Network


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

Hotspot


Dive into the research topics where Mako Ohshima is active.

Publication


Featured researches published by Mako Ohshima.


Journal of the American College of Cardiology | 2009

Ischemic Pre-Conditioning Enhances the Mobilization and Recruitment of Bone Marrow Stem Cells to Protect Against Ischemia/Reperfusion Injury in the Late Phase

Takahiro Kamota; Tao-Sheng Li; Noriyasu Morikage; Masanori Murakami; Mako Ohshima; Masayuki Kubo; Toshiro Kobayashi; Akihito Mikamo; Yasuhiro Ikeda; Masunori Matsuzaki; Kimikazu Hamano

OBJECTIVES The aim of this study was to investigate whether the mobilization and recruitment of bone marrow stem cells (BMSCs) contribute to cardioprotection in the late phase after ischemic pre-conditioning (IPC). BACKGROUND IPC is an innate phenomenon in which brief exposure to sublethal ischemia provides tissue protection from subsequent ischemia/reperfusion (I/R) injury. A delayed cardioprotection also occurs after IPC, but the precise mechanism is unclear. METHODS IPC was created with 4 cycles of 5-min occlusion and reperfusion of the abdominal aorta in mice. Heart I/R injury was induced by occluding the left anterior descending artery for 30 min immediately (early phase) or 24 h (late phase) after IPC. RESULTS Serum vascular endothelial growth factor and stromal cell-derived factor-1alpha levels were increased significantly 1 and 3 h after IPC, but CD34+ and CD34+/flk-1+ stem cells in the peripheral blood were increased significantly 12 and 24 h after IPC (p < 0.05). Compared with the control treatment, both the early and late phases of IPC protected the heart against I/R injury. However, the recruitment of BMSCs was significantly greater in the heart when I/R injury was induced in late phase than in the early phase of IPC (p < 0.01). Interestingly, the blockade of the recruitment of BMSCs significantly attenuated the cardioprotective effect of IPC in the late phase (p < 0.01) but did not change in the early phase. CONCLUSIONS Cardioprotection was observed in the early and late phases of IPC; however, the enhanced mobilization and recruitment of BMSCs played an important role in the late phase of IPC.


Journal of Cellular Physiology | 2009

Increased Expression of CXCR4 and Integrin αM in Hypoxia-Preconditioned Cells Contributes to Improved Cell Retention and Angiogenic Potency

Masayuki Kubo; Tao-Sheng Li; Takahiro Kamota; Mako Ohshima; Shu Lan Qin; Kimikazu Hamano

Cell‐based angiogenesis is a promising method for the treatment of ischemic diseases, but the poor retention of implanted cells in targeted tissues is a major drawback. We tested whether hypoxic preconditioning increased retention and angiogenic potency of implanted cells in ischemic tissue. Hypoxic preconditioning of mouse peripheral blood mononuclear cells (PBMNCs) was done with 24 h of culture under 2% O2. Normoxia‐cultured PBMNCs were used as a control. Hypoxic preconditioning increased the adhesion capacity of the PBMNCs. Moreover, the expression of integrin αM and CXCR4 was significantly higher in the hypoxia‐preconditioned PBMNCs than in the normoxia‐cultured PBMNCs. Interestingly, the expression of intercellular adhesion molecule‐1 (ICAM‐1), a ligand of integrin αM, and stromal cell‐derived factor‐1 (SDF‐1), a chemokine for CXCR4, were remarkably increased in the ischemic hindlimbs. The retention of the hypoxia‐preconditioned PBMNCs was significantly higher than that of the normoxia‐cultured PBMNCs, 3 days after their intramuscular implantation into ischemic hindlimbs. We also noted better blood flow in the ischemic hindlimbs implanted with the hypoxia‐preconditioned PBMNCs than in those implanted with the normoxia‐cultured PBMNCs, 14 days after treatment. Furthermore, antibody neutralization of integrin αM and CXCR4 abolished completely the increased cell retention and angiogenic potency of the hypoxia‐preconditioned PBMNCs after implantation into the ischemic hindlimbs. These results indicate that hypoxic preconditioning of implanted cells is a feasible method of enhancing therapeutic angiogenesis by increasing their retention. J. Cell. Physiol. 220: 508–514, 2009.


