Network


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

Hotspot


Dive into the research topics where Hai-Kun Liu is active.

Publication


Featured researches published by Hai-Kun Liu.


Nature | 2015

Brain tumour cells interconnect to a functional and resistant network

Matthias Osswald; Erik Jung; Felix Sahm; Gergely Solecki; Varun Venkataramani; Jonas Blaes; Sophie Weil; Heinz Horstmann; Benedikt Wiestler; Mustafa Syed; Lulu Huang; Miriam Ratliff; Kianush Karimian Jazi; Felix T. Kurz; Torsten Schmenger; Dieter Lemke; Miriam Gömmel; Martin Pauli; Yunxiang Liao; Peter Häring; Stefan Pusch; Verena Herl; Christian Steinhäuser; Damir Krunic; Mostafa Jarahian; Hrvoje Miletic; Anna Sophie Berghoff; Oliver Griesbeck; Georgios Kalamakis; Olga Garaschuk

Astrocytic brain tumours, including glioblastomas, are incurable neoplasms characterized by diffusely infiltrative growth. Here we show that many tumour cells in astrocytomas extend ultra-long membrane protrusions, and use these distinct tumour microtubes as routes for brain invasion, proliferation, and to interconnect over long distances. The resulting network allows multicellular communication through microtube-associated gap junctions. When damage to the network occurred, tumour microtubes were used for repair. Moreover, the microtube-connected astrocytoma cells, but not those remaining unconnected throughout tumour progression, were protected from cell death inflicted by radiotherapy. The neuronal growth-associated protein 43 was important for microtube formation and function, and drove microtube-dependent tumour cell invasion, proliferation, interconnection, and radioresistance. Oligodendroglial brain tumours were deficient in this mechanism. In summary, astrocytomas can develop functional multicellular network structures. Disconnection of astrocytoma cells by targeting their tumour microtubes emerges as a new principle to reduce the treatment resistance of this disease.


Genes & Development | 2010

The nuclear receptor tailless induces long-term neural stem cell expansion and brain tumor initiation

Hai-Kun Liu; Ying Wang; Thorsten Belz; Dagmar Bock; Andrea Takacs; Bernhard Radlwimmer; Sebastian Barbus; Guido Reifenberger; Peter Lichter; Giinther Schütz

Malignant gliomas are the most common primary brain tumors, and are associated with frequent resistance to therapy as well as poor prognosis. Here we demonstrate that the nuclear receptor tailless (Tlx), which in the adult is expressed exclusively in astrocyte-like B cells of the subventricular zone, acts as a key regulator of neural stem cell (NSC) expansion and brain tumor initiation from NSCs. Overexpression of Tlx antagonizes age-dependent exhaustion of NSCs in mice and leads to migration of stem/progenitor cells from their natural niche. The increase of NSCs persists with age, and leads to efficient production of newborn neurons in aged brain tissues. These cells initiate the development of glioma-like lesions and gliomas. Glioma development is accelerated upon loss of the tumor suppressor p53. Tlx-induced NSC expansion and gliomagenesis are associated with increased angiogenesis, which allows for the migration and maintenance of brain tumor stem cells in the perivascular niche. We also demonstrate that Tlx transcripts are overexpressed in human primary glioblastomas in which Tlx expression is restricted to a subpopulation of nestin-positive perivascular tumor cells. Our study clearly demonstrates how NSCs contribute to brain tumorgenesis driven by a stem cell-specific transcription factor, thus providing novel insights into the histogenesis and molecular pathogenesis of primary brain tumors.


Genes & Development | 2008

The nuclear receptor tailless is required for neurogenesis in the adult subventricular zone

Hai-Kun Liu; Thorsten Belz; Dagmai Bock; Andrea Takacs; Hui Wu; Peter Lichter; Minqiang Chai; Günther Schütz

The tailless (Tlx) gene encodes an orphan nuclear receptor that is expressed by neural stem/progenitor cells in the adult brain of the subventricular zone (SVZ) and the dentate gyrus (DG). The function of Tlx in neural stem cells of the adult SVZ remains largely unknown. We show here that in the SVZ of the adult brain Tlx is exclusively expressed in astrocyte-like B cells. An inducible mutation of the Tlx gene in the adult brain leads to complete loss of SVZ neurogenesis. Furthermore, analysis indicates that Tlx is required for the transition from radial glial cells to astrocyte-like neural stem cells. These findings demonstrate the crucial role of Tlx in the generation and maintenance of NSCs in the adult SVZ in vivo.


Cell Stem Cell | 2014

Targeting Self-Renewal in High-Grade Brain Tumors Leads to Loss of Brain Tumor Stem Cells and Prolonged Survival

Zhe Zhu; Muhammad Amir Khan; Markus Weiler; Jonas Blaes; Leonie Jestaedt; Madeleine Geibert; Peng Zou; Jan Gronych; Olga Bernhardt; Andrey Korshunov; Verena Bugner; Peter Lichter; Bernhard Radlwimmer; Sabine Heiland; Martin Bendszus; Wolfgang Wick; Hai-Kun Liu

Cancer stem cells (CSCs) have been suggested as potential therapeutic targets for treating malignant tumors, but the in vivo supporting evidence is still missing. Using a GFP reporter driven by the promoter of the nuclear receptor tailless (Tlx), we demonstrate that Tlx(+) cells in primary brain tumors are mostly quiescent. Lineage tracing demonstrates that single Tlx(+) cells can self-renew and generate Tlx(-) tumor cells in primary tumors, suggesting that they are brain tumor stem cells (BTSCs). After introducing a BTSC-specific knock-out of the Tlx gene in primary mouse tumors, we observed a loss of self-renewal of BTSCs and prolongation of animal survival, accompanied by induction of essential signaling pathways mediating cell-cycle arrest, cell death, and neural differentiation. Our study demonstrates the feasibility of targeting glioblastomas and indicates the suitability of BTSCs as therapeutic targets, thereby supporting the CSC hypothesis.


Nature Communications | 2017

Chd7 is indispensable for mammalian brain development through activation of a neuronal differentiation programme

Weijun Feng; Daisuke Kawauchi; Huiqin Körkel-Qu; Huan Deng; Elisabeth Serger; Laura Sieber; Jenna Ariel Lieberman; Silvia Jimeno-González; Sander Lambo; Bola Hanna; Yassin Harim; Malin Jansen; Anna Neuerburg; Olga Friesen; Marc Zuckermann; Vijayanad Rajendran; Jan Gronych; Olivier Ayrault; Andrey Korshunov; David T. W. Jones; Marcel Kool; Paul A. Northcott; Peter Lichter; Felipe Cortés-Ledesma; Stefan M. Pfister; Hai-Kun Liu

Mutations in chromatin modifier genes are frequently associated with neurodevelopmental diseases. We herein demonstrate that the chromodomain helicase DNA-binding protein 7 (Chd7), frequently associated with CHARGE syndrome, is indispensable for normal cerebellar development. Genetic inactivation of Chd7 in cerebellar granule neuron progenitors leads to cerebellar hypoplasia in mice, due to the impairment of granule neuron differentiation, induction of apoptosis and abnormal localization of Purkinje cells, which closely recapitulates known clinical features in the cerebella of CHARGE patients. Combinatory molecular analyses reveal that Chd7 is required for the maintenance of open chromatin and thus activation of genes essential for granule neuron differentiation. We further demonstrate that both Chd7 and Top2b are necessary for the transcription of a set of long neuronal genes in cerebellar granule neurons. Altogether, our comprehensive analyses reveal a mechanism with chromatin remodellers governing brain development via controlling a core transcriptional programme for cell-specific differentiation.


Neuro-oncology | 2012

Expression of podoplanin in human astrocytic brain tumors is controlled by the PI3K-AKT-AP-1 signaling pathway and promoter methylation

Heike Peterziel; Julia Müller; Andreas Danner; Sebastian Barbus; Hai-Kun Liu; Bernhard Radlwimmer; Torsten Pietsch; Peter Lichter; Günther Schütz; Jochen Hess; Peter Angel

Recently, we found strong overexpression of the mucin-type glycoprotein podoplanin (PDPN) in human astrocytic brain tumors, specifically in primary glioblastoma multiforme (GB). In the current study, we show an inverse correlation between PDPN expression and PTEN levels in primary human GB and glioma cell lines, and we report elevated PDPN protein levels in the subventricular zone of brain tissue sections of PTEN-deficient mice. In human glioma cells lacking functional PTEN, reintroduction of wild-type PTEN, inhibition of the PTEN downstream target protein kinase B/AKT, or interference with transcription factor AP-1 function resulted in efficient downregulation of PDPN expression. In addition, we observed hypoxia-dependent PDPN transcriptional control and demonstrated that PDPN expression is subject to negative transcriptional regulation by promoter methylation in human GB and in glioma cell lines. Treatment of PTEN-negative glioma cells with demethylating agents induced expression of PDPN. Together, our findings show that increased PDPN expression in human GB is caused by loss of PTEN function and activation of the PI3K-AKT-AP-1 signaling pathway, accompanied by epigenetic regulation of PDPN promoter activity. Silencing of PDPN expression leads to reduced proliferation and migration of glioma cells, suggesting a functional role of PDPN in glioma progression and malignancy. Thus, specific targeting of PDPN expression and/or function could be a promising strategy for the treatment of patients with primary GB.


European Journal of Neuroscience | 2007

Inactivation of the gene for the nuclear receptor tailless in the brain preserving its function in the eye

Thorsten Belz; Hai-Kun Liu; Dagmar Bock; Andrea Takacs; Miriam A. Vogt; Tim Wintermantel; Christiane Brandwein; Peter Gass; Erich Greiner; Günther Schütz

During embryogenesis, tailless, an orphan member of the nuclear receptor family, is expressed in the germinal zones of the brain and the developing retina, and is involved in regulating the cell cycle of progenitor cells. Consequently, a deletion of the tailless gene leads to decreased cell number with associated anatomical defects in the limbic system, the cortex and the eye. These structural abnormalities are associated with blindness, increased aggressiveness, poor performance in learning paradigms and reduced anxiousness. In order to assess the contribution of blindness to the behavioural changes, we established tailless mutant mice with intact visual abilities. We generated a mouse line in which the second exon of the tailless gene is flanked by loxP sites and crossed these animals with a transgenic line expressing the Cre recombinase in the neurogenic area of the developing brain, but not in the eye. The resulting animals have anatomically indistinguishable brains compared with tailless germline mutants, but are not blind. They are less anxious and much more aggressive than controls, like tailless germline mutants. In contrast to germline mutants, the conditional mutants are not impaired in fear conditioning. Furthermore, they show good performance in the Morris water‐maze despite severely reduced hippocampal structures. Thus, the pathological aggressiveness and reduced anxiety found in tailless germline mutants are due to malformations caused by inactivation of the tailless gene in the brain, but the poor performance of tailless null mice in learning and memory paradigms is dependent on the associated blindness.


Mechanisms of Development | 2013

Role of the nuclear receptor Tailless in adult neural stem cells.

Yu Wang; Hai-Kun Liu; Günther Schütz

Tailless (Tlx) is an orphan nuclear receptor which is specifically expressed in the neural stem cells of the two largest germinal neurogenesis zones in the adult mouse brain, the subventricular zone (SVZ) of the lateral ventricle and the subgranular zone (SGZ) of the dentate gyrus. By interacting with its cofactors, Tlx represses its target genes and plays an important role in the maintenance of adult NSCs. This review provides a snapshot of current knowledge about Tlx function in adult NSCs.


Progress in Neuro-psychopharmacology & Biological Psychiatry | 2018

CRISPR-engineered genome editing for the next generation neurological disease modeling

Weijun Feng; Hai-Kun Liu; Daisuke Kawauchi

&NA; Neurological disorders often occur because of failure of proper brain development and/or appropriate maintenance of neuronal circuits. In order to understand roles of causative factors (e.g. genes, cell types) in disease development, generation of solid animal models has been one of straight‐forward approaches. Recent next generation sequencing studies on human patient‐derived clinical samples have identified various types of recurrent mutations in individual neurological diseases. While these discoveries have prompted us to evaluate impact of mutated genes on these neurological diseases, a feasible but flexible genome editing tool had remained to be developed. An advance of genome editing technology using the clustered regularly interspaced short palindromic repeats (CRISPR) with the CRISPR‐associated protein (Cas) offers us a tremendous potential to create a variety of mutations in the cell, leading to “next generation” disease models carrying disease‐associated mutations. We will here review recent progress of CRISPR‐based brain disease modeling studies and discuss future requirement to tackle current difficulties in usage of these technologies. HighlightsGenome editing using CRISPR/Cas technology allows for neurological disease‐associated mutagenesis.CRISPR‐based disease modeling requires a reasonable choice of Cas nucleases and gene delivery methods.CRISPR‐induced off‐target effects and intercellular mosaic mutations should be considered for disease modeling studies.


Aging Cell | 2018

Molecular profiling of aged neural progenitors identifies Dbx2 as a candidate regulator of age-associated neurogenic decline

Giuseppe Lupo; Paola S. Nisi; Pilar Esteve; Yu-Lee Paul; Clara Lopes Novo; Ben Sidders; Muhammad A. Khan; Stefano Biagioni; Hai-Kun Liu; Paola Bovolenta; Emanuele Cacci; Peter J. Rugg-Gunn

Adult neurogenesis declines with aging due to the depletion and functional impairment of neural stem/progenitor cells (NSPCs). An improved understanding of the underlying mechanisms that drive age‐associated neurogenic deficiency could lead to the development of strategies to alleviate cognitive impairment and facilitate neuroregeneration. An essential step towards this aim is to investigate the molecular changes that occur in NSPC aging on a genomewide scale. In this study, we compare the transcriptional, histone methylation and DNA methylation signatures of NSPCs derived from the subventricular zone (SVZ) of young adult (3 months old) and aged (18 months old) mice. Surprisingly, the transcriptional and epigenomic profiles of SVZ‐derived NSPCs are largely unchanged in aged cells. Despite the global similarities, we detect robust age‐dependent changes at several hundred genes and regulatory elements, thereby identifying putative regulators of neurogenic decline. Within this list, the homeobox gene Dbx2 is upregulated in vitro and in vivo, and its promoter region has altered histone and DNA methylation levels, in aged NSPCs. Using functional in vitro assays, we show that elevated Dbx2 expression in young adult NSPCs promotes age‐related phenotypes, including the reduced proliferation of NSPC cultures and the altered transcript levels of age‐associated regulators of NSPC proliferation and differentiation. Depleting Dbx2 in aged NSPCs caused the reverse gene expression changes. Taken together, these results provide new insights into the molecular programmes that are affected during mouse NSPC aging, and uncover a new functional role for Dbx2 in promoting age‐related neurogenic decline.

Collaboration


Dive into the Hai-Kun Liu's collaboration.

Top Co-Authors

Avatar

Peter Lichter

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Weijun Feng

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Daisuke Kawauchi

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Günther Schütz

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andrey Korshunov

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Bernhard Radlwimmer

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Jan Gronych

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Stefan M. Pfister

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

Andrea Takacs

German Cancer Research Center

View shared research outputs
Top Co-Authors

Avatar

David T. W. Jones

German Cancer Research Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge