Network


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

Hotspot


Dive into the research topics where Yohei Hayashi is active.

Publication


Featured researches published by Yohei Hayashi.


Cell | 2010

Direct Reprogramming of Fibroblasts into Functional Cardiomyocytes by Defined Factors

Masaki Ieda; Ji Dong Fu; Paul Delgado-Olguin; Vasanth Vedantham; Yohei Hayashi; Benoit G. Bruneau; Deepak Srivastava

The reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs) raises the possibility that a somatic cell could be reprogrammed to an alternative differentiated fate without first becoming a stem/progenitor cell. A large pool of fibroblasts exists in the postnatal heart, yet no single master regulator of direct cardiac reprogramming has been identified. Here, we report that a combination of three developmental transcription factors (i.e., Gata4, Mef2c, and Tbx5) rapidly and efficiently reprogrammed postnatal cardiac or dermal fibroblasts directly into differentiated cardiomyocyte-like cells. Induced cardiomyocytes expressed cardiac-specific markers, had a global gene expression profile similar to cardiomyocytes, and contracted spontaneously. Fibroblasts transplanted into mouse hearts one day after transduction of the three factors also differentiated into cardiomyocyte-like cells. We believe these findings demonstrate that functional cardiomyocytes can be directly reprogrammed from differentiated somatic cells by defined factors. Reprogramming of endogenous or explanted fibroblasts might provide a source of cardiomyocytes for regenerative approaches.The reprogramming of fibroblasts to induced pluripotent stem cells (iPSCs) raises the possibility that a somatic cell could be reprogrammed to an alternative differentiated fate without first becoming a stem/progenitor cell. A large pool of fibroblasts exists in the postnatal heart, yet no single master regulator of direct cardiac reprogramming has been identified. Here, we report that a combination of three developmental transcription factors (i.e., Gata4, Mef2c, and Tbx5) rapidly and efficiently reprogrammed postnatal cardiac or dermal fibroblasts directly into differentiated cardiomyocyte-like cells. Induced cardiomyocytes expressed cardiac-specific markers, had a global gene expression profile similar to cardiomyocytes, and contracted spontaneously. Fibroblasts transplanted into mouse hearts one day after transduction of the three factors also differentiated into cardiomyocyte-like cells. We believe these findings demonstrate that functional cardiomyocytes can be directly reprogrammed from differentiated somatic cells by defined factors. Reprogramming of endogenous or explanted fibroblasts might provide a source of cardiomyocytes for regenerative approaches.


Stem cell reports | 2013

Direct Reprogramming of Human Fibroblasts toward a Cardiomyocyte-like State

Ji Dong Fu; Nicole R. Stone; Lei Liu; C. Ian Spencer; Li Qian; Yohei Hayashi; Paul Delgado-Olguin; Sheng Ding; Benoit G. Bruneau; Deepak Srivastava

Summary Direct reprogramming of adult somatic cells into alternative cell types has been shown for several lineages. We previously showed that GATA4, MEF2C, and TBX5 (GMT) directly reprogrammed nonmyocyte mouse heart cells into induced cardiomyocyte-like cells (iCMs) in vitro and in vivo. However, GMT alone appears insufficient in human fibroblasts, at least in vitro. Here, we show that GMT plus ESRRG and MESP1 induced global cardiac gene-expression and phenotypic shifts in human fibroblasts derived from embryonic stem cells, fetal heart, and neonatal skin. Adding Myocardin and ZFPM2 enhanced reprogramming, including sarcomere formation, calcium transients, and action potentials, although the efficiency remained low. Human iCM reprogramming was epigenetically stable. Furthermore, we found that transforming growth factor β signaling was important for, and improved the efficiency of, human iCM reprogramming. These findings demonstrate that human fibroblasts can be directly reprogrammed toward the cardiac lineage, and lay the foundation for future refinements in vitro and in vivo.


Cell Stem Cell | 2014

The let-7/LIN-41 Pathway Regulates Reprogramming to Human Induced Pluripotent Stem Cells by Controlling Expression of Prodifferentiation Genes

Kathleen A. Worringer; Tim A. Rand; Yohei Hayashi; Salma Sami; Kazutoshi Takahashi; Koji Tanabe; Megumi Narita; Deepak Srivastava; Shinya Yamanaka

Reprogramming differentiated cells into induced pluripotent stem cells (iPSCs) promotes a broad array of cellular changes. Here we show that the let-7 family of microRNAs acts as an inhibitory influence on the reprogramming process through a regulatory pathway involving prodifferentiation factors, including EGR1. Inhibiting let-7 in human cells promotes reprogramming to a comparable extent to c-MYC when combined with OCT4, SOX2, and KLF4, and persistence of let-7 inhibits reprogramming. Inhibiting let-7 during reprogramming leads to an increase in the level of the let-7 target LIN-41/TRIM71, which in turn promotes reprogramming and is important for overcoming the let-7 barrier to reprogramming. Mechanistic studies revealed that LIN-41 regulates a broad array of differentiation genes, and more specifically, inhibits translation of EGR1 through binding its cognate mRNA. Together our findings outline a let-7-based pathway that counteracts the activity of reprogramming factors through promoting the expression of prodifferentiation genes.


Orphanet Journal of Rare Diseases | 2013

Induced pluripotent stem cells from patients with human fibrodysplasia ossificans progressiva show increased mineralization and cartilage formation

Yoshihisa Matsumoto; Yohei Hayashi; Christopher R. Schlieve; Makoto Ikeya; Hannah Kim; Trieu Nguyen; Salma Sami; Shiro Baba; Emilie Barruet; Akira Nasu; Isao Asaka; Takanobu Otsuka; Shinya Yamanaka; Bruce R. Conklin; Junya Toguchida; Edward C. Hsiao

BackgroundAbnormal activation of endochondral bone formation in soft tissues causes significant medical diseases associated with disability and pain. Hyperactive mutations in the bone morphogenetic protein (BMP) type 1 receptor ACVR1 lead to fibrodysplasia ossificans progressiva (FOP), a rare genetic disorder characterized by progressive ossification in soft tissues. However, the specific cellular mechanisms are unclear. In addition, the difficulty obtaining tissue samples from FOP patients and the limitations in mouse models of FOP hamper our ability to dissect the pathogenesis of FOP.MethodsTo address these challenges and develop a “disease model in a dish”, we created human induced pluripotent stem cells (iPS cells) derived from normal and FOP dermal fibroblasts by two separate methods, retroviral integration or integration-free episomal vectors. We tested if the ability to contribute to different steps of endochondral bone formation was different in FOP vs. control iPS cells.ResultsRemarkably, FOP iPS cells showed increased mineralization and enhanced chondrogenesis in vitro. The mineralization phenotypes could be suppressed with a small-molecule inhibitor of BMP signaling, DMH1. Our results indicate that the FOP ACVR1 R206H mutation favors chondrogenesis and increases mineral deposition in vitro.ConclusionsOur findings establish a FOP disease cell model for in vitro experimentation and provide a proof-of-concept for using human iPS cell models to understand human skeletal disorders.


Stem cell reports | 2014

Calcium Transients Closely Reflect Prolonged Action Potentials in iPSC Models of Inherited Cardiac Arrhythmia

C. Ian Spencer; Shiro Baba; Kenta Nakamura; Ethan A. Hua; Marie A.F. Sears; Chi-cheng Fu; Jianhua Zhang; Sadguna Y. Balijepalli; Kiichiro Tomoda; Yohei Hayashi; Paweena Lizarraga; Julianne Wojciak; Melvin M. Scheinman; Katriina Aalto-Setälä; Jonathan C. Makielski; Craig T. January; Kevin E. Healy; Timothy J. Kamp; Shinya Yamanaka; Bruce R. Conklin

Summary Long-QT syndrome mutations can cause syncope and sudden death by prolonging the cardiac action potential (AP). Ion channels affected by mutations are various, and the influences of cellular calcium cycling on LQTS cardiac events are unknown. To better understand LQTS arrhythmias, we performed current-clamp and intracellular calcium ([Ca2+]i) measurements on cardiomyocytes differentiated from patient-derived induced pluripotent stem cells (iPS-CM). In myocytes carrying an LQT2 mutation (HERG-A422T), APs and [Ca2+]i transients were prolonged in parallel. APs were abbreviated by nifedipine exposure and further lengthened upon releasing intracellularly stored Ca2+. Validating this model, control iPS-CM treated with HERG-blocking drugs recapitulated the LQT2 phenotype. In LQT3 iPS-CM, expressing NaV1.5-N406K, APs and [Ca2+]i transients were markedly prolonged. AP prolongation was sensitive to tetrodotoxin and to inhibiting Na+-Ca2+ exchange. These results suggest that LQTS mutations act partly on cytosolic Ca2+ cycling, potentially providing a basis for functionally targeted interventions regardless of the specific mutation site.


Nature | 2014

Cell-autonomous correction of ring chromosomes in human induced pluripotent stem cells

Marina Bershteyn; Yohei Hayashi; Guillaume Desachy; Edward C. Hsiao; Salma Sami; Kathryn Tsang; Lauren A. Weiss; Arnold R. Kriegstein; Shinya Yamanaka; Anthony Wynshaw-Boris

Ring chromosomes are structural aberrations commonly associated with birth defects, mental disabilities and growth retardation. Rings form after fusion of the long and short arms of a chromosome, and are sometimes associated with large terminal deletions. Owing to the severity of these large aberrations that can affect multiple contiguous genes, no possible therapeutic strategies for ring chromosome disorders have been proposed. During cell division, ring chromosomes can exhibit unstable behaviour leading to continuous production of aneuploid progeny with low viability and high cellular death rate. The overall consequences of this chromosomal instability have been largely unexplored in experimental model systems. Here we generated human induced pluripotent stem cells (iPSCs) from patient fibroblasts containing ring chromosomes with large deletions and found that reprogrammed cells lost the abnormal chromosome and duplicated the wild-type homologue through the compensatory uniparental disomy (UPD) mechanism. The karyotypically normal iPSCs with isodisomy for the corrected chromosome outgrew co-existing aneuploid populations, enabling rapid and efficient isolation of patient-derived iPSCs devoid of the original chromosomal aberration. Our results suggest a fundamentally different function for cellular reprogramming as a means of ‘chromosome therapy’ to reverse combined loss-of-function across many genes in cells with large-scale aberrations involving ring structures. In addition, our work provides an experimentally tractable human cellular system for studying mechanisms of chromosomal number control, which is of critical relevance to human development and disease.


Proceedings of the National Academy of Sciences of the United States of America | 2016

BMP-SMAD-ID promotes reprogramming to pluripotency by inhibiting p16/INK4A-dependent senescence

Yohei Hayashi; Edward C. Hsiao; Salma Sami; Mariselle Lancero; Christopher R. Schlieve; Trieu Nguyen; Koyori Yano; Ayako Nagahashi; Makoto Ikeya; Yoshihisa Matsumoto; Ken Nishimura; Aya Fukuda; Koji Hisatake; Kiichiro Tomoda; Isao Asaka; Junya Toguchida; Bruce R. Conklin; Shinya Yamanaka

Significance The development of iPSCs provides unprecedented opportunities for life sciences, drug discovery, and regenerative medicine. The efficiency of iPSC generation is quite low: typically less than 1% of human primary somatic cells that have received reprogramming factors turn into iPSCs. Previous studies revealed that cellular senescence was a major barrier to iPSC generation. In this study using human FOP mutant cells, we provide evidence that the BMP-SMAD-ID signaling suppressed p16/INK4A-mediated cellular senescence during the early phase in iPSC generation. These results are unexpected because BMP-SMAD signaling has negative effects on the self-renewal of human iPSCs. Here, we show that a human natural mutation increases the efficiency of iPSC generation. Fibrodysplasia ossificans progressiva (FOP) patients carry a missense mutation in ACVR1 [617G > A (R206H)] that leads to hyperactivation of BMP-SMAD signaling. Contrary to a previous study, here we show that FOP fibroblasts showed an increased efficiency of induced pluripotent stem cell (iPSC) generation. This positive effect was attenuated by inhibitors of BMP-SMAD signaling (Dorsomorphin or LDN1931890) or transducing inhibitory SMADs (SMAD6 or SMAD7). In normal fibroblasts, the efficiency of iPSC generation was enhanced by transducing mutant ACVR1 (617G > A) or SMAD1 or adding BMP4 protein at early times during the reprogramming. In contrast, adding BMP4 at later times decreased iPSC generation. ID genes, transcriptional targets of BMP-SMAD signaling, were critical for iPSC generation. The BMP-SMAD-ID signaling axis suppressed p16/INK4A-mediated cell senescence, a major barrier to reprogramming. These results using patient cells carrying the ACVR1 R206H mutation reveal how cellular signaling and gene expression change during the reprogramming processes.


Stem Cells International | 2016

Biological Effects of Culture Substrates on Human Pluripotent Stem Cells

Yohei Hayashi; Miho K. Furue

In recent years, as human pluripotent stem cells (hPSCs) have been commonly cultured in feeder-free conditions, a number of cell culture substrates have been applied or developed. However, the functional roles of these substrates in maintaining hPSC self-renewal remain unclear. Here in this review, we summarize the types of these substrates and their effect on maintaining hPSC self-renewal. Endogenous extracellular matrix (ECM) protein expression has been shown to be crucial in maintaining hPSC self-renewal. These ECM molecules interact with integrin cell-surface receptors and transmit their cellular signaling. We discuss the possible effect of integrin-mediated signaling pathways on maintaining hPSC self-renewal. Activation of integrin-linked kinase (ILK), which transmits ECM-integrin signaling to AKT (also known as protein kinase B), has been shown to be critical in maintaining hPSC self-renewal. Also, since naïve pluripotency has been widely recognized as an alternative pluripotent state of hPSCs, we discuss the possible effects of culture substrates and integrin signaling on naïve hPSCs based on the studies of mouse embryonic stem cells. Understanding the role of culture substrates in hPSC self-renewal and differentiation enables us to control hPSC behavior precisely and to establish scalable or microfabricated culture technologies for regenerative medicine and drug development.


Stem cell reports | 2017

A Role for KLF4 in Promoting the Metabolic Shift via TCL1 during Induced Pluripotent Stem Cell Generation

Ken Nishimura; Shiho Aizawa; Fransiska Liliani Nugroho; Emi Shiomitsu; Yen Thi Hai Tran; Phuong Linh Bui; Evgeniia Borisova; Yuta Sakuragi; Hitomi Takada; Akira Kurisaki; Yohei Hayashi; Aya Fukuda; Mahito Nakanishi; Koji Hisatake

Summary Reprogramming of somatic cells into induced pluripotent stem cells (iPSCs) is accompanied by morphological, functional, and metabolic alterations before acquisition of full pluripotency. Although the genome-wide effects of the reprogramming factors on gene expression are well documented, precise mechanisms by which gene expression changes evoke phenotypic responses remain to be determined. We used a Sendai virus-based system that permits reprogramming to progress in a strictly KLF4-dependent manner to screen for KLF4 target genes that are critical for the progression of reprogramming. The screening identified Tcl1 as a critical target gene that directs the metabolic shift from oxidative phosphorylation to glycolysis. KLF4-induced TCL1 employs a two-pronged mechanism, whereby TCL1 activates AKT to enhance glycolysis and counteracts PnPase to diminish oxidative phosphorylation. These regulatory mechanisms described here highlight a central role for a reprogramming factor in orchestrating the metabolic shift toward the acquisition of pluripotency during iPSC generation.


Oral Diseases | 2017

In Vitro Models of Cranial Neural Crest Development toward Toxicity Tests: Frog, Mouse and Human

Mika Suga; Yohei Hayashi; Miho K. Furue

During craniofacial development, cranial neural crest (NC)-derived mesenchymal cells migrate to pharyngeal arches and contribute extensively to neurons, Schwann cells, smooth muscle cells, osteoblasts, chondrocytes, and odontoblasts, forming maxillofacial structures. In vitro models using model organism cells, such as African clawed frog (Xenopus Laevis) and mouse (Mus Musculus), were developed to understand cellular and molecular mechanisms of cranial NC development. Recent studies using human embryonic stem cells (hESCs) and human-induced pluripotent stem cells (hiPSCs) have enabled the generation of human NC cells (NCCs) inxa0vitro to provide insight into human NC development. Understanding molecular mechanisms underlying craniofacial development will contribute to develop novel embryotoxicity tests and to decrease the incidence of drug-induced congenital anomalies in the craniofacial region, such as cleft lip or cleft palate. Here, we review culture methods to derive NCCs inxa0vitro from Xenopus presumptive ectoderm (animal caps), mouse embryonic stem cells (mESCs), and human pluripotent stem cells (hPSCs) and discuss how these inxa0vitro models can be used to help clarify the mechanisms underlying craniofacial development and for developing embryotoxicity tests predicting drug-induced congenital anomalies in the craniofacial region.

Collaboration


Dive into the Yohei Hayashi's collaboration.

Top Co-Authors

Avatar

Aya Fukuda

Saitama Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Salma Sami

University of California

View shared research outputs
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