Network


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

Hotspot


Dive into the research topics where Emi Aizawa is active.

Publication


Featured researches published by Emi Aizawa.


Nature | 2016

In vivo genome editing via CRISPR/Cas9 mediated homology-independent targeted integration

Keiichiro Suzuki; Yuji Tsunekawa; Reyna Hernández-Benítez; Jun Wu; Jie Zhu; Euiseok J. Kim; Fumiyuki Hatanaka; Mako Yamamoto; Toshikazu Araoka; Zhe Li; Masakazu Kurita; Tomoaki Hishida; Mo Li; Emi Aizawa; Shicheng Guo; Song Chen; April Goebl; Rupa Devi Soligalla; Jing Qu; Tingshuai Jiang; Xin Fu; Maryam Jafari; Concepcion Rodriguez Esteban; W. Travis Berggren; Jeronimo Lajara; Estrella Núñez-Delicado; Pedro Guillen; Josep M. Campistol; Fumio Matsuzaki; Guang-Hui Liu

Targeted genome editing via engineered nucleases is an exciting area of biomedical research and holds potential for clinical applications. Despite rapid advances in the field, in vivo targeted transgene integration is still infeasible because current tools are inefficient, especially for non-dividing cells, which compose most adult tissues. This poses a barrier for uncovering fundamental biological principles and developing treatments for a broad range of genetic disorders. Based on clustered regularly interspaced short palindromic repeat/Cas9 (CRISPR/Cas9) technology, here we devise a homology-independent targeted integration (HITI) strategy, which allows for robust DNA knock-in in both dividing and non-dividing cells in vitro and, more importantly, in vivo (for example, in neurons of postnatal mammals). As a proof of concept of its therapeutic potential, we demonstrate the efficacy of HITI in improving visual function using a rat model of the retinal degeneration condition retinitis pigmentosa. The HITI method presented here establishes new avenues for basic research and targeted gene therapies.


Science | 2015

Aging stem cells. A Werner syndrome stem cell model unveils heterochromatin alterations as a driver of human aging.

Weiqi Zhang; Jingyi Li; Keiichiro Suzuki; Jing Qu; Ping Wang; J. Zhou; Xiaomeng Liu; Ruotong Ren; Xiuling Xu; Alejandro Ocampo; Tingting Yuan; Jiping Yang; Ying Li; Liang Shi; Dee Guan; Huize Pan; Shunlei Duan; Zhichao Ding; Mo Li; Fei Yi; Yayu Wang; Chang Chen; Fuquan Yang; Xiaoyu Li; Zimei Wang; Emi Aizawa; April Goebl; Rupa Devi Soligalla; Pradeep Reddy; Concepcion Rodriguez Esteban

Heterochromatin in aging stem cells Analysis of human aging syndromes, such as Werner syndrome (WS), may lead to greater understanding of both premature and normal aging. Zhang et al. generated isogenic WS-specific human embryonic stem cell lines (see the Perspective by Brunauer and Kennedy). WS-mesenchymal stem cells displayed features characteristic of premature aging, including heterochromatin disorganization. WRN protein thus functions in the maintenance of heterochromatin, and heterochromatin alterations may represent a driving force of human aging. Science, this issue p. 1160; see also p. 1093 Stabilization of heterochromatin by WRN protein safeguards human mesenchymal stem cells from aging. [Also see Perspective by Brunauer and Kennedy] Werner syndrome (WS) is a premature aging disorder caused by WRN protein deficiency. Here, we report on the generation of a human WS model in human embryonic stem cells (ESCs). Differentiation of WRN-null ESCs to mesenchymal stem cells (MSCs) recapitulates features of premature cellular aging, a global loss of H3K9me3, and changes in heterochromatin architecture. We show that WRN associates with heterochromatin proteins SUV39H1 and HP1α and nuclear lamina–heterochromatin anchoring protein LAP2β. Targeted knock-in of catalytically inactive SUV39H1 in wild-type MSCs recapitulates accelerated cellular senescence, resembling WRN-deficient MSCs. Moreover, decrease in WRN and heterochromatin marks are detected in MSCs from older individuals. Our observations uncover a role for WRN in maintaining heterochromatin stability and highlight heterochromatin disorganization as a potential determinant of human aging.


Cell Stem Cell | 2014

Targeted Gene Correction Minimally Impacts Whole-Genome Mutational Load in Human-Disease-Specific Induced Pluripotent Stem Cell Clones

Keiichiro Suzuki; Chang Yu; Jing Qu; Mo Li; Xiaotian Yao; Tingting Yuan; April Goebl; Senwei Tang; Ruotong Ren; Emi Aizawa; Fan Zhang; Xiuling Xu; Rupa Devi Soligalla; Feng Chen; Jessica Kim; Na Young Kim; Hsin-Kai Liao; Christopher Benner; Concepcion Rodriguez Esteban; Yabin Jin; Guang-Hui Liu; Yingrui Li; Juan Carlos Izpisua Belmonte

The utility of genome editing technologies for disease modeling and developing cellular therapies has been extensively documented, but the impact of these technologies on mutational load at the whole-genome level remains unclear. We performed whole-genome sequencing to evaluate the mutational load at single-base resolution in individual gene-corrected human induced pluripotent stem cell (hiPSC) clones in three different disease models. In single-cell clones, gene correction by helper-dependent adenoviral vector (HDAdV) or Transcription Activator-Like Effector Nuclease (TALEN) exhibited few off-target effects and a low level of sequence variation, comparable to that accumulated in routine hiPSC culture. The sequence variants were randomly distributed and unique to individual clones. We also combined both technologies and developed a TALEN-HDAdV hybrid vector, which significantly increased gene-correction efficiency in hiPSCs. Therefore, with careful monitoring via whole-genome sequencing it is possible to apply genome editing to human pluripotent cells with minimal impact on genomic mutational load.


Nature | 2015

An alternative pluripotent state confers interspecies chimaeric competency

Jun Wu; Daiji Okamura; Mo Li; Keiichiro Suzuki; Chongyuan Luo; Li Ma; Yupeng He; Zhongwei Li; Christopher Benner; Isao Tamura; Marie N. Krause; Joseph R. Nery; Tingting Du; Zhuzhu Zhang; Tomoaki Hishida; Yuta Takahashi; Emi Aizawa; Na Young Kim; Jeronimo Lajara; Pedro Guillen; Josep M. Campistol; Concepcion Rodriguez Esteban; Pablo J. Ross; Alan Saghatelian; Bing Ren; Joseph R. Ecker; Juan Carlos Izpisua Belmonte

Pluripotency, the ability to generate any cell type of the body, is an evanescent attribute of embryonic cells. Transitory pluripotent cells can be captured at different time points during embryogenesis and maintained as embryonic stem cells or epiblast stem cells in culture. Since ontogenesis is a dynamic process in both space and time, it seems counterintuitive that these two temporal states represent the full spectrum of organismal pluripotency. Here we show that by modulating culture parameters, a stem-cell type with unique spatial characteristics and distinct molecular and functional features, designated as region-selective pluripotent stem cells (rsPSCs), can be efficiently obtained from mouse embryos and primate pluripotent stem cells, including humans. The ease of culturing and editing the genome of human rsPSCs offers advantages for regenerative medicine applications. The unique ability of human rsPSCs to generate post-implantation interspecies chimaeric embryos may facilitate our understanding of early human development and evolution.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Highly efficient transient gene expression and gene targeting in primate embryonic stem cells with helper-dependent adenoviral vectors

Keiichiro Suzuki; Kaoru Mitsui; Emi Aizawa; Kouichi Hasegawa; Eihachiro Kawase; Toshiyuki Yamagishi; Yoshihiko Shimizu; Hirofumi Suemori; Norio Nakatsuji; Kohnosuke Mitani

Human embryonic stem (hES) cells are regarded as a potentially unlimited source of cellular materials for regenerative medicine. For biological studies and clinical applications using primate ES cells, the development of a general strategy to obtain efficient gene delivery and genetic manipulation, especially gene targeting via homologous recombination (HR), would be of paramount importance. However, unlike mouse ES (mES) cells, efficient strategies for transient gene delivery and HR in hES cells have not been established. Here, we report that helper-dependent adenoviral vectors (HDAdVs) were able to transfer genes in hES and cynomolgus monkey (Macaca fasicularis) ES (cES) cells efficiently. Without losing the undifferentiated state of the ES cells, transient gene transfer efficiency was ≈100%. Using HDAdVs with homology arms, approximately one out of 10 chromosomal integrations of the vector was via HR, whereas the rate was only ≈1% with other gene delivery methods. Furthermore, in combination with negative selection, ≈45% of chromosomal integrations of the vector were targeted integrations, indicating that HDAdVs would be a powerful tool for genetic manipulation in hES cells and potentially in other types of human stem cells, such as induced pluripotent stem (iPS) cells.


Cell | 2017

Interspecies Chimerism with Mammalian Pluripotent Stem Cells

Jun Wu; Aida Platero-Luengo; Masahiro Sakurai; Atsushi Sugawara; M.A. Gil; Takayoshi Yamauchi; Keiichiro Suzuki; Y. S. Bogliotti; C. Cuello; Mariana Morales Valencia; Daiji Okumura; Jingping Luo; Marcela Vilarino; I. Parrilla; Delia Alba Soto; Cristina A. Martinez; Tomoaki Hishida; Sonia Sánchez-Bautista; M. Llanos Martinez-Martinez; Huili Wang; A. Nohalez; Emi Aizawa; Paloma Martínez-Redondo; Alejandro Ocampo; Pradeep Reddy; Jordi Roca; Elizabeth A. Maga; Concepcion Rodriguez Esteban; W. Travis Berggren; Estrella Nuñez Delicado

Interspecies blastocyst complementation enables organ-specific enrichment of xenogenic pluripotent stem cell (PSC) derivatives. Here, we establish a versatile blastocyst complementation platform based on CRISPR-Cas9-mediated zygote genome editing and show enrichment of rat PSC-derivatives in several tissues of gene-edited organogenesis-disabled mice. Besides gaining insights into species evolution, embryogenesis, and human disease, interspecies blastocyst complementation might allow human organ generation in animals whose organ size, anatomy, and physiology are closer to humans. To date, however, whether human PSCs (hPSCs) can contribute to chimera formation in non-rodent species remains unknown. We systematically evaluate the chimeric competency of several types of hPSCs using a more diversified clade of mammals, the ungulates. We find that naïve hPSCs robustly engraft in both pig and cattle pre-implantation blastocysts but show limited contribution to post-implantation pig embryos. Instead, an intermediate hPSC type exhibits higher degree of chimerism and is able to generate differentiated progenies in post-implantation pig embryos.


Nature Communications | 2014

Modelling Fanconi anemia pathogenesis and therapeutics using integration-free patient-derived iPSCs

Guang Hui Liu; Keiichiro Suzuki; Mo Li; Jing Qu; Nuria Montserrat; Carolina Tarantino; Ying Gu; Fei Yi; Xiuling Xu; Weiqi Zhang; Sergio Ruiz; Nongluk Plongthongkum; Kun Zhang; Shigeo Masuda; Emmanuel Nivet; Yuji Tsunekawa; Rupa Devi Soligalla; April Goebl; Emi Aizawa; Na Young Kim; Jessica Kim; Ilir Dubova; Ying Li; Ruotong Ren; Christopher Benner; Antonio del Sol; Juan A. Bueren; Juan P. Trujillo; Jordi Surrallés; Enrico Cappelli

Fanconi anaemia (FA) is a recessive disorder characterized by genomic instability, congenital abnormalities, cancer predisposition and bone marrow (BM) failure. However, the pathogenesis of FA is not fully understood partly due to the limitations of current disease models. Here, we derive integration free-induced pluripotent stem cells (iPSCs) from an FA patient without genetic complementation and report in situ gene correction in FA-iPSCs as well as the generation of isogenic FANCA-deficient human embryonic stem cell (ESC) lines. FA cellular phenotypes are recapitulated in iPSCs/ESCs and their adult stem/progenitor cell derivatives. By using isogenic pathogenic mutation-free controls as well as cellular and genomic tools, our model serves to facilitate the discovery of novel disease features. We validate our model as a drug-screening platform by identifying several compounds that improve hematopoietic differentiation of FA-iPSCs. These compounds are also able to rescue the hematopoietic phenotype of FA patient BM cells.


Biochemical and Biophysical Research Communications | 2009

Gene targeting in human pluripotent stem cells with adeno-associated virus vectors

Kaoru Mitsui; Keiichiro Suzuki; Emi Aizawa; Eihachiro Kawase; Hirofumi Suemori; Norio Nakatsuji; Kohnosuke Mitani

Human pluripotent stem cells, such as embryonic stem cells (hESCs) and induced pluripotent stem cells (hiPSCs), have the ability to differentiate into various cell types, and will become a potential source of cellular materials for regenerative medicine. To make full use of hESCs or hiPSCs for both basic and clinical research, genetic modification, especially gene targeting via homologous recombination (HR), would be an essential technique. This report describes the successful gene targeting of the hypoxanthine phosphoribosyl transferase 1 (HPRT1) and the NANOG loci in human pluripotent stem cells with adeno-associated virus (AAV) vectors. At the HPRT1 locus, up to 1% of stable transformants were targeted via HR with an AAV-HPRT1 targeting vector, without loss of pluripotency. On the other hand, 20-87% of stable transformants were targeted using an AAV-NANOG-targeting vector designed for the promoter-trap strategy. In the KhES-3 cell line, which shows particularly high fragility to experimental manipulation, gene targeting was successful only by using an AAV vector but not by electroporation. In addition to hESC, gene targeting was achieved in hiPSC lines at similar frequencies. These data indicate that AAV vectors may therefore be a useful tool to introduce genetic modifications in hESCs and hiPSCs.


Molecular Therapy | 2012

Efficient and Accurate Homologous Recombination in hESCs and hiPSCs Using Helper-dependent Adenoviral Vectors

Emi Aizawa; Yuka Hirabayashi; Yuzuru Iwanaga; Keiichiro Suzuki; Kenji Sakurai; Miho Shimoji; Kazuhiro Aiba; Tamaki Wada; Norie Tooi; Eihachiro Kawase; Hirofumi Suemori; Norio Nakatsuji; Kohnosuke Mitani

Low efficiencies of gene targeting via homologous recombination (HR) have limited basic research and applications using human embryonic stem cells (hESCs) and human induced pluripotent stem cells (hiPSCs). Here, we show highly and equally efficient gene knockout and knock-in at both transcriptionally active (HPRT1, KU80, LIG1, LIG3) and inactive (HB9) loci in these cells using high-capacity helper-dependent adenoviral vectors (HDAdVs). Without the necessity of introducing artificial DNA double-strand breaks, 7-81% of drug-resistant colonies were gene-targeted by accurate HR, which were not accompanied with additional ectopic integrations. Even at the motor neuron-specific HB9 locus, the enhanced green fluorescent protein (EGFP) gene was accurately knocked in in 23-57% of drug-resistant colonies. In these clones, induced differentiation into the HB9-positive motor neuron correlated with EGFP expression. Furthermore, HDAdV infection had no detectable adverse effects on the undifferentiated state and pluripotency of hESCs and hiPSCs. These results suggest that HDAdV is one of the best methods for efficient and accurate gene targeting in hESCs and hiPSCs and might be especially useful for therapeutic applications.


Journal of Gene Medicine | 2009

Correction of mutant Fanconi anemia gene by homologous recombination in human hematopoietic cells using adeno-associated virus vector.

Kittiphong Paiboonsukwong; Fumi Ohbayashi; Haruka Shiiba; Emi Aizawa; Takayuki Yamashita; Kohnosuke Mitani

Adeno‐associated virus (AAV) vectors have been shown to correct a variety of mutations in human cells by homologous recombination (HR) at high rates, which can overcome insertional mutagenesis and transgene silencing, two of the major hurdles in conventional gene addition therapy of inherited diseases. We examined an ability of AAV vectors to repair a mutation in human hematopoietic cells by HR.

Collaboration


Dive into the Emi Aizawa's collaboration.

Top Co-Authors

Avatar

Concepcion Rodriguez Esteban

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Mo Li

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

April Goebl

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Rupa Devi Soligalla

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Jing Qu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kohnosuke Mitani

Saitama Medical University

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jun Wu

Salk Institute for Biological Studies

View shared research outputs
Researchain Logo
Decentralizing Knowledge