Network


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

Hotspot


Dive into the research topics where Tomoki Abe is active.

Publication


Featured researches published by Tomoki Abe.


Journal of Applied Physiology | 2012

Unloading stress disturbs muscle regeneration through perturbed recruitment and function of macrophages.

Shohei Kohno; Yui Yamashita; Tomoki Abe; Katsuya Hirasaka; Motoko Oarada; Ayako Ohno; Shigetada Teshima-Kondo; Akira Higashibata; Inho Choi; Edward M. Mills; Yuushi Okumura; Junji Terao; Takeshi Nikawa

Skeletal muscle is one of the most sensitive tissues to mechanical loading, and unloading inhibits the regeneration potential of skeletal muscle after injury. This study was designed to elucidate the specific effects of unloading stress on the function of immunocytes during muscle regeneration after injury. We examined immunocyte infiltration and muscle regeneration in cardiotoxin (CTX)-injected soleus muscles of tail-suspended (TS) mice. In CTX-injected TS mice, the cross-sectional area of regenerating myofibers was smaller than that of weight-bearing (WB) mice, indicating that unloading delays muscle regeneration following CTX-induced skeletal muscle damage. Delayed infiltration of macrophages into the injured skeletal muscle was observed in CTX-injected TS mice. Neutrophils and macrophages in CTX-injected TS muscle were presented over a longer period at the injury sites compared with those in CTX-injected WB muscle. Disturbance of activation and differentiation of satellite cells was also observed in CTX-injected TS mice. Further analysis showed that the macrophages in soleus muscles were mainly Ly-6C-positive proinflammatory macrophages, with high expression of tumor necrosis factor-α and interleukin-1β, indicating that unloading causes preferential accumulation and persistence of proinflammatory macrophages in the injured muscle. The phagocytic and myotube formation properties of macrophages from CTX-injected TS skeletal muscle were suppressed compared with those from CTX-injected WB skeletal muscle. We concluded that the disturbed muscle regeneration under unloading is due to impaired macrophage function, inhibition of satellite cell activation, and their cooperation.


Diabetes | 2013

Cbl-b Is a Critical Regulator of Macrophage Activation Associated With Obesity-Induced Insulin Resistance in Mice

Tomoki Abe; Katsuya Hirasaka; Sachiko Kagawa; Shohei Kohno; Arisa Ochi; Kenro Utsunomiya; Atsuko Sakai; Ayako Ohno; Shigetada Teshima-Kondo; Yuushi Okumura; Motoko Oarada; Yoichi Maekawa; Junji Terao; Edward M. Mills; Takeshi Nikawa

We previously reported the potential involvement of casitas B-cell lymphoma-b (Cbl-b) in aging-related murine insulin resistance. Because obesity also induces macrophage recruitment into adipose tissue, we elucidated here the role of Cbl-b in obesity-related insulin resistance. Cbl-b+/+ and Cbl-b−/− mice were fed a high-fat diet (HFD) and then examined for obesity-related changes in insulin signaling. The HFD caused recruitment of macrophages into adipose tissue and increased inflammatory reaction in Cbl-b−/− compared with Cbl-b+/+ mice. Peritoneal macrophages from Cbl-b−/− mice and Cbl-b–overexpressing RAW264.7 macrophages were used to examine the direct effect of saturated fatty acids (FAs) on macrophage activation. In macrophages, Cbl-b suppressed saturated FA-induced Toll-like receptor 4 (TLR4) signaling by ubiquitination and degradation of TLR4. The physiological role of Cbl-b in vivo was also examined by bone marrow transplantation and Eritoran, a TLR4 antagonist. Hematopoietic cell-specific depletion of the Cbl-b gene induced disturbed responses on insulin and glucose tolerance tests. Blockade of TLR4 signaling by Eritoran reduced fasting blood glucose and serum interleukin-6 levels in obese Cbl-b−/− mice. These results suggest that Cbl-b deficiency could exaggerate HFD-induced insulin resistance through saturated FA-mediated macrophage activation. Therefore, inhibition of TLR4 signaling is an attractive therapeutic strategy for treatment of obesity-related insulin resistance.


Muscle & Nerve | 2011

RANTES SECRETED FROM MACROPHAGES DISTURBS SKELETAL MUSCLE REGENERATION AFTER CARDIOTOXIN INJECTION IN Cbl-b-DEFICIENT MICE

Shohei Kohno; Tatsuya Ueji; Tomoki Abe; Reiko Nakao; Katsuya Hirasaka; Motoko Oarada; Akiko Harada-Sukeno; Ayako Ohno; Akira Higashibata; Rie Mukai; Junji Terao; Yuushi Okumura; Takeshi Nikawa

Deficiency of the Cbl‐b ubiquitin ligase gene activates macrophages in mice. This study aimed to elucidate the pathophysiological roles of macrophages in muscle degeneration/regeneration in Cbl‐b‐deficient mice. We examined immune cell infiltration and cytokine expression in cardiotoxin‐injected tibialis anterior muscle of Cbl‐b‐deficient mice. Ablation of the Cbl‐b gene expression delayed regeneration of cardiotoxin‐induced skeletal muscle damage compared with wild‐type mice. CD8‐positive T cells were still present in the damaged muscle on day 14 after cardiotoxin injection in Cbl‐b‐deficient mice, but there was dispersal of the same cells over that time‐frame in wild‐type mice. Infiltrating macrophages in Cbl‐b‐deficient mice showed strong expression of RANTES (regulated‐on‐activation, normal T cell expressed and secreted), a chemokine for CD8‐positive T cells. In turn, a neutralizing antibody against RANTES significantly suppressed the infiltration of CD8‐positive T cells into the muscle, resulting in restoration of the disturbed muscle regeneration. Cbl‐b is an important regulatory factor for cytotoxic T‐cell infiltration via RANTES production in macrophages. Muscle Nerve, 2011


Journal of Nutritional Science and Vitaminology | 2015

Flavones Inhibit LPS-Induced Atrogin-1/MAFbx Expression in Mouse C2C12 Skeletal Myotubes

Chieko Shiota; Tomoki Abe; Nobuhiko Kawai; Ayako Ohno; Shigetada Teshima-Kondo; Hiroyo Mori; Junji Terao; Eiji Tanaka; Takeshi Nikawa

Muscle atrophy is a complex process that occurs as a consequence of various stress events. Muscle atrophy-associated genes (atrogenes) such as atrogin-1/MAFbx and MuRF-1 are induced early in the atrophy process, and the increase in their expression precedes the loss of muscle weight. Although antioxidative nutrients suppress atrogene expression in skeletal muscle cells, the inhibitory effects of flavonoids on inflammation-induced atrogin-1/MAFbx expression have not been clarified. Here, we investigated the inhibitory effects of flavonoids on lipopolysaccharide (LPS)-induced atrogin-1/MAFbx expression. We examined whether nine flavonoids belonging to six flavonoid categories inhibited atrogin-1/MAFbx expression in mouse C2C12 myotubes. Two major flavones, apigenin and luteolin, displayed potent inhibitory effects on atrogin-1/MAFbx expression. The pretreatment with apigenin and luteolin significantly prevented the decrease in C2C12 myotube diameter caused by LPS stimulation. Importantly, the pretreatment of LPS-stimulated myoblasts with these flavones significantly inhibited LPS-induced JNK phosphorylation in C2C12 myotubes, resulting in the significant suppression of atrogin-1/MAFbx promoter activity. These results suggest that apigenin and luteolin, prevent LPS-mediated atrogin-1/MAFbx expression through the inhibition of the JNK signaling pathway in C2C12 myotubes. Thus, these flavones, apigenin and luteolin, may be promising agents to prevent LPS-induced muscle atrophy.


Bioscience, Biotechnology, and Biochemistry | 2012

An intracellular fragment of osteoactivin formed by ectodomain shedding translocated to the nucleoplasm and bound to RNA binding proteins.

Kenro Utsunomiya; Kanako Owaki; Yuushi Okumura; Momoko Yano; Takahiro Oto; Eri Suzuki; Seiko Tamura; Tomoki Abe; Shohei Kohno; Ayako Ohno; Katsuya Hirasaka; Shigetada Teshima-Kondoh; Takeshi Nikawa

Osteoactivin is a type I transmembrane protein upregulated by unloading stresses, including denervation, prolonged bed rest, and space flight, but the regulatory mechanisms of its expression and activation under these conditions remain undefined. Here we report that osteoactivin protein exists in two forms: an intact transmembrane form and a secreted form. The secreted form, the extracellular fragment of osteoactivin, was produced by ectodomain shedding and was released into a culture medium. Amino acid sequence analysis of the carboxy-terminal fragment of osteoactivin (OA-CTF) revealed that cleavage of osteoactivin by proteases occurred both at the cell surface and within the cell membrane. Localization analysis demonstrated translocalization of OA-CTF to the nucleus and the endoplasmic reticulum. Moreover, RNA binding proteins, which regulate pre-mRNA splicing, were identified as OA-CTF binding proteins. These results suggest that OA-CTF formed by ectodomain shedding is involved in the regulation of pre-mRNA splicing.


The Journal of Medical Investigation | 2015

Chronic exposure of VEGF inhibitors promotes the malignant phenotype of colorectal cancer cells

Chisato Tomida; Naoko Yamagishi; Kana Aibara; Chiaki Yano; Takayuki Uchida; Tomoki Abe; Ayako Ohno; Katsuya Hirasaka; Takeshi Nikawa; Shigetada Teshima-Kondo

VEGF-targeting anti-angiogenic drugs have enabled significant advances in cancer therapy. However, acquired resistance to VEGF-targeting drugs occurs, leading to disease progression. How tumors become the resistance remains fully uncertain. One of possible mechanisms for the resistance may be the direct effect of VEGF inhibitors on tumor cells expressing VEGF receptors (VEGF-R). We investigated here the direct effect of chronic VEGF inhibition on phenotype changes in cancer cells. To chronically inhibit cancer cell-derived VEGF, human colon cancer HCT116 cells were chronically exposed (3 months) to anti-VEGF neutralizing monoclonal antibody (HCT/mAb cells, blockade of VEGF alone) or VEGF-R tyrosine kinase inhibitor foretinib (HCT/fore cells, blockade of all VEGF family). HCT/mAb cells redundantly increased VEGF family member (VEGF, PlGF, VEGF-B, VEGF-R1 and VEGF-R2) and induced a resistance to hypoxia-induced apoptosis. By contrast, HCT/fore cells did not show the redundant increase in VEGF family member, but significantly increased a VEGF-independent pro-angiogenic factor FGF-2. HCT/fore cells showed increased migration and invasion activities in addition to a resistance to hypoxia-induced apoptosis. The resistance to apoptosis was significantly suppressed by inhibition of hypoxia-inducible factor-1α in HCT/mAb cells, but not in HCT/fore cells. These findings suggest that chronic inhibition of VEGF/VEGF-R accelerates malignant phenotypes of colon cancer cells. J. Med. Invest. 62: 75-79, February, 2015.


Biochimica et Biophysica Acta | 2015

Prevention of skeletal muscle atrophy in vitro using anti-ubiquitination oligopeptide carried by atelocollagen

Nobuhiko Kawai; Katsuya Hirasaka; Tasuku Maeda; Marie Haruna; Chieko Shiota; Arisa Ochi; Tomoki Abe; Shohei Kohno; Ayako Ohno; Sigetada Teshima-Kondo; Hiroyo Mori; Eiji Tanaka; Takeshi Nikawa

Skeletal muscle atrophy occurs when the rate of protein degradation exceeds that of protein synthesis in various catabolic conditions, such as fasting, disuse, aging, and chronic diseases. Insulin-like growth factor-1 (IGF-1) signaling stimulates muscle growth and suppresses muscle protein breakdown. In atrophied muscles, ubiquitin ligase, Cbl-b, increases and stimulates the ubiquitination and degradation of IRS-1, an intermediate in IGF-1 signaling pathway, resulting in IGF-1 resistance. In this study, we evaluated the efficacy of atelocollagen (ATCOL)-transported anti-ubiquitination oligopeptide (Cblin: Cbl-b inhibitor) (consisting of tyrosine phosphorylation domain of IRS-1) in starved C2C12 myotubes. The amount of IRS-1 protein was lower in starved versus unstarved myotubes. The Cblin-ATCOL complex inhibited IRS-1 degradation in a concentration-dependent manner. Myotubes incubated with Cblin-ATCOL complex showed significant resistance to starvation-induced atrophy (p<0.01). Furthermore, the Cblin-ATCOL complex significantly inhibited any decrease in Akt phosphorylation (p<0.01) and localization of FOXO3a to the nucleus in starved myotubes. These results suggest that Cblin prevented starvation-induced C2C12 myotube atrophy by maintaining the IGF-1/Akt/FOXO signaling. Therefore, attachment of anti-ubiquitination oligopeptide, Cblin, to ATCOL enhances its delivery to myotubes and could be a potentially effective strategy in the treatment of atrophic myopathies.


BMC Cell Biology | 2015

A novel myogenic function residing in the 5′ non-coding region of Insulin receptor substrate-1 ( Irs-1 ) transcript

Hikaru Nagano; Naoko Yamagishi; Chisato Tomida; Chiaki Yano; Kana Aibara; Shohei Kohno; Tomoki Abe; Ayako Ohno; Katsuya Hirasaka; Yuushi Okumura; Edward M. Mills; Takeshi Nikawa; Shigetada Teshima-Kondo

