Network


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

Hotspot


Dive into the research topics where Jun Asai is active.

Publication


Featured researches published by Jun Asai.


Nature Medicine | 2005

Sonic hedgehog myocardial gene therapy: tissue repair through transient reconstitution of embryonic signaling

Kengo Kusano; Roberto Pola; Toshinori Murayama; Cynthia Curry; Atsuhiko Kawamoto; Atsushi Iwakura; Satoshi Shintani; Masaaki; Jun Asai; Tengiz Tkebuchava; Tina Thorne; Hideya Takenaka; Ryuichi Aikawa; David A. Goukassian; Patrick von Samson; Hiromichi Hamada; Young-sup Yoon; Marcy Silver; Elizabeth Eaton; Hong Ma; Lindsay Heyd; Marianne Kearney; William Munger; Jeffery A Porter; Raj Kishore; Douglas W. Losordo

Sonic hedgehog (Shh) is a crucial regulator of organ development during embryogenesis. We investigated whether intramyocardial gene transfer of naked DNA encoding human Shh (phShh) could promote a favorable effect on recovery from acute and chronic myocardial ischemia in adult animals, not only by promoting neovascularization, but by broader effects, consistent with the role of this morphogen in embryogenesis. After Shh gene transfer, the hedgehog pathway was upregulated in mammalian fibroblasts and cardiomyocytes. This resulted in preservation of left ventricular function in both acute and chronic myocardial ischemia by enhanced neovascularization, and reduced fibrosis and cardiac apoptosis. Shh gene transfer also enhanced the contribution of bone marrow–derived endothelial progenitor cells to myocardial neovascularization. These data suggest that Shh gene therapy may have considerable therapeutic potential in individuals with acute and chronic myocardial ischemia by triggering expression of multiple trophic factors and engendering tissue repair in the adult heart.


Circulation | 2006

Topical Sonic Hedgehog Gene Therapy Accelerates Wound Healing in Diabetes by Enhancing Endothelial Progenitor Cell–Mediated Microvascular Remodeling

Jun Asai; Hideya Takenaka; Kengo Kusano; Masaaki; Corinne Luedemann; Cynthia Curry; Elizabeth Eaton; Atsushi Iwakura; Yoshiaki Tsutsumi; Hiromichi Hamada; Saburo Kishimoto; Tina Thorne; Raj Kishore; Douglas W. Losordo

Background— Sonic hedgehog (Shh) is a prototypical morphogen known to regulate epithelial-mesenchymal interaction during embryonic development. Recent observations indicate that exogenous administration of Shh can induce angiogenesis and may accelerate repair of ischemic myocardium and skeletal muscle. Because angiogenesis plays a pivotal role in wound repair, we hypothesized that activation of the hedgehog pathway may promote a favorable effect on microvascular remodeling during cutaneous wound healing and thereby accelerate wound closure. Because diabetes is associated with impaired wound healing, we tested this hypothesis in a diabetic model of cutaneous wound repair. Methods and Results— In Ptc1-LacZ mice, cutaneous injury resulted in LacZ expression, indicating that expression of the Shh receptor Patched was induced and therefore that the Shh signaling pathway was intact postnatally and upregulated in the process of wound repair. In diabetic mice, topical gene therapy with the use of naked DNA encoding for Shh resulted in significant local gene expression and acceleration of wound recovery. The acceleration in wound healing was notable for increased wound vascularity. In bone marrow transplantation models, the enhanced vascularity of the wound was shown to be mediated, at least in part, by enhanced recruitment of bone marrow–derived endothelial progenitor cells. In vitro, Shh promoted production of angiogenic cytokines from fibroblasts as well as proliferation of dermal fibroblasts. Furthermore, Shh directly promoted endothelial progenitor cell proliferation, migration, adhesion, and tube formation. Conclusions— These findings suggest that a simple strategy of topically applied Shh gene therapy may have significant therapeutic potential for enhanced wound healing in patients with impaired microcirculation such as occurs in diabetes. (Circulation. 2006;113:2413-2424.)


