Network


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

Hotspot


Dive into the research topics where Kenichi Nagano is active.

Publication


Featured researches published by Kenichi Nagano.


Proceedings of the National Academy of Sciences of the United States of America | 2014

Engineered nanomedicine for myeloma and bone microenvironment targeting.

Archana Swami; Michaela R. Reagan; Pamela Basto; Yuji Mishima; Nazila Kamaly; Siobhan Glavey; Sufeng Zhang; Michele Moschetta; Dushanth Seevaratnam; Yong Zhang; Jinhe Liu; Masoumeh Memarzadeh; Jun Wu; Salomon Manier; Jinjun Shi; Nicolas Bertrand; Zhi Ning Lu; Kenichi Nagano; Roland Baron; Antonio Sacco; Aldo M. Roccaro; Omid C. Farokhzad; Irene M. Ghobrial

Significance Limited treatment options exist for cancer within the bone, as demonstrated by the inevitable, pernicious course of metastatic breast, prostate, and blood cancers. The difficulty of eliminating bone-residing cancer necessitates novel, alternative treatments to manipulate the tumor cells and their microenvironment, with minimal off-target effects. To this end, we engineered bone-homing, stealth nanoparticles to deliver anticancer, bone-stimulatory drugs, and demonstrated their utility with bortezomib (a model drug) and multiple myeloma (a model cancer). To test our hypothesis that increasing bone volume and strength inhibits tumor growth, mice were treated with these nanoparticles before being injected with cancer cells. Results demonstrated significantly slower myeloma growth and prolonged survival. Our research demonstrates the potential of bone-homing nanomedicine as an efficacious cancer treatment mechanism. Bone is a favorable microenvironment for tumor growth and a frequent destination for metastatic cancer cells. Targeting cancers within the bone marrow remains a crucial oncologic challenge due to issues of drug availability and microenvironment-induced resistance. Herein, we engineered bone-homing polymeric nanoparticles (NPs) for spatiotemporally controlled delivery of therapeutics to bone, which diminish off-target effects and increase local drug concentrations. The NPs consist of poly(d,l-lactic-co-glycolic acid) (PLGA), polyethylene glycol (PEG), and bisphosphonate (or alendronate, a targeting ligand). The engineered NPs were formulated by blending varying ratios of the synthesized polymers: PLGA-b-PEG and alendronate-conjugated polymer PLGA-b-PEG-Ald, which ensured long circulation and targeting capabilities, respectively. The bone-binding ability of Ald-PEG-PLGA NPs was investigated by hydroxyapatite binding assays and ex vivo imaging of adherence to bone fragments. In vivo biodistribution of fluorescently labeled NPs showed higher retention, accumulation, and bone homing of targeted Ald-PEG-PLGA NPs, compared with nontargeted PEG-PLGA NPs. A library of bortezomib-loaded NPs (bone-targeted Ald-Bort-NPs and nontargeted Bort-NPs) were developed and screened for optimal physiochemical properties, drug loading, and release profiles. Ald-Bort-NPs were tested for efficacy in mouse models of multiple myeloma (MM). Results demonstrated significantly enhanced survival and decreased tumor burden in mice pretreated with Ald-Bort-NPs versus Ald-Empty-NPs (no drug) or the free drug. We also observed that bortezomib, as a pretreatment regimen, modified the bone microenvironment and enhanced bone strength and volume. Our findings suggest that NP-based anticancer therapies with bone-targeting specificity comprise a clinically relevant method of drug delivery that can inhibit tumor progression in MM.


Nature Medicine | 2014

Osteoblast-derived WNT16 represses osteoclastogenesis and prevents cortical bone fragility fractures.

Sofia Movérare-Skrtic; Petra Henning; Xianwen Liu; Kenichi Nagano; Hiroaki Saito; Anna E. Börjesson; Klara Sjögren; Sara H. Windahl; Helen H. Farman; Bert Kindlund; Cecilia Engdahl; Antti Koskela; Fu-Ping Zhang; Emma Eriksson; Farasat Zaman; Ann Hammarstedt; Hanna Isaksson; Marta Bally; Ali Kassem; Catharina Lindholm; Olof Sandberg; Per Aspenberg; Lars Sävendahl; Jian Q. Feng; Jan Tuckermann; Juha Tuukkanen; Matti Poutanen; Roland Baron; Ulf H. Lerner; Francesca Gori

The WNT16 locus is a major determinant of cortical bone thickness and nonvertebral fracture risk in humans. The disability, mortality and costs caused by osteoporosis-induced nonvertebral fractures are enormous. We demonstrate here that Wnt16-deficient mice develop spontaneous fractures as a result of low cortical thickness and high cortical porosity. In contrast, trabecular bone volume is not altered in these mice. Mechanistic studies revealed that WNT16 is osteoblast derived and inhibits human and mouse osteoclastogenesis both directly by acting on osteoclast progenitors and indirectly by increasing expression of osteoprotegerin (Opg) in osteoblasts. The signaling pathway activated by WNT16 in osteoclast progenitors is noncanonical, whereas the pathway activated in osteoblasts is both canonical and noncanonical. Conditional Wnt16 inactivation revealed that osteoblast-lineage cells are the principal source of WNT16, and its targeted deletion in osteoblasts increases fracture susceptibility. Thus, osteoblast-derived WNT16 is a previously unreported key regulator of osteoclastogenesis and fracture susceptibility. These findings open new avenues for the specific prevention or treatment of nonvertebral fractures, a substantial unmet medical need.


Journal of Bone and Mineral Research | 2015

HDAC5 Controls MEF2C‐Driven Sclerostin Expression in Osteocytes

Marc N. Wein; Jordan Spatz; Shigeki Nishimori; John G. Doench; David E. Root; Philip Babij; Kenichi Nagano; Roland Baron; Daniel J. Brooks; Mary L. Bouxsein; Paola Divieti Pajevic; Henry M. Kronenberg

Osteocytes secrete paracrine factors that regulate the balance between bone formation and destruction. Among these molecules, sclerostin (encoded by the gene SOST) inhibits osteoblastic bone formation and is an osteoporosis drug target. The molecular mechanisms underlying SOST expression remain largely unexplored. Here, we report that histone deacetylase 5 (HDAC5) negatively regulates sclerostin levels in osteocytes in vitro and in vivo. HDAC5 shRNA increases, whereas HDAC5 overexpression decreases SOST expression in the novel murine Ocy454 osteocytic cell line. HDAC5 knockout mice show increased levels of SOST mRNA, more sclerostin‐positive osteocytes, decreased Wnt activity, low trabecular bone density, and reduced bone formation by osteoblasts. In osteocytes, HDAC5 binds and inhibits the function of MEF2C, a crucial transcription factor for SOST expression. Using chromatin immunoprecipitation, we have mapped endogenous MEF2C binding in the SOST gene to a distal intergenic enhancer 45 kB downstream from the transcription start site. HDAC5 deficiency increases SOST enhancer MEF2C chromatin association and H3K27 acetylation and decreases recruitment of corepressors NCoR and HDAC3. HDAC5 associates with and regulates the transcriptional activity of this enhancer, suggesting direct regulation of SOST gene expression by HDAC5 in osteocytes. Finally, increased sclerostin production achieved by HDAC5 shRNA is abrogated by simultaneous knockdown of MEF2C, indicating that MEF2C is a major target of HDAC5 in osteocytes.


Molecular and Cellular Biology | 2014

Direct Transcriptional Repression of Zfp423 by Zfp521 Mediates a Bone Morphogenic Protein-Dependent Osteoblast versus Adipocyte Lineage Commitment Switch

William N. Addison; Martin Ming-Jen Fu; Helen X. Yang; Zhao Lin; Kenichi Nagano; Francesca Gori; Roland Baron

ABSTRACT Osteoblasts and adipocytes arise from a common mesenchymal precursor cell. The cell fate decision of a mesenchymal precursor cell is under the influence of molecular cues and signaling pathways that lead to the activation or repression of lineage-specific transcription factors. The molecular mechanisms determining osteoblast versus adipocyte lineage specificity in response to bone morphogenic protein (BMP) remain unclear. In this study, we describe the mechanism through which Zfp521 (ZNF521), a regulator of lineage progression in multiple immature cell populations, regulates lineage specification of mesenchymal progenitor cells during BMP-induced differentiation events. In vivo deletion or in vitro knockdown of Zfp521 in mesenchymal precursors resulted in increased expression of the adipocyte determinant factor Zfp423 (ZNF423). This was concurrent with the loss of histone H3K9 methylation and an increase in histone H3K9 acetylation at the Zfp423 promoter, which together are indicative of decreased gene repression. Indeed, we found that Zfp521 occupies and represses the promoter and intronic enhancer regions of Zfp423. Accordingly, conditional deletion of Zfp521 inhibited heterotopic bone formation in response to local injection of BMP2. In contrast, marrow adiposity within BMP2-induced bone was markedly enhanced in Zfp521-deficient mice, suggesting that precursor cells lacking Zfp521 differentiate preferentially into adipocytes instead of osteoblasts in response to BMP2. Consistent with a cell-autonomous role of Zfp521 in mesenchymal precursors, knockdown of Zfp521 in stromal cells prevented BMP2-induced osteoblast marker expression and simultaneously enhanced lipid accumulation and expression of adipocyte-related genes. Taken together, the data suggest that Zfp521 is a cell fate switch critical for BMP-induced osteoblast commitment and identify Zfp521 as the intrinsic repressor of Zfp423 and hence of adipocyte commitment during BMP-induced mesenchymal precursor differentiation.


Nature Communications | 2016

SIKs control osteocyte responses to parathyroid hormone

Marc N. Wein; Yanke Liang; Olga Göransson; Thomas B. Sundberg; Jinhua Wang; Elizabeth A. Williams; Maureen J. O'Meara; Nicolas Govea; Belinda Beqo; Shigeki Nishimori; Kenichi Nagano; Daniel J. Brooks; Janaina S. Martins; Braden Corbin; Anthony Anselmo; Ruslan I. Sadreyev; Joy Y. Wu; Kei Sakamoto; Marc Foretz; Ramnik J. Xavier; Roland Baron; Mary L. Bouxsein; Thomas J. Gardella; Paola Divieti-Pajevic; Nathanael S. Gray; Henry M. Kronenberg

Parathyroid hormone (PTH) activates receptors on osteocytes to orchestrate bone formation and resorption. Here we show that PTH inhibition of SOST (sclerostin), a WNT antagonist, requires HDAC4 and HDAC5, whereas PTH stimulation of RANKL, a stimulator of bone resorption, requires CRTC2. Salt inducible kinases (SIKs) control subcellular localization of HDAC4/5 and CRTC2. PTH regulates both HDAC4/5 and CRTC2 localization via phosphorylation and inhibition of SIK2. Like PTH, new small molecule SIK inhibitors cause decreased phosphorylation and increased nuclear translocation of HDAC4/5 and CRTC2. SIK inhibition mimics many of the effects of PTH in osteocytes as assessed by RNA-seq in cultured osteocytes and following in vivo administration. Once daily treatment with the small molecule SIK inhibitor YKL-05-099 increases bone formation and bone mass. Therefore, a major arm of PTH signalling in osteocytes involves SIK inhibition, and small molecule SIK inhibitors may be applied therapeutically to mimic skeletal effects of PTH.


Kidney International | 2017

Klotho expression in osteocytes regulates bone metabolism and controls bone formation

Hirotaka Komaba; Jovana Kaludjerovic; Dorothy Hu; Kenichi Nagano; Katsuhiko Amano; Noriko Ide; Tadatoshi Sato; Michael Densmore; Jun-ichi Hanai; Hannes Olauson; Teresita Bellido; Tobias E. Larsson; Roland Baron; Beate Lanske

Osteocytes within the mineralized bone matrix control bone remodeling by regulating osteoblast and osteoclast activity. Osteocytes express the aging suppressor Klotho, but the functional role of this protein in skeletal homeostasis is unknown. Here we identify Klotho expression in osteocytes as a potent regulator of bone formation and bone mass. Targeted deletion of Klotho from osteocytes led to a striking increase in bone formation and bone volume coupled with enhanced osteoblast activity, in sharp contrast to what is observed in Klotho hypomorphic (kl/kl) mice. Conversely, overexpression of Klotho in cultured osteoblastic cells inhibited mineralization and osteogenic activity during osteocyte differentiation. Further, the induction of chronic kidney disease with high-turnover renal osteodystrophy led to downregulation of Klotho in bone cells. This appeared to offset the skeletal impact of osteocyte-targeted Klotho deletion. Thus, our findings establish a key role of osteocyte-expressed Klotho in regulating bone metabolism and indicate a new mechanism by which osteocytes control bone formation.


Molecular Endocrinology | 2014

Inhibition of BMP2-Induced Bone Formation by the p65 Subunit of NF-κB via an Interaction With Smad4

Shizu Hirata-Tsuchiya; Hidefumi Fukushima; Takenobu Katagiri; Satoshi Ohte; Masashi Shin; Kenichi Nagano; Kazuhiro Aoki; Takahiko Morotomi; Goro Sugiyama; Chihiro Nakatomi; Shoichiro Kokabu; Takahiro Doi; Hiroshi Takeuchi; Keiichi Ohya; Masamichi Terashita; Masato Hirata; Chiaki Kitamura; Eijiro Jimi

Bone morphogenic proteins (BMPs) stimulate bone formation in vivo and osteoblast differentiation in vitro via a Smad signaling pathway. Recent findings revealed that the activation of nuclear factor-κB (NF-κB) inhibits BMP-induced osteoblast differentiation. Here, we show that NF-κB inhibits BMP signaling by directly targeting the Smad pathway. A selective inhibitor of the classic NF-κB pathway, BAY11-770682, enhanced BMP2-induced ectopic bone formation in vivo. In mouse embryonic fibroblasts (MEFs) prepared from mice deficient in p65, the main subunit of NF-κB, BMP2, induced osteoblastic differentiation via the Smad complex to a greater extent than that in wild-type MEFs. In p65(-/-) MEFs, the BMP2-activated Smad complex bound much more stably to the target element than that in wild-type MEFs without affecting the phosphorylation levels of Smad1/5/8. Overexpression of p65 inhibited BMP2 activity by decreasing the DNA binding of the Smad complex. The C-terminal region, including the TA2 domain, of p65 was essential for inhibiting the BMP-Smad pathway. The C-terminal TA2 domain of p65 associated with the MH1 domain of Smad4 but not Smad1. Taken together, our results suggest that p65 inhibits BMP signaling by blocking the DNA binding of the Smad complex via an interaction with Smad4. Our study also suggests that targeting the association between p65 and Smad4 may help to promote bone regeneration in the treatment of bone diseases.


Journal of Bone and Mineral Research | 2013

Disruption of NF‐κB1 prevents bone loss caused by mechanical unloading

Hitomi Nakamura; Kazuhiro Aoki; Wataru Masuda; Neil Alles; Kenichi Nagano; Hidefumi Fukushima; Kenji Osawa; Hisataka Yasuda; Ichiro Nakamura; Yuko Mikuni-Takagaki; Keiichi Ohya; Kenshi Maki; Eijiro Jimi

Mechanical unloading, such as in a microgravity environment in space or during bed rest (for patients who require prolonged bed rest), leads to a decrease in bone mass because of the suppression of bone formation and the stimulation of bone resorption. To address the challenges presented by a prolonged stay in space and the forthcoming era of a super‐aged society, it will be important to prevent the bone loss caused by prolonged mechanical unloading. Nuclear factor κB (NF‐κB) transcription factors are activated by mechanical loading and inflammatory cytokines. Our objective was to elucidate the role of NF‐κB pathways in bone loss that are caused by mechanical unloading. Eight‐week‐old wild‐type (WT) and NF‐κB1‐deficient mice were randomly assigned to a control or mechanically unloaded with tail suspension group. After 2 weeks, a radiographic analysis indicated a decrease in bone mass in the tibias and femurs of the unloaded WT mice but not in the NF‐κB1–deficient mice. An NF‐κB1 deficiency suppressed the unloading‐induced reduction in bone formation by maintaining the proportion and/or potential of osteoprogenitors or immature osteoblasts, and by suppression of bone resorption through the inhibition of intracellular signaling through the receptor activator of NF‐κB ligand (RANKL) in osteoclast precursors. Thus, NF‐κB1 is involved in two aspects of rapid reduction in bone mass that are induced by disuse osteoporosis in space or bed rest.


Journal of Bone and Mineral Metabolism | 2011

The tumor necrosis factor type 2 receptor plays a protective role in tumor necrosis factor-α-induced bone resorption lacunae on mouse calvariae.

Kenichi Nagano; Neil Alles; Anower Hussain Mian; Asako Shimoda; Nobuyuki Morimoto; Yukihiko Tamura; Hitoyata Shimokawa; Kazunari Akiyoshi; Keiichi Ohya; Kazuhiro Aoki

Tumor necrosis factor (TNF)-α exerts its biological function via TNF type 1 and type 2 receptors (TNFR1 and TNFR2). We have previously reported that bone resorption induced by lipopolysaccharide (LPS) in TNFR2-deficient mice is accelerated compared to that in wild-type (WT) mice. Although these results suggested that TNFR2 might have a protective role in bone resorption, we could not exclude the possibility that TNFR2 has no role in bone resorption. To clarify the role of TNFR2, we developed a TNF-α-induced bone resorption model using cholesterol-bearing pullulan nanogel as a TNF-α carrier to minimize the influence of inflammatory cytokines other than TNF-α. Injections of human TNF-α (hTNF), an agonist of mouse TNFR1, stimulated bone resorption lacunae on the calvariae in WT mice, but mouse TNF-α (mTNF), an agonist of both mouse TNFR1 and TNFR2, could not. To eliminate the possibility that the TNFR1 agonistic effects of hTNF were stronger than those of mTNF, we used the same model in TNFR2-deficient mice. Injection of mTNF resulted in clear bone resorption lacunae to the same extent observed after using hTNF in the TNFR2-deficient mice. Histomorphometric analysis of osteoclast number supported the observed changes in bone resorption lacunae. These data suggest that TNFR2 has a protective role in TNF-α-induced bone resorption.


PLOS ONE | 2015

The Crosstalk between Osteoclasts and Osteoblasts Is Dependent upon the Composition and Structure of Biphasic Calcium Phosphates.

Yukari Shiwaku; Lynn Neff; Kenichi Nagano; Ken-Ichi Takeyama; Joost de Bruijn; Michel Dard; Francesca Gori; Roland Baron

Biphasic calcium phosphates (BCPs), consisting of hydroxyapatite (HA) and β-tricalcium phosphate (β-TCP), exhibit good biocompatibility and osteoconductivity, maintaining a balance between resorption of the biomaterial and formation of new bone. We tested whether the chemical composition and/or the microstructure of BCPs affect osteoclasts (OCs) differentiation and/or their ability to crosstalk with osteoblasts (OBs). To this aim, OCs were cultured on BCPs with HA content of 5, 20 or 60% and their differentiation and activity were assessed. We found that OC differentiation is partially impaired by increased HA content, but not by the presence of micropores within BCP scaffolds, as indicated by TRAP staining and gene profile expression. We then investigated whether the biomaterial-induced changes in OC differentiation also affect their ability to crosstalk with OBs and regulate OB function. We found that BCPs with low percentage of HA favored the expression of positive coupling factors, including sphingosine-kinase 1 (SPHK1) and collagen triple helix repeat containing 1 (Cthrc1). In turn, the increase of these secreted coupling factors promotes OB differentiation and function. All together our studies suggest that the chemical composition of biomaterials affects not only the differentiation and activity of OCs but also their potential to locally regulate bone formation.

Collaboration


Dive into the Kenichi Nagano's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kazuhiro Aoki

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar

Keiichi Ohya

Tokyo Medical and Dental University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Daniel J. Brooks

Beth Israel Deaconess Medical Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Neil Alles

Tokyo Medical and Dental University

View shared research outputs
Researchain Logo
Decentralizing Knowledge