BackgroundThere is evidence that several messenger RNAs (mRNAs) are bifunctional RNAs, i.e. RNA transcript carrying both protein-coding capacity and activity as functional non-coding RNA via 5′ and 3′ untranslated regions (UTRs).ResultsIn this study, we identified a novel bifunctional RNA that is transcribed from insulin receptor substrate-1 (Irs-1) gene with full-length 5′UTR sequence (FL-Irs-1 mRNA). FL-Irs-1 mRNA was highly expressed only in skeletal muscle tissue. In cultured skeletal muscle C2C12 cells, the FL-Irs-1 transcript functioned as a bifunctional mRNA. The FL-Irs-1 transcript produced IRS-1 protein during differentiation of myoblasts into myotubes; however, this transcript functioned as a regulatory RNA in proliferating myoblasts. The FL-Irs-1 5′UTR contains a partial complementary sequence to Rb mRNA, which is a critical factor for myogenic differentiation. The overexpression of the 5′UTR markedly reduced Rb mRNA expression, and this reduction was fully dependent on the complementary element and was not compensated by IRS-1 protein. Conversely, knockdown of FL-Irs-1 mRNA increased Rb mRNA expression and enhanced myoblast differentiation into myotubes.ConclusionsOur findings suggest that the FL-Irs-1 transcript regulates myogenic differentiation as a regulatory RNA in myoblasts.


Archives of Biochemistry and Biophysics | 2015

N-myristoylated ubiquitin ligase Cbl-b inhibitor prevents on glucocorticoid-induced atrophy in mouse skeletal muscle.

Arisa Ochi; Tomoki Abe; Reiko Nakao; Yoriko Yamamoto; Kanako Kitahata; Marina Takagi; Katsuya Hirasaka; Ayako Ohno; Shigetada Teshima-Kondo; Gwag Taesik; Inho Choi; Tomoyuki Kawamura; Hisao Nemoto; Rie Mukai; Junji Terao; Takeshi Nikawa

A DGpYMP peptide mimetic of tyrosine(608)-phosphorylated insulin receptor substrate-1 (IRS-1), named Cblin, was previously shown to significantly inhibit Cbl-b-mediated IRS-1 ubiquitination. In the present study, we developed N-myristoylated Cblin and investigated whether it was effective in preventing glucocorticoid-induced muscle atrophy. Using HEK293 cells overexpressing Cbl-b, IRS-1 and ubiquitin, we showed that the 50% inhibitory concentrations of Cbl-b-mediated IRS-1 ubiquitination by N-myristoylated Cblin and Cblin were 30 and 120 μM, respectively. Regarding the DEX-induced atrophy of C2C12 myotubes, N-myristoylated Cblin was more effective than Cblin for inhibiting the DEX-induced decreases in C2C12 myotube diameter and IRS-1 degradation. The inhibitory efficacy of N-myristoylated Cblin on IRS-1 ubiquitination in C2C12 myotubes was approximately fourfold larger than that of Cblin. Furthermore, N-myristoylation increased the incorporation of Cblin into HEK293 cells approximately 10-folds. Finally, we demonstrated that N-myristoylated Cblin prevented the wet weight loss, IRS-1 degradation, and MAFbx/atrogin-1 and MuRF-1 expression in gastrocnemius muscle of DEX-treated mice approximately fourfold more effectively than Cblin. Taken together, these results suggest that N-myristoylated Cblin prevents DEX-induced skeletal muscle atrophy in vitro and in vivo, and that N-myristoylated Cblin more effectively prevents muscle atrophy than unmodified Cblin.


European Journal of Immunology | 2014

Novel CD3‐specific antibody induces immunosuppression via impaired phosphorylation of LAT and PLCγ1 following T‐cell stimulation

Hirokazu Shiheido; Takane Aoyama; Honami Takahashi; Kaori Hanaoka; Tomoki Abe; Emi Nishida; Chen Chen; Orie Koga; Masaki Hikida; Yoshio Shibagaki; Akimichi Morita; Takeshi Nikawa; Seisuke Hattori; Takeshi Watanabe; Jun Shimizu

The activation of T cells is known to be accompanied by the temporary downmodulation of the TCR/CD3 complex on the cell surface. Here, we established a novel monoclonal antibody, Dow2, that temporarily induces downmodulation of the TCR/CD3 complex in mouse CD4+ T cells without activating T cells. Dow2 recognized the determinant on CD3ε; however, differences were observed in the binding mode between Dow2 and the agonistic anti‐CD3ε Ab, 145–2C11. An injection of Dow2 in vivo resulted in T‐cell anergy, and prolonged the survival of cardiac allografts without a marked increase in cytokine release. The phosphorylated forms of the signaling proteins PLC‐γ1 and LAT in Dow2‐induced anergic T cells were markedly decreased upon stimulation. However, the levels of phosphorylated LAT and PLCγ1 in Dow2‐induced anergic T cells could be rescued in the presence of the proteasome inhibitor MG‐132. These results suggest that proteasome‐mediated degradation is involved in hypophosphorylated LAT and PLCγ1 in Dow2‐induced anergic T cells. The novel CD3‐specific Ab, Dow2, may provide us with a unique tool for inducing immunosuppression.

Collaboration


Dive into the Tomoki Abe's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Ayako Ohno

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar

Shohei Kohno

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Junji Terao

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar

Arisa Ochi

University of Tokushima

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge