoline-2-amine, could be further enhanced by “designing in” functional groups that would mimic key intermolecular interactions that we had observed in the crystal structures. We performed a focused exploration of decorating the quinoline core with alkylamino groups at all possible positions. These studies have led to the identification of a novel TLR8 agonist that was ∼20-fold more potent than the parent compound and displays prominent adjuvantic activity in a rabbit model of immunization.


PLOS ONE | 2015

The Imidazoquinoline Toll-Like Receptor-7/8 Agonist Hybrid-2 Potently Induces Cytokine Production by Human Newborn and Adult Leukocytes

Lakshmi Ganapathi; Simon D. van Haren; David J. Dowling; Ilana Bergelson; Nikunj M. Shukla; Subbalakshmi S. Malladi; Rajalakshmi Balakrishna; Hiromi Tanji; Umeharu Ohto; Toshiyuki Shimizu; Sunil A. David; Ofer Levy

Background Newborns and young infants are at higher risk for infections than adults, and manifest suboptimal vaccine responses, motivating a search for novel immunomodulators and/or vaccine adjuvants effective in early life. In contrast to most TLR agonists (TLRA), TLR8 agonists such as imidazoquinolines (IMQs) induce adult-level Th1-polarizing cytokine production from human neonatal cord blood monocytes and are candidate early life adjuvants. We assessed whether TLR8-activating IMQ congeners may differ in potency and efficacy in inducing neonatal cytokine production in vitro, comparing the novel TLR7/8-activating IMQ analogues Hybrid-2, Meta-amine, and Para-amine to the benchmark IMQ resiquimod (R848). Methods TLRA-induced NF-κB activation was measured in TLR-transfected HEK cells. Cytokine production in human newborn cord and adult peripheral blood and in monocyte-derived dendritic cell cultures were measured by ELISA and multiplex assays. X-ray crystallography characterized the interaction of human TLR8 with Hybrid-2. Results Hybrid-2 selectively activated both TLR7 and 8 and was more potent than R848 in inducing adult-like levels of TNF-α, and IL-1β. Consistent with its relatively high in vitro activity, crystallographic studies suggest that absence in Hybrid-2 of an ether oxygen of the C2-ethoxymethyl substituent, which can engage in unfavorable electrostatic and/or dipolar interactions with the carbonyl oxygen of Gly572 in human TLR8, may confer greater efficacy and potency compared to R848. Conclusions Hybrid-2 is a selective and potent TLR7/8 agonist that is a candidate adjuvant for early life immunization.


Proceedings of the National Academy of Sciences of the United States of America | 2016

Autoinhibition and relief mechanism by the proteolytic processing of Toll-like receptor 8

Hiromi Tanji; Umeharu Ohto; Yuji Motoi; Takuma Shibata; Kensuke Miyake; Toshiyuki Shimizu

Significance Toll-like receptors (TLRs) activate the innate immune system in response to invading pathogens. The single-stranded RNA (ssRNA) sensor TLR8 requires proteolytic cleavage at the loop region (Z-loop) for its activation. Biochemical and biophysical studies have revealed that the uncleaved Z-loop prevents formation of the TLR8 dimer, which is essential for its activation. Crystallographic analysis demonstrated that the uncleaved Z-loop located on the ascending lateral face prevents the approach of the dimerization partner by steric hindrance. Like TLR8, TLR7 and TLR9 also contain the Z-loop, and thus these proteins also might have this autoinhibition mechanism. Toll-like receptor 8 (TLR8) senses single-stranded RNA (ssRNA) and initiates innate immune responses. TLR8 requires proteolytic cleavage at the loop region (Z-loop) between leucine-rich repeat (LRR) 14 and LRR15 for its activation. However, the molecular basis of Z-loop processing remains unknown. To elucidate the mechanism of Z-loop processing, we performed biochemical and structural studies of how the Z-loop affects the function of TLR8. TLR8 with the uncleaved Z-loop is unable to form a dimer, which is essential for activation, irrespective of the presence of agonistic ligands. Crystallographic analysis revealed that the uncleaved Z-loop located on the ascending lateral face prevents the approach of the dimerization partner by steric hindrance. This autoinhibition mechanism of dimerization by the Z-loop might be occurring in the proteins of the same subfamily, TLR7 and TLR9.

Collaboration


Dive into the Hiromi Tanji's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge