Network


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

Hotspot


Dive into the research topics where Andreas Reik is active.

Publication


Featured researches published by Andreas Reik.


Cell | 2012

Controlling Long-Range Genomic Interactions at a Native Locus by Targeted Tethering of a Looping Factor

Wulan Deng; Jongjoo Lee; Hongxin Wang; Jeffrey C. Miller; Andreas Reik; Philip D. Gregory; Ann Dean; Gerd A. Blobel

Chromatin loops juxtapose distal enhancers with active promoters, but their molecular architecture and relationship with transcription remain unclear. In erythroid cells, the locus control region (LCR) and β-globin promoter form a chromatin loop that requires transcription factor GATA1 and the associated molecule Ldb1. We employed artificial zinc fingers (ZF) to tether Ldb1 to the β-globin promoter in GATA1 null erythroblasts, in which the β-globin locus is relaxed and inactive. Remarkably, targeting Ldb1 or only its self-association domain to the β-globin promoter substantially activated β-globin transcription in the absence of GATA1. Promoter-tethered Ldb1 interacted with endogenous Ldb1 complexes at the LCR to form a chromatin loop, causing recruitment and phosphorylation of RNA polymerase II. ZF-Ldb1 proteins were inactive at alleles lacking the LCR, demonstrating that their activities depend on long-range interactions. Our findings establish Ldb1 as a critical effector of GATA1-mediated loop formation and indicate that chromatin looping causally underlies gene regulation.


Nature Medicine | 2012

Editing T cell specificity towards leukemia by zinc finger nucleases and lentiviral gene transfer

Elena Provasi; Pietro Genovese; Angelo Lombardo; Zulma Magnani; Pei Qi Liu; Andreas Reik; Victoria Chu; David Paschon; Lei Zhang; Jürgen Kuball; Barbara Camisa; Attilio Bondanza; Giulia Casorati; Maurilio Ponzoni; Fabio Ciceri; Claudio Bordignon; Philip D. Greenberg; Michael C. Holmes; Philip D. Gregory; Luigi Naldini; Chiara Bonini

The transfer of high-avidity T cell receptor (TCR) genes isolated from rare tumor-specific lymphocytes into polyclonal T cells is an attractive cancer immunotherapy strategy. However, TCR gene transfer results in competition for surface expression and inappropriate pairing between the exogenous and endogenous TCR chains, resulting in suboptimal activity and potentially harmful unpredicted antigen specificities of the resultant TCRs. We designed zinc-finger nucleases (ZFNs) that promoted the disruption of endogenous TCR β- and α-chain genes. Lymphocytes treated with ZFNs lacked surface expression of CD3-TCR and expanded with the addition of interleukin-7 (IL-7) and IL-15. After lentiviral transfer of a TCR specific for the Wilms tumor 1 (WT1) antigen, these TCR-edited cells expressed the new TCR at high levels, were easily expanded to near purity and were superior at specific antigen recognition compared to donor-matched, unedited TCR-transferred cells. In contrast to unedited TCR-transferred cells, the TCR-edited lymphocytes did not mediate off-target reactivity while maintaining their anti-tumor activity in vivo, thus showing that complete editing of T cell specificity generates tumor-specific lymphocytes with improved biosafety profiles.


Blood | 2015

Correction of the sickle-cell disease mutation in human hematopoietic stem/progenitor cells

Megan D. Hoban; Gregory J. Cost; Matthew C. Mendel; Zulema Romero; Michael L. Kaufman; Alok V. Joglekar; Michelle Ho; Dianne Lumaquin; David Gray; Georgia R. Lill; Aaron R. Cooper; Fabrizia Urbinati; Shantha Senadheera; Allen Zhu; Pei-Qi Liu; David Paschon; Lei Zhang; Edward J. Rebar; Andrew Wilber; Xiaoyan Wang; Philip D. Gregory; Michael C. Holmes; Andreas Reik; Roger P. Hollis; Donald B. Kohn

Sickle cell disease (SCD) is characterized by a single point mutation in the seventh codon of the β-globin gene. Site-specific correction of the sickle mutation in hematopoietic stem cells would allow for permanent production of normal red blood cells. Using zinc-finger nucleases (ZFNs) designed to flank the sickle mutation, we demonstrate efficient targeted cleavage at the β-globin locus with minimal off-target modification. By co-delivering a homologous donor template (either an integrase-defective lentiviral vector or a DNA oligonucleotide), high levels of gene modification were achieved in CD34(+) hematopoietic stem and progenitor cells. Modified cells maintained their ability to engraft NOD/SCID/IL2rγ(null) mice and to produce cells from multiple lineages, although with a reduction in the modification levels relative to the in vitro samples. Importantly, ZFN-driven gene correction in CD34(+) cells from the bone marrow of patients with SCD resulted in the production of wild-type hemoglobin tetramers.


Biotechnology and Bioengineering | 2010

Generation of a triple-gene knockout mammalian cell line using engineered zinc-finger nucleases†

Pei-Qi Liu; Edmond Chan; Gregory J. Cost; Lin Zhang; Jianbin Wang; Jeffrey C. Miller; Dmitry Guschin; Andreas Reik; Michael C. Holmes; John Mott; Trevor N. Collingwood; Philip D. Gregory

Mammalian cells with multi‐gene knockouts could be of considerable utility in research, drug discovery, and cell‐based therapeutics. However, existing methods for targeted gene deletion require sequential rounds of homologous recombination and drug selection to isolate rare desired events—a process sufficiently laborious to limit application to individual loci. Here we present a solution to this problem. Firstly, we report the development of zinc‐finger nucleases (ZFNs) targeted to cleave three independent genes with known null phenotypes. Mammalian cells exposed to each ZFN pair in turn resulted in the generation of cell lines harboring single, double, and triple gene knockouts, that is, the successful disruption of two, four, and six alleles. All three biallelic knockout events were obtained at frequencies of >1% without the use of selection, displayed the expected knockout phenotype(s), and harbored DNA mutations centered at the ZFN binding sites. These data demonstrate the utility of ZFNs in multi‐locus genome engineering. Biotechnol. Bioeng. 2010; 106: 97–105.


Nature Methods | 2015

Functional footprinting of regulatory DNA

Jeff Vierstra; Andreas Reik; Kai Hsin Chang; Sandra Stehling-Sun; Yuanyue Zhou; Sarah J. Hinkley; David Paschon; Lei Zhang; Nikoletta Psatha; Yuri R. Bendana; Colleen M. O'Neil; Alexander Song; Andrea Mich; Pei Qi Liu; Gary Lee; Daniel E. Bauer; Michael C. Holmes; Stuart H. Orkin; Thalia Papayannopoulou; George Stamatoyannopoulos; Edward J. Rebar; Philip D. Gregory; Fyodor D. Urnov; John A. Stamatoyannopoulos

Regulatory regions harbor multiple transcription factor (TF) recognition sites; however, the contribution of individual sites to regulatory function remains challenging to define. We describe an approach that exploits the error-prone nature of genome editing–induced double-strand break repair to map functional elements within regulatory DNA at nucleotide resolution. We demonstrate the approach on a human erythroid enhancer, revealing single TF recognition sites that gate the majority of downstream regulatory function.


Molecular Therapy | 2015

Clinical Scale Zinc Finger Nuclease-mediated Gene Editing of PD-1 in Tumor Infiltrating Lymphocytes for the Treatment of Metastatic Melanoma

Joal D. Beane; Gary Lee; Zhili Zheng; Matthew C. Mendel; Daniel Abate-Daga; Mini Bharathan; Mary A. Black; Nimisha Gandhi; Zhiya Yu; Smita S. Chandran; Martin A. Giedlin; Dale Ando; Jeffrey C. Miller; David Paschon; Dmitry Guschin; Edward J. Rebar; Andreas Reik; Michael C. Holmes; Philip D. Gregory; Nicholas P. Restifo; Steven A. Rosenberg; Richard A. Morgan; Steven A. Feldman

Programmed cell death-1 (PD-1) is expressed on activated T cells and represents an attractive target for gene-editing of tumor targeted T cells prior to adoptive cell transfer (ACT). We used zinc finger nucleases (ZFNs) directed against the gene encoding human PD-1 (PDCD-1) to gene-edit melanoma tumor infiltrating lymphocytes (TIL). We show that our clinical scale TIL production process yielded efficient modification of the PD-1 gene locus, with an average modification frequency of 74.8% (n = 3, range 69.9-84.1%) of the alleles in a bulk TIL population, which resulted in a 76% reduction in PD-1 surface-expression. Forty to 48% of PD-1 gene-edited cells had biallelic PD-1 modification. Importantly, the PD-1 gene-edited TIL product showed improved in vitro effector function and a significantly increased polyfunctional cytokine profile (TNFα, GM-CSF, and IFNγ) compared to unmodified TIL in two of the three donors tested. In addition, all donor cells displayed an effector memory phenotype and expanded approximately 500-2,000-fold in vitro. Thus, further study to determine the efficiency and safety of adoptive cell transfer using PD-1 gene-edited TIL for the treatment of metastatic melanoma is warranted.


Current Opinion in Genetics & Development | 2002

Biotechnologies and therapeutics: chromatin as a target.

Andreas Reik; Philip D. Gregory; Fyodor D. Urnov

As alterations in gene expression underlie a considerable proportion of human diseases, correcting such aberrant transcription in vivo is expected to provide therapeutic benefit to the patient. Attempts to control endogenous mammalian genes, however, face a significant obstacle in the form of chromatin. Aberrant gene repression can be alleviated by using small-molecule inhibitors that exert nucleus-wide effects on chromatin-based repressors. Genome-wide chromatin remodeling also occurs during cloning via nuclear transfer, and causes the deregulation of epigenetically controlled genes. Regulation of genes in vivo can be accomplished via the use of designed transcription factors - these result from a fusion of a designed DNA-binding domain based on the zinc finger protein motif to a functional domain of choice.


Molecular therapy. Methods & clinical development | 2017

Long-Term Engraftment and Fetal Globin Induction upon BCL11A Gene Editing in Bone-Marrow-Derived CD34+ Hematopoietic Stem and Progenitor Cells

Kai-Hsin Chang; Sarah Smith; Timothy Sullivan; Kai Chen; Qianhe Zhou; Jason A. West; Mei Liu; Yingchun Liu; Benjamin F. Vieira; Chao Sun; Vu P. Hong; Mingxuan Zhang; Xiao Yang; Andreas Reik; Fyodor D. Urnov; Edward J. Rebar; Michael C. Holmes; Olivier Danos; Haiyan Jiang; Siyuan Tan

To develop an effective and sustainable cell therapy for sickle cell disease (SCD), we investigated the feasibility of targeted disruption of the BCL11A gene, either within exon 2 or at the GATAA motif in the intronic erythroid-specific enhancer, using zinc finger nucleases in human bone marrow (BM) CD34+ hematopoietic stem and progenitor cells (HSPCs). Both targeting strategies upregulated fetal globin expression in erythroid cells to levels predicted to inhibit hemoglobin S polymerization. However, complete inactivation of BCL11A resulting from bi-allelic frameshift mutations in BCL11A exon 2 adversely affected erythroid enucleation. In contrast, bi-allelic disruption of the GATAA motif in the erythroid enhancer of BCL11A did not negatively impact enucleation. Furthermore, BCL11A exon 2-edited BM-CD34+ cells demonstrated a significantly reduced engraftment potential in immunodeficient mice. Such an adverse effect on HSPC function was not observed upon BCL11A erythroid-enhancer GATAA motif editing, because enhancer-edited CD34+ cells achieved robust long-term engraftment and gave rise to erythroid cells with elevated levels of fetal globin expression when chimeric BM was cultured ex vivo. Altogether, our results support further clinical development of the BCL11A erythroid-specific enhancer editing in BM-CD34+ HSPCs as an autologous stem cell therapy in SCD patients.


EMBO Reports | 2002

Designed transcription factors as structural, functional and therapeutic probes of chromatin in vivo

Fyodor D. Urnov; Edward J. Rebar; Andreas Reik; Pier Paolo Pandolfi

Despite its central importance in gene regulation, chromatin in mammalian cells remains relatively poorly understood—a predicament due to the paucity of robust genetic tools in mammals, the complexity of the chromatin remodeling machinery, and the dynamic properties of chromatin in vivo. Here we review recent developments in understanding endogenous mammalian gene regulation via the use of designed transcription factors (TFs). These include mutated forms of naturally occurring TFs that exhibit dominant‐negative activity, and designed proteins with novel, predetermined DNA‐binding specificities. Systematic targeting of designed TFs to particular promoters is helping to illuminate the complex rules that chromatin imposes on TF access and action in vivo. We evaluate the potential applications of these proteins as probes of mammalian chromatin‐based regulatory pathways and their potential for the therapy of human disease, highlighting leukemia in particular.


Journal of Biomolecular Screening | 2005

Isogenic Human Cell Lines for Drug Discovery: Regulation of Target Gene Expression by Engineered Zinc-Finger Protein Transcription Factors

Pei-Qi Liu; Siyuan Tan; Matthew C. Mendel; Richard J. Murrills; Bheem M. Bhat; Brian Schlag; Rachelle L. Samuel; Jeanne J. Matteo; Ragan de la Rosa; Katherine Howes; Andreas Reik; Casey C. Case; Frederick J. Bex; Kathleen H. Young; Philip D. Gregory

Isogenic cell lines differing only in the expression of the protein of interest provide the ideal platform for cell-based screening. However, related natural lines differentially expressing the therapeutic target of choice are rare. Here the authors report a strategy for drug screening employing isogenic human cell lines in which the expression of the target protein is regulated by a gene-specific engineered zinc-finger protein (ZFP) transcription factor (TF). To demonstrate this approach, a ZFP TF activator of the human parathyroid hormone receptor 1 (PTHR1) gene was identified and introduced into HEK293 cells (negative for PTHR1). Following induction of ZFP TF expression, this cell line produced functional PTHR1 protein, resulting in a robust and ligand-specific cyclic adenosine monophosphate (cAMP) response. Reciprocally, the natural expression of PTHR1 observed in SAOS2 cells was dramatically reduced by the introduction of the appropriate PTHR1-specific ZFP TF repressor. Moreover, this ZFP-driven PTHR1 repression selectively eliminated the functional cAMP response invoked by known ligands of PTHR1. These data establish ZFP TF–generated isogenic lines as a general approach for the identification of therapeutic agents specific for the target gene of interest.

Collaboration


Dive into the Andreas Reik'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