1
|
Moazzeni S, Kyker-Snowman K, Cohen RI, Wang H, Li R, Shreiber DI, Zahn JD, Shi Z, Lin H. N-Cadherin based adhesion and Rac1 activity regulate tension polarization in the actin cortex. Sci Rep 2025; 15:4296. [PMID: 39905109 PMCID: PMC11794589 DOI: 10.1038/s41598-025-88537-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2024] [Accepted: 01/29/2025] [Indexed: 02/06/2025] Open
Abstract
Tension-adhesion interplay is a crucial mechanism in multicellular organisms that determines the tension differential among internal and external interfaces, which in turn, mediates tissue surface tension and cell sorting, morphogenesis and remodeling, and cancer progression. Cadherins are widely believed to be involved, yet key aspects of the process are neither well characterized nor quantified. This study demonstrates the critical role of N-cadherin in driving tension polarization throughout the actin cortical network. N-cadherin regulates both tension increase at the cell-medium (external) interface and decrease at the cell-cell (internal) interface, and their quantitative magnitudes, both absolute and relative, strongly depend on the surface density of N-cadherin. Furthermore, the strength of tension polarization also increases with respect to the number of cell-cell interfaces for cells within a multicellular cluster. The cadherin-actin contractility linkage is mediated by Rac1, which serves as a molecular switch to trigger cortex remodeling and contraction via myosin II. Inhibition of Rac1 activity decreases tension polarization and leads to reduced coherence in both small clusters and spheroids. These results provide a pathway to reconcile opposing theories for tissue surface tension generation and perspectives in cancer treatment.
Collapse
Affiliation(s)
- Seyedsajad Moazzeni
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, 98 Brett Rd, Piscataway, NJ, 08854, USA
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Kelly Kyker-Snowman
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Rick I Cohen
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Huan Wang
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Rd, Piscataway, NJ, 08854, USA
| | - Ran Li
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, 98 Brett Rd, Piscataway, NJ, 08854, USA
| | - David I Shreiber
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Jeffrey D Zahn
- Department of Biomedical Engineering, Rutgers, The State University of New Jersey, 599 Taylor Rd, Piscataway, NJ, 08854, USA
| | - Zheng Shi
- Department of Chemistry and Chemical Biology, Rutgers, The State University of New Jersey, 123 Bevier Rd, Piscataway, NJ, 08854, USA.
| | - Hao Lin
- Department of Mechanical & Aerospace Engineering, Rutgers, The State University of New Jersey, 98 Brett Rd, Piscataway, NJ, 08854, USA.
| |
Collapse
|
2
|
Matsubara J, Li YF, Koul S, Mukohyama J, Valencia Salazar LE, Isobe T, Qian D, Clarke MF, Sahoo D, Altman RB, Dalerba P. The E2F4 transcriptional repressor is a key mechanistic regulator of colon cancer resistance to irinotecan (CPT-11). BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.22.633435. [PMID: 39896677 PMCID: PMC11785039 DOI: 10.1101/2025.01.22.633435] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2025]
Abstract
Background. Colorectal carcinomas (CRCs) are seldom eradicated by cytotoxic chemotherapy. Cancer cells with stem-like functional properties, often referred to as "cancer stem cells" (CSCs), display preferential resistance to several anti-tumor agents used in cancer chemotherapy, but the molecular mechanisms underpinning their selective survival remain only partially understood. Methods. In this study, we used Transcription Factor Target Genes (TFTG) enrichment analysis to identify transcriptional regulators (activators or repressors) that undergo preferential activation by chemotherapy in CRC cells with a "bottom-of-the-crypt" phenotype (EPCAM + /CD44 + /CD166 + ; CSC-enriched) as compared to CRC cells with a "top-of-the-crypt" phenotype (EPCAM + /CD44 neg /CD166 neg ; CSC-depleted). The two cell populations were purified in parallel by fluorescence-activated cell sorting (FACS) from a patient-derived xenograft (PDX) line representative of a moderately differentiated human CRC, following in vivo chemotherapy with irinotecan (CPT-11). The transcriptional regulators identified as differentially activated were tested for differential expression in normal vs. cancer tissues, and in cell populations enriched in stem/progenitor cell-types as compared to differentiated lineages (goblet cells, enterocytes) in the mouse colon epithelium. Finally, the top candidate was tested for mechanistic contribution to drug-resistance by selective down-regulation using short-hairpin RNAs (shRNAs). Results. Our analysis identified E2F4 and TFDP1, two core components of the DREAM transcriptional repression complex, as transcriptional modulators preferentially activated by irinotecan in EPCAM + /CD44 + /CD166 + as compared to EPCAM + /CD44 neg /CD166 neg cancer cells. The expression levels of both genes ( E2F4 , TFDP1 ) were found up-regulated in CRCs as compared to human normal colon tissues, and in a sub-population of mouse colon epithelial cells enriched in stem/progenitor elements (Epcam + /Cd44 + /Cd66a low /Kit neg ) as compared to other sub-populations enriched in either goblet cells (Epcam + /Cd44 + /Cd66a low /Kit + ) or enterocytes (Epcam + /Cd44 neg /Cd66a high ). Most importantly, E2F4 down-regulation using shRNAs dramatically enhanced the sensitivity of human CRCs to in vivo treatment with irinotecan , across three independent PDX models. Conclusions. Our data identified E2F4 and the DREAM repressor complex as critical regulators of human CRC resistance to irinotecan , and as candidate targets for the development of chemo-sensitizing agents.
Collapse
|
3
|
Díaz-Maneh A, Pérez-Rubio P, Granes CR, Bosch-Molist L, Lavado-García J, Gòdia F, Cervera L. Targeted knockdown of ATM, ATR, and PDEδ increases Gag HIV-1 VLP production in HEK293 cells. Appl Microbiol Biotechnol 2025; 109:1. [PMID: 39747723 PMCID: PMC11695449 DOI: 10.1007/s00253-024-13389-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2024] [Revised: 12/16/2024] [Accepted: 12/18/2024] [Indexed: 01/04/2025]
Abstract
Several strategies have been developed in recent years to improve virus-like particle (VLP)-based vaccine production processes. Among these, the metabolic engineering of cell lines has been one of the most promising approaches. Based on previous work and a proteomic analysis of HEK293 cells producing Human Immunodeficiency Virus-1 (HIV-1) Gag VLPs under transient transfection, four proteins susceptible of enhancing VLP production were identified: ataxia telangiectasia mutated (ATM), ataxia telangiectasia and rad3-related (ATR), DNA-dependent protein kinase catalytic subunit (DNA-PKcs), and retinal rod rhodopsin-sensitive cGMP 3',5'-cyclic phosphodiesterase subunit delta (PDEδ). The knockdown of ATM, ATR, and PDEδ in HEK293 cells increased HIV-1 VLP titers in the supernatant by 3.4-, 2.1-, and 2.2-fold, respectively. Also, possible metabolic synergies between plasmids were investigated by statistical design of experiments (DoE), enabling us to identify the optimal production strategy, that was further demonstrated at lab-scale stirred tank bioreactor operated in perfusion, significantly increasing both VLPs specific and volumetric productivities to 8.3 × 103 VLPs/cellxday and 7.5 × 1012 VLPs/Lxday, respectively. KEY POINTS: • ATM, ATR, and PDEδ knockdowns increased VLP production in HEK293 cells. • Knockdown of ATM increased budding efficiency and extracellular vesicle concentration. • ATM knockdown could be intensified to bioreactor scale operated in perfusion.
Collapse
Affiliation(s)
- Andy Díaz-Maneh
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.
