Network


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

Hotspot


Dive into the research topics where Guang-Hui Liu is active.

Publication


Featured researches published by Guang-Hui Liu.


Nature | 2011

Recapitulation of premature ageing with iPSCs from Hutchinson-Gilford progeria syndrome

Guang-Hui Liu; Basam Z. Barkho; Sergio Ruiz; Dinh Diep; Jing Qu; Sheng-Lian Yang; Athanasia D. Panopoulos; Keiichiro Suzuki; Leo Kurian; Christopher A. Walsh; James Thompson; Stéphanie Boué; Ho Lim Fung; Ignacio Sancho-Martinez; Kun Zhang; John R. Yates; Juan Carlos Izpisua Belmonte

Hutchinson–Gilford progeria syndrome (HGPS) is a rare and fatal human premature ageing disease, characterized by premature arteriosclerosis and degeneration of vascular smooth muscle cells (SMCs). HGPS is caused by a single point mutation in the lamin A (LMNA) gene, resulting in the generation of progerin, a truncated splicing mutant of lamin A. Accumulation of progerin leads to various ageing-associated nuclear defects including disorganization of nuclear lamina and loss of heterochromatin. Here we report the generation of induced pluripotent stem cells (iPSCs) from fibroblasts obtained from patients with HGPS. HGPS-iPSCs show absence of progerin, and more importantly, lack the nuclear envelope and epigenetic alterations normally associated with premature ageing. Upon differentiation of HGPS-iPSCs, progerin and its ageing-associated phenotypic consequences are restored. Specifically, directed differentiation of HGPS-iPSCs to SMCs leads to the appearance of premature senescence phenotypes associated with vascular ageing. Additionally, our studies identify DNA-dependent protein kinase catalytic subunit (DNAPKcs, also known as PRKDC) as a downstream target of progerin. The absence of nuclear DNAPK holoenzyme correlates with premature as well as physiological ageing. Because progerin also accumulates during physiological ageing, our results provide an in vitro iPSC-based model to study the pathogenesis of human premature and physiological vascular ageing.


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.


Cell Metabolism | 2013

Mitochondrial Regulation in Pluripotent Stem Cells

Xiuling Xu; Shunlei Duan; Fei Yi; Alejandro Ocampo; Guang-Hui Liu; Juan Carlos Izpisua Belmonte

Due to their fundamental role in energy production, mitochondria have been traditionally known as the powerhouse of the cell. Recent discoveries have suggested crucial roles of mitochondria in the maintenance of pluripotency, differentiation, and reprogramming of induced pluripotent stem cells (iPSCs). While glycolytic energy production is observed at pluripotent states, an increase in mitochondrial oxidative phosphorylation is necessary for cell differentiation. Consequently, a transition from somatic mitochondrial oxidative metabolism to glycolysis seems to be required for successful reprogramming. Future research aiming to dissect the roles of mitochondria in the establishment and homeostasis of pluripotency, as well as combining cell reprogramming with gene editing technologies, may unearth novel insights into our understanding of mitochondrial diseases and aging.


Cell Stem Cell | 2011

Targeted Gene Correction of Laminopathy-Associated LMNA Mutations in Patient-Specific iPSCs

Guang-Hui Liu; Keiichiro Suzuki; Jing Qu; Ignacio Sancho-Martinez; Fei Yi; Mo Li; Sachin Kumar; Emmanuel Nivet; Jessica Kim; Rupa Devi Soligalla; Ilir Dubova; April Goebl; Nongluk Plongthongkum; Ho-Lim Fung; Kun Zhang; Jeanne F. Loring; Louise C. Laurent; Juan Carlos Izpisua Belmonte

Combination of stem cell-based approaches with gene-editing technologies represents an attractive strategy for studying human disease and developing therapies. However, gene-editing methodologies described to date for human cells suffer from technical limitations including limited target gene size, low targeting efficiency at transcriptionally inactive loci, and off-target genetic effects that could hamper broad clinical application. To address these limitations, and as a proof of principle, we focused on homologous recombination-based gene correction of multiple mutations on lamin A (LMNA), which are associated with various degenerative diseases. We show that helper-dependent adenoviral vectors (HDAdVs) provide a highly efficient and safe method for correcting mutations in large genomic regions in human induced pluripotent stem cells and can also be effective in adult human mesenchymal stem cells. This type of approach could be used to generate genotype-matched cell lines for disease modeling and drug discovery and potentially also in therapeutics.


Nature | 2012

Progressive degeneration of human neural stem cells caused by pathogenic LRRK2

Guang-Hui Liu; Jing Qu; Keiichiro Suzuki; Emmanuel Nivet; MeiZhi Li; Nuria Montserrat; Fei Yi; Xiuling Xu; Sergio Ruiz; Weiqi Zhang; Ulrich Wagner; Audrey Kim; Bing Ren; Ying Li; April Goebl; Jessica Kim; Rupa Devi Soligalla; Ilir Dubova; James Thompson; John R. Yates; Concepcion Rodriguez Esteban; Ignacio Sancho-Martinez; Juan Carlos Izpisua Belmonte

Nuclear-architecture defects have been shown to correlate with the manifestation of a number of human diseases as well as ageing. It is therefore plausible that diseases whose manifestations correlate with ageing might be connected to the appearance of nuclear aberrations over time. We decided to evaluate nuclear organization in the context of ageing-associated disorders by focusing on a leucine-rich repeat kinase 2 (LRRK2) dominant mutation (G2019S; glycine-to-serine substitution at amino acid 2019), which is associated with familial and sporadic Parkinson’s disease as well as impairment of adult neurogenesis in mice. Here we report on the generation of induced pluripotent stem cells (iPSCs) derived from Parkinson’s disease patients and the implications of LRRK2(G2019S) mutation in human neural-stem-cell (NSC) populations. Mutant NSCs showed increased susceptibility to proteasomal stress as well as passage-dependent deficiencies in nuclear-envelope organization, clonal expansion and neuronal differentiation. Disease phenotypes were rescued by targeted correction of the LRRK2(G2019S) mutation with its wild-type counterpart in Parkinson’s disease iPSCs and were recapitulated after targeted knock-in of the LRRK2(G2019S) mutation in human embryonic stem cells. Analysis of human brain tissue showed nuclear-envelope impairment in clinically diagnosed Parkinson’s disease patients. Together, our results identify the nucleus as a previously unknown cellular organelle in Parkinson’s disease pathology and may help to open new avenues for Parkinson’s disease diagnoses as well as for the potential development of therapeutics targeting this fundamental cell structure.


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.


Cell | 2015

Selective Elimination of Mitochondrial Mutations in the Germline by Genome Editing

Pradeep Reddy; Alejandro Ocampo; Keiichiro Suzuki; Jinping Luo; Sandra R. Bacman; Sion L. Williams; Atsushi Sugawara; Daiji Okamura; Yuji Tsunekawa; Jun Wu; David Lam; Xiong Xiong; Nuria Montserrat; Concepcion Rodriguez Esteban; Guang-Hui Liu; Ignacio Sancho-Martinez; Dolors Manau; Salva Civico; Francesc Cardellach; Maria del Mar O’Callaghan; Jaime Campistol; Huimin Zhao; Josep M. Campistol; Carlos T. Moraes; Juan Carlos Izpisua Belmonte

Mitochondrial diseases include a group of maternally inherited genetic disorders caused by mutations in mtDNA. In most of these patients, mutated mtDNA coexists with wild-type mtDNA, a situation known as mtDNA heteroplasmy. Here, we report on a strategy toward preventing germline transmission of mitochondrial diseases by inducing mtDNA heteroplasmy shift through the selective elimination of mutated mtDNA. As a proof of concept, we took advantage of NZB/BALB heteroplasmic mice, which contain two mtDNA haplotypes, BALB and NZB, and selectively prevented their germline transmission using either mitochondria-targeted restriction endonucleases or TALENs. In addition, we successfully reduced human mutated mtDNA levels responsible for Lebers hereditary optic neuropathy (LHOND), and neurogenic muscle weakness, ataxia, and retinitis pigmentosa (NARP), in mammalian oocytes using mitochondria-targeted TALEN (mito-TALENs). Our approaches represent a potential therapeutic avenue for preventing the transgenerational transmission of human mitochondrial diseases caused by mutations in mtDNA. PAPERCLIP.


Journal of Biological Chemistry | 2014

A Cut above the Rest: Targeted Genome Editing Technologies in Human Pluripotent Stem Cells

Mo Li; Keiichiro Suzuki; Na Young Kim; Guang-Hui Liu; Juan Carlos Izpisua Belmonte

Human pluripotent stem cells (hPSCs) offer unprecedented opportunities to study cellular differentiation and model human diseases. The ability to precisely modify any genomic sequence holds the key to realizing the full potential of hPSCs. Thanks to the rapid development of novel genome editing technologies driven by the enormous interest in the hPSC field, genome editing in hPSCs has evolved from being a daunting task a few years ago to a routine procedure in most laboratories. Here, we provide an overview of the mainstream genome editing tools, including zinc finger nucleases, transcription activator-like effector nucleases, clustered regularly interspaced short palindromic repeat/CAS9 RNA-guided nucleases, and helper-dependent adenoviral vectors. We discuss the features and limitations of these technologies, as well as how these factors influence the utility of these tools in basic research and therapies.


Embo Molecular Medicine | 2013

Autophagic control of cell 'stemness'

Huize Pan; Ning Cai; Mo Li; Guang-Hui Liu; Juan Carlos Izpisua Belmonte

Stem cells have the ability to self‐renew and differentiate into various cell types. Both cell‐intrinsic and extrinsic factors may contribute to aging‐related decline in stem cell function and loss of stemness. The maintenance of cellular homeostasis requires timely removal of toxic proteins and damaged organelles that accumulate with age or in pathological conditions. Autophagy is one of the main strategies to eliminate unwanted cytoplasmic materials thereby ultimately preventing cellular damage. Here, we shall discuss the accumulating evidence suggesting that autophagy plays a critical role in the homeostatic control of stem cell functions during aging, tissue regeneration, and cellular reprogramming.

Collaboration


Dive into the Guang-Hui Liu's collaboration.

Top Co-Authors

Avatar

Jing Qu

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Juan Carlos Izpisua Belmonte

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Fei Yi

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Weiqi Zhang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Ruotong Ren

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Mo Li

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jiping Yang

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Keiichiro Suzuki

Salk Institute for Biological Studies

View shared research outputs
Top Co-Authors

Avatar

Xiuling Xu

Scripps Research Institute

View shared research outputs
Researchain Logo
Decentralizing Knowledge