Network


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

Hotspot


Dive into the research topics where Masaki Kinehara is active.

Publication


Featured researches published by Masaki Kinehara.


PLOS ONE | 2013

Protein Kinase C Regulates Human Pluripotent Stem Cell Self-Renewal

Masaki Kinehara; Suguru Kawamura; Daiki Tateyama; Mika Suga; Hiroko Matsumura; Sumiyo Mimura; Noriko Hirayama; Mitsuhi Hirata; Kozue Uchio-Yamada; Arihiro Kohara; Kana Yanagihara; Miho K. Furue

Background The self-renewal of human pluripotent stem (hPS) cells including embryonic stem and induced pluripotent stem cells have been reported to be supported by various signal pathways. Among them, fibroblast growth factor-2 (FGF-2) appears indispensable to maintain self-renewal of hPS cells. However, downstream signaling of FGF-2 has not yet been clearly understood in hPS cells. Methodology/Principal Findings In this study, we screened a kinase inhibitor library using a high-throughput alkaline phosphatase (ALP) activity-based assay in a minimal growth factor-defined medium to understand FGF-2-related molecular mechanisms regulating self-renewal of hPS cells. We found that in the presence of FGF-2, an inhibitor of protein kinase C (PKC), GF109203X (GFX), increased ALP activity. GFX inhibited FGF-2-induced phosphorylation of glycogen synthase kinase-3β (GSK-3β), suggesting that FGF-2 induced PKC and then PKC inhibited the activity of GSK-3β. Addition of activin A increased phosphorylation of GSK-3β and extracellular signal-regulated kinase-1/2 (ERK-1/2) synergistically with FGF-2 whereas activin A alone did not. GFX negated differentiation of hPS cells induced by the PKC activator, phorbol 12-myristate 13-acetate whereas Gö6976, a selective inhibitor of PKCα, β, and γ isoforms could not counteract the effect of PMA. Intriguingly, functional gene analysis by RNA interference revealed that the phosphorylation of GSK-3β was reduced by siRNA of PKCδ, PKCε, and ζ, the phosphorylation of ERK-1/2 was reduced by siRNA of PKCε and ζ, and the phosphorylation of AKT was reduced by PKCε in hPS cells. Conclusions/Significance Our study suggested complicated cross-talk in hPS cells that FGF-2 induced the phosphorylation of phosphatidylinositol-3 kinase (PI3K)/AKT, mitogen-activated protein kinase/ERK-1/2 kinase (MEK), PKC/ERK-1/2 kinase, and PKC/GSK-3β. Addition of GFX with a MEK inhibitor, U0126, in the presence of FGF-2 and activin A provided a long-term stable undifferentiated state of hPS cells even though hPS cells were dissociated into single cells for passage. This study untangles the cross-talk between molecular mechanisms regulating self-renewal and differentiation of hPS cells.


Scientific Reports | 2016

Parametric analysis of colony morphology of non-labelled live human pluripotent stem cells for cell quality control

Ryuji Kato; Megumi Matsumoto; Hiroto Sasaki; Risako Joto; Mai Okada; Yurika Ikeda; Kei Kanie; Mika Suga; Masaki Kinehara; Kana Yanagihara; Yujung Liu; Kozue Uchio-Yamada; Takayuki Fukuda; Hiroaki Kii; Takayuki Uozumi; Hiroyuki Honda; Yasujiro Kiyota; Miho K. Furue

Given the difficulties inherent in maintaining human pluripotent stem cells (hPSCs) in a healthy state, hPSCs should be routinely characterized using several established standard criteria during expansion for research or therapeutic purposes. hPSC colony morphology is typically considered an important criterion, but it is not evaluated quantitatively. Thus, we designed an unbiased method to evaluate hPSC colony morphology. This method involves a combination of automated non-labelled live-cell imaging and the implementation of morphological colony analysis algorithms with multiple parameters. To validate the utility of the quantitative evaluation method, a parent cell line exhibiting typical embryonic stem cell (ESC)-like morphology and an aberrant hPSC subclone demonstrating unusual colony morphology were used as models. According to statistical colony classification based on morphological parameters, colonies containing readily discernible areas of differentiation constituted a major classification cluster and were distinguishable from typical ESC-like colonies; similar results were obtained via classification based on global gene expression profiles. Thus, the morphological features of hPSC colonies are closely associated with cellular characteristics. Our quantitative evaluation method provides a biological definition of ‘hPSC colony morphology’, permits the non-invasive monitoring of hPSC conditions and is particularly useful for detecting variations in hPSC heterogeneity.


Stem Cells and Development | 2014

Protein Kinase C-Induced Early Growth Response Protein-1 Binding to SNAIL Promoter in Epithelial–Mesenchymal Transition of Human Embryonic Stem Cells

Masaki Kinehara; Suguru Kawamura; Sumiyo Mimura; Mika Suga; Akiko Hamada; Mari Wakabayashi; Hiroki Nikawa; Miho K. Furue

Epithelial-mesenchymal transition (EMT) has been thought to occur during early embryogenesis, and also the differentiation process of human embryonic stem (hES) cells. Spontaneous differentiation is sometimes observed at the peripheral of the hES cell colonies in conventional culture conditions, indicating that EMT occurs in hES cell culture. However, the triggering mechanism of EMT is not yet fully understood. The balance between self-renewal and differentiation of human pluripotent stem (hPS) cells is controlled by various signal pathways, including the fibroblast growth factor (FGF)-2. However, FGF-2 has a complex role for self-renewal of hES cells. FGF-2 activates phosphatidylinositol-3 kinase/AKT, mitogen-activated protein kinase/extracellular signal-regulated kinase-1/2 kinase, and also protein kinase C (PKC). Here, we showed that a PKC rapidly induced an early growth response protein-1 (EGR-1) in hES cells, which was followed by upregulation of EMT-related genes. Before the induction of EMT-related genes, EGR-1 was translocated into the nucleus, and then bound directly to the promoter region of SNAIL, which is a master regulator of EMT. SNAIL expression was attenuated by knockdown of EGR-1, but upregulated by ectopic expression of EGR-1. EGR-1 as the downstream signal of PKC might play a key role in EMT initiation during early differentiation of hES cells. This study would lead to a more robust understanding of the mechanisms underlying the balance between self-renewal and initiation of differentiation in hPS cells.


In Vitro Cellular & Developmental Biology – Animal | 2015

Synergistic effects of FGF-2 and Activin A on early neural differentiation of human pluripotent stem cells.

Sumiyo Mimura; Mika Suga; Yujung Liu; Masaki Kinehara; Kana Yanagihara; Kiyoshi Ohnuma; Hiroki Nikawa; Miho K. Furue

Neural differentiation is an important target of human embryonic stem cells, which provide a source for cell-based therapy, developmental biology, and pharmaceutical research. Previous studies revealed that inhibition of the bone morphogenetic protein is required for neural induction from human embryonic stem cells. On the contrary, the functions of fibroblast growth factors and Activin/Nodal signaling are controversial. Fibroblast growth factor-2 and Activin/Nodal pathways exert divergent influences on human embryonic stem cell concerning the maintenance of both pluripotency and cellular differentiation. We hypothesized that the combination of fibroblast growth factor-2 and Activin A at various concentrations synergistically exerts diverse effects on cell differentiation. To determine the effects of fibroblast growth factor-2 and Activin A on cellular differentiation into neural lineages, we examined the expression of neural differentiation markers in human embryonic stem cells treated with fibroblast growth factor-2 and/or Activin A at various concentrations in a growth factor-defined serum-free medium in short-term culture. In this study, we provide evidence that fibroblast growth factor-2 and Activin A synergistically regulated the initiation of human embryonic stem cell differentiation into neural cell lineages even though human embryonic stem cells autonomously differentiate into neural cell lineages.


The International Journal of Developmental Biology | 2015

Bone morphogenetic protein 4 promotes craniofacial neural crest induction from human pluripotent stem cells

Sumiyo Mimura; Mika Suga; Kaori Okada; Masaki Kinehara; Hiroki Nikawa; Miho K. Furue

Neural crest (NC) cells are a group of cells located in the neural folds at the boundary between the neural and epidermal ectoderm. Cranial NC cells migrate to the branchial arches and give rise to the majority of the craniofacial region, whereas trunk and tail NC cells contribute to the heart, enteric ganglia of the gut, melanocytes, sympathetic ganglia, and adrenal chromaffin cells. Positional information is indispensable for the regulation of cranial or trunk and tail NC cells. However, the mechanisms underlying the regulation of positional information during human NC induction have yet to be fully elucidated. In the present study, supplementation of bone morphogenetic protein (BMP) 4 in defined serum-free culture conditions including fibroblast growth factor-2 and Wnt3a from day 8 after NC specification induced the expression of cranial NC markers, AP2alpha, MSX1, and DLX1, during NC cell differentiation from human pluripotent stem cells. On the other hand, the proportion of cells expressing p75(NTR) or HNK1 decreased compared with that of cells cultured without BMP4, whereas gene expression analysis demonstrated that the expression levels of cranial NC-associated genes increased in BMP4-treated NC cells. These BMP4-treated NC cells were capable of differentiation into osteocytes and chondrocytes. The results of the present study indicate that BMP4 regulates cranial positioning during NC development.


Archive | 2017

DNA and Histone Modifications in Cancer Diagnosis

Masaki Kinehara; Yuki Yamamoto; Yoshitomo Shiroma; Mariko Ikuo; Akira Shimamoto; Hidetoshi Tahara

A number of epigenetic alterations occur during carcinogenesis, and inactivation of tumor suppressor genes is a major determinant of cancer development. Hypermethylation of tumor suppressor genes, histone modification, DNA methylation are major epigenetic alteration in cancers. These alterations may cause a change in gene expression in cells as well as in secretory factors, which include proteins and nucleic acid. Aberrant miRNA expression associated with promoter methylation has been found in body fluids such as plasma and serum, and liquid biopsy can therefore be used to identify significant biomarkers in the early detection of cancer. In addition, telomeres are a key regulator of chromosome stability in cancer. Epigenetic modification by histone methylation is associated with the maintenance of telomere length. This maintenance is essential for cancer to maintain an immortal phenotype. Telomeric repeat-containing RNA (TERRA) is also a regulator of epigenetic modification. In this review, we describe recent advances in our understanding of epigenetic regulation in cancers, including DNA and histone modifications, as well as regulation by non-coding RNAs.


In Vitro Cellular & Developmental Biology – Animal | 2010

Advantages and difficulties in culturing human pluripotent stem cells in growth factor-defined serum-free medium

Miho K. Furue; Daiki Tateyama; Masaki Kinehara; Jie Na; Tetsuji Okamoto; J. Denry Sato


Archive | 2011

METHOD FOR CULTURING HUMAN PLURIPOTENT STEM CELLS

Miho K. Furue; Masaki Kinehara


Biochemical and Biophysical Research Communications | 2018

Levels of tight junction protein CLDND1 are regulated by microRNA-124 in the cerebellum of stroke-prone spontaneously hypertensive rats

Hiroshi Matsuoka; Aki Tamura; Masaki Kinehara; Akiho Shima; Arisa Uda; Hidetoshi Tahara; Akihiro Michihara


The Japanese Biochemical Society/The Molecular Biology Society of Japan | 2017

Development of inhibitor of telomere binding protein using DSE-FRET assay

Yoshitomo Shiroma; Go Fujita; Masaki Kinehara; Mariko Ikuo; Akira Shimamoto; Hidetoshi Tahara

Collaboration


Dive into the Masaki Kinehara'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