Circulation Research | 2006

Endothelial Progenitor Thrombospondin-1 Mediates Diabetes-Induced Delay in Reendothelialization Following Arterial Injury

Masaaki; Hideya Takenaka; Jun Asai; Kayoko Ibusuki; Yusuke Mizukami; Kazuichi Maruyama; Young-sup Yoon; Andrea Wecker; Corinne Luedemann; Elizabeth Eaton; Marcy Silver; Tina Thorne; Douglas W. Losordo

Delayed reendothelialization contributes to restenosis after angioplasty and stenting in diabetes. Prior data have shown that bone marrow (BM)-derived endothelial progenitor cells (EPCs) contribute to endothelial recovery after arterial injury. We investigated the hypothesis that the EPC contribution to reendothelialization may be impaired in diabetes, resulting in delayed reendothelialization. Reendothelialization was significantly reduced in diabetic mice compared with nondiabetic mice in a wire-induced carotid denudation model. The EPC contribution to neoendothelium was significantly reduced in Tie2/LacZ BM-transplanted diabetic versus nondiabetic mice. BM from diabetic and nondiabetic mice was transplanted into nondiabetic mice, revealing that reendothelialization was impaired in the recipients of diabetic BM. To examine the relative roles of denuded artery versus EPCs in diabetes, we injected diabetic and nondiabetic EPCs intravenously after arterial injury in diabetic and nondiabetic mice. Diabetic EPCs recruitment to the neoendothelium was significantly reduced, regardless of the diabetic status of the recipient mice. In vitro, diabetic EPCs exhibited decreased migration and adhesion activities. Vascular endothelial growth factor and endothelial NO synthase expressions were also significantly reduced in diabetic EPCs. Notably, thrombospondin-1 mRNA expression was significantly upregulated in diabetic EPCs, associating with the decreased EPC adhesion activity in vitro and in vivo. Reendothelialization is impaired by malfunctioning EPCs in diabetes. Diabetic EPCs have phenotypic differences involving thrombospondin-1 expression compared with nondiabetic EPCs, revealing potential novel mechanistic insights and therapeutic targets to improve reendothelialization and reduce restenosis in diabetes.


Circulation | 2006

Estrogen Receptors α and β Mediate Contribution of Bone Marrow–Derived Endothelial Progenitor Cells to Functional Recovery After Myocardial Infarction

Hiromichi Hamada; Myeong Kon Kim; Atsushi Iwakura; Masaaki; Tina Thorne; Gangjian Qin; Jun Asai; Yoshiaki Tsutsumi; Haruki Sekiguchi; Marcy Silver; Andrea Wecker; Evelyn Bord; Yan Zhu; Raj Kishore; Douglas W. Losordo

Background— Estradiol (E2) modulates the kinetics of circulating endothelial progenitor cells (EPCs) and favorably affects neovascularization after ischemic injury. However, the roles of estrogen receptors &agr; (ER&agr;) and &bgr; (ER&bgr;) in EPC biology are largely unknown. Methods and Results— In response to E2, migration, tube formation, adhesion, and estrogen-responsive element–dependent gene transcription activities were severely impaired in EPCs obtained from ER&agr;-knockout mice (ER&agr;KO) and moderately impaired in ER&bgr;KO EPCs. The number of ER&agr;&Kgr;&Ogr; EPCs (42.4±1.5; P<0.001) and ER&bgr;KO EPCs (55.4±1.8; P= 0.03) incorporated into the ischemic border zone was reduced as compared with wild-type (WT) EPCs (72.5±1.3). In bone marrow transplantation (BMT) models, the number of mobilized endogenous EPCs in E2-treated mice was significantly reduced in ER&agr;KO BMT (WT mice transplanted with ER&agr;KO bone marrow) (2.03±0.18%; P= 0.004 versus WT BMT) and ER&bgr;KO BMT (2.62±0.07%; P= 0.02 versus WT) compared with WT BMT (2.87±0.13%) (WT to WT BMT as control) mice. Capillary density at the border zone of ischemic myocardium also was significantly reduced in ER&agr;KO BMT and ER&bgr;KO BMT compared with WT mice (WT BMT, 1718±75/mm2; ER&agr;KO BMT, 1107±48/mm2; ER&bgr;KO BMT, 1567±50/mm2). ER&agr; mRNA was expressed more abundantly on EPCs compared with ER&bgr;. Moreover, vascular endothelial growth factor was significantly downregulated on ER&agr;KO EPCs compared with WT EPCs both in vitro and in vivo. Conclusions— Both ER&agr; and ER&bgr; contribute to E2-mediated EPC activation and tissue incorporation and to preservation of cardiac function after myocardial infarction. ER&agr; plays a more prominent role in this process. Moreover, ER&agr; contributes to upregulation of vascular endothelial growth factor, revealing possible mechanisms of an effect of E2 on EPC biology. Finally, these data provide additional evidence of the importance of bone marrow–derived EPC phenotype in ischemic tissue repair.


The FASEB Journal | 2006

IL-10-induced TNF-alpha mRNA destabilization is mediated via IL-10 suppression of p38 MAP kinase activation and inhibition of HuR expression

Johnson Rajasingh; Evelyn Bord; Corinne Luedemann; Jun Asai; Hiromichi Hamada; Tina Thorne; Gangjian Qin; David A. Goukassian; Yan Zhu; Douglas W. Losordo; Raj Kishore

Inflammation plays an essential role in vascular injury and repair. Mononuclear phagocytes are important contributors in these processes, in part, via adhesive interactions and secretion of proinflammatory cytokines. The antiinflammatory cytokine interleukin (IL)‐10 suppresses such responses via deactivation of monocytes/macrophages and repression of inflammatory cytokine expression. The mechanisms of IL‐10s suppressive action are, however, incompletely characterized. Here, we report that systemic IL‐10 treatment after carotid artery denudation in mice blunts inflammatory cell infiltration and arterial tumor necrosis factor (TNF) expression. At the molecular level, in a human monocytic cell line, U937 IL‐10 suppressed LPS‐induced mRNA expression of a number of inflammatory cytokines, mainly via posttranscriptional mRNA destabilization. Detailed studies on IL‐10 regulation of TNF‐ mRNA expression identified AU‐rich elements (ARE) in the 3 untranslated region as a necessary determinant of IL‐10mediated TNF‐α mRNA destabilization. IL‐10 sensitivity to TNF depends on the ability of IL‐10 to inhibit the expression and mRNA‐stabilizing protein HuR and via IL‐10 mediated repression of p38 mitogen‐activated protein (MAP) kinase activation. Because IL‐10 function and signaling are important components for control of inflammatory responses, these results may provide insights necessary to develop strategies for modulating vascular repair and other accelerated arteriopathies, including transplant vasculopathy and vein graft hyperplasia.—Johnson Rajasingh, Evelyn Bord, Corinne Luedemann, Jun Asai, Hiromichi Hamada, Tina Thorne, Gangjian Qin, David Goukassian, Yan Zhu, Douglas W. Losordo, and Raj Kishore. IL‐10‐induced TNF‐αalpha mRNA destabilization is mediated via IL‐10 suppression of p38 MAP kinase activation and inhibition of HuR expression. FASEB J. 20, E1393–E1403 (2006)


Journal of Investigative Dermatology | 2012

CXCR4 Antagonist AMD3100 Accelerates Impaired Wound Healing in Diabetic Mice

Yukihide Nishimura; Masaaki; Gangjian Qin; Hiromichi Hamada; Jun Asai; Hideya Takenaka; Haruki Sekiguchi; Marie-Ange Renault; Kentaro Jujo; Norito Katoh; Saburo Kishimoto; Aiko Ito; Christine Kamide; John Kenny; Meredith Millay; Sol Misener; Tina Thorne; Douglas W. Losordo

The antagonism of CXC-chemokine receptor 4 (CXCR4) with AMD3100 improves cardiac performance after myocardial infarction by augmenting the recruitment of endothelial progenitor cells (EPCs) from the bone marrow to the regenerating vasculature. We investigated whether AMD3100 may accelerate diabetes-impaired wound healing through a similar mechanism. Skin wounds were made on the backs of leptin-receptor–deficient mice and treated with AMD3100 or saline. Fourteen days after treatment, wound closure was significantly more complete in AMD3100-treated mice (AMD3100: 87.0±2.6%, Saline: 33.1±1.8%; P<0.0001) and was accompanied by greater collagen-fiber formation, capillary density, smooth-muscle-containing vessel density, and monocyte/macrophage infiltration. On day 7 after treatment, AMD3100 was associated with higher circulating EPC and macrophage counts and with significantly upregulated mRNA levels of stromal-cell–derived factor 1 and platelet-derived growth-factor B in the wound bed. AMD3100 also promoted macrophage proliferation and phagocytosis and the migration and proliferation of diabetic mouse primary dermal fibroblasts and 3T3 fibroblasts, which express very little CXCR4. In conclusion, a single topical application of AMD3100 promoted wound healing in diabetic mice by increasing cytokine production, mobilizing bone-marrow EPCs, and enhancing the activity of fibroblasts and monocytes/macrophages, thereby increasing both angiogenesis and vasculogenesis. Not all of the AMD3100-mediated effects evolved through CXCR4 antagonism.


American Journal of Pathology | 2012

Topical Simvastatin Accelerates Wound Healing in Diabetes by Enhancing Angiogenesis and Lymphangiogenesis

Jun Asai; Hideya Takenaka; Satoshi Hirakawa; Jun-ichi Sakabe; Asami Hagura; Saburo Kishimoto; Kazuichi Maruyama; Kentaro Kajiya; Shigeru Kinoshita; Yoshiki Tokura; Norito Katoh

Impaired wound healing is a major complication of diabetes. Recent studies have reported reduced lymphangiogenesis and angiogenesis during diabetic wound healing, which are thought to be new therapeutic targets. Statins have effects beyond cholesterol reduction and can stimulate angiogenesis when used systemically. However, the effects of topically applied statins on wound healing have not been well investigated. The present study tested the hypothesis that topical application of simvastatin would promote lymphangiogenesis and angiogenesis during wound healing in genetically diabetic mice. A full-thickness skin wound was generated on the back of the diabetic mice and treated with simvastatin or vehicle topically. Simvastatin administration resulted in significant acceleration of wound recovery, which was notable for increases in both angiogenesis and lymphangiogenesis. Furthermore, simvastatin promoted infiltration of macrophages, which produced vascular endothelial growth factor C in granulation tissues. In vitro, simvastatin directly promoted capillary morphogenesis and exerted an antiapoptotic effect on lymphatic endothelial cells. These results suggest that the favorable effects of simvastatin on lymphangiogenesis are due to both a direct influence on lymphatics and indirect effects via macrophages homing to the wound. In conclusion, a simple strategy of topically applied simvastatin may have significant therapeutic potential for enhanced wound healing in patients with impaired microcirculation such as that in diabetes.


International Wound Journal | 2013

Topical application of ex vivo expanded endothelial progenitor cells promotes vascularisation and wound healing in diabetic mice.

Jun Asai; Hideya Takenaka; Masaaki; Michio Asahi; Saburo Kishimoto; Norito Katoh; Douglas W. Losordo

Impaired wound healing leading to skin ulceration is a serious complication of diabetes and may be caused by defective angiogenesis. Endothelial progenitor cells (EPCs) can augment neovascularisation in the ischaemic tissue. Experiments were performed to test the hypothesis that locally administered EPCs can promote wound healing in diabetes. Full‐thickness skin wounds were created on the dorsum of diabetic mice. EPCs were obtained from bone marrow mononuclear cells (BMMNCs) and applied topically to the wound immediately after surgery. Vehicle and non‐selective BMMNCs were used as controls. Wound size was measured on days 5, 10 and 14 after treatment, followed by resection, histological analysis and quantification of vascularity. Topical application of EPCs significantly promoted wound healing, as assessed by closure rate and wound vascularity. Immunostaining revealed that transplanted EPCs induced increased expression of vascular endothelial growth factor and basic fibroblast growth factor. Few EPCs were observed in the neovasculature based on in vivo staining of the functional vasculature. Ex vivo expanded EPCs promote wound healing in diabetic mice via mechanisms involving increased local cytokine expression and enhanced neovascularisation of the wound. This strategy exploiting the therapeutic capacity of autologously derived EPCs may be a novel approach to skin repair in diabetes.


OncoImmunology | 2016

Intratumoral expression levels of PD-L1, GZMA, and HLA-A along with oligoclonal T cell expansion associate with response to nivolumab in metastatic melanoma.

Hiroyuki Inoue; Jae-Hyun Park; Kazuma Kiyotani; Makda Zewde; Azusa Miyashita; Masatoshi Jinnin; Yukiko Kiniwa; Ryuhei Okuyama; Ryota Tanaka; Yasuhiro Fujisawa; Hiroshi Kato; Akimichi Morita; Jun Asai; Norito Katoh; Kenji Yokota; Masashi Akiyama; Hironobu Ihn; Satoshi Fukushima; Yusuke Nakamura

ABSTRACT Immune checkpoint inhibitors blocking the interaction between programmed death-1 (PD-1) and PD-1 ligand-1 (PD-L1) are revolutionizing the cancer immunotherapies with durable clinical responses. Although high expression of PD-L1 in tumor tissues has been implicated to correlate with the better response to the anti-PD-1 therapies, this association has been controversial. In this study, to characterize immune microenvironment in tumors, we examined mRNA levels of immune-related genes and characterized T cell repertoire in the tumors of 13 melanoma patients before and after nivolumab treatment. We found that, in addition to the PD-L1 (p = 0.03), expression levels of PD-1 ligand-2 (PD-L2), granzyme A (GZMA) and human leukocyte antigen-A (HLA-A) in the pre-treatment tumors were significantly higher (p = 0.04, p = 0.01 and p = 0.006, respectively) in responders (n = 5) than in non-responders (n = 8). With nivolumab treatment, tumors in responders exhibited a substantial increase of CD8, GZMA and perforin 1 (PRF1) expression levels as well as increased ratio of TBX21/GATA3, suggesting dominancy of helper T cell type 1 (Th1) response to type 2 (Th2) response. T cell receptor β (TCR-β) repertoire analysis revealed oligoclonal expansion of tumor-infiltrating T lymphocytes (TILs) in the tumor tissues of the responders. Our findings suggest that melanoma harboring high PD-1 ligands (PD-L1 and PD-L2), GZMA and HLA-A expression may respond preferentially to nivolumab treatment, which can enhance Th1-skewed cellular immunity with oligoclonal expansion of TILs.


Journal of Dermatology | 2006

Vaccination of Japanese patients with advanced melanoma with peptide, tumor lysate or both peptide and tumor lysate-pulsed mature, monocyte-derived dendritic cells

Noriaki Nakai; Jun Asai; Eiichiro Ueda; Hideya Takenaka; Norito Katoh; Saburo Kishimoto

We performed a clinical trial to assess the feasibility and efficacy of immunotherapy with peptides, tumor lysate or both peptides and tumor lysate‐pulsed mature, monocyte‐derived dendritic cells (DC) for advanced malignant melanoma patients that are resistant to conventional therapies. Sixteen patients were enrolled in this trial. All patients received DC vaccines i.d. in the proximal thigh, close to the inguinal lymph nodes, one treatment per week or 2 weeks. Several factors such as clinical findings, computed tomography (CT) images, delayed type hypersensitivity (DTH) response, enzyme‐linked immunosorbent spot (ELISPOT) assay, and immunohistochemistry in primary, metastatic lesions and the DTH site were evaluated. Clinical results through DC vaccination were as follows: in 11 evaluable cases, three stable disease, six progression of disease and two disease‐free from the time of study entry to the completion of one vaccination course. One patient showed reduction of the tumors in the metastases on chest CT during the first and second course of DC vaccination. Ten out of 14 evaluable cases showed positive DTH responses to more than one treatment with melanoma peptides or tumor lysate. Eight out of 13 evaluable cases showed positive immunological responses to more than one treatment with melanoma peptides or tumor lysate in an ELISPOT assay. As for the experiences with toxicity and adverse reactions, autosensitization dermatitis‐like eruptions appeared in five cases during DC vaccination. No severe adverse effects were seen in any of the patients. In our study, the clinical efficacy in prolongation of the patients’ survival was confirmed. At the same time, cancer immunoediting of the tumor was also found. It will be necessary to improve the tumor‐specificity of this therapeutic approach and to analyze the mechanism(s) of tumor escape from immunosurveillance in melanoma.

Collaboration


Dive into the Jun Asai's collaboration.

Top Co-Authors

Avatar

Norito Katoh

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Hideya Takenaka

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Makoto Wada

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Saburo Kishimoto

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Tina Thorne

Northwestern University

View shared research outputs
Top Co-Authors

Avatar

Satoshi Komori

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Eiichi Konishi

Kyoto Prefectural University of Medicine

View shared research outputs
Top Co-Authors

Avatar

Keiji Hanada

Kyoto Prefectural University of Medicine

View shared research outputs
Researchain Logo
Decentralizing Knowledge