PLOS ONE | 2011

The effects of mechanical stress on the growth, differentiation, and paracrine factor production of cardiac stem cells

Hiroshi Kurazumi; Masayuki Kubo; Mako Ohshima; Yumi Yamamoto; Yoshihiro Takemoto; Ryo Suzuki; Shigeru Ikenaga; Akihito Mikamo; Koichi Udo; Kimikazu Hamano; Tao-Sheng Li

Stem cell therapies have been clinically employed to repair the injured heart, and cardiac stem cells are thought to be one of the most potent stem cell candidates. The beating heart is characterized by dynamic mechanical stresses, which may have a significant impact on stem cell therapy. The purpose of this study is to investigate how mechanical stress affects the growth and differentiation of cardiac stem cells and their release of paracrine factors. In this study, human cardiac stem cells were seeded in a silicon chamber and mechanical stress was then induced by cyclic stretch stimulation (60 cycles/min with 120% elongation). Cells grown in non-stretched silicon chambers were used as controls. Our result revealed that mechanical stretching significantly reduced the total number of surviving cells, decreased Ki-67-positive cells, and increased TUNEL-positive cells in the stretched group 24 hrs after stretching, as compared to the control group. Interestingly, mechanical stretching significantly increased the release of the inflammatory cytokines IL-6 and IL-1β as well as the angiogenic growth factors VEGF and bFGF from the cells in 12 hrs. Furthermore, mechanical stretching significantly reduced the percentage of c-kit-positive stem cells, but increased the expressions of cardiac troponin-I and smooth muscle actin in cells 3 days after stretching. Using a traditional stretching model, we demonstrated that mechanical stress suppressed the growth and proliferation of cardiac stem cells, enhanced their release of inflammatory cytokines and angiogenic factors, and improved their myogenic differentiation. The development of this in vitro approach may help elucidate the complex mechanisms of stem cell therapy for heart failure.


Cancer Biology & Therapy | 2010

DNA damage signaling is activated during cancer progression in human colorectal carcinoma

Kazuhito Oka; Toshiki Tanaka; Tadahiko Enoki; Koichi Yoshimura; Mako Ohshima; Masayuki Kubo; Tomoyuki Murakami; Toshikazu Gondou; Yoshihide Minami; Yoshihiro Takemoto; Eijirou Harada; Takaaki Tsushimi; Tao-Sheng Li; Frank Traganos; Zbigniew Darzynkiewicz; Kimikazu Hamano

Purpose: Recent studies have shown that the DNA damage response (DDR) is activated in precancerous lesions, suggesting that neoplastic cells may avoid apoptosis by impairing the DDR which acts as a barrier against tumor progression. To define the role of the DDR pathway in human colorectal carcinoma, we investigated the level of phosphorylated proteins of the DDR pathway. Experimental Design: Colorectal tissue samples were obtained at the time of surgery, from 55 patients at two hospitals. The tissues were classified into four groups according to pathology: normal mucosa, adenoma, early carcinoma and advanced carcinoma. We evaluated phosphorylated ataxia telangiectasia mutated (pATM), phosphorylated H2AX (γH2AX) and Chk2 (pChk2) protein levels by immunohistochemistry and Western blot analysis. We also evaluated apoptosis by the TUNEL assay. Results: Immunostaining for pATM, γH2AX and pChk2 revealed that all were significantly expressed during tumor progression in advanced carcinoma (vs. normal tissue for pATM [p


Circulation | 2009

Identification of Risk Factors Related to Poor Angiogenic Potency of Bone Marrow Cells From Different Patients

Tao-Sheng Li; Masayuki Kubo; Kazuhiro Ueda; Masanori Murakami; Mako Ohshima; Toshiro Kobayashi; Toshiki Tanaka; Bungo Shirasawa; Akihito Mikamo; Kimikazu Hamano

Background— Therapeutic angiogenesis induced by the implantation of autologous bone marrow–derived cells has been used for the treatment of ischemic diseases. However, as the outcomes of cell implantation obviously vary among patients, it is essential to identify patients that would benefit the most from this treatment. Methods and Results— We collected clinical and laboratory data from 25 patients scheduled to undergo sternotomy for various surgical procedures. Then, we aspirated bone marrow cells from the sternum during the operation and investigated the cell quality in vitro by cultivation, and their angiogenic potency in vivo using an ischemic limb model of mice. The angiogenic potency of bone marrow cells differed among patients. Aging, renal failure, anemia, and high serum levels of triglyceride, C-reactive protein, interleukin-6, and type I collagen cross-linked N-telopeptide (NTX) significantly correlated with poor angiogenic potency of bone marrow cells. We assigned scores to these risk factors, and found a strong correlation between the risk scores of patients and the angiogenic potency of their bone marrow cells (r=−0.883, P<0.001). These risk scores can predict the angiogenic potency of bone marrow cells for inducing therapeutic angiogenesis with an accuracy of 80%. Conclusions— We have identified the risk factors related to poor angiogenic potency of bone marrow cells and developed a new scoring system to predict their angiogenic potency for the treatment of ischemic diseases. Our results may help select patients for this treatment in future clinical trials.


Cell Transplantation | 2008

Myocardial Repair Achieved by the Intramyocardial Implantation of Adult Cardiomyocytes in Combination with Bone Marrow Cells

Tao-Sheng Li; Masaya Takahashi; Mako Ohshima; Shu Lan Qin; Masayuki Kubo; Keichi Muramatsu; Kimikazu Hamano

Various cytokines produced by bone marrow cells can protect adult cardiomyocytes against apoptosis. Thus, we investigated the feasibility of implanting adult cardiomyocytes in combination with bone marrow cells for myocardial repair. Ventricular cardiomyocytes were isolated from adult rats and cocultured with bone marrow cells. Using a rat model of doxorubicin-induced cardiomyopathy, we injected 6 × 105 adult cardiomyocytes, 3 × 107 bone marrow cells, or both into damaged hearts, for myocardial repair. Coculture of the cardiomyocytes with the bone marrow cells enhanced the expression of integrin-β1D and focal adhesion kinase in cardiomyocytes, resulting in increased survival and decreased apoptosis of the cardiomyocytes after 7 days of culture. Compared with the baseline levels, cardiac function was preserved by the implantation of bone marrow cells alone and by the implantation of cardiomyocytes in combination with bone marrow cells, but it was decreased significantly 28 days after the implantation of cardiomyocytes alone. Furthermore, apoptosis of the host cardiomyocytes was decreased significantly after the implantation of bone marrow cells alone, or in combination with cardiomyocytes, compared with that after the implantation of cardiomyocytes alone (p < 0.01). Interestingly, the implantation of adult cardiomyocytes in combination with bone marrow cells resulted in a dramatic increase in the survival of donor cardiomyocytes, and induced the myogenic differentiation of donor bone marrow stem cells. Our findings indicate that cardiomyocytes and bone marrow cells can assist and compliment each other; thus, the implantation of adult cardiomyocytes in combination with bone marrow cells shows promise as a feasible new strategy for myocardial repair.


Surgery | 2011

Operative injury accelerates tumor growth by inducing mobilization and recruitment of bone marrow-derived stem cells.

Yoshihiro Takemoto; Tao-Sheng Li; Masayuki Kubo; Mako Ohshima; Kazuhiro Ueda; Eijirou Harada; Tadahiko Enoki; Mariko Okamoto; Yoichi Mizukami; Tomoaki Murata; Kimikazu Hamano

BACKGROUND Although operative injury is thought generally to worsen the prognosis of cancer patients, the relevant mechanisms are not yet understood fully. We tested the hypothesis that operative injury induces mobilization and recruitment of bone marrow stem cells, thereby enhancing angiogenesis and accelerating tumor growth. METHODS Mice were subjected to an open gastrotomy, and naïve mice were used as controls. The mobilization of bone marrow stem cells was monitored after operation. Using an established tumor model in green fluorescent protein (GFP)(+) bone marrow-transplanted chimera mice, we investigated further whether the mobilized stem cells affected tumor growth. RESULTS Compared with the control, gastrotomy increased the populations of CD34(+) cells (6.9 ± 4.5 % vs 3.3 ± 0.4%, P < .05) and CD34(+)/Flk-1(+) cells (0.08 ± 0.02% vs 0.05 ± 0.01%, P < .05) in peripheral blood 12 h after operation. Twelve days after operation, the tumor volume almost doubled in mice after gastrotomy compared with control (580 ± 106 mm(3) vs 299 ± 162 mm(3), P < .05). A histologic analysis of tumor tissue revealed that the microvessel density and number of proliferating cells were significantly greater, but those of apoptotic cells were significantly less, in mice after gastrotomy as compared with control. Furthermore, the number of GFP(+) cells found in tumor tissue was significantly greater in mice that underwent gastrotomy than in controls. Some of the stained GFP(+) cells were positive for CD34 and had been incorporated into microvessels. Administration of AMD3100, which is an antagonist of stromal-cell-derived factor (SDF)-1/CXCR4 signaling pathway, inhibited the recruitment of GFP(+) cells and negated completely the acceleration in tumor growth after operation (345 ± 172 mm(3), P < .05). CONCLUSION Operative injury may induce the mobilization and recruitment of bone marrow stem cells, thereby enhancing angiogenesis and accelerating tumor growth. Inhibition of the SDF-1/CXCR4 signals may represent a new therapeutic strategy for preventing acceleration of tumor growth after operation.


Journal of Vascular Surgery | 2010

Extracorporeal shock wave therapy ameliorates secondary lymphedema by promoting lymphangiogenesis

Masayuki Kubo; Tao-Sheng Li; Takahiro Kamota; Mako Ohshima; Bungo Shirasawa; Kimikazu Hamano

OBJECTIVE Although secondary lymphedema is a common complication after surgical and radiation therapy for cancer, the treatment options for lymphedema remain limited and largely ineffective. We thus studied the effect of extracorporeal shock wave therapy on promoting lymphangiogenesis and improving secondary lymphedema. METHODS A rabbit ear model of lymphedema was created by disruption of lymphatic vessels. Two weeks after surgery, the lymphedematous ear was treated with or without low-energy shock waves (0.09 mJ/mm(2), 200 shots), three times per week for 4 weeks. RESULTS Western blot analysis showed that the expression of vascular endothelial growth factor (VEGF)-C (1.23-fold, P < .05) and VEGF receptor 3 (VEGFR3; 1.53-fold, P < .05) was significantly increased in the ears treated with shock wave than in the untreated lymphedematous ears. Compared with the control group, shock wave treatment led to a significant decrease in the thickness of lymphedematous ears (3.80 +/- 0.25 mm vs 4.54 +/- 0.18 mm, P < .05). Immunohistochemistry for VEGFR3 showed the density of lymphatic vessels was significantly increased by shock wave treatment (P < .05). CONCLUSION Extracorporeal shock wave therapy promotes lymphangiogenesis and ameliorates secondary lymphedema, suggesting that extracorporeal shock wave therapy may be a novel, feasible, effective, and noninvasive treatment for lymphedema.


PLOS ONE | 2011

Diabetic Impairment of C-Kit+ Bone Marrow Stem Cells Involves the Disorders of Inflammatory Factors, Cell Adhesion and Extracellular Matrix Molecules

Tao-Sheng Li; Satoshi Ikeda; Masayuki Kubo; Mako Ohshima; Hiroshi Kurazumi; Yoshihiro Takemoto; Kazuhiro Ueda; Kimikazu Hamano

Bone marrow stem cells from diabetes mellitus patients exhibit functional impairment, but the relative molecular mechanisms responsible for this impairment are poorly understood. We investigated the mechanisms responsible for diabetes-related functional impairment of bone marrow stem cells by extensively screening the expression levels of inflammatory factors, cell cycle regulating molecules, extracellular matrix molecules and adhesion molecules. Bone marrow cells were collected from type 2 diabetic (db/db) and healthy control (db/m+) mice, and c-kit+ stem cells were purified (purity>85%) for experiments. Compared with the healthy control mice, diabetic mice had significantly fewer c-kit+ stem cells, and these cells had a lower potency of endothelial differentiation; however, the production of the angiogenic growth factor VEGF did not differ between groups. A pathway-focused array showed that the c-kit+ stem cells from diabetic mice had up-regulated expression levels of many inflammatory factors, including Tlr4, Cxcl9, Il9, Tgfb1, Il4, and Tnfsf5, but no obvious change in the expression levels of cell cycle molecules. Interestingly, diabetes-related alterations of the extracellular matrix and adhesion molecules were varied; Pecam, Mmp10, Lamc1, Itgb7, Mmp9, and Timp4 were up-regulated, but Col11a1, Fn1, Admts2, and Itgav were down-regulated. Some of these changes were also confirmed at the protein level by flow cytometry analysis. In conclusion, c-kit+ bone marrow stem cells from diabetic mice exhibited an extensive enhancement of inflammatory factors and disorders of the extracellular matrix and adhesion molecules. Further intervention studies are required to determine the precise role of each molecule in the diabetes-related functional impairment of c-kit+ bone marrow stem cells.


PLOS ONE | 2012

Heat shock factor 1 contributes to ischemia-induced angiogenesis by regulating the mobilization and recruitment of bone marrow stem/progenitor cells.

Masayuki Kubo; Tao-Sheng Li; Hiroshi Kurazumi; Yoshihiro Takemoto; Mako Ohshima; Yumi Yamamoto; Arata Nishimoto; Akihito Mikamo; Mitsuaki Fujimoto; Akira Nakai; Kimikazu Hamano

Bone marrow (BM)-derived stem/progenitor cells play an important role in ischemia-induced angiogenesis in cardiovascular diseases. Heat shock factor 1 (HSF1) is known to be induced in response to hypoxia and ischemia. We examined whether HSF1 contributes to ischemia-induced angiogenesis through the mobilization and recruitment of BM-derived stem/progenitor cells using HSF1-knockout (KO) mice. After the induction of ischemia, blood flow and microvessel density in the ischemic hindlimb were significantly lower in the HSF1-KO mice than in the wild-type (WT) mice. The mobilization of BM-derived Sca-1- and c-kit-positive cells in peripheral blood after ischemia was significantly lower in the HSF1-KO mice than in the WT mice. BM stem/progenitor cells from HSF1-KO mice showed a significant decrease in their recruitment to ischemic tissue and in migration, adhesion, and survival when compared with WT mice. Blood flow recovery in the ischemic hindlimb significantly decreased in WT mice receiving BM reconstitution with donor cells from HSF1-KO mice. Conversely, blood flow recovery in the ischemic hindlimb significantly increased in HSF1-KO mice receiving BM reconstitution with donor cells from WT mice. These findings suggest that HSF1 contributes to ischemia-induced angiogenesis by regulating the mobilization and recruitment of BM-derived stem/progenitor cells.

Collaboration


Dive into the Mako Ohshima'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