101
|
Lang L, Ding HF, Chen X, Sun SY, Liu G, Yan C. Internal Ribosome Entry Site-Based Bicistronic In Situ Reporter Assays for Discovery of Transcription-Targeted Lead Compounds. ACTA ACUST UNITED AC 2015; 22:957-64. [PMID: 26144883 DOI: 10.1016/j.chembiol.2015.06.009] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2014] [Revised: 06/04/2015] [Accepted: 06/09/2015] [Indexed: 12/27/2022]
Abstract
Although transgene-based reporter gene assays have been used to discover small molecules targeting expression of cancer-driving genes, the success is limited due to the fact that reporter gene expression regulated by incomplete cis-acting elements and foreign epigenetic environments does not faithfully reproduce chemical responses of endogenous genes. Here, we present an internal ribosome entry site-based strategy for bicistronically co-expressing reporter genes with an endogenous gene in the native gene locus, yielding an in situ reporter assay closely mimicking endogenous gene expression without disintegrating its function. This strategy combines the CRISPR-Cas9-mediated genome-editing tool with the recombinase-mediated cassette-exchange technology, and allows for rapid development of orthogonal assays for excluding false hits generated from primary screens. We validated this strategy by developing a screening platform for identifying compounds targeting oncogenic eIF4E, and demonstrated that the novel reporter assays are powerful in searching for transcription-targeted lead compounds with high confidence.
Collapse
Affiliation(s)
- Liwei Lang
- GRU Cancer Center, Georgia Regents University, CN2134, 1410 Laney Walker Boulevard, Augusta, GA 30912, USA; Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | - Han-Fei Ding
- GRU Cancer Center, Georgia Regents University, CN2134, 1410 Laney Walker Boulevard, Augusta, GA 30912, USA; Department of Pathology, Georgia Regents University, Augusta, GA 30912, USA
| | - Xiaoguang Chen
- Department of Pharmacology, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100031, China
| | - Shi-Yong Sun
- Department of Hematology and Medical Oncology, Emory University School of Medicine and Winship Cancer Institute, Atlanta, GA 30322, USA
| | - Gang Liu
- Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA
| | - Chunhong Yan
- GRU Cancer Center, Georgia Regents University, CN2134, 1410 Laney Walker Boulevard, Augusta, GA 30912, USA; Department of Biochemistry and Molecular Biology, Georgia Regents University, Augusta, GA 30912, USA; Center for Cell Biology & Cancer Research, Albany Medical College, Albany, NY 12208, USA.
| |
Collapse
|
102
|
Camaj P, Primo S, Wang Y, Heinemann V, Zhao Y, Laubender RP, Stintzing S, Giessen-Jung C, Jung A, Gamba S, Bruns CJ, Modest DP. KRAS exon 2 mutations influence activity of regorafenib in an SW48-based disease model of colorectal cancer. Future Oncol 2015; 11:1919-29. [DOI: 10.2217/fon.15.97] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
ABSTRACT Aim: To investigate the impact of KRAS mutation variants on the activity of regorafenib in SW48 colorectal cancer cells. Materials & methods: Activity of regorafenib was evaluated in isogenic SW48 KRAS wild-type (WT) and mutant cells. Subcutaneous xenografts (KRAS WT and G12C mutant variants) in NOD/SCID mice were analyzed to elucidate the effect of regorafenib treatment in vivo. Results: Compared with KRAS WT cells, all mutant variants seemed associated with some degree of resistance to regorafenib-treatment in vitro. In vivo, activation of apoptosis (TUNEL) and reduction of proliferation (Ki67) after treatment with regorafenib were more pronounced in KRAS WT tumors as compared with G12C variants. Conclusion: In SW48 cells, exon 2 mutations of the KRAS gene may influence antitumor effects of regorafenib.
Collapse
Affiliation(s)
- Peter Camaj
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-University, Magdeburg, Germany
| | - Stefano Primo
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Yan Wang
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-University, Magdeburg, Germany
| | - Volker Heinemann
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Medicine III, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Yue Zhao
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-University, Magdeburg, Germany
| | - Ruediger Paul Laubender
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Institute of Medical Informatics, Biometry & Epidemiology, University of Munich, Munich, Germany
| | - Sebastian Stintzing
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Medicine III, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Clemens Giessen-Jung
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Medicine III, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Andreas Jung
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Institute of Pathology, University of Munich, Munich, Germany
| | - Sebastian Gamba
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
| | - Christiane Josephine Bruns
- Department of Surgery, University Hospital Grosshadern, University of Munich, Munich, Germany
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Division of Experimental Surgery, Department of Surgery, Otto-von-Guericke-University, Magdeburg, Germany
| | - Dominik Paul Modest
- German Cancer Consortium (DKTK), Heidelberg, Germany
- Department of Medicine III, University Hospital Grosshadern, University of Munich, Munich, Germany
| |
Collapse
|
103
|
FoxO1-negative cells are cancer stem-like cells in pancreatic ductal adenocarcinoma. Sci Rep 2015; 5:10081. [PMID: 26068418 PMCID: PMC4464177 DOI: 10.1038/srep10081] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/20/2015] [Indexed: 12/31/2022] Open
Abstract
Flow cytometry assays using aldehyde dehydrogenase (ALDH) activity or CD133 positivity to isolate cancer stem cells (CSCs) are widely applied but have limitations. Thus, characterization of CSC makers for a specific cancer is potentially important. We have previously shown that miR-21 regulates cancer cell growth via FoxO1 in pancreatic ductal adenocarcinoma (PDAC). Here, we areported evidence of FoxO1-negative PDAC cells as CSCs in PDAC. Both ALDH-high and CD133-high cell fractions isolated from PDAC of the patients expressed high levels of miR-21 and null FoxO1. Cultured PDAC cells were virally transduced with GFP under FoxO1 promoter. GFP (FoxO1)-null PDAC cells expressed high levels of miR-21, and grew more quickly than FoxO1-positive PDAC cells. Moreover, the fold increases in growth of FoxO1-negative vs FoxO1-positive cells were greater than CD133-high vs CD133-low cells, or ALDH-high vs ALDH-low cells. Further, FoxO1-negative cells formed tumor spheres in culture and developed tumors after serial adoptive transplantation into NOD/SCID mice, while the FoxO1-positive cells did not. Finally, selective elimination of FoxO1-negative cells completely inhibited the growth of PDAC cells. Together, these data suggest that FoxO1-negative cells as CSCs in PDAC, and targeting FoxO1-negative cells in PDAC may provide better therapeutic outcome.
Collapse
|
104
|
Application of chemokine receptor antagonist with stents reduces local inflammation and suppresses cancer growth. Tumour Biol 2015; 36:8637-43. [DOI: 10.1007/s13277-015-3557-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2015] [Accepted: 05/11/2015] [Indexed: 12/19/2022] Open
|
105
|
Zhao Z, Xi H, Xu D, Li C. Transforming growth factor β receptor signaling restrains growth of pancreatic carcinoma cells. Tumour Biol 2015; 36:7711-6. [PMID: 25934336 DOI: 10.1007/s13277-015-3466-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2015] [Accepted: 04/15/2015] [Indexed: 01/04/2023] Open
Abstract
Pancreatic ductal adenocarcinoma (PDAC) is extremely malignant. Efficient control of cancer growth may substantially improve the survival of PDAC patients. However, no efficient treatments are so far available. Here, we inhibited transforming growth factor β (TGFβ) receptor signaling by overexpression of a key inhibitor of this pathway, SMAD7, in the mouse pancreas, using a recently developed intraductal infusion method. Overexpression of SMAD7 significantly increased growth of both implanted PDAC and PDAC by K-ras modification. Our data thus suggest that TGFβ receptor signaling restrains growth of PDAC, and modulation of TGFβ receptor signaling may be an effective treatment for PDAC.
Collapse
Affiliation(s)
- Zhiming Zhao
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China. .,Department of Surgical Oncology, Chinese PLA General Hospital, 28 Fuxing Road, Beijing, 100853, China.
| | - Hao Xi
- Department of Hepatobiliary Surgery, The Hospital of Shunyi District, Beijing, 101300, China
| | - Dabin Xu
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China
| | - Chenggang Li
- Department of Surgical Oncology, Chinese PLA General Hospital, Beijing, 100853, China
| |
Collapse
|
106
|
Ding H, Wang C. Role of Lgr5-positive cells in colorectal cancer. Tumour Biol 2015; 36:6759-64. [PMID: 25835970 DOI: 10.1007/s13277-015-3357-7] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2014] [Accepted: 03/18/2015] [Indexed: 01/09/2023] Open
Abstract
The molecular regulation of the growth of colorectal cancer (CRC) cells is not completely understood. Here, we report expression of Lgr5, a stem cell marker for the intestine and hair follicle, in some of the CRC cells in the patients. To determine the role of Lgr5-positive cells in the tumorigenesis of CRCs, we prepared an adeno-associated virus (AAV) that carries diphtheria toxin fragment A (DTA) under the control of Lgr5 promoter (AAV-pLgr5-DTA). Transduction of several CRC cell lines with this virus selectively killed Lgr5-positive cells, resulting in significant inhibition of the CRC cell growth in vitro and in vivo. Thus, our data highlight a potential role of Lgr5-positive cells in the tumorigenesis of CRCs, and suggest that treating these Lgr5-positive cells in CRCs may substantially improve the outcome of CRC therapy.
Collapse
Affiliation(s)
- Honghua Ding
- Department of Oncology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, 200080, China
| | - Chungang Wang
- Department of Radiation Oncology, Shanghai First People's Hospital, School of Medicine, Shanghai Jiao Tong University, 100 Haining Road, Shanghai, 200080, China.
| |
Collapse
|
107
|
|
108
|
Lu Y, Guo Y, Xu L, Li Y, Cao L. Foxp3 regulates ratio of Treg and NKT cells in a mouse model of asthma. Mol Cell Biochem 2015; 403:25-31. [PMID: 25636804 DOI: 10.1007/s11010-015-2333-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 01/16/2015] [Indexed: 12/19/2022]
Abstract
Chronic inflammatory disorder of the airways causes asthma. Regulatory T cells (Treg cells) and Natural killer T cells (NKT cells) both play critical roles in the pathogenesis of asthma. Activation of Treg cells requires Foxp3, whereas whether Foxp3 may regulate the ratio of Treg and NKT cells to affect asthma is uncertain. In an ovalbumin (OVA)-induced mouse model of asthma, we either increased Treg cells by lentivirus-mediated forced expression of exogenous Foxp3, or increased NKT cells by stimulation with its activator α-GalCer. We found that the CD4+CD25+ Treg cells increased by forced Foxp3 expression, and decreased by α-GalCer, while the CD3+CD161+ NKT cells decreased by forced Foxp3 expression, and increased by α-GalCer. Moreover, forced Foxp3 expression, but not α-GalCer, significantly alleviated the hallmarks of asthma. Furthermore, forced Foxp3 increased levels of IL_10 and TGFβ1, and α-GalCer increased levels of IL_4 and INFγ in the OVA-treated lung. Taken together, our study suggests that Foxp3 may activate Treg cells and suppress NKT cells in asthma. Treg and NKT cells may antagonize the effects of each other in asthma.
Collapse
Affiliation(s)
- Yanming Lu
- Department of Pediatrics, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 145 Shandongzhong Road, Shanghai, 200001, China,
| | | | | | | | | |
Collapse
|
109
|
Xu K, Stewart AF, Porter AC. Stimulation of oligonucleotide-directed gene correction by Redβ expression and MSH2 depletion in human HT1080 cells. Mol Cells 2015; 38:33-9. [PMID: 25431426 PMCID: PMC4314130 DOI: 10.14348/molcells.2015.2163] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 10/14/2014] [Accepted: 10/15/2014] [Indexed: 01/30/2023] Open
Abstract
The correction of disease-causing mutations by single-strand oligonucleotide-templated DNA repair (ssOR) is an attractive approach to gene therapy, but major improvements in ssOR efficiency and consistency are needed. The mechanism of ssOR is poorly understood but may involve annealing of oligonucleotides to transiently exposed single-stranded regions in the target duplex. In bacteria and yeast it has been shown that ssOR is promoted by expression of Redβ, a single-strand DNA annealing protein from bacteriophage lambda. Here we show that Redβ expression is well tolerated in a human cell line where it consistently promotes ssOR. By use of short interfering RNA, we also show that ssOR is stimulated by the transient depletion of the endogenous DNA mismatch repair protein MSH2. Furthermore, we find that the effects of Redβ expression and MSH2 depletion on ssOR can be combined with a degree of cooperativity. These results suggest that oligonucleotide annealing and mismatch recognition are distinct but interdependent events in ssOR that can be usefully modulated in gene correction strategies.
Collapse
Affiliation(s)
- Ke Xu
- Tianjin Key Laboratory of Lung Cancer Metastasis and Tumor Microenviroment, Tianjin Lung Cancer Institute, Tianjin Medical University General Hospital, Tianjin 300052,
China
- Gene Targeting Group, Department of Hematology, Faculty of Medicine, Imperial College London, London W12 0NN,
UK
| | - A. Francis Stewart
- Genomics, Bio Innovations Zentrum, Technische Universitaet Dresden, 01307 Dresden,
Germany
| | - Andrew C.G. Porter
- Gene Targeting Group, Department of Hematology, Faculty of Medicine, Imperial College London, London W12 0NN,
UK
| |
Collapse
|
110
|
Alexander IE, Russell DW. The Potential of AAV-Mediated Gene Targeting for Gene and Cell Therapy Applications. CURRENT STEM CELL REPORTS 2015. [DOI: 10.1007/s40778-014-0001-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
|
111
|
Abstract
The ability to edit the genome of cell lines has provided valuable insights into biological processes and the contribution of specific mutations to disease biology. These techniques fall into two categories based on the DNA repair mechanism that is used to incorporate the genetic change. Nuclease-based technologies, such as Zinc-Finger Nucleases, TALENS, and Crispr/Cas9, rely on non-homologous end-joining (NHEJ) and homology directed repair (HDR) to generate a range of genetic modifications. Adeno-Associated Virus (AAV) utilizes homologous recombination to generate precise and predictable genetic modifications directly at the target locus. AAV has been used to create over 500 human isogenic cell lines comprising a wide range of genetic alterations from gene knockouts, insertions of point mutations, indels, epitope tags, and reporter genes. Here we describe the generation and use of AAV gene targeting vectors and viruses to create targeted isogenic cell lines.
Collapse
Affiliation(s)
- Rob Howes
- MedImmune, Milstein Building, Granta Park, Cambridge, CB21 6GH, UK,
| | | |
Collapse
|
112
|
Stoimenov I, Ali MA, Pandzic T, Sjöblom T. Computational and molecular tools for scalable rAAV-mediated genome editing. Nucleic Acids Res 2014; 43:e30. [PMID: 25488813 PMCID: PMC4357690 DOI: 10.1093/nar/gku1286] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023] Open
Abstract
The rapid discovery of potential driver mutations through large-scale mutational analyses of human cancers generates a need to characterize their cellular phenotypes. Among the techniques for genome editing, recombinant adeno-associated virus (rAAV)-mediated gene targeting is suited for knock-in of single nucleotide substitutions and to a lesser degree for gene knock-outs. However, the generation of gene targeting constructs and the targeting process is time-consuming and labor-intense. To facilitate rAAV-mediated gene targeting, we developed the first software and complementary automation-friendly vector tools to generate optimized targeting constructs for editing human protein encoding genes. By computational approaches, rAAV constructs for editing ~71% of bases in protein-coding exons were designed. Similarly, ~81% of genes were predicted to be targetable by rAAV-mediated knock-out. A Gateway-based cloning system for facile generation of rAAV constructs suitable for robotic automation was developed and used in successful generation of targeting constructs. Together, these tools enable automated rAAV targeting construct design, generation as well as enrichment and expansion of targeted cells with desired integrations.
Collapse
Affiliation(s)
- Ivaylo Stoimenov
- Science For Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Muhammad Akhtar Ali
- Science For Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Tatjana Pandzic
- Science For Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| | - Tobias Sjöblom
- Science For Life Laboratory, Department of Immunology, Genetics and Pathology, Rudbeck Laboratory, Uppsala University, SE-751 85 Uppsala, Sweden
| |
Collapse
|
113
|
Xiao X, Guo P, Prasadan K, Shiota C, Peirish L, Fischbach S, Song Z, Gaffar I, Wiersch J, El-Gohary Y, Husain SZ, Gittes GK. Pancreatic cell tracing, lineage tagging and targeted genetic manipulations in multiple cell types using pancreatic ductal infusion of adeno-associated viral vectors and/or cell-tagging dyes. Nat Protoc 2014; 9:2719-24. [PMID: 25356582 PMCID: PMC4734891 DOI: 10.1038/nprot.2014.183] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Genetic manipulations, with or without lineage tracing for specific pancreatic cell types, are very powerful tools for studying diabetes, pancreatitis and pancreatic cancer. Nevertheless, the use of Cre/loxP systems to conditionally activate or inactivate the expression of genes in a cell type- and/or temporal-specific manner is not applicable to cell tracing and/or gene manipulations in more than one lineage at a time. Here we report a technique that allows efficient delivery of dyes for cell tagging into the mouse pancreas through the duct system, and that also delivers viruses carrying transgenes or siRNA under a specific promoter. When this technique is applied in genetically modified mice, it enables the investigator to perform either double lineage tracing or cell lineage tracing combined with gene manipulation in a second lineage. The technique requires <40 min.
Collapse
Affiliation(s)
- Xiangwei Xiao
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Ping Guo
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Krishna Prasadan
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Chiyo Shiota
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Lauren Peirish
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Shane Fischbach
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Zewen Song
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Iljana Gaffar
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - John Wiersch
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Yousef El-Gohary
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - Sohail Z Husain
- Division of Pediatrics, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| | - George K Gittes
- Division of Pediatric Surgery, Children's Hospital of Pittsburgh, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA
| |
Collapse
|
114
|
Deyle DR, Hansen RS, Cornea AM, Li LB, Burt AA, Alexander IE, Sandstrom RS, Stamatoyannopoulos JA, Wei CL, Russell DW. A genome-wide map of adeno-associated virus-mediated human gene targeting. Nat Struct Mol Biol 2014; 21:969-75. [PMID: 25282150 PMCID: PMC4405182 DOI: 10.1038/nsmb.2895] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2014] [Accepted: 08/27/2014] [Indexed: 02/03/2023]
Abstract
To determine which genomic features promote homologous recombination, we created a genome-wide map of gene targeting sites. We used an adeno-associated virus vector to target identical loci introduced as transcriptionally active retroviral vectors. A comparison of ~2,000 targeted and untargeted sites showed that targeting occurred throughout the human genome and was not influenced by the presence of nearby CpG islands, sequence repeats or DNase I-hypersensitive sites. Targeted sites were preferentially located within transcription units, especially when the target loci were transcribed in the opposite orientation to their surrounding chromosomal genes. We determined the impact of DNA replication by mapping replication forks, which revealed a preference for recombination at target loci transcribed toward an incoming fork. Our results constitute the first genome-wide screen of gene targeting in mammalian cells and demonstrate a strong recombinogenic effect of colliding polymerases.
Collapse
Affiliation(s)
- David R Deyle
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - R Scott Hansen
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Anda M Cornea
- Department of Molecular and Cellular Biology, University of Washington, Seattle, Washington, USA
| | - Li B Li
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Amber A Burt
- Department of Medicine, University of Washington, Seattle, Washington, USA
| | - Ian E Alexander
- Gene Therapy Research Unit, Children's Medical Research Institute, Westmead, New South Wales, Australia
| | - Richard S Sandstrom
- Department of Genome Sciences, University of Washington, Seattle, Washington, USA
| | | | - Chia-Lin Wei
- Genomic Technologies Department, Joint Genome Institute, Walnut Creek, California, USA
| | - David W Russell
- 1] Department of Medicine, University of Washington, Seattle, Washington, USA. [2] Department of Biochemistry, University of Washington, Seattle, Washington, USA
| |
Collapse
|
115
|
Arandjelovic P, Kaur P. The regenerative potential of epithelial stem cells in tissue repair. Int J Biochem Cell Biol 2014; 56:107-10. [PMID: 25467018 DOI: 10.1016/j.biocel.2014.10.017] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2014] [Revised: 10/03/2014] [Accepted: 10/09/2014] [Indexed: 10/24/2022]
Abstract
Acute and chronic wounds encompass devastating injuries with significant physical, emotional and economic costs at both the individual and societal level. The pathogenesis of chronic wounds is as varied as the potential causes; however, contributing factors include repetitive ischaemia/reperfusion injury coupled with bacterial infection, inflammation and matrix degradation at the wound site. Similarly, the acute physical damage of burns may leave patients vulnerable to dehydration and infection, and in certain cases this may be followed by a body-wide systemic response with debilitating consequences. Epithelial stem cells provide a promising avenue for the treatment of burns and chronic wounds. This is exemplified by recent achievements such as the restoration of corneal epithelium using limbal stem cells, and the treatment of epidermolysis bullosa via a gene therapy approach. Nevertheless, many technical and regulatory challenges remain to be addressed. This article is part of a Directed Issue entitled: Regenerative Medicine: the challenge of translation.
Collapse
Affiliation(s)
- Philip Arandjelovic
- Epithelial Stem Cell Biology Laboratory, Research Division, Peter MacCallum Cancer Centre, St Andrew's Place, Melbourne, Victoria, Australia.
| | - Pritinder Kaur
- Epithelial Stem Cell Biology Laboratory, Research Division, Peter MacCallum Cancer Centre, St Andrew's Place, Melbourne, Victoria, Australia; Sir Peter MacCallum Department of Oncology, University of Melbourne, Melbourne, Victoria, Australia; Department of Anatomy and Neuroscience, University of Melbourne, Melbourne, Victoria, Australia
| |
Collapse
|
116
|
Holkers M, Maggio I, Henriques SFD, Janssen JM, Cathomen T, Gonçalves MAFV. Adenoviral vector DNA for accurate genome editing with engineered nucleases. Nat Methods 2014; 11:1051-7. [PMID: 25152084 DOI: 10.1038/nmeth.3075] [Citation(s) in RCA: 104] [Impact Index Per Article: 10.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2014] [Accepted: 07/17/2014] [Indexed: 02/08/2023]
Abstract
Engineered sequence-specific nucleases and donor DNA templates can be customized to edit mammalian genomes via the homologous recombination (HR) pathway. Here we report that the nature of the donor DNA greatly affects the specificity and accuracy of the editing process following site-specific genomic cleavage by transcription activator-like effector nucleases (TALENs) and clustered, regularly interspaced, short palindromic repeats (CRISPR)-Cas9 nucleases. By applying these designer nucleases together with donor DNA delivered as protein-capped adenoviral vector (AdV), free-ended integrase-defective lentiviral vector or nonviral vector templates, we found that the vast majority of AdV-modified human cells underwent scarless homology-directed genome editing. In contrast, a significant proportion of cells exposed to free-ended or to covalently closed HR substrates were subjected to random and illegitimate recombination events. These findings are particularly relevant for genome engineering approaches aiming at high-fidelity genetic modification of human cells.
Collapse
Affiliation(s)
- Maarten Holkers
- 1] Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands. [2]
| | - Ignazio Maggio
- 1] Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands. [2]
| | - Sara F D Henriques
- 1] Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands. [2] Departamento de Biologia Animal, Faculdade de Ciências da Universidade de Lisboa, Lisbon, Portugal
| | - Josephine M Janssen
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| | - Toni Cathomen
- 1] Institute for Cell and Gene Therapy, University Medical Center Freiburg, Freiburg, Germany. [2] Center for Chronic Immunodeficiency, University Medical Center Freiburg, Freiburg, Germany
| | - Manuel A F V Gonçalves
- Department of Molecular Cell Biology, Leiden University Medical Center, Leiden, the Netherlands
| |
Collapse
|
117
|
Sherman SL, Curnow EC, Easley CA, Jin P, Hukema RK, Tejada MI, Willemsen R, Usdin K. Use of model systems to understand the etiology of fragile X-associated primary ovarian insufficiency (FXPOI). J Neurodev Disord 2014; 6:26. [PMID: 25147583 PMCID: PMC4139715 DOI: 10.1186/1866-1955-6-26] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 08/13/2014] [Indexed: 01/04/2023] Open
Abstract
Fragile X-associated primary ovarian insufficiency (FXPOI) is among the family of disorders caused by the expansion of a CGG repeat sequence in the 5' untranslated region of the X-linked gene FMR1. About 20% of women who carry the premutation allele (55 to 200 unmethylated CGG repeats) develop hypergonadotropic hypogonadism and cease menstruating before age 40. Some proportion of those who are still cycling show hormonal profiles indicative of ovarian dysfunction. FXPOI leads to subfertility and an increased risk of medical conditions associated with early estrogen deficiency. Little progress has been made in understanding the etiology of this clinically significant disorder. Understanding the molecular mechanisms of FXPOI requires a detailed knowledge of ovarian FMR1 mRNA and FMRP’s function. In humans, non-invasive methods to discriminate the mechanisms of the premutation on ovarian function are not available, thus necessitating the development of model systems. Vertebrate (mouse and rat) and invertebrate (Drosophila melanogaster) animal studies for the FMR1 premutation and ovarian function exist and have been instrumental in advancing our understanding of the disease phenotype. For example, rodent models have shown that FMRP is highly expressed in oocytes where it is important for folliculogenesis. The two premutation mouse models studied to date show evidence of ovarian dysfunction and, together, suggest that the long repeat in the transcript itself may have some pathological effect quite apart from any effect of the toxic protein. Further, ovarian morphology in young animals appears normal and the primordial follicle pool size does not differ from that of wild-type animals. However, there is a progressive premature decline in the levels of most follicle classes. Observations also include granulosa cell abnormalities and altered gene expression patterns. Further comparisons of these models are now needed to gain insight into the etiology of the ovarian dysfunction. Premutation model systems in non-human primates and those based on induced pluripotent stem cells show particular promise and will complement current models. Here, we review the characterization of the current models and describe the development and potential of the new models. Finally, we will discuss some of the molecular mechanisms that might be responsible for FXPOI.
Collapse
Affiliation(s)
- Stephanie L Sherman
- Department of Human Genetics, Emory University, 615 Michael St, Emory University, Atlanta, GA 30322, USA
| | - Eliza C Curnow
- Washington National Primate Center, University of Washington, Seattle, WA, USA
| | - Charles A Easley
- Laboratory of Translational Cell Biology, Department of Cell Biology, Emory University, Atlanta, GA, USA
| | - Peng Jin
- Department of Human Genetics, Emory University, 615 Michael St, Emory University, Atlanta, GA 30322, USA
| | - Renate K Hukema
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Maria Isabel Tejada
- Molecular Genetics Laboratory, Genetics Service, BioCruces Health Research Institute, Hospital Universitario Cruces, Barakaldo, Biscay, Spain
| | - Rob Willemsen
- Department of Clinical Genetics, Erasmus MC, Rotterdam, The Netherlands
| | - Karen Usdin
- Laboratory of Molecular and Cellular Biology, NIDDK, National Institutes of Health, Bethesda, MD, USA
| |
Collapse
|
118
|
Jones RE, Oh S, Grimstead JW, Zimbric J, Roger L, Heppel NH, Ashelford KE, Liddiard K, Hendrickson EA, Baird DM. Escape from telomere-driven crisis is DNA ligase III dependent. Cell Rep 2014; 8:1063-76. [PMID: 25127141 DOI: 10.1016/j.celrep.2014.07.007] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 05/28/2014] [Accepted: 07/10/2014] [Indexed: 12/27/2022] Open
Abstract
Short dysfunctional telomeres are capable of fusion, generating dicentric chromosomes and initiating breakage-fusion-bridge cycles. Cells that escape the ensuing cellular crisis exhibit large-scale genomic rearrangements that drive clonal evolution and malignant progression. We demonstrate that there is an absolute requirement for fully functional DNA ligase III (LIG3), but not ligase IV (LIG4), to facilitate the escape from a telomere-driven crisis. LIG3- and LIG4-dependent alternative (A) and classical (C) nonhomologous end-joining (NHEJ) pathways were capable of mediating the fusion of short dysfunctional telomeres, both displaying characteristic patterns of microhomology and deletion. Cells that failed to escape crisis exhibited increased proportions of C-NHEJ-mediated interchromosomal fusions, whereas those that escaped displayed increased proportions of intrachromosomal fusions. We propose that the balance between inter- and intrachromosomal telomere fusions dictates the ability of human cells to escape crisis and is influenced by the relative activities of A- and C-NHEJ at short dysfunctional telomeres.
Collapse
Affiliation(s)
- Rhiannon E Jones
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Sehyun Oh
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Julia W Grimstead
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Jacob Zimbric
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Laureline Roger
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Nicole H Heppel
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Kevin E Ashelford
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Kate Liddiard
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK
| | - Eric A Hendrickson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | - Duncan M Baird
- Institute of Cancer and Genetics, School of Medicine, Cardiff University, Heath Park, Cardiff CF14 4XN, UK.
| |
Collapse
|
119
|
Zacchigna S, Zentilin L, Giacca M. Adeno-associated virus vectors as therapeutic and investigational tools in the cardiovascular system. Circ Res 2014; 114:1827-46. [PMID: 24855205 DOI: 10.1161/circresaha.114.302331] [Citation(s) in RCA: 106] [Impact Index Per Article: 10.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
The use of vectors based on the small parvovirus adeno-associated virus has gained significant momentum during the past decade. Their high efficiency of transduction of postmitotic tissues in vivo, such as heart, brain, and retina, renders these vectors extremely attractive for several gene therapy applications affecting these organs. Besides functional correction of different monogenic diseases, the possibility to drive efficient and persistent transgene expression in the heart offers the possibility to develop innovative therapies for prevalent conditions, such as ischemic cardiomyopathy and heart failure. Therapeutic genes are not only restricted to protein-coding complementary DNAs but also include short hairpin RNAs and microRNA genes, thus broadening the spectrum of possible applications. In addition, several spontaneous or engineered variants in the virus capsid have recently improved vector efficiency and expanded their tropism. Apart from their therapeutic potential, adeno-associated virus vectors also represent outstanding investigational tools to explore the function of individual genes or gene combinations in vivo, thus providing information that is conceptually similar to that obtained from genetically modified animals. Finally, their single-stranded DNA genome can drive homology-directed gene repair at high efficiency. Here, we review the main molecular characteristics of adeno-associated virus vectors, with a particular view to their applications in the cardiovascular field.
Collapse
Affiliation(s)
- Serena Zacchigna
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.)
| | - Lorena Zentilin
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.)
| | - Mauro Giacca
- From the Molecular Medicine Laboratory, International Centre for Genetic Engineering and Biotechnology, Trieste, Italy (S.Z., L.Z., M.G.); and Department of Medical, Surgical, and Health Sciences, University of Trieste, Trieste, Italy (S.Z., M.G.).
| |
Collapse
|
120
|
Oh S, Harvey A, Zimbric J, Wang Y, Nguyen T, Jackson PJ, Hendrickson EA. DNA ligase III and DNA ligase IV carry out genetically distinct forms of end joining in human somatic cells. DNA Repair (Amst) 2014; 21:97-110. [PMID: 24837021 DOI: 10.1016/j.dnarep.2014.04.015] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2014] [Revised: 04/15/2014] [Accepted: 04/24/2014] [Indexed: 12/11/2022]
Abstract
Ku-dependent C-NHEJ (classic non-homologous end joining) is the primary DNA EJing (end joining) repair pathway in mammals. Recently, an additional EJing repair pathway (A-NHEJ; alternative-NHEJ) has been described. Currently, the mechanism of A-NHEJ is obscure although a dependency on LIGIII (DNA ligase III) is often implicated. To test the requirement for LIGIII in A-NHEJ we constructed a LIGIII conditionally-null human cell line using gene targeting. Nuclear EJing activity appeared unaffected by a deficiency in LIGIII as, surprisingly, so were random gene targeting integration events. In contrast, LIGIII was required for mitochondrial function and this defined the gene's essential activity. Human Ku:LIGIII and Ku:LIGIV (DNA ligase IV) double knockout cell lines, however, demonstrated that LIGIII is required for the enhanced A-NHEJ activity that is observed in Ku-deficient cells. Most unexpectedly, however, the majority of EJing events remained LIGIV-dependent. In conclusion, although human LIGIII has an essential function in mitochondrial maintenance, it is dispensable for most types of nuclear DSB repair, except for the A-NHEJ events that are normally suppressed by Ku. Moreover, we describe that a robust Ku-independent, LIGIV-dependent repair pathway exists in human somatic cells.
Collapse
Affiliation(s)
- Sehyun Oh
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States.
| | - Adam Harvey
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Jacob Zimbric
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States.
| | - Yongbao Wang
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States.
| | - Thanh Nguyen
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Pauline J Jackson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Eric A Hendrickson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States.
| |
Collapse
|
121
|
Kan Y, Ruis B, Lin S, Hendrickson EA. The mechanism of gene targeting in human somatic cells. PLoS Genet 2014; 10:e1004251. [PMID: 24699519 PMCID: PMC3974634 DOI: 10.1371/journal.pgen.1004251] [Citation(s) in RCA: 40] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2013] [Accepted: 02/03/2014] [Indexed: 12/24/2022] Open
Abstract
Gene targeting in human somatic cells is of importance because it can be used to either delineate the loss-of-function phenotype of a gene or correct a mutated gene back to wild-type. Both of these outcomes require a form of DNA double-strand break (DSB) repair known as homologous recombination (HR). The mechanism of HR leading to gene targeting, however, is not well understood in human cells. Here, we demonstrate that a two-end, ends-out HR intermediate is valid for human gene targeting. Furthermore, the resolution step of this intermediate occurs via the classic DSB repair model of HR while synthesis-dependent strand annealing and Holliday Junction dissolution are, at best, minor pathways. Moreover, and in contrast to other systems, the positions of Holliday Junction resolution are evenly distributed along the homology arms of the targeting vector. Most unexpectedly, we demonstrate that when a meganuclease is used to introduce a chromosomal DSB to augment gene targeting, the mechanism of gene targeting is inverted to an ends-in process. Finally, we demonstrate that the anti-recombination activity of mismatch repair is a significant impediment to gene targeting. These observations significantly advance our understanding of HR and gene targeting in human cells. Gene targeting is important for basic research and clinical applications. In the laboratory, gene targeting is used to knockout genes so that loss-of-function phenotypes can be assessed. In the clinic, gene targeting is the gold standard to which most gene therapy approaches aspire. One of the most promising tools for gene targeting in humans is recombinant adeno-associated virus (rAAV). The mechanism by which rAAV performs gene targeting has, however, remained obscure. Here, we surprisingly demonstrate that the normally single-stranded rAAV performs gene targeting via double-stranded intermediates, which are mechanistically indistinguishable from standard plasmid-mediated gene targeting. Moreover, we establish the double-strand break (DSB) repair model as the paradigm to describe human gene targeting, and delineate the dynamics of crossovers in this model. Most unexpectedly, we demonstrate that when a meganuclease is used to introduce a chromosomal DSB to augment gene targeting, the mechanism of gene targeting is inverted such that the chromosome becomes the “attacker” instead of the “attackee”. Finally, we confirm that the anti-recombination activity of mismatch repair is a significant impediment to gene targeting. These observations advance our understanding of the mechanism of human gene targeting and should readily lend themselves to developing improvements to existing methodologies.
Collapse
Affiliation(s)
- Yinan Kan
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Brian Ruis
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Sherry Lin
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
| | - Eric A. Hendrickson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, Minnesota, United States of America
- * E-mail:
| |
Collapse
|
122
|
Ojala DS, Amara DP, Schaffer DV. Adeno-associated virus vectors and neurological gene therapy. Neuroscientist 2014; 21:84-98. [PMID: 24557878 DOI: 10.1177/1073858414521870] [Citation(s) in RCA: 86] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Gene therapy has strong potential for treating a variety of genetic disorders, as demonstrated in recent clinical trials. There is unfortunately no scarcity of disease targets, and the grand challenge in this field has instead been the development of safe and efficient gene delivery platforms. To date, approximately two thirds of the 1800 gene therapy clinical trials completed worldwide have used viral vectors. Among these, adeno-associated virus (AAV) has emerged as particularly promising because of its impressive safety profile and efficiency in transducing a wide range of cell types. Gene delivery to the CNS involves both considerable promise and unique challenges, and better AAV vectors are thus needed to translate CNS gene therapy approaches to the clinic. This review discusses strategies for vector design, potential routes of administration, immune responses, and clinical applications of AAV in the CNS.
Collapse
Affiliation(s)
- David S Ojala
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA
| | - Dominic P Amara
- Department of Molecular and Cell Biology, University of California, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, CA, USA Department of Bioengineering, University of California, Berkeley, CA, USA The Helen Wills Neuroscience Institute, University of California, Berkeley, CA, USA
| |
Collapse
|
123
|
Galli A, Cervelli T. Inverted terminal repeats of adeno-associated virus decrease random integration of a gene targeting fragment in Saccharomyces cerevisiae. BMC Mol Biol 2014; 15:5. [PMID: 24521444 PMCID: PMC3925961 DOI: 10.1186/1471-2199-15-5] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2013] [Accepted: 02/06/2014] [Indexed: 12/03/2022] Open
Abstract
Background Homologous recombination mediated gene targeting is still too inefficient to be applied extensively in genomics and gene therapy. Although sequence-specific nucleases could greatly stimulate gene targeting efficiency, the off-target cleavage sites of these nucleases highlighted the risk of this strategy. Adeno-associated virus (AAV)-based vectors are used for specific gene knockouts, since several studies indicate that these vectors are able to induce site-specific genome alterations at high frequency. Since each targeted event is accompanied by at least ten random integration events, increasing our knowledge regarding the mechanisms behind these events is necessary in order to understand the potential of AAV-mediated gene targeting for therapy application. Moreover, the role of AAV regulatory proteins (Rep) and inverted terminal repeated sequences (ITRs) in random and homologous integration is not completely known. In this study, we used the yeast Saccharomyces cerevisiae as a genetic model system to evaluate whether the presence of ITRs in the integrating plasmid has an effect on gene targeting and random integration. Results We have shown that the presence of ITRs flanking a gene targeting vector containing homology to its genomic target decreased the frequency of random integration, leading to an increase in the gene targeting/random integration ratio. On the other hand, the expression of Rep proteins, which produce a nick in the ITR, significantly increased non-homologous integration of a DNA fragment sharing no homology to the genome, but had no effect on gene targeting or random integration when the DNA fragment shared homology with the genome. Molecular analysis showed that ITRs are frequently conserved in the random integrants, and that they induce rearrangements. Conclusions Our results indicate that ITRs may be a useful tool for decreasing random integration, and consequently favor homologous gene targeting.
Collapse
Affiliation(s)
- Alvaro Galli
- Yeast Genetics and Genomics Group, Institute of Clinical Physiology, CNR, via Moruzzi 1, 56125 Pisa, Italy.
| | | |
Collapse
|
124
|
Fattah FJ, Kweon J, Wang Y, Lee EH, Kan Y, Lichter N, Weisensel N, Hendrickson EA. A role for XLF in DNA repair and recombination in human somatic cells. DNA Repair (Amst) 2014; 15:39-53. [PMID: 24461734 DOI: 10.1016/j.dnarep.2013.12.006] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2013] [Revised: 11/03/2013] [Accepted: 12/10/2013] [Indexed: 01/08/2023]
Abstract
Classic non-homologous end-joining (C-NHEJ) is required for the repair of radiation-induced DNA double-strand breaks (DSBs) in mammalian cells and plays a critical role in lymphoid V(D)J recombination. A core C-NHEJ component is the DNA ligase IV co-factor, Cernunnos/XLF (hereafter XLF). In patients, mutations in XLF cause predicted increases in radiosensitivity and deficits in immune function, but also cause other less well-understood pathologies including neural disorders. To characterize XLF function(s) in a defined genetic system, we used a recombinant adeno-associated virus-mediated gene targeting strategy to inactivate both copies of the XLF locus in the human HCT116 cell line. Analyses of XLF-null cells (which were viable) showed that they were highly sensitive to ionizing radiation and a radiomimetic DNA damaging agent, etoposide. XLF-null cells had profound DNA DSB repair defects as measured by in vivo plasmid end-joining assays and were also dramatically impaired in their ability to form either V(D)J coding or signal joints on extrachromosomal substrates. Thus, our somatic XLF-null cell line recapitulates many of the phenotypes expected from XLF patient cell lines. Subsequent structure:function experiments utilizing the expression of wild-type and mutant XLF cDNAs demonstrated that all of the phenotypes of an XLF deficiency could be rescued by the overexpression of a wild-type XLF cDNA. Unexpectedly, mutant forms of XLF bearing point mutations at amino acid positions L115 and L179, also completely complemented the null phenotype suggesting, in contrast to predictions to the contrary, that these mutations do not abrogate XLF function. Finally, we demonstrate that the absence of XLF causes a small, but significant, increase in homologous recombination, implicating XLF in DSB pathway choice regulation. We conclude that human XLF is a non-essential, but critical, C-NHEJ-repair factor.
Collapse
Affiliation(s)
- Farjana Jahan Fattah
- Departments of Pharmacology and Radiation Oncology, Simmons Cancer Center, University of Texas Southwestern Medical Center, Dallas, TX 75390, United States.
| | - Junghun Kweon
- Department of Pediatrics, Section of Cardiology, University of Chicago, 900 East 57th Street, KCBD Room 5240, Chicago, IL 60637, United States.
| | - Yongbao Wang
- Cancer Diagnostics Service, Quest Diagnostics Nichols Institute, Chantilly, VA 20151, United States.
| | - Eu Han Lee
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Yinan Kan
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States
| | - Natalie Lichter
- University of ND School of Medicine, 501 Columbia Road, Grand Forks, ND 58203, United States.
| | - Natalie Weisensel
- University of Wisconsin School of Medicine and Public Health, Health Sciences Learning Center, 750 Highland Ave., Madison, WI 53705, United States.
| | - Eric A Hendrickson
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, United States.
| |
Collapse
|
125
|
Somatic correction of junctional epidermolysis bullosa by a highly recombinogenic AAV variant. Mol Ther 2014; 22:725-33. [PMID: 24390279 PMCID: PMC3982486 DOI: 10.1038/mt.2013.290] [Citation(s) in RCA: 53] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Accepted: 12/17/2013] [Indexed: 12/29/2022] Open
Abstract
Definitive correction of disease causing mutations in somatic cells by homologous recombination (HR) is an attractive therapeutic approach for the treatment of genetic diseases. However, HR-based somatic gene therapy is limited by the low efficiency of gene targeting in mammalian cells and replicative senescence of primary cells ex vivo, forcing investigators to explore alternative strategies such as retro- and lentiviral gene transfer, or genome editing in induced pluripotent stem cells. Here, we report correction of mutations at the LAMA3 locus in primary keratinocytes derived from a patient affected by recessive inherited Herlitz junctional epidermolysis bullosa (H-JEB) disorder using recombinant adenoassociated virus (rAAV)-mediated HR. We identified a highly recombinogenic AAV serotype, AAV-DJ, that mediates efficient gene targeting in keratinocytes at clinically relevant frequencies with a low rate of random integration. Targeted H-JEB patient cells were selected based on restoration of adhesion phenotype, which eliminated the need for foreign sequences in repaired cells, enhancing the clinical use and safety profile of our approach. Corrected pools of primary cells assembled functional laminin-332 heterotrimer and fully reversed the blistering phenotype both in vitro and in skin grafts. The efficient targeting of the LAMA3 locus by AAV-DJ using phenotypic selection, together with the observed low frequency of off-target events, makes AAV-DJ based somatic cell targeting a promising strategy for ex vivo therapy for this severe and often lethal epithelial disorder.
Collapse
|
126
|
Deyle DR, Li LB, Ren G, Russell DW. The effects of polymorphisms on human gene targeting. Nucleic Acids Res 2013; 42:3119-24. [PMID: 24371280 PMCID: PMC3950700 DOI: 10.1093/nar/gkt1303] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/03/2022] Open
Abstract
DNA mismatches that occur between vector homology arms and chromosomal target sequences reduce gene targeting frequencies in several species; however, this has not been reported in human cells. Here we demonstrate that even a single mismatched base pair can significantly decrease human gene targeting frequencies. In addition, we show that homology arm polymorphisms can be used to direct allele-specific targeting or to improve unfavorable vector designs that introduce deletions.
Collapse
Affiliation(s)
- David R Deyle
- Department of Medicine, University of Washington, Seattle, WA, 98195, USA and Department of Biochemistry, University of Washington, Seattle, WA, 98195, USA
| | | | | | | |
Collapse
|
127
|
Smuder AJ, Falk DJ, Sollanek KJ, Nelson WB, Powers SK. Delivery of recombinant adeno-associated virus vectors to rat diaphragm muscle via direct intramuscular injection. Hum Gene Ther Methods 2013; 24:364-71. [PMID: 24006956 PMCID: PMC3869534 DOI: 10.1089/hgtb.2013.055] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Accepted: 09/04/2013] [Indexed: 01/14/2023] Open
Abstract
The diaphragm is the most important inspiratory muscle in all mammals, and ventilatory insufficiency caused by diaphragm dysfunction is the leading cause of morbidity and mortality in many genetic and acquired diseases affecting skeletal muscle. Currently, pharmacological inhibitors, genetically modified animals, and invasive procedures are used to study disorders affecting the diaphragm. However, these methodologies can be problematic because of off-target drug effects and the possible nonphysiological consequences of lifelong genetic alterations. Therefore, alternative methods to study this important respiratory muscle are needed. To resolve this, we have developed a methodology to deliver recombinant adeno-associated virus (rAAV) vectors to the rat diaphragm via direct intramuscular injection. We hypothesized that by direct injection of rAAV into the muscle we can selectively target the diaphragm and establish a novel experimental method for studying signaling pathways and also provide a strategy for effectively using rAAV to protect the diaphragm against disease. This report describes the methods and evidence to support the use of rAAV as a therapeutic intervention to study rat diaphragm biology during conditions that promote diaphragm dysfunction.
Collapse
Affiliation(s)
- Ashley J. Smuder
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL 32611
| | - Darin J. Falk
- Department of Pediatrics, Powell Gene Therapy Center, University of Florida, Gainesville, FL 32611
| | - Kurt J. Sollanek
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL 32611
| | - W. Bradley Nelson
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL 32611
| | - Scott K. Powers
- Department of Applied Physiology and Kinesiology, Center for Exercise Science, University of Florida, Gainesville, FL 32611
| |
Collapse
|
128
|
Burbacher TM, Grant KS, Worlein J, Ha J, Curnow E, Juul S, Sackett GP. Four decades of leading-edge research in the reproductive and developmental sciences: the Infant Primate Research Laboratory at the University of Washington National Primate Research Center. Am J Primatol 2013; 75:1063-83. [PMID: 23873400 PMCID: PMC5452618 DOI: 10.1002/ajp.22175] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2013] [Revised: 05/31/2013] [Accepted: 05/31/2013] [Indexed: 12/20/2022]
Abstract
The Infant Primate Research Laboratory (IPRL) was established in 1970 at the University of Washington as a visionary project of Dr. Gene (Jim) P. Sackett. Supported by a collaboration between the Washington National Primate Research Center and the Center on Human Development and Disability, the IPRL operates under the principle that learning more about the causes of abnormal development in macaque monkeys will provide important insights into the origins and treatment of childhood neurodevelopmental disabilities. Over the past 40 years, a broad range of research projects have been conducted at the IPRL. Some have described the expression of normative behaviors in nursery-reared macaques while others have focused on important biomedical themes in child health and development. This article details the unique scientific history of the IPRL and the contributions produced by research conducted in the laboratory. Past and present investigations have explored the topics of early rearing effects, low-birth-weight, prematurity, birth injury, epilepsy, prenatal neurotoxicant exposure, viral infection (pediatric HIV), diarrheal disease, vaccine safety, and assisted reproductive technologies. Data from these studies have helped advance our understanding of both risk and resiliency in primate development. New directions of research at the IPRL include the production of transgenic primate models using our embryonic stem cell-based technology to better understand and treat heritable forms of human intellectual disabilities such as fragile X.
Collapse
Affiliation(s)
- Thomas M. Burbacher
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, 98195 USA
- Center on Human Development and Disability, University of Washington, Seattle, WA, 98195 USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
| | - Kimberly S. Grant
- Department of Environmental and Occupational Health Sciences, School of Public Health, University of Washington, Seattle, WA, 98195 USA
- Center on Human Development and Disability, University of Washington, Seattle, WA, 98195 USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
| | - Julie Worlein
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
| | - James Ha
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
- Department of Psychology, School of Arts and Sciences, University of Washington, Seattle, WA, 98195 USA
| | - Eliza Curnow
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
| | - Sandra Juul
- Center on Human Development and Disability, University of Washington, Seattle, WA, 98195 USA
- Department of Pediatrics, School of Medicine, University of Washington, Seattle, WA, 98195 USA
| | - Gene P. Sackett
- Center on Human Development and Disability, University of Washington, Seattle, WA, 98195 USA
- Washington National Primate Research Center, University of Washington, Seattle, WA 98195 USA
- Department of Psychology, School of Arts and Sciences, University of Washington, Seattle, WA, 98195 USA
| |
Collapse
|
129
|
Brafman DA, Moya N, Allen-Soltero S, Fellner T, Robinson M, McMillen ZL, Gaasterland T, Willert K. Analysis of SOX2-expressing cell populations derived from human pluripotent stem cells. Stem Cell Reports 2013; 1:464-78. [PMID: 24286033 PMCID: PMC3841266 DOI: 10.1016/j.stemcr.2013.09.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/22/2013] [Revised: 09/24/2013] [Accepted: 09/25/2013] [Indexed: 01/20/2023] Open
Abstract
SOX2 is involved in several cell and developmental processes, including maintenance of embryonic stem cells, differentiation of neural progenitor cells, and patterning of gut endoderm. To study its role in a human system, we generated a human embryonic stem cell (hESC) line harboring a reporter gene encoding GFP in the SOX2 locus. This SOX2 reporter line faithfully recapitulates expression of the SOX2 gene in undifferentiated human pluripotent stem cells (hPSCs), neural progenitor cells (NPCs), and anterior foregut endoderm (AFE). In undifferentiated hESCs, GFP expression corresponds to those cells with highest levels of expression of genes associated with the pluripotent state. In NPCs, expression of GFP can be employed to isolate cells expressing markers associated with NPC multipotency. In AFE, we used transcriptome-wide expression analysis to identify cell surface markers with elevated expression in this population, thereby facilitating isolation and purification of this hPSC-derived cell population. A SOX2-GFP hESC line is used to isolate and characterize multiple cell types A cell surface marker signature allows for the purification of endodermal progeny Targeted gene insertion with adeno-associated virus (AAV) is highly efficient
Collapse
Affiliation(s)
- David A Brafman
- Stem Cell Program, Department of Cellular and Molecular Medicine, UCSD, 9500 Gilman Drive, La Jolla, CA 92093-0695, USA
| | | | | | | | | | | | | | | |
Collapse
|
130
|
Ramamoorthi K, Curtis D, Asuri P. Advances in homology directed genetic engineering of human pluripotent and adult stem cells. World J Stem Cells 2013; 5:98-105. [PMID: 24179598 PMCID: PMC3812527 DOI: 10.4252/wjsc.v5.i4.98] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2013] [Revised: 09/06/2013] [Accepted: 09/17/2013] [Indexed: 02/06/2023] Open
Abstract
The ability to introduce precise genomic modifications in human cells has profound implications for both basic and applied research in stem cells, ranging from identification of genes regulating stem cell self-renewal and multilineage differentiation to therapeutic gene correction and creation of in vitro models of human diseases. However, the overall efficiency of this process is challenged by several factors including inefficient gene delivery into stem cells and low rates of homology directed site-specific targeting. Recent studies report the development of novel techniques to improve gene targeting efficiencies in human stem cells; these methods include molecular engineering of viral vectors to efficiently deliver episomal genetic sequences that can participate in homology directed targeting, as well as the design of synthetic proteins that can introduce double-stranded breaks in DNA to initiate such recombination events. This review focuses on the potential of these new technologies to precisely alter the human stem cell genome and also highlights the possibilities offered by the combination of these complementary strategies.
Collapse
|
131
|
Opportunities and challenges of pluripotent stem cell neurodegenerative disease models. Nat Neurosci 2013; 16:780-9. [PMID: 23799470 DOI: 10.1038/nn.3425] [Citation(s) in RCA: 152] [Impact Index Per Article: 13.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2013] [Accepted: 05/05/2013] [Indexed: 02/06/2023]
Abstract
Human neurodegenerative disorders are among the most difficult to study. In particular, the inability to readily obtain the faulty cell types most relevant to these diseases has impeded progress for decades. Recent advances in pluripotent stem cell technology now grant access to substantial quantities of disease-pertinent neurons both with and without predisposing mutations. While this suite of technologies has revolutionized the field of 'in vitro disease modeling', great care must be taken in their deployment if robust, durable discoveries are to be made. Here we review what we perceive to be several of the stumbling blocks in the use of stem cells for the study of neurological disease and offer strategies to overcome them.
Collapse
|
132
|
Abstract
The clinical use of human pluripotent stem cells and their derivatives is limited by the rejection of transplanted cells due to differences in their human leukocyte antigen (HLA) genes. This has led to the proposed use of histocompatible, patient-specific stem cells; however, the preparation of many different stem cell lines for clinical use is a daunting task. Here, we develop two distinct genetic engineering approaches that address this problem. First, we use a combination of gene targeting and mitotic recombination to derive HLA-homozygous embryonic stem cell (ESC) subclones from an HLA-heterozygous parental line. A small bank of HLA-homozygous stem cells with common haplotypes would match a significant proportion of the population. Second, we derive HLA class I-negative cells by targeted disruption of both alleles of the Beta-2 Microglobulin (B2M) gene in ESCs. Mixed leukocyte reactions and peptide-specific HLA-restricted CD8(+) T cell responses were reduced in class I-negative cells that had undergone differentiation in embryoid bodies. These B2M(-/-) ESCs could act as universal donor cells in applications where the transplanted cells do not express HLA class II genes. Both approaches used adeno-associated virus (AAV) vectors for efficient gene targeting in the absence of potentially genotoxic nucleases, and produced pluripotent, transgene-free cell lines.
Collapse
|
133
|
Modest DP, Camaj P, Heinemann V, Schwarz B, Jung A, Laubender RP, Gamba S, Haertl C, Stintzing S, Primo S, Bruns CJ. KRAS allel-specific activity of sunitinib in an isogenic disease model of colorectal cancer. J Cancer Res Clin Oncol 2013; 139:953-61. [DOI: 10.1007/s00432-013-1401-9] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2012] [Accepted: 02/11/2013] [Indexed: 01/13/2023]
|
134
|
Nuclease Mediated Targeted Genome Modification in Mammalian Cells. SITE-DIRECTED INSERTION OF TRANSGENES 2013. [DOI: 10.1007/978-94-007-4531-5_13] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
135
|
Oh S, Wang Y, Zimbric J, Hendrickson EA. Human LIGIV is synthetically lethal with the loss of Rad54B-dependent recombination and is required for certain chromosome fusion events induced by telomere dysfunction. Nucleic Acids Res 2012; 41:1734-49. [PMID: 23275564 PMCID: PMC3561972 DOI: 10.1093/nar/gks1326] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Classic non-homologous end joining (C-NHEJ) is the predominant DNA double-strand break repair pathway in humans. Although seven genes Ku70, Ku86, DNA-PKcs, Artemis, DNA Ligase IV (LIGIV), X-ray cross-complementing group 4 and XRCC4-like factor are required for C-NHEJ, several of them also have ancillary functions. For example, Ku70:Ku86 possesses an essential telomere maintenance activity. In contrast, LIGIV is believed to function exclusively in C-NHEJ. Moreover, a viable LIGIV-null human B-cell line and LIGIV-reduced patient cell lines have been described. Together, these observations suggest that LIGIV (and hence C-NHEJ), albeit important, is nonetheless dispensable, whereas Ku70:Ku86 and telomere maintenance are essential. To confirm this hypothesis, we inactivated LIGIV in the epithelial human cell line, HCT116. The resulting LIGIV-null cell line was viable, verifying that the gene and C-NHEJ are not essential. However, functional inactivation of RAD54B, a key homologous recombination factor, in the LIGIV-null background yielded no viable clones, suggesting that the combined absence of RAD54B/homologous recombination and C-NHEJ is synthetically lethal. Finally, we demonstrate that LIGIV is differentially required for certain chromosome fusion events induced by telomere dysfunction—used for those owing to the overexpression of a dominant negative version of telomere recognition factor 2, but not used for those owing to absence of Ku70:Ku86.
Collapse
Affiliation(s)
- Sehyun Oh
- Department of Biochemistry, Molecular Biology, and Biophysics, University of Minnesota Medical School, Minneapolis, MN 55455, USA
| | | | | | | |
Collapse
|
136
|
Yan C, Higgins PJ. Drugging the undruggable: transcription therapy for cancer. Biochim Biophys Acta Rev Cancer 2012; 1835:76-85. [PMID: 23147197 DOI: 10.1016/j.bbcan.2012.11.002] [Citation(s) in RCA: 68] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2012] [Revised: 10/30/2012] [Accepted: 11/01/2012] [Indexed: 11/19/2022]
Abstract
Transcriptional regulation is often the convergence point of oncogenic signaling. It is not surprising, therefore, that aberrant gene expression is a hallmark of cancer. Transformed cells often develop a dependency on such a reprogramming highlighting the therapeutic potential of rectifying cancer-associated transcriptional abnormalities in malignant cells. Although transcription is traditionally considered as undruggable, agents have been developed that target various levels of transcriptional regulation including DNA binding by transcription factors, protein-protein interactions, and epigenetic alterations. Some of these agents have been approved for clinical use or entered clinical trials. While artificial transcription factors have been developed that can theoretically modulate expression of any given gene, the emergence of reliable reporter assays greatly facilitates the search for transcription-targeted agents. This review provides a comprehensive overview of these developments, and discusses various strategies applicable for developing transcription-targeted therapeutic agents.
Collapse
Affiliation(s)
- Chunhong Yan
- Center for Cell Biology and Cancer Research, Albany Medical College, MC-165, 47 New Scotland Avenue, Albany, NY 12208, USA.
| | | |
Collapse
|
137
|
Karnan S, Konishi Y, Ota A, Takahashi M, Damdindorj L, Hosokawa Y, Konishi H. Simple monitoring of gene targeting efficiency in human somatic cell lines using the PIGA gene. PLoS One 2012; 7:e47389. [PMID: 23056640 PMCID: PMC3466256 DOI: 10.1371/journal.pone.0047389] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2012] [Accepted: 09/11/2012] [Indexed: 12/31/2022] Open
Abstract
Gene targeting in most of human somatic cell lines has been labor-intensive because of low homologous recombination efficiency. The development of an experimental system that permits a facile evaluation of gene targeting efficiency in human somatic cell lines is the first step towards the improvement of this technology and its application to a broad range of cell lines. In this study, we utilized phosphatidylinositol glycan anchor biosynthesis class A (PIGA), a gene essential for the synthesis of glycosylphosphatidyl inositol (GPI) anchors, as a reporter of gene targeting events in human somatic cell lines. Targeted disruption of PIGA was quantitatively detected with FLAER, a reagent that specifically binds to GPI anchors. Using this PIGA-based reporter system, we successfully detected adeno-associated virus (AAV)-mediated gene targeting events both with and without promoter-trap enrichment of gene-targeted cell population. The PIGA-based reporter system was also capable of reproducing previous findings that an AAV-mediated gene targeting achieves a remarkably higher ratio of homologous versus random integration (H/R ratio) of targeting vectors than a plasmid-mediated gene targeting. The PIGA-based system also detected an approximately 2-fold increase in the H/R ratio achieved by a small negative selection cassette introduced at the end of the AAV-based targeting vector with a promoter-trap system. Thus, our PIGA-based system is useful for monitoring AAV-mediated gene targeting and will assist in improving gene targeting technology in human somatic cell lines.
Collapse
Affiliation(s)
- Sivasundaram Karnan
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yuko Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Akinobu Ota
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Miyuki Takahashi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Lkhagvasuren Damdindorj
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Yoshitaka Hosokawa
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | - Hiroyuki Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
- * E-mail:
| |
Collapse
|
138
|
Konishi Y, Karnan S, Takahashi M, Ota A, Damdindorj L, Hosokawa Y, Konishi H. A system for the measurement of gene targeting efficiency in human cell lines using an antibiotic resistance-GFP fusion gene. Biotechniques 2012; 53:141-52. [PMID: 22963476 DOI: 10.2144/0000113911] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2012] [Accepted: 07/25/2012] [Indexed: 11/23/2022] Open
Abstract
Gene targeting in a broad range of human somatic cell lines has been hampered by inefficient homologous recombination. To improve this technology and facilitate its widespread application, it is critical to first have a robust and efficient research system for measuring gene targeting efficiency. Here, using a fusion gene consisting of hygromycin B phosphotransferase and 3'-truncated enhanced GFP (HygR-5' EGFP) as a reporter gene, we created a molecular system monitoring the ratio of homologous to random integration (H/R ratio) of targeting vectors into the genome. Cell clones transduced with a reporter vector containing HygR-5' EGFP were efficiently established from two human somatic cell lines. Established HygR-5' EGFP reporter clones retained their capacity to monitor gene targeting efficiency for a longer duration than a conventional reporter system using an unfused 5' EGFP gene. With the HygR-5' EGFP reporter system, we reproduced previous findings of gene targeting frequency being up-regulated by the use of an adeno-associated viral (AAV) backbone, a promoter-trap system, or a longer homology arm in a targeting vector, suggesting that this system accurately monitors H/R ratio. Thus, our HygR-5' EGFP reporter system will assist in the development of an efficient AAV-based gene targeting technology.
Collapse
Affiliation(s)
- Yuko Konishi
- Department of Biochemistry, Aichi Medical University School of Medicine, Nagakute, Aichi, Japan
| | | | | | | | | | | | | |
Collapse
|
139
|
Affiliation(s)
- William G Kaelin
- Department of Adult Oncology, Office M457, Dana-Farber Cancer Institute, 450 Brookline Avenue, Boston, MA 02215, USA.
| |
Collapse
|
140
|
Lin SY, Li TY, Liu Q, Zhang C, Li X, Chen Y, Zhang SM, Lian G, Liu Q, Ruan K, Wang Z, Zhang CS, Chien KY, Wu J, Li Q, Han J, Lin SC. GSK3-TIP60-ULK1 Signaling Pathway Links Growth Factor Deprivation to Autophagy. Science 2012; 336:477-81. [DOI: 10.1126/science.1217032] [Citation(s) in RCA: 265] [Impact Index Per Article: 22.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
141
|
Mata JF, Lopes T, Gardner R, Jansen LET. A rapid FACS-based strategy to isolate human gene knockin and knockout clones. PLoS One 2012; 7:e32646. [PMID: 22393430 PMCID: PMC3290580 DOI: 10.1371/journal.pone.0032646] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2011] [Accepted: 01/28/2012] [Indexed: 11/18/2022] Open
Abstract
Gene targeting protocols for mammalian cells remain inefficient and labor intensive. Here we describe FASTarget, a rapid, fluorescent cell sorting based strategy to isolate rare gene targeting events in human somatic cells. A fluorescent protein is used as a means for direct selection of targeted clones obviating the need for selection and outgrowth of drug resistant clones. Importantly, the use of a promoter-less, ATG-less construct greatly facilitates the recovery of correctly targeted cells. Using this method we report successful gene targeting in up to 94% of recovered human somatic cell clones. We create functional EYFP-tagged knockin clones in both transformed and non-transformed human somatic cell lines providing a valuable tool for mammalian cell biology. We further demonstrate the use of this technology to create gene knockouts. Using this generally applicable strategy we can recover gene targeted clones within approximately one month from DNA construct delivery to obtaining targeted monoclonal cell lines.
Collapse
Affiliation(s)
- João F. Mata
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Telma Lopes
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | - Rui Gardner
- Instituto Gulbenkian de Ciência, Oeiras, Portugal
| | | |
Collapse
|
142
|
Bordignon V, Cordiali-Fei P, Rinaldi M, Signori E, Cottarelli A, Zonfrillo M, Ensoli F, Rasi G, Fuggetta MP. Evaluation of antigen specific recognition and cell mediated cytotoxicity by a modified lysispot assay in a rat colon carcinoma model. JOURNAL OF EXPERIMENTAL & CLINICAL CANCER RESEARCH : CR 2012; 31:9. [PMID: 22296726 PMCID: PMC3395825 DOI: 10.1186/1756-9966-31-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/02/2011] [Accepted: 02/01/2012] [Indexed: 12/29/2022]
Abstract
Background Antigen-specific CD8+ cytotoxic T lymphocytes represent potent effector cells of the adaptive immune response against viruses as well as tumours. Therefore assays capable at exploring the generation and function of cytotoxic T lymphocytes represent an important objective for both clinical and experimental settings. Methods Here we show a simple and reproducible assay for the evaluation of antigen-specific CD8+ cytotoxic T lymphocytes based on a LysiSpot technique for the simultaneous determination of antigen-specific IFN-γ production and assessment of tumor cytolysis. The assay was developed within an experimental model of colorectal carcinoma, induced by the colorectal tumor cell line DHD-K12 that induces tumors in BDIX rats and, in turn, elicits a tumor- specific immune response. Results Using DHD-K12 cells transfected to express Escherichia coli β-galactosidase as target cells, and by the fine setting of spot colours detection, we have developed an in vitro assay that allows the recognition of cytotoxic T lymphocytes induced in BDIX rats as well as the assessment of anti-tumour cytotoxicity. The method highlighted that in the present experimental model the tumour antigen-specific immune response was bound to killing target cells in the proportion of 55%, while 45% of activated cells were not cytotoxic but released IFN-γ. Moreover in this model by an ELISPOT assay we demonstrated the specific recognition of a nonapeptide epitope called CSH-275 constitutionally express in DHD-K12 cells. Conclusions The assay proved to be highly sensitive and specific, detecting even low frequencies of cytotoxic/activated cells and providing the evaluation of cytokine-expressing T cells as well as the extent of cytotoxicity against the target cells as independent functions. This assay may represent an important tool to be adopted in experimental settings including the development of vaccines or immune therapeutic strategies
Collapse
Affiliation(s)
- Valentina Bordignon
- Laboratory of Clinical Pathology and Microbiology, San Gallicano Dermatologic Institute, Via Elio Chianesi, 53, 00144 Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
143
|
Asuri P, Bartel MA, Vazin T, Jang JH, Wong TB, Schaffer DV. Directed evolution of adeno-associated virus for enhanced gene delivery and gene targeting in human pluripotent stem cells. Mol Ther 2012; 20:329-38. [PMID: 22108859 PMCID: PMC3277219 DOI: 10.1038/mt.2011.255] [Citation(s) in RCA: 97] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2011] [Accepted: 10/27/2011] [Indexed: 12/12/2022] Open
Abstract
Efficient approaches for the precise genetic engineering of human pluripotent stem cells (hPSCs) can enhance both basic and applied stem cell research. Adeno- associated virus (AAV) vectors are of particular interest for their capacity to mediate efficient gene delivery to and gene targeting in various cells. However, natural AAV serotypes offer only modest transduction of human embryonic and induced pluripotent stem cells (hESCs and hiPSCs), which limits their utility for efficiently manipulating the hPSC genome. Directed evolution is a powerful means to generate viral vectors with novel capabilities, and we have applied this approach to create a novel AAV variant with high gene delivery efficiencies (~50%) to hPSCs, which are importantly accompanied by a considerable increase in gene-targeting frequencies, up to 0.12%. While this level is likely sufficient for numerous applications, we also show that the gene-targeting efficiency mediated by an evolved AAV variant can be further enhanced (>1%) in the presence of targeted double- stranded breaks (DSBs) generated by the co-delivery of artificial zinc finger nucleases (ZFNs). Thus, this study demonstrates that under appropriate selective pressures, AAV vectors can be created to mediate efficient gene targeting in hPSCs, alone or in the presence of ZFN- mediated double-stranded DNA breaks.
Collapse
Affiliation(s)
- Prashanth Asuri
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, California 94720-1462, USA
| | | | | | | | | | | |
Collapse
|
144
|
Abstract
Transcription activator-like effectors (TALEs) are a class of naturally occurring DNA-binding proteins found in the plant pathogen Xanthomonas sp. The DNA-binding domain of each TALE consists of tandem 34-amino acid repeat modules that can be rearranged according to a simple cipher to target new DNA sequences. Customized TALEs can be used for a wide variety of genome engineering applications, including transcriptional modulation and genome editing. Here we describe a toolbox for rapid construction of custom TALE transcription factors (TALE-TFs) and nucleases (TALENs) using a hierarchical ligation procedure. This toolbox facilitates affordable and rapid construction of custom TALE-TFs and TALENs within 1 week and can be easily scaled up to construct TALEs for multiple targets in parallel. We also provide details for testing the activity in mammalian cells of custom TALE-TFs and TALENs using quantitative reverse-transcription PCR and Surveyor nuclease, respectively. The TALE toolbox described here will enable a broad range of biological applications.
Collapse
|
145
|
Chun YS, Byun K, Lee B. Induced pluripotent stem cells and personalized medicine: current progress and future perspectives. Anat Cell Biol 2011; 44:245-55. [PMID: 22254153 PMCID: PMC3254878 DOI: 10.5115/acb.2011.44.4.245] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2011] [Revised: 12/16/2011] [Accepted: 12/21/2011] [Indexed: 01/26/2023] Open
Abstract
Generation of induced pluripotent stem cells (iPSCs) has revolutionized the field of regenerative medicine by providing researchers with a unique tool to derive disease-specific stem cells for study. iPSCs can self-renew and can differentiate into many cell types, offering a potentially unlimited source of cells for targeted differentiation into somatic effector cells. Hence, iPSCs are likely to be invaluable for therapeutic applications and disease-related research. In this review, we summarize the recent progress of iPSC generation that has been made with an emphasis on both basic and clinical applications including disease modeling, drug toxicity screening/drug discovery and cell replacement therapy.
Collapse
Affiliation(s)
- Yong Soon Chun
- Department of Surgery, Gachon University Gil Hospital, Incheon, Korea
| | | | | |
Collapse
|
146
|
Efficient and accurate homologous recombination in hESCs and hiPSCs using helper-dependent adenoviral vectors. Mol Ther 2011; 20:424-31. [PMID: 22146343 DOI: 10.1038/mt.2011.266] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
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.
Collapse
|
147
|
Mansilla-Soto J, Rivière I, Sadelain M. Genetic strategies for the treatment of sickle cell anaemia. Br J Haematol 2011; 154:715-27. [DOI: 10.1111/j.1365-2141.2011.08773.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
|