Network


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

Hotspot


Dive into the research topics where Kentaro Akiyama is active.

Publication


Featured researches published by Kentaro Akiyama.


Stem Cells | 2009

Mesenchymal Stem Cell Transplantation Reverses Multiorgan Dysfunction in Systemic Lupus Erythematosus Mice and Humans

Lingyun Sun; Kentaro Akiyama; Huayong Zhang; Takayoshi Yamaza; Yayi Hou; Shengnan Zhao; Ting Xu; Anh Le; Songtao Shi

Systemic lupus erythematosus (SLE) is a multisystem autoimmune disease that, despite the advances in immunosuppressive medical therapies, remains potentially fatal in some patients, especially in treatment‐refractory patients. Here, we reported that impairment of bone marrow mesenchymal stem cells (BMMSCs) and their associated osteoblastic niche deficiency contribute in part to the pathogenesis of SLE‐like disease in MRL/lpr mice. Interestingly, allogenic BMMSC transplantation (MSCT) is capable of reconstructing the bone marrow osteoblastic niche and more effectively reverses multiorgan dysfunction when compared with medical immunosuppression with cyclophosphamide (CTX). At the cellular level, MSCT, not CTX treatment, was capable to induce osteoblastic niche reconstruction, possibly contributing to the recovery of regulatory T‐cells and reestablishment of the immune homeostasis. On the basis of the promising clinical outcomes in SLE mice, we treated four CTX/glucocorticoid treatment‐refractory SLE patients using allogenic MSCT and showed a stable 12–18 months disease remission in all treated patients. The patients benefited an amelioration of disease activity, improvement in serologic markers and renal function. These early evidences suggest that allogenic MSCT may be a feasible and safe salvage therapy in refractory SLE patients. STEM CELLS 2009;27:1421–1432


Nature Medicine | 2011

Mesenchymal stem cell–based tissue regeneration is governed by recipient T lymphocytes via IFN-γ and TNF-α

Yi Liu; Lei Wang; Takashi Kikuiri; Kentaro Akiyama; Chider Chen; Xingtian Xu; Ruili Yang; WanJun Chen; Songlin Wang; Songtao Shi

Stem cell–based regenerative medicine is a promising approach in tissue reconstruction. Here we show that proinflammatory T cells inhibit the ability of exogenously added bone marrow mesenchymal stem cells (BMMSCs) to mediate bone repair. This inhibition is due to interferon γ (IFN-γ)–induced downregulation of the runt-related transcription factor 2 (Runx-2) pathway and enhancement of tumor necrosis factor α (TNF-α) signaling in the stem cells. We also found that, through inhibition of nuclear factor κB (NF-κB), TNF-α converts the signaling of the IFN-γ–activated, nonapoptotic form of TNF receptor superfamily member 6 (Fas) in BMMSCs to a caspase 3– and caspase 8–associated proapoptotic cascade, resulting in the apoptosis of these cells. Conversely, reduction of IFN-γ and TNF-α concentrations by systemic infusion of Foxp3+ regulatory T cells, or by local administration of aspirin, markedly improved BMMSC-based bone regeneration and calvarial defect repair in C57BL/6 mice. These data collectively show a previously unrecognized role of recipient T cells in BMMSC-based tissue engineering.


Cell Stem Cell | 2012

Mesenchymal-stem-cell-induced immunoregulation involves FAS-ligand-/FAS-mediated T cell apoptosis

Kentaro Akiyama; Chider Chen; Dandan Wang; Xingtian Xu; Cunye Qu; Takayoshi Yamaza; Tao Cai; WanJun Chen; Lingyun Sun; Songtao Shi

Systemic infusion of bone marrow mesenchymal stem cells (BMMSCs) yields therapeutic benefit for a variety of autoimmune diseases, but the underlying mechanisms are poorly understood. Here we show that in mice systemic infusion of BMMSCs induced transient T cell apoptosis via the FAS ligand (FASL)-dependent FAS pathway and could ameliorate disease phenotypes in fibrillin-1 mutated systemic sclerosis (SS) and dextran-sulfate-sodium-induced experimental colitis. FASL(-/-) BMMSCs did not induce T cell apoptosis in recipients, and could not ameliorate SS and colitis. Mechanistic analysis revealed that FAS-regulated monocyte chemotactic protein 1 (MCP-1) secretion by BMMSCs recruited T cells for FASL-mediated apoptosis. The apoptotic T cells subsequently triggered macrophages to produce high levels of TGFβ, which in turn led to the upregulation of CD4(+)CD25(+)Foxp3(+) regulatory T cells and, ultimately, immune tolerance. These data therefore demonstrate a previously unrecognized mechanism underlying BMMSC-based immunotherapy involving coupling via FAS/FASL to induce T cell apoptosis.


Journal of Prosthodontic Research | 2012

Stem cells in dentistry - Part I: Stem cell sources

Hiroshi Egusa; Wataru Sonoyama; Masahiro Nishimura; Ikiru Atsuta; Kentaro Akiyama

Stem cells can self-renew and produce different cell types, thus providing new strategies to regenerate missing tissues and treat diseases. In the field of dentistry, adult mesenchymal stem/stromal cells (MSCs) have been identified in several oral and maxillofacial tissues, which suggests that the oral tissues are a rich source of stem cells, and oral stem and mucosal cells are expected to provide an ideal source for genetically reprogrammed cells such as induced pluripotent stem (iPS) cells. Furthermore, oral tissues are expected to be not only a source but also a therapeutic target for stem cells, as stem cell and tissue engineering therapies in dentistry continue to attract increasing clinical interest. Part I of this review outlines various types of intra- and extra-oral tissue-derived stem cells with regard to clinical availability and applications in dentistry. Additionally, appropriate sources of stem cells for regenerative dentistry are discussed with regard to differentiation capacity, accessibility and possible immunomodulatory properties.


PLOS ONE | 2008

Pharmacologic Stem Cell Based Intervention as a New Approach to Osteoporosis Treatment in Rodents

Takayoshi Yamaza; Yasuo Miura; Yanming Bi; Yongzhong Liu; Kentaro Akiyama; Wataru Sonoyama; Voymesh Patel; Silvio Gutkind; M.F. Young; Stan Gronthos; Anh Le; Cun-Yu Wang; Wanjun Chen; Songtao Shi

Background Osteoporosis is the most prevalent skeletal disorder, characterized by a low bone mineral density (BMD) and bone structural deterioration, leading to bone fragility fractures. Accelerated bone resorption by osteoclasts has been established as a principal mechanism in osteoporosis. However, recent experimental evidences suggest that inappropriate apoptosis of osteoblasts/osteocytes accounts for, at least in part, the imbalance in bone remodeling as occurs in osteoporosis. The aim of this study is to examine whether aspirin, which has been reported as an effective drug improving bone mineral density in human epidemiology studies, regulates the balance between bone resorption and bone formation at stem cell levels. Methods and Findings We found that T cell-mediated bone marrow mesenchymal stem cell (BMMSC) impairment plays a crucial role in ovariectomized-induced osteoporosis. Ex vivo mechanistic studies revealed that T cell-mediated BMMSC impairment was mainly attributed to the apoptosis of BMMSCs via the Fas/Fas ligand pathway. To explore potential of using pharmacologic stem cell based intervention as an approach for osteoporosis treatment, we selected ovariectomy (OVX)-induced ostoeporosis mouse model to examine feasibility and mechanism of aspirin-mediated therapy for osteoporosis. We found that aspirin can inhibit T cell activation and Fas ligand induced BMMSC apoptosis in vitro. Further, we revealed that aspirin increases osteogenesis of BMMSCs by aiming at telomerase activity and inhibits osteoclast activity in OVX mice, leading to ameliorating bone density. Conclusion Our findings have revealed a novel osteoporosis mechanism in which activated T cells induce BMMSC apoptosis via Fas/Fas ligand pathway and suggested that pharmacologic stem cell based intervention by aspirin may be a new alternative in osteoporosis treatment including activated osteoblasts and inhibited osteoclasts.


Journal of Bone and Mineral Research | 2010

Cell-based immunotherapy with mesenchymal stem cells cures bisphosphonate-related osteonecrosis of the jaw–like disease in mice

Takashi Kikuiri; Takyoshi Yamaza; Kentaro Akiyama; Qunzhou Zhang; Yunsheng Li; Chider Chen; Wanjun Chen; Songlin Wang; Anh D. Le; Songtao Shi

Patients on high‐dose bisphosphonate and immunosuppressive therapy have an increased risk of bisphosphonate‐related osteonecrosis of the jaw (BRONJ); despite the disease severity, its pathophysiology remains unknown, and appropriate therapy is not established. Here we have developed a mouse model of BRONJ‐like disease that recapitulates major clinical and radiographic manifestations of the human disease, including characteristic features of an open alveolar socket, exposed necrotic bone or sequestra, increased inflammatory infiltrates, osseous sclerosis, and radiopaque alveolar bone. We show that administration of zoledronate, a potent aminobisphosphonate, and dexamethasone, an immunosuppressant drug, causes BRONJ‐like disease in mice in part by suppressing the adaptive regulatory T cells, Tregs, and activating the inflammatory T‐helper‐producing interleukin 17 cells, Th17. Most interestingly, we demonstrate that systemic infusion with mesenchymal stem cells (MSCs) prevents and cures BRONJ‐like disease possibly via induction of peripheral tolerance, shown as an inhibition of Th17 and increase in Treg cells. The suppressed Tregs/Th17 ratio in zoledronate‐ and dexamethasone‐treated mice is restored in mice undergoing salvage therapy with Tregs. These findings provide evidence of an immunity‐based mechanism of BRONJ‐like disease and support the rationale for in vivo immunomodulatory therapy using Tregs or MSCs to treat BRONJ.


Journal of Prosthodontic Research | 2012

Stem cells in dentistry - Part II: Clinical applications

Hiroshi Egusa; Wataru Sonoyama; Masahiro Nishimura; Ikiru Atsuta; Kentaro Akiyama

New technologies that facilitate solid alveolar ridge augmentation are receiving considerable attention in the field of prosthodontics because of the growing requirement for esthetic and functional reconstruction by dental implant treatments. Recently, several studies have demonstrated potential advantages for stem-cell-based therapies in regenerative treatments. Mesenchymal stem/stromal cells (MSCs) are now an excellent candidate for tissue replacement therapies, and tissue engineering approaches and chair-side cellular grafting approaches using autologous MSCs represent the clinical state of the art for stem-cell-based alveolar bone regeneration. Basic studies have revealed that crosstalk between implanted donor cells and recipient immune cells plays a key role in determining clinical success that may involve the recently observed immunomodulatory properties of MSCs. Part II of this review first overviews progress in regenerative dentistry to consider the implications of the stem cell technology in dentistry and then highlights cutting-edge stem-cell-based alveolar bone regenerative therapies. Factors that affect stem-cell-based bone regeneration as related to the local immune response are then discussed. Additionally, pre-clinical stem cell studies for the regeneration of teeth and other oral organs as well as possible applications of MSC-based immunotherapy in dentistry are outlined. Finally, the marketing of stem cell technology in dental stem cell banks with a view toward future regenerative therapies is introduced.


Biomaterials | 2013

Co-encapsulation of anti-BMP2 monoclonal antibody and mesenchymal stem cells in alginate microspheres for bone tissue engineering

Alireza Moshaverinia; Sahar Ansari; Chider Chen; Xingtian Xu; Kentaro Akiyama; Malcolm L. Snead; Homayoun H. Zadeh; Songtao Shi

Recently, it has been shown that tethered anti-BMP2 monoclonal antibodies (mAbs) can trap BMP ligands and thus provide BMP inductive signals for osteo-differentiation of progenitor cells. The objectives of this study were to: (1) develop a co-delivery system based on murine anti-BMP2 mAb-loaded alginate microspheres encapsulating human bone marrow mesenchymal stem cells (hBMMSCs); and (2) investigate osteogenic differentiation of encapsulated stem cells in alginate microspheres in vitro and in vivo. Alginate microspheres of 1 ± 0.1 mm diameter were fabricated with 2 × 10(6) hBMMSCs per mL of alginate. Critical-size calvarial defects (5 mm diameter) were created in immune-compromised mice and alginate microspheres preloaded with anti-BMP mAb encapsulating hBMMSCs were transplanted into defect sites. Alginate microspheres pre-loaded with isotype-matched non-specific antibody were used as the negative control. After 8 weeks, micro CT and histologic analyses were used to analyze bone formation. In vitro analysis demonstrated that anti-BMP2 mAbs tethered BMP2 ligands that can activate the BMP receptors on hBMMSCs. The co-delivery system described herein, significantly enhanced hBMMSC-mediated osteogenesis, as confirmed by the presence of BMP signal pathway-activated osteoblast determinants Runx2 and ALP. Our results highlight the importance of engineering the microenvironment for stem cells, and particularly the value of presenting inductive signals for osteo-differentiation of hBMMSCs by tethering BMP ligands using mAbs. This strategy of engineering the microenvironment with captured BMP signals is a promising modality for repair and regeneration of craniofacial, axial and appendicular bone defects.


Journal of Dental Research | 2013

Gingivae Contain Neural-crest- and Mesoderm-derived Mesenchymal Stem Cells

Xingtian Xu; Chider Chen; Kentaro Akiyama; Yang Chai; Anh D. Le; Z. Wang; Songtao Shi

Gingivae represent a unique soft tissue that serves as a biological barrier to cover the oral cavity side of the maxilla and mandible. Recently, the gingivae were identified as containing mesenchymal stem cells (GMSCs). However, it is unknown whether the GMSCs are derived from cranial neural crest cells (CNCC) or the mesoderm. In this study, we show that around 90% of GMSCs are derived from CNCC and 10% from the mesoderm. In comparison with mesoderm MSCs (M-GMSCs), CNCC-derived GMSCs (N-GMSCs) show an elevated capacity to differentiate into neural cells and chondrocytes and induce activated T-cell apoptosis in vitro. When transplanted into mice with dextran sulfate sodium (DSS)-induced colitis, N-GMSCs showed superior effects in ameliorating inflammatory-related disease phenotype in comparison with the M-GMSC treatment group. Mechanistically, the increased immunomodulatory effect of N-GMSCs is associated with up-regulated expression of FAS ligand (FASL), a transmembrane protein that plays an important role in MSC-based immunomodulation. In summary, our study indicates that the gingivae contain both neural-crest- and mesoderm-derived MSCs with distinctive stem cell properties.


Stem Cells | 2013

IFN‐γ and TNF‐α Synergistically Induce Mesenchymal Stem Cell Impairment and Tumorigenesis via NFκB Signaling

Lei Wang; Yinghua Zhao; Yi Liu; Kentaro Akiyama; Chider Chen; Cunye Qu; Yan Jin; Songtao Shi

An inflammatory microenvironment may cause organ degenerative diseases and malignant tumors. However, the precise mechanisms of inflammation‐induced diseases are not fully understood. Here, we show that the proinflammatory cytokines interferon‐γ (IFN‐γ) and tumor necrosis factor α (TNF‐α) synergistically impair self‐renewal and differentiation of mesenchymal stem cells (MSCs) via nuclear factor κB (NFκB)‐mediated activation of mothers against decapentaplegic homolog 7 (SMAD7) in ovariectomized (OVX) mice. More interestingly, a long‐term elevated levels of IFN‐γ and TNF‐α result in significantly increased susceptibility to malignant transformation in MSCs through NFκB‐mediated upregulation of the oncogenes c‐Fos and c‐Myc. Depletion of either IFN‐γ or TNF‐α in OVX mice abolishes MSC impairment and the tendency toward malignant transformation with no NFκB‐mediated oncogene activation. Systemic administration of aspirin, which significantly reduces the levels of IFN‐γ and TNF‐α, results in blockage of MSC deficiency and tumorigenesis by inhibition of NFκB/SMAD7 and NFκB/c‐FOS and c‐MYC pathways in OVX mice. In summary, this study reveals that inflammation factors, such as IFN‐γ and TNF‐α, synergistically induce MSC deficiency via NFκB/SMAD7 signaling and tumorigenesis via NFκB‐mediated oncogene activation. STEM Cells2013;31:1383–1395

Collaboration


Dive into the Kentaro Akiyama's collaboration.

Top Co-Authors

Avatar

Songtao Shi

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar

Chider Chen

University of Pennsylvania

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Xingtian Xu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Cunye Qu

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Ruili Yang

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Wataru Sonoyama

University of Southern California

View shared research outputs
Top Co-Authors

Avatar

Yi Liu

Capital Medical University

View shared research outputs
Top Co-Authors

Avatar

Alireza Moshaverinia

University of Southern California

View shared research outputs
Researchain Logo
Decentralizing Knowledge