- Aglaris Cell, C/ Santiago Grisolía, 2, Tres Cantos, 28760, Madrid, Spain.
| | - Pol Pérez-Rubio
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Cristina Rigau Granes
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Asklepios Biopharmaceutical, Inc, 20 TW Alexander Dr #110, Research Triangle Park, Chapel Hill, NC, 27709, USA
| | - Laia Bosch-Molist
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Jesús Lavado-García
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, 2800, Kgs. Lyngby, Denmark
| | - Francesc Gòdia
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Laura Cervera
- Grup d'Enginyeria de Bioprocessos i Biocatàlisi Aplicada, ENG4BIO, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
- Serra Hunter, Catalonia, Barcelona, Spain
| |
Collapse
|
4
|
Habisch R, Neubauer P, Soza-Ried J, Puschmann E. Repeated harvest enables efficient production of VSV-GP. Front Bioeng Biotechnol 2024; 12:1505338. [PMID: 39703791 PMCID: PMC11656157 DOI: 10.3389/fbioe.2024.1505338] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2024] [Accepted: 11/19/2024] [Indexed: 12/21/2024] Open
Abstract
Viral products keep gaining importance in multiple therapeutic fields. Considering the scale and production slot limitations, optimizing the outcome of every manufacturing batch is essential to minimize costs and make this therapeutic modality broadly available to patients. Most manufacturing processes for oncolytic viruses currently in clinical studies are based on a batch process. Here, we evaluated the benefits in terms of titer increase of a repeated harvest approach and compared it to the classical batch production process. While no effect on cell density was observed, the cumulated infectious titer following repeated harvest was over 400 times higher than the evaluated batch process yield. This shows that repeated harvests or perfusion have the potential to boost viral yields and should be considered when deciding on a process format for production.
Collapse
Affiliation(s)
- Rebecca Habisch
- Boehringer Ingelheim, Viral Therapeutics Center, Ochsenhausen, Germany
| | - Peter Neubauer
- Department of Bioprocess Engineering, Institute of Biotechnology, Technische Universität Berlin, Berlin, Germany
| | - Jorge Soza-Ried
- Boehringer Ingelheim, Viral Therapeutics Center, Ochsenhausen, Germany
| | - Eva Puschmann
- Boehringer Ingelheim, Viral Therapeutics Center, Ochsenhausen, Germany
| |
Collapse
|
5
|
Alekseeva ON, Hoa LT, Vorobyev PO, Kochetkov DV, Gumennaya YD, Naberezhnaya ER, Chuvashov DO, Ivanov AV, Chumakov PM, Lipatova AV. Receptors and Host Factors for Enterovirus Infection: Implications for Cancer Therapy. Cancers (Basel) 2024; 16:3139. [PMID: 39335111 PMCID: PMC11430599 DOI: 10.3390/cancers16183139] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 08/29/2024] [Accepted: 09/06/2024] [Indexed: 09/30/2024] Open
Abstract
Enteroviruses, with their diverse clinical manifestations ranging from mild or asymptomatic infections to severe diseases such as poliomyelitis and viral myocarditis, present a public health threat. However, they can also be used as oncolytic agents. This review shows the intricate relationship between enteroviruses and host cell factors. Enteroviruses utilize specific receptors and coreceptors for cell entry that are critical for infection and subsequent viral replication. These receptors, many of which are glycoproteins, facilitate virus binding, capsid destabilization, and internalization into cells, and their expression defines virus tropism towards various types of cells. Since enteroviruses can exploit different receptors, they have high oncolytic potential for personalized cancer therapy, as exemplified by the antitumor activity of certain enterovirus strains including the bioselected non-pathogenic Echovirus type 7/Rigvir, approved for melanoma treatment. Dissecting the roles of individual receptors in the entry of enteroviruses can provide valuable insights into their potential in cancer therapy. This review discusses the application of gene-targeting techniques such as CRISPR/Cas9 technology to investigate the impact of the loss of a particular receptor on the attachment of the virus and its subsequent internalization. It also summarizes the data on their expression in various types of cancer. By understanding how enteroviruses interact with specific cellular receptors, researchers can develop more effective regimens of treatment, offering hope for more targeted and efficient therapeutic strategies.
Collapse
Affiliation(s)
- Olga N Alekseeva
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Le T Hoa
- Department of Molecular Microbiology and Immunology, Norris Comprehensive Cancer Center, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Pavel O Vorobyev
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Dmitriy V Kochetkov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Yana D Gumennaya
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | | | - Denis O Chuvashov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Alexander V Ivanov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Peter M Chumakov
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| | - Anastasia V Lipatova
- Engelhardt Institute of Molecular Biology, Russian Academy of Sciences, 119991 Moscow, Russia
| |
Collapse
|
6
|
Champeil J, Mangion M, Gilbert R, Gaillet B. Improved Manufacturing Methods of Extracellular Vesicles Pseudotyped with the Vesicular Stomatitis Virus Glycoprotein. Mol Biotechnol 2024; 66:1116-1131. [PMID: 38182864 DOI: 10.1007/s12033-023-01007-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2023] [Accepted: 11/27/2023] [Indexed: 01/07/2024]
Abstract
Extracellular vesicles (EV), which expose the vesicular stomatitis virus glycoprotein (VSVG) on their surface, are used for delivery of nucleic acids and proteins in human cell lines. These particles are biomanufactured using methods that are difficult to scale up. Here, we describe the development of the first EV-VSVG production process in serum-free media using polyethylenimine (PEI)-based transient transfection of HEK293 suspension cells, as well as the first EV-VSVG purification process to utilize both ultracentrifugation and chromatography. Three parameters were investigated for EV-VSVG production: cell density, DNA concentration, and DNA:PEI ratio. The best production titer was obtained with 3 × 106 cells/mL, a plasmid concentration of 2 µg/mL, and a DNA:PEI ratio of 1:4. The production kinetics of VSVG was performed and showed that the highest amount of VSVG was obtained 3 days after transfection. Addition of cell culture supplements during the transfection resulted in an increase in VSVG production, with a maximum yield obtained with 2 mM of sodium butyrate added 18 h after transfection. Moreover, the absence of EV-VSVG during cell transfection with a GFP-coding plasmid revealed to be ineffective, with no fluorescent cells. An efficient EV-VSVG purification procedure consisting of a two-step concentration by low-speed centrifugation and sucrose cushion ultracentrifugation followed by a heparin affinity chromatography purification was also developed. Purified bioactive EV-VSVG preparations were characterized and revealed that EV-VSVG are spherical particles of 176.4 ± 88.32 nm with 91.4% of protein similarity to exosomes.
Collapse
Affiliation(s)
- Juliette Champeil
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
| | - Mathias Mangion
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
| | - Rénald Gilbert
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada
- Human Health Therapeutics Research Center, National Research Council Canada, 6100, Avenue Royalmount, Montréal, Québec, H4P 2R2, Canada
| | - Bruno Gaillet
- Chemical Engineering Department, Université Laval, 1065, Avenue de la Médecine, Pavillon Pouliot, Québec, QC, G1V 0A6, Canada.
- PROTEO: The Quebec Network for Research on Protein Function, Structure, and Engineering, Université du Québec à Montréal, 201 Avenue du Président Kennedy, Montréal, QC, H2X 3Y7, Canada.
- ThéCell: FRQS Cell, Tissue and Gene Therapy Network, Laboratoire d'organogénèse expérimentale - LOEX, 1401, 18E rue, Québec, QC, G1J 1Z4, Canada.
| |
Collapse
|
7
|
Kim Y, Lee S, Cho S, Park J, Chae D, Park T, Minna JD, Kim HH. High-throughput functional evaluation of human cancer-associated mutations using base editors. Nat Biotechnol 2022; 40:874-884. [PMID: 35411116 PMCID: PMC10243181 DOI: 10.1038/s41587-022-01276-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2021] [Accepted: 03/10/2022] [Indexed: 12/26/2022]
Abstract
Comprehensive phenotypic characterization of the many mutations found in cancer tissues is one of the biggest challenges in cancer genomics. In this study, we evaluated the functional effects of 29,060 cancer-related transition mutations that result in protein variants on the survival and proliferation of non-tumorigenic lung cells using cytosine and adenine base editors and single guide RNA (sgRNA) libraries. By monitoring base editing efficiencies and outcomes using surrogate target sequences paired with sgRNA-encoding sequences on the lentiviral delivery construct, we identified sgRNAs that induced a single primary protein variant per sgRNA, enabling linking those mutations to the cellular phenotypes caused by base editing. The functions of the vast majority of the protein variants (28,458 variants, 98%) were classified as neutral or likely neutral; only 18 (0.06%) and 157 (0.5%) variants caused outgrowing and likely outgrowing phenotypes, respectively. We expect that our approach can be extended to more variants of unknown significance and other tumor types.
Collapse
Affiliation(s)
- Younggwang Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Seungho Lee
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Soohyuk Cho
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Jinman Park
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
- Graduate School of Medical Science, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Dongwoo Chae
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea
| | - Taeyoung Park
- Department of Applied Statistics, Yonsei University, Seoul, Republic of Korea
| | - John D Minna
- Hamon Center for Therapeutic Oncology Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Hyongbum Henry Kim
- Department of Pharmacology, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Graduate School of Medical Science, Brain Korea 21 Plus Project for Medical Sciences, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Severance Biomedical Science Institute, Yonsei University College of Medicine, Seoul, Republic of Korea.
- Center for Nanomedicine, Institute for Basic Science (IBS), Seoul, Republic of Korea.
- Yonsei-IBS Institute, Yonsei University, Seoul, Republic of Korea.
- Institute for Immunology and Immunological Diseases, Yonsei University College of Medicine, Seoul, Republic of Korea.
| |
Collapse
|
8
|
Yadav M, Atala A, Lu B. Developing all-in-one virus-like particles for Cas9 mRNA/single guide RNA co-delivery and aptamer-containing lentiviral vectors for improved gene expression. Int J Biol Macromol 2022; 209:1260-1270. [PMID: 35461863 DOI: 10.1016/j.ijbiomac.2022.04.114] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/13/2022] [Accepted: 04/15/2022] [Indexed: 01/10/2023]
Abstract
Lentiviral vectors (LVs) are widely used for delivering foreign genes for long-term expression. Recently, virus-like particles (VLPs) were developed for mRNA or ribonucleoprotein (RNP) delivery for short-term endonuclease expression. Generating large amount of LVs or VLPs is challenging. On the other hand, methods for using VLPs to co-deliver Cas9 mRNA and single guide RNA (sgRNA) are limited. Fusing aptamer-binding protein (ABP) to the N-terminus of HIV Gag protein is currently the successful way to develop hybrid particles for co-delivering Cas9 mRNA and sgRNA. The effects of modifying Gag protein this way on particle assembly are unknown. Previously we found that adding an ABP after the second zinc finger domain of nucleocapsid (NC) protein had minimal effects on particle assembly. Based on these observations, here we developed hybrid particles for Cas9 mRNA and sgRNA co-delivery with normal capsid assembly efficiency. We further improved LVs for integrated gene expression by including an aptamer sequence in lentiviral genomic RNA, which improved lentiviral particle production and enhanced LV genomic RNA packaging. In summary, here we describe the development of new all-in-one VLPs for co-delivery of Cas9 mRNA and sgRNA, and new LVs for enhanced vector production and gene expression.
Collapse
Affiliation(s)
- Manish Yadav
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Anthony Atala
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA
| | - Baisong Lu
- Wake Forest Institute for Regenerative Medicine, Wake Forest University Health Sciences, Winston-Salem, NC, USA.
| |
Collapse
|
9
|
Transition from serum-supplemented monolayer to serum-free suspension lentiviral vector production for generation of chimeric antigen receptor T cells. Cytotherapy 2022; 24:850-860. [DOI: 10.1016/j.jcyt.2022.03.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 03/07/2022] [Accepted: 03/28/2022] [Indexed: 11/21/2022]
|
10
|
Yang HL, Xu C, Yang YK, Tang WQ, Hong M, Pan L, Chen HY. ZNF750 exerted its Antitumor Action in Oral Squamous Cell Carcinoma by regulating E2F2. J Cancer 2022; 12:7266-7276. [PMID: 35003347 PMCID: PMC8734408 DOI: 10.7150/jca.63919] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2021] [Accepted: 10/07/2021] [Indexed: 11/21/2022] Open
Abstract
Cell cycle activator E2F transcription factor 2 (E2F2) play a key role in tumor development and metastasis. Previous RNA sequence analysis (GSE134835) revealed E2F2 was significantly reduced by Zinc-finger protein 750 (ZNF750) in oral squamous cell carcinoma (OSCC). This study was aimed to determine the involvement of E2F2 in antitumor action of ZNF750. The nude mouse xenograft model was established by subcutaneously injection of stable cell line CAL-27oeZNF750 or CAL-27shZNF750. Xenograft tumor volume and tumor weight was measured. The expression of E2F2, transcriptional repressors such as enhancer of zeste 2 (Ezh2), PHD finger protein 19 (PHF19), and the genes related to cell proliferation or metastasis was studied in vivo or in vitro. Luciferase assay was performed to investigate regulation effect of ZNF750 on E2F2 luciferase activity. The involvement of E2F2 in the antitumor action of ZNF750 was studied by cotransduced ZNF750 with E2F2 lentivirus. The tumor growth and metastasis was repressed by ZNF750 manifested by reduced tumor size, tumor weight and the genes related to cell proliferation and metastasis. However, all of these were reversed by knockdown of the ZNF750 gene. Furthermore, E2F2 luciferase activity was inhibited by ZNF750. E2F2 partly blocked the antitumor action of ZNF750 manifested by increased self-renewal, invasion, migration, elevated Ezh2 and MMP13 protein expression in ZNF750 + E2F2 groups. However, silenced E2F2 further enhanced the antitumor action of ZNF750. ZNF750 depressed E2F2 activity and played a critical role in regulating transcriptional repressors for inhibiting the OSCC growth and metastasis in OSCC.
Collapse
Affiliation(s)
- Hong-Li Yang
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| | - Cong Xu
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| | - Yi-Kun Yang
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| | - Wen-Qiang Tang
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| | - Min Hong
- School of Chemistry and Chemical Engineering, Liaocheng University, Liaocheng 252059, P. R. China
| | - Li Pan
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| | - Hai-Ying Chen
- Central laboratory of Liaocheng People's Hospital, Liaocheng, 252000, P.R. China
| |
Collapse
|
11
|
Xu C, Yang HL, Yang YK, Pan L, Chen HY. Zinc-finger protein 750 mitigates malignant biological behavior of oral CSC-like cells enriched from parental CAL-27 cells. Oncol Lett 2022; 23:28. [PMID: 34868365 PMCID: PMC8630818 DOI: 10.3892/ol.2021.13146] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2021] [Accepted: 11/05/2021] [Indexed: 11/06/2022] Open
Abstract
Oral squamous cell carcinoma (OSCC) is the most commonly occurring oral malignancy. Cancer stem cells (CSCs) are known to be responsible for cancer recurrence and metastasis. Zinc-finger protein 750 (ZNF750) has been reported to inhibit OSCC cell proliferation and invasion. The present study aimed to elucidate the role of ZNF750 in the inhibition of the renewal ability of CSCs derived from the OSCC cell line, CAL-27. The effects of ZNF750 on CSC-like properties were examined using aldehyde dehydrogenase (ALDH), tumor sphere formation and colony formation assays. Reverse transcription-quantitative PCR and western blotting were performed to detect the expression levels of octamer-binding transcription factor 4, sex-determining region Y-box 2, the enhancer of zeste homolog 2 (Ezh2), embryonic ectoderm development (EED) and SUZ12 polycomb repressive complex 2 subunit (SUZ12), and for the identification of genes associated with metastasis. ZNF750 effectively attenuated CSC-like cell self-renewal abilities; ZNF750 decreased the ALDH-positive cell population, tumor sphere and colony formation abilities, cell viability and stemness factors. Furthermore, the expression levels of Ezh2, EED and SUZ12 were decreased by ZNF750. ZNF750 inhibited MMP1, 3, 9 and 13 expression levels, and decreased the cell invasion and migratory abilities. Moreover, the expression of tissue inhibitors of matrix metalloproteinases-1 was increased by ZNF750. However, opposite effects were observed following the knockdown of the ZNF750 gene. Overall, the present study demonstrated that ZNF750 has the potential to inhibit the renewal of CSC-like cells enriched from parental CAL-27 cells.
Collapse
Affiliation(s)
- Cong Xu
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Hong-Li Yang
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yi-Kun Yang
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Li Pan
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Hai-Ying Chen
- Central Laboratory of Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
12
|
Zheng F, Kawabe Y, Murakami M, Takahashi M, Nishihata K, Yoshida S, Ito A, Kamihira M. LINE-1 vectors mediate recombinant antibody gene transfer by retrotransposition in Chinese hamster ovary cells. Biotechnol J 2021; 16:e2000620. [PMID: 33938150 DOI: 10.1002/biot.202000620] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2020] [Revised: 04/26/2021] [Accepted: 04/28/2021] [Indexed: 11/09/2022]
Abstract
Retrotransposons, such as long interspersed element-1 (LINE-1), can copy themselves to other genomic loci via a transposition event (termed retrotransposition). Retrotransposons, therefore, have potential use as an efficient gene delivery tool to integrate multiple copies of a target gene into a host genome. Here, we developed a retrotransposon vector based on LINE-1 that achieves target gene integration of multiple transgene copies. The retrotransposon vector contains a neomycin resistance gene split by an intron as a marker gene, and a gene encoding an antibody single-chain variable fragment (Fv) fused with the constant antibody region (Fc) (scFv-Fc) as a model target gene. G418-resistant Chinese hamster ovary cells were generated using this retrotransposon vector, and scFv-Fc was produced in the culture medium. To regulate retrotransposition, we developed a retrotransposon vector system that separately expressed the two open reading frames (ORF1 and ORF2) of LINE-1. Genomic PCR analysis detected the transgene sequence in almost all tested clones. Compared with clones established using the intact LINE-1 vector, clones generated with the split ORF1 and ORF2 system showed similar specific scFv-Fc productivity and retrotransposition efficiency. This approach of using a retrotransposon-based vector system has the potential to provide a new gene delivery tool for mammalian cells.
Collapse
Affiliation(s)
- Feiyang Zheng
- Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Mai Murakami
- Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Mamika Takahashi
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Kyoka Nishihata
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Souichiro Yoshida
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| | - Masamichi Kamihira
- Graduate School of Systems Life Sciences, Kyushu University, Nishi-ku, Fukuoka, Japan.,Department of Chemical Engineering, Faculty of Engineering, Kyushu University, Nishi-ku, Fukuoka, Japan
| |
Collapse
|
13
|
Perry C, Rayat ACME. Lentiviral Vector Bioprocessing. Viruses 2021; 13:268. [PMID: 33572347 PMCID: PMC7916122 DOI: 10.3390/v13020268] [Citation(s) in RCA: 86] [Impact Index Per Article: 21.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2021] [Revised: 02/02/2021] [Accepted: 02/03/2021] [Indexed: 12/11/2022] Open
Abstract
Lentiviral vectors (LVs) are potent tools for the delivery of genes of interest into mammalian cells and are now commonly utilised within the growing field of cell and gene therapy for the treatment of monogenic diseases and adoptive therapies such as chimeric antigen T-cell (CAR-T) therapy. This is a comprehensive review of the individual bioprocess operations employed in LV production. We highlight the role of envelope proteins in vector design as well as their impact on the bioprocessing of lentiviral vectors. An overview of the current state of these operations provides opportunities for bioprocess discovery and improvement with emphasis on the considerations for optimal and scalable processing of LV during development and clinical production. Upstream culture for LV generation is described with comparisons on the different transfection methods and various bioreactors for suspension and adherent producer cell cultivation. The purification of LV is examined, evaluating different sequences of downstream process operations for both small- and large-scale production requirements. For scalable operations, a key focus is the development in chromatographic purification in addition to an in-depth examination of the application of tangential flow filtration. A summary of vector quantification and characterisation assays is also presented. Finally, the assessment of the whole bioprocess for LV production is discussed to benefit from the broader understanding of potential interactions of the different process options. This review is aimed to assist in the achievement of high quality, high concentration lentiviral vectors from robust and scalable processes.
Collapse
Affiliation(s)
- Christopher Perry
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
- Division of Advanced Therapies, National Institute for Biological Standards and Control, South Mimms EN6 3QG, UK
| | - Andrea C. M. E. Rayat
- The Advanced Centre for Biochemical Engineering, Department of Biochemical Engineering, University College London, Gower St, London WC1E 6BT, UK;
| |
Collapse
|
14
|
Liu X, Yang Y, Xu C, Yang H, Chen S, Chen H. RNA sequencing analysis of the CAL-27 cell response to over-expressed ZNF750 gene revealed an extensive regulation on cell cycle. Biomed Pharmacother 2019; 118:109377. [DOI: 10.1016/j.biopha.2019.109377] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2019] [Revised: 08/16/2019] [Accepted: 08/22/2019] [Indexed: 02/08/2023] Open
|
15
|
Hara T, Verma IM. Modeling Gliomas Using Two Recombinases. Cancer Res 2019; 79:3983-3991. [PMID: 31315836 PMCID: PMC6677610 DOI: 10.1158/0008-5472.can-19-0717] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Revised: 05/15/2019] [Accepted: 05/30/2019] [Indexed: 01/21/2023]
Abstract
Development of animal models to investigate the complex ecosystem of malignant gliomas using the Cre/loxP recombination system has significantly contributed to our understanding of the molecular underpinnings of this deadly disease. In these model systems, once the tumor is induced by activation of Cre-recombinase in a tissue-specific manner, further genetic manipulations to explore the progression of tumorigenesis are limited. To expand the application of mouse models for gliomas, we developed glial fibrillary acidic protein (GFAP)-FLP recombinase (FLPo) mice that express FLPo recombinase specifically in GFAP-positive cells. Lentivirus-based in vivo delivery of cancer genes conditioned by FLP/FRT-mediated recombination initiated gliomas in GFAP-FLPo mice. Using the Cre-mediated multifluorescent protein-expressing system, we demonstrated that the GFAP-FLPo mouse model enables the analysis of various stages of gliomagenesis. Collectively, we present a new mouse model that will expand our ability to dissect developmental processes of gliomagenesis and to provide new avenues for therapeutic approaches. SIGNIFICANCE: This study presents a new glioma mouse model derived using lentiviral vectors and two recombination systems that will expand the ability to dissect developmental processes of gliomagenesis.
Collapse
Affiliation(s)
- Toshiro Hara
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California.
| | - Inder M Verma
- Laboratory of Genetics, Salk Institute for Biological Studies, La Jolla, California
| |
Collapse
|
16
|
Abstract
The combination of single-cell RNA-seq and CRISPR allows for efficient interrogation of possibly any number of genes, only limited by the sequencing capability. Here we describe the current protocols for CRISPR screening in single cells, from cloning and virus production to generating sequencing data.
Collapse
Affiliation(s)
- Johan Henriksson
- Molecular Infection Medicine Sweden, Umeå University, Umeå, Sweden.
| |
Collapse
|
17
|
Robust Enhancement of Lentivirus Production by Promoter Activation. Sci Rep 2018; 8:15036. [PMID: 30310119 PMCID: PMC6181906 DOI: 10.1038/s41598-018-33042-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2018] [Accepted: 08/22/2018] [Indexed: 12/15/2022] Open
Abstract
Lentiviral vectors are a valuable tool to deliver exogenous genes for stable expression in cells. While much progress has been made in processing lentiviral vector-containing culture medium, it remains to be explored how the production of lentiviral vector from producer cells can be increased. We initially found that co-expression of the SPRY domain-containing SOCS box protein 1 (SPSB1) promotes the production of human immunodeficiency virus type 1 (HIV-1) and lentiviral vector with increased expression of the Gag and envelope proteins and activation of the HIV-1 LTR and CMV promoter. The presence of AP-1, NF-κB and CREB/ATF recognition sites in these promoters prompted us to utilize human T-lymphotropic virus type 1 (HTLV-1) Tax for lentiviral vector production because Tax activates all these transcription factors. Co-expression of a small amount of Tax markedly increased both the expression of viral structural proteins in producer cells and release of lentiviral vector particles, resulting in a more than 10-fold enhancement of transduction efficiency. Of note, the Tax protein was not detected in the lentiviral vector particles concentrated by ultracentrifugation, supporting the safety of this preparation. Collectively, these results indicate that promoter activation in producer cells represents a promising approach to preparing high-titer lentiviral vectors.
Collapse
|
18
|
ZNF750 inhibited the malignant progression of oral squamous cell carcinoma by regulating tumor vascular microenvironment. Biomed Pharmacother 2018; 105:566-572. [DOI: 10.1016/j.biopha.2018.06.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 06/02/2018] [Accepted: 06/02/2018] [Indexed: 12/19/2022] Open
|
19
|
Ramsey J, Butnor K, Peng Z, Leclair T, van der Velden J, Stein G, Lian J, Kinsey CM. Loss of RUNX1 is associated with aggressive lung adenocarcinomas. J Cell Physiol 2018; 233:3487-3497. [PMID: 28926105 PMCID: PMC5989135 DOI: 10.1002/jcp.26201] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2017] [Accepted: 09/08/2017] [Indexed: 12/25/2022]
Abstract
The mammalian runt-related factor 1 (RUNX1) is a master transcription factor that regulates lineage specification of hematopoietic stem cells. RUNX1 translocations result in the development of myeloid leukemias. Recently, RUNX1 has been implicated as a tumor suppressor in other cancers. We postulated RUNX1 expression may be associated with lung adenocarcinoma etiology and/or progression. We evaluated the association of RUNX1 mRNA expression with overall survival data from The Cancer Genome Atlas (TCGA), a publically available database. Compared to high expression levels, Low RUNX1 levels from lung adenocarcinomas were associated with a worse overall survival (Hazard Ratio = 2.014 (1.042-3.730 95% confidence interval), log-rank p = 0.035) compared to those that expressed high RUNX1 levels. Further immunohistochemical examination of 85 surgical specimens resected at the University of Vermont Medical Center identified that low RUNX1 protein expression was associated with larger tumors (p = 0.038). Gene expression network analysis was performed on the same subset of TCGA cases that demonstrated differential survival by RUNX1 expression. This analysis, which reveals regulatory relationships, showed that reduced RUNX1 levels were closely linked to upregulation of the transcription factor E2F1. To interrogate this relationship, RUNX1 was depleted in a lung cancer cell line that expresses high levels of RUNX1. Loss of RUNX1 resulted in enhanced proliferation, migration, and invasion. RUNX1 depletion also resulted in increased mRNA expression of E2F1 and multiple E2F1 target genes. Our data implicate loss of RUNX1 as driver of lung adenocarcinoma aggression, potentially through deregulation of the E2F1 pathway.
Collapse
Affiliation(s)
- Jon Ramsey
- Department of Biochemistry, University of Vermont, Burlington VT
| | - Kelly Butnor
- Department of Pathology, University of Vermont Medical Center, Burlington VT
| | - Zhihua Peng
- Department of Biochemistry, University of Vermont, Burlington VT
| | - Tim Leclair
- Department of Thoracic Surgery and Interventional Pulmonology, Beth Israel Deaconess Medical Center, Boston MA
| | - Jos van der Velden
- Department of Pathology, University of Vermont Medical Center, Burlington VT
| | - Gary Stein
- Department of Biochemistry, University of Vermont, Burlington VT
| | - Jane Lian
- Department of Biochemistry, University of Vermont, Burlington VT
| | - C. Matthew Kinsey
- Pulmonary and Critical Care, University of Vermont Medical Center, Burlington VT
| |
Collapse
|
20
|
Fuenmayor J, Cervera L, Rigau C, Gòdia F. Enhancement of HIV-1 VLP production using gene inhibition strategies. Appl Microbiol Biotechnol 2018; 102:4477-4487. [DOI: 10.1007/s00253-018-8930-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2018] [Revised: 03/06/2018] [Accepted: 03/10/2018] [Indexed: 10/17/2022]
|
21
|
Vijayraghavan S, Kantor B. A Protocol for the Production of Integrase-deficient Lentiviral Vectors for CRISPR/Cas9-mediated Gene Knockout in Dividing Cells. J Vis Exp 2017. [PMID: 29286484 DOI: 10.3791/56915] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Lentiviral vectors are an ideal choice for delivering gene-editing components to cells due to their capacity for stably transducing a broad range of cells and mediating high levels of gene expression. However, their ability to integrate into the host cell genome enhances the risk of insertional mutagenicity and thus raises safety concerns and limits their usage in clinical settings. Further, the persistent expression of gene-editing components delivered by these integration-competent lentiviral vectors (ICLVs) increases the probability of promiscuous gene targeting. As an alternative, a new generation of integrase-deficient lentiviral vectors (IDLVs) has been developed that addresses many of these concerns. Here the production protocol of a new and improved IDLV platform for CRISPR-mediated gene editing and list the steps involved in the purification and concentration of such vectors is described and their transduction and gene-editing efficiency using HEK-293T cells was demonstrated. This protocol is easily scalable and can be used to generate high titer IDLVs that are capable of transducing cells in vitro and in vivo. Moreover, this protocol can be easily adapted for the production of ICLVs.
Collapse
Affiliation(s)
- Sriram Vijayraghavan
- Duke Viral Vector Core, Department of Neurobiology, Duke University School of Medicine
| | - Boris Kantor
- Duke Viral Vector Core, Department of Neurobiology, Duke University School of Medicine;
| |
Collapse
|
22
|
Pathway-focused PCR array profiling of CAL-27 cell with over-expressed ZNF750. Oncotarget 2017; 9:566-575. [PMID: 29416636 PMCID: PMC5787490 DOI: 10.18632/oncotarget.23075] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2017] [Accepted: 11/14/2017] [Indexed: 02/01/2023] Open
Abstract
Zinc-finger protein 750 (ZNF750) is the potential anti-cancer gene in oral squamous cell carcinoma (OSCC). The present study was to investigate the expression changes of ZNF750 in OSCC tissue and to reveal the induction of altered mRNA expression profiles caused by over-expressed ZNF750 in CAL-27 cell. The expression level of ZNF750 in tissue specimens from OSCC patients was detected by immunohistochemistry. Gene expression profiling was performed using Human Signal Transduction PathwayFinder RT2 Profiler™ PCR Array. The expression changes of 84 key genes representing 10 signal transduction pathways in human following over-expressed ZNF750 in CAL-27 cell was examined. The expression of ZNF750 protein was reduced in OSCC tissues. The R2 PCR Array analysis revealed that 39 of the 84 examined genes that changed at least a two-fold between control and ZNF750 groups. These genes related to oxidative stress, WNT, JAK/STAT, TGFβ, NF-kappaB (NFκB), p53, Notch, Hedgehog, PPAR and Hypoxia signaling. ZNF750 could inhibit the candidate genes ATF4, SQSTM1, HMOX1, CCND1, TNF-alpha, TNFSF10 and FOSL1 but activate CDKN1A and EMP1. Our studies suggest that ZNF750 can regulate signaling pathways that related to proliferation, cell cycle, inflammation and oxidative stress in CAL-27 cell.
Collapse
|
23
|
Yang H, Pan L, Xu C, Zhang Y, Li K, Chen S, Zhang B, Liu Z, Wang LX, Chen H. Overexpression of tumor suppressor gene ZNF750 inhibits oral squamous cell carcinoma metastasis. Oncol Lett 2017; 14:5591-5596. [PMID: 29113187 DOI: 10.3892/ol.2017.6908] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2017] [Accepted: 06/09/2017] [Indexed: 11/06/2022] Open
Abstract
Zinc-finger protein 750 (ZNF750) encodes a putative C2H2 zinc finger protein and is typically mutated or deleted in squamous cell carcinoma. The role of ZNF750 in oral squamous cell carcinoma (OSCC) remains unknown. The aim of the present study was to investigate the effects of ZNF750 overexpression in CAL-27 cells. Cell viability, and the expression of genes associated with proliferation, differentiation and the epithelial-mesenchymal transition were investigated in CAL-27 cells following ZNF750 overexpression, using Cell Counting kit-8, reverse transcription-quantitative polymerase chain reaction and western blot analysis, respectively. In addition, scratch wound, invasion and migration assays were performed. Cell viability, matrix metalloproteinase 28 expression, cyclin B1 expression and mesenchymal marker neural cadherin expression were decreased following ZNF750 overexpression compared with the control groups. ZNF750 overexpression induced the differentiation-associated genes late cornified envelope 3A and small proline-rich protein 1A and upregulated the expression of late epidermal differentiation factor Kruppel-like factor 4. Overexpression of ZNF750 in CAL-27 cells resulted in inhibition of cell invasion and migration. Taken together, these data suggest that ZNF750 may inhibit the metastasis of OSCC.
Collapse
Affiliation(s)
- Hongli Yang
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Li Pan
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Cong Xu
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Yingxin Zhang
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Keyi Li
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China.,Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Shuangfeng Chen
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China
| | - Bin Zhang
- Department of Oral and Maxillofacial Surgery, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China.,Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| | - Zhijun Liu
- Department of Microbiology, Weifang Medical University, Weifang, Shandong 261053, P.R. China
| | - Le-Xin Wang
- School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2650, Australia
| | - Haiying Chen
- Oral Maxillofacial Head-Neck Key Laboratory of Medical Biology and Central Laboratory, Liaocheng People's Hospital, Liaocheng, Shandong 252000, P.R. China.,Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong 252000, P.R. China
| |
Collapse
|
24
|
Gélinas JF, Davies LA, Gill DR, Hyde SC. Assessment of selected media supplements to improve F/HN lentiviral vector production yields. Sci Rep 2017; 7:10198. [PMID: 28860488 PMCID: PMC5579034 DOI: 10.1038/s41598-017-07893-3] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Accepted: 05/31/2017] [Indexed: 12/01/2022] Open
Abstract
The development of lentiviral-based therapeutics is challenged by the high cost of current Good Manufacturing Practices (cGMP) production. Lentiviruses are enveloped viruses that capture a portion of the host cell membrane during budding, which then constitutes part of the virus particle. This process might lead to lipid and protein depletion in the cell membrane and affect cell viability. Furthermore, growth in suspension also causes stresses that can affect virus production yields. To assess the impact of these issues, selected supplements (Cholesterol Lipid Concentrate, Chemically Defined Lipid Concentrate, Lipid Mixture 1, Gelatin Peptone N3, N-Acetyl-L-Cysteine and Pluronic F-68) were assayed in order to improve production yields in a transient transfection production of a Sendai virus F/HN-pseudotyped HIV-1-based third generation lentiviral vector in FreeStyle 293 (serum-free media) in suspension. None of the supplements tested had a significant positive impact on lentiviral vector yields, but small non-significant improvements could be combined to increase vector production in a cell line where other conditions have been optimised.
Collapse
Affiliation(s)
- Jean-François Gélinas
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK
| | - Lee A Davies
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK.,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK
| | - Deborah R Gill
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK.,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK
| | - Stephen C Hyde
- Gene Medicine Research Group, NDCLS, Radcliffe Department of Medicine, John Radcliffe Hospital, Oxford University, Oxford, UK. .,United Kingdom Cystic Fibrosis Gene Therapy Consortium, Oxford, Edinburgh, London, UK.
| |
Collapse
|
25
|
Lentiviral vectors can be used for full-length dystrophin gene therapy. Sci Rep 2017; 7:44775. [PMID: 28303972 PMCID: PMC5356018 DOI: 10.1038/srep44775] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 02/13/2017] [Indexed: 12/13/2022] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is caused by a lack of dystrophin expression in patient muscle fibres. Current DMD gene therapy strategies rely on the expression of internally deleted forms of dystrophin, missing important functional domains. Viral gene transfer of full-length dystrophin could restore wild-type functionality, although this approach is restricted by the limited capacity of recombinant viral vectors. Lentiviral vectors can package larger transgenes than adeno-associated viruses, yet lentiviral vectors remain largely unexplored for full-length dystrophin delivery. In our work, we have demonstrated that lentiviral vectors can package and deliver inserts of a similar size to dystrophin. We report a novel approach for delivering large transgenes in lentiviruses, in which we demonstrate proof-of-concept for a ‘template-switching’ lentiviral vector that harnesses recombination events during reverse-transcription. During this work, we discovered that a standard, unmodified lentiviral vector was efficient in delivering full-length dystrophin to target cells, within a total genomic load of more than 15,000 base pairs. We have demonstrated gene therapy with this vector by restoring dystrophin expression in DMD myoblasts, where dystrophin was expressed at the sarcolemma of myotubes after myogenic differentiation. Ultimately, our work demonstrates proof-of-concept that lentiviruses can be used for permanent full-length dystrophin gene therapy, which presents a significant advancement in developing an effective treatment for DMD.
Collapse
|
26
|
Counsell JR, Asgarian Z, Meng J, Ferrer V, Vink CA, Howe SJ, Waddington SN, Thrasher AJ, Muntoni F, Morgan JE, Danos O. Lentiviral vectors can be used for full-length dystrophin gene therapy. Sci Rep 2017; 7:79. [PMID: 28250438 PMCID: PMC5427806 DOI: 10.1038/s41598-017-00152-5] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2016] [Accepted: 02/13/2017] [Indexed: 01/08/2023] Open
Abstract
Duchenne Muscular Dystrophy (DMD) is caused by a lack of dystrophin expression in patient muscle fibres. Current DMD gene therapy strategies rely on the expression of internally deleted forms of dystrophin, missing important functional domains. Viral gene transfer of full-length dystrophin could restore wild-type functionality, although this approach is restricted by the limited capacity of recombinant viral vectors. Lentiviral vectors can package larger transgenes than adeno-associated viruses, yet lentiviral vectors remain largely unexplored for full-length dystrophin delivery. In our work, we have demonstrated that lentiviral vectors can package and deliver inserts of a similar size to dystrophin. We report a novel approach for delivering large transgenes in lentiviruses, in which we demonstrate proof-of-concept for a 'template-switching' lentiviral vector that harnesses recombination events during reverse-transcription. During this work, we discovered that a standard, unmodified lentiviral vector was efficient in delivering full-length dystrophin to target cells, within a total genomic load of more than 15,000 base pairs. We have demonstrated gene therapy with this vector by restoring dystrophin expression in DMD myoblasts, where dystrophin was expressed at the sarcolemma of myotubes after myogenic differentiation. Ultimately, our work demonstrates proof-of-concept that lentiviruses can be used for permanent full-length dystrophin gene therapy, which presents a significant advancement in developing an effective treatment for DMD.
Collapse
Affiliation(s)
- John R Counsell
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK.
- UCL Cancer Institute, Paul O 'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK.
- Molecular and Cellular Immunology, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK.
- Gene Transfer Technology Group, Institute for Womens Health, University College London, 86-96, Chenies Mews, London, UK.
| | - Zeinab Asgarian
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Jinhong Meng
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Veronica Ferrer
- UCL Cancer Institute, Paul O 'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK
| | - Conrad A Vink
- Molecular and Cellular Immunology, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Steven J Howe
- Molecular and Cellular Immunology, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Simon N Waddington
- Gene Transfer Technology Group, Institute for Womens Health, University College London, 86-96, Chenies Mews, London, UK
- MRC Antiviral Gene Therapy Research Unit, Faculty of Health Sciences, University of the Witswatersrand, Johannesburg, South Africa
| | - Adrian J Thrasher
- Molecular and Cellular Immunology, Institute of Child Health, University College London, 30 Guilford Street, London, WC1N 1EH, UK
| | - Francesco Muntoni
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Jennifer E Morgan
- The Dubowitz Neuromuscular Centre, Molecular Neurosciences Section, Developmental Neurosciences Programme, UCL Great Ormond Street Institute of Child Health, 30 Guilford Street, London, WC1N 1EH, UK
| | - Olivier Danos
- UCL Cancer Institute, Paul O 'Gorman Building, University College London, 72 Huntley Street, London, WC1E 6BT, UK
- Biogen, 14 Cambridge Center, Cambridge, MA, 02142, USA
| |
Collapse
|
27
|
Chen H, Yang H, Xu C, Yue H, Xia P, Strappe PM, Wang L, Pan L, Tang W, Chen S, Wang L. Gene expression profiling of common signal transduction pathways affected by rBMSCs/F92A-Cav1 in the lungs of rat with pulmonary arterial hypertension. Biomed Pharmacother 2016; 83:100-106. [PMID: 27470556 DOI: 10.1016/j.biopha.2016.06.028] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2016] [Revised: 05/25/2016] [Accepted: 06/14/2016] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND Pulmonary arterial hypertension (PAH) is associated with sustained vasoconstriction, inflammation and suppressed apoptosis of smooth muscle cells. Our previous studies have found that rat bone marrow-derived mesenchymal stem cells (rBMSCs) transduced with a mutant caveolin-1(F92A-Cav1) could enhance endothelial nitric oxide synthase (eNOS) activity and improve pulmonary vascular remodeling, but the potential mechanism is not yet fully explored. The present study was to investigate the gene expression profile upon rBMSCs/F92A-Cav1delivered to PAH rat to evaluate the role of F92A-Cav1 in its regulation. METHODS PAH was induced with monocrotaline (MCT, 60mg/kg) prior to delivery of lentiviral vector transduced rBMSCs expressing Cav1 or F92A-Cav1. Gene expression profiling was performed using Rat Signal Transduction PathwayFinder array. The expression changes of 84 key genes representing 10 signal transduction pathways in rat following rBMSCs/F92A-Cav1 treatment was examined. RESULTS Screening with the Rat Signal Transduction PathwayFinder R2 PCR Array system and subsequent western blot, immunohistochemistry or real time PCR analysis revealed that F92A-Cav1 modified rBMSCs can inhibit the inflammation factors (TNF-alpha, Icam1 and C/EBPdelta), pro-proliferation genes (c-Myc, Bcl2a1d, Notch1and Hey2), oxidative stress gene (Hmox1) and activate cell cycle arrested gene Cdkn1a, ameliorating inflammation and inhibiting cell proliferation in PAH rat. CONCLUSION rBMSCs/F92A-Cav1 inhibits inflammation and cell proliferation by regulating signaling pathways that related to inflammation, proliferation, cell cycle and oxidative stress.
Collapse
Affiliation(s)
- Haiying Chen
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China; Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong, China
| | - Hongli Yang
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Chong Xu
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Hongmei Yue
- Liaocheng People's Hospital and Clinical School of Taishan Medical University, Liaocheng, Shandong, China; Department of Cardiology, Liaocheng People's Hospital and Affiliated Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong, 252000, China
| | - Peng Xia
- Department of Cardiology, Liaocheng People's Hospital and Affiliated Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong, 252000, China
| | | | - Lei Wang
- Department of Cardiology, Liaocheng People's Hospital and Affiliated Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong, 252000, China
| | - Li Pan
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Wenqiang Tang
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Shuangfeng Chen
- Central laboratory, and key Laboratory of Oral and Maxillofacial-Head and Neck Medical Biology, Liaocheng People's Hospital, Liaocheng, Shandong, 252000, China
| | - Lexin Wang
- Department of Cardiology, Liaocheng People's Hospital and Affiliated Liaocheng People's Hospital of Shandong University, Liaocheng, Shandong, 252000, China; School of Biomedical Sciences, Charles Sturt University, Wagga Wagga, NSW 2650, Australia.
| |
Collapse
|
28
|
Valdez CM, Murphy GG, Beg AA. The Rac-GAP alpha2-chimaerin regulates hippocampal dendrite and spine morphogenesis. Mol Cell Neurosci 2016; 75:14-26. [PMID: 27297944 DOI: 10.1016/j.mcn.2016.06.002] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/29/2016] [Revised: 05/09/2016] [Accepted: 06/07/2016] [Indexed: 12/01/2022] Open
Abstract
Dendritic spines are fine neuronal processes where spatially restricted input can induce activity-dependent changes in one spine, while leaving neighboring spines unmodified. Morphological spine plasticity is critical for synaptic transmission and is thought to underlie processes like learning and memory. Significantly, defects in dendritic spine stability and morphology are common pathogenic features found in several neurodevelopmental and neuropsychiatric disorders. The remodeling of spines relies on proteins that modulate the underlying cytoskeleton, which is primarily composed of filamentous (F)-actin. The Rho-GTPase Rac1 is a major regulator of F-actin and is essential for the development and plasticity of dendrites and spines. However, the key molecules and mechanisms that regulate Rac1-dependent pathways at spines and synapses are not well understood. We have identified the Rac1-GTPase activating protein, α2-chimaerin, as a critical negative regulator of Rac1 in hippocampal neurons. The loss of α2-chimaerin significantly increases the levels of active Rac1 and induces the formation of aberrant polymorphic dendritic spines. Further, disruption of α2-chimaerin signaling simplifies dendritic arbor complexity and increases the presence of dendritic spines that appear poly-innervated. Our data suggests that α2-chimaerin serves as a "brake" to constrain Rac1-dependent signaling to ensure that the mature morphology of spines is maintained in response to network activity.
Collapse
Affiliation(s)
- Chris M Valdez
- Interdepartmental Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, United States
| | - Geoffrey G Murphy
- Interdepartmental Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, United States; Molecular and Behavioral Neuroscience Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, United States
| | - Asim A Beg
- Interdepartmental Program in Neuroscience, University of Michigan, Ann Arbor, MI 48109, United States; Department of Pharmacology, University of Michigan, Ann Arbor, MI 48109, United States.
| |
Collapse
|
29
|
Fricano-Kugler CJ, Williams MR, Salinaro JR, Li M, Luikart B. Designing, Packaging, and Delivery of High Titer CRISPR Retro and Lentiviruses via Stereotaxic Injection. J Vis Exp 2016:53783. [PMID: 27285851 PMCID: PMC4927708 DOI: 10.3791/53783] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Replication defective lentiviruses or retroviruses are capable of stably integrating transgenes into the genome of an infected host cell. This technique has been widely used to encode fluorescent proteins, opto- or chemo-genetic controllers of cell activity, or heterologous expression of human genes in model organisms. These viruses have also successfully been used to deliver recombinases to relevant target sites in transgenic animals, or even deliver small hairpin or micro RNAs in order to manipulate gene expression. While these techniques have been fruitful, they rely on transgenic animals (recombinases) or frequently lack high efficacy and specificity (shRNA/miRNA). In contrast, the CRISPR/Cas system uses an exogenous Cas nuclease which targets specific sites in an organism's genome via an exogenous guide RNA in order to induce double stranded breaks in DNA. These breaks are then repaired by non-homologous end joining (NHEJ), producing insertion and deletion (indel) mutations that can result in deleterious missense or nonsense mutations. This manuscript provides detailed methods for the design, production, injection, and validation of single lenti/retro virus particles that can stably transduce neurons to express a fluorescent reporter, Cas9, and sgRNAs to knockout genes in a model organism.
Collapse
Affiliation(s)
| | - Michael R Williams
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College
| | - Julia R Salinaro
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College
| | - Meijie Li
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College
| | - Bryan Luikart
- Department of Physiology and Neurobiology, Geisel School of Medicine at Dartmouth College;
| |
Collapse
|
30
|
Humbert O, Gisch DW, Wohlfahrt ME, Adams AB, Greenberg PD, Schmitt TM, Trobridge GD, Kiem HP. Development of Third-generation Cocal Envelope Producer Cell Lines for Robust Lentiviral Gene Transfer into Hematopoietic Stem Cells and T-cells. Mol Ther 2016; 24:1237-46. [PMID: 27058824 DOI: 10.1038/mt.2016.70] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Accepted: 04/04/2016] [Indexed: 12/19/2022] Open
Abstract
Lentiviral vectors (LVs) pseudotyped with vesicular stomatitis virus envelope glycoprotein (VSV-G) have demonstrated great promise in gene therapy trials employing hematopoietic stem cell and T-cells. The VSV-G envelope confers broad tropism and stability to the vector but is toxic when constitutively expressed, which has impeded efforts to generate stable producer cell lines. We previously showed that cocal pseudotyped LVs offer an excellent alternative to VSV-G vectors because of their broad tropism and resistance to human serum inactivation. In this study, we demonstrate that cocal LVs transduce CD34(+) and CD4(+) T-cells more efficiently than VSV-G LVs and share the same receptor(s) for cell entry. 293T-cells stably expressing the cocal envelope produced significantly higher LV titers than VSV-G expressing cells. We developed cocal pseudotyped, third-generation, self-inactivating LV producer cell lines for a GFP reporter and for a WT1 tumor-specific T-cell receptor, which achieved concentrated titers above 10(8) IU/ml and were successfully adapted for growth in suspension, serum-free culture. The resulting LVs were at least as effective as standard LVs in transducing CD34(+) and CD4(+) T-cells. Our stable cocal LV producer cell lines should facilitate the production of large-scale, high titer clinical grade vectors.
Collapse
Affiliation(s)
- Olivier Humbert
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Don W Gisch
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Martin E Wohlfahrt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Amie B Adams
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Phil D Greenberg
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Clinical Research Division, Program in Immunology, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Immunology, University of Washington, Seattle, Washington, USA.,Department of Medicine, Division of Medical Oncology, University of Washington School of Medicine, Seattle, Washington, USA
| | - Tom M Schmitt
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA
| | - Grant D Trobridge
- Department of Pharmaceutical Sciences, Washington State University, Spokane, Washington, USA.,School of Molecular Biosciences, Washington State University, Pullman, Washington, USA
| | - Hans-Peter Kiem
- Clinical Research Division, Fred Hutchinson Cancer Research Center, Seattle, Washington, USA.,Department of Medicine, University of Washington, Seattle, Washington, USA.,Department of Pathology, University of Washington, Seattle, Washington, USA
| |
Collapse
|
31
|
Kawabe Y, Shimomura T, Huang S, Imanishi S, Ito A, Kamihira M. Targeted transgene insertion into the CHO cell genome using Cre recombinase-incorporating integrase-defective retroviral vectors. Biotechnol Bioeng 2016; 113:1600-10. [DOI: 10.1002/bit.25923] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2015] [Revised: 12/18/2015] [Accepted: 12/29/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Yoshinori Kawabe
- Department of Chemical Engineering, Faculty of Engineering; Kyushu University; Nishi-ku Fukuoka Japan
| | - Takuya Shimomura
- Department of Chemical Engineering, Faculty of Engineering; Kyushu University; Nishi-ku Fukuoka Japan
| | - Shuohao Huang
- Graduate School of Systems Life Sciences; Kyushu University; 744 Motooka Nishi-ku Fukuoka 819-0395 Japan
| | - Suguru Imanishi
- Department of Chemical Engineering, Faculty of Engineering; Kyushu University; Nishi-ku Fukuoka Japan
| | - Akira Ito
- Department of Chemical Engineering, Faculty of Engineering; Kyushu University; Nishi-ku Fukuoka Japan
| | - Masamichi Kamihira
- Department of Chemical Engineering, Faculty of Engineering; Kyushu University; Nishi-ku Fukuoka Japan
- Graduate School of Systems Life Sciences; Kyushu University; 744 Motooka Nishi-ku Fukuoka 819-0395 Japan
| |
Collapse
|
32
|
Petersen GF, Hilbert BJ, Trope GD, Kalle WHJ, Strappe PM. Direct Conversion of Equine Adipose-Derived Stem Cells into Induced Neuronal Cells Is Enhanced in Three-Dimensional Culture. Cell Reprogram 2015; 17:419-26. [PMID: 26579833 DOI: 10.1089/cell.2015.0046] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
The ability to culture neurons from horses may allow further investigation into equine neurological disorders. In this study, we demonstrate the generation of induced neuronal cells from equine adipose-derived stem cells (EADSCs) using a combination of lentiviral vector expression of the neuronal transcription factors Brn2, Ascl1, Myt1l (BAM) and NeuroD1 and a defined chemical induction medium, with βIII-tubulin-positive induced neuronal cells displaying a distinct neuronal morphology of rounded and compact cell bodies, extensive neurite outgrowth, and branching of processes. Furthermore, we investigated the effects of dimensionality on neuronal transdifferentiation, comparing conventional two-dimensional (2D) monolayer culture against three-dimensional (3D) culture on a porous polystyrene scaffold. Neuronal transdifferentiation was enhanced in 3D culture, with evenly distributed cells located on the surface and throughout the scaffold. Transdifferentiation efficiency was increased in 3D culture, with an increase in mean percent conversion of more than 100% compared to 2D culture. Additionally, induced neuronal cells were shown to transit through a Nestin-positive precursor state, with MAP2 and Synapsin 2 expression significantly increased in 3D culture. These findings will help to increase our understanding of equine neuropathogenesis, with prospective roles in disease modeling, drug screening, and cellular replacement for treatment of equine neurological disorders.
Collapse
Affiliation(s)
- Gayle F Petersen
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Bryan J Hilbert
- 2 School of Animal and Veterinary Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Gareth D Trope
- 2 School of Animal and Veterinary Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Wouter H J Kalle
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| | - Padraig M Strappe
- 1 School of Biomedical Sciences, Charles Sturt University , Wagga Wagga, New South Wales, Australia
| |
Collapse
|
33
|
Cervera L, Fuenmayor J, González-Domínguez I, Gutiérrez-Granados S, Segura MM, Gòdia F. Selection and optimization of transfection enhancer additives for increased virus-like particle production in HEK293 suspension cell cultures. Appl Microbiol Biotechnol 2015; 99:9935-49. [PMID: 26278533 DOI: 10.1007/s00253-015-6842-4] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2015] [Revised: 07/08/2015] [Accepted: 07/11/2015] [Indexed: 01/09/2023]
Abstract
The manufacturing of biopharmaceuticals in mammalian cells typically relies on the use of stable producer cell lines. However, in recent years, transient gene expression has emerged as a suitable technology for rapid production of biopharmaceuticals. Transient gene expression is particularly well suited for early developmental phases, where several potential therapeutic targets need to be produced and tested in vivo. As a relatively new bioprocessing modality, a number of opportunities exist for improving cell culture productivity upon transient transfection. For instance, several compounds have shown positive effects on transient gene expression. These transfection enhancers either facilitate entry of PEI/DNA transfection complexes into the cell or nucleus or increase levels of gene expression. In this work, the potential of combining transfection enhancers to increase Gag-based virus-like particle production levels upon transfection of suspension-growing HEK 293 cells is evaluated. Using Plackett-Burman design of experiments, it is first tested the effect of eight transfection enhancers: trichostatin A, valproic acid, sodium butyrate, dimethyl sulfoxide (DMSO), lithium acetate, caffeine, hydroxyurea, and nocodazole. An optimal combination of compounds exhibiting the highest effect on gene expression levels was subsequently identified using a surface response experimental design. The optimal consisted on the addition of 20 mM lithium acetate, 3.36 mM valproic acid, and 5.04 mM caffeine which increased VLP production levels 3.8-fold, while maintaining cell culture viability at 94%.
Collapse
Affiliation(s)
- Laura Cervera
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain.
| | - Javier Fuenmayor
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Irene González-Domínguez
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Sonia Gutiérrez-Granados
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| | - Maria Mercedes Segura
- Bluebird Bio Pharmaceutical Sciences, Process Development Group, 150 2nd Street, Cambridge, MA, 02141, USA
| | - Francesc Gòdia
- Grup d'Enginyeria Cellular i Bioprocés, Escola d'Enginyeria, Universitat Autònoma de Barcelona, Campus de Bellaterra, Cerdanyola del Vallès, 08193, Barcelona, Spain
| |
Collapse
|
34
|
Optimization of methods for the genetic modification of human T cells. Immunol Cell Biol 2015; 93:896-908. [PMID: 26027856 PMCID: PMC4659746 DOI: 10.1038/icb.2015.59] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2015] [Revised: 05/06/2015] [Accepted: 05/13/2015] [Indexed: 12/18/2022]
Abstract
CD4+ T cells are critical in the fight against parasitic, bacterial, and viral infections, but are also involved in many autoimmune and pathological disorders. Studies of protein function in human T cells are confined to techniques such as RNAi due to ethical reasons and relative simplicity of these methods. However, introduction of RNAi or genes into primary human T cells is often hampered by toxic effects from transfection or transduction methods that yield cell numbers inadequate for downstream assays. Additionally, the efficiency of recombinant DNA expression is frequently low due to multiple factors including efficacy of the method and strength of the targeting RNAs. Here, we describe detailed protocols that will aid in the study of primary human CD4+ T cells. First, we describe a method for development of effective microRNA/shRNAs using available online algorithms. Second, we illustrate an optimized protocol for high efficacy retroviral or lentiviral transduction of human T cell lines. Importantly, we demonstrate that activated primary human CD4+ T cells can be transduced efficiently with lentiviruses, with a highly activated population of T cells receiving the largest number of copies of integrated DNA. We also illustrate a method for efficient lentiviral transduction of hard-to-transduce un-activated primary human CD4+ T cells. These protocols will significantly assist in understanding the activation and function of human T cells and will ultimately aid in the development or improvement of current drugs that target human CD4+ T cells.
Collapse
|
35
|
Increase in the titer of lentiviral vectors expressing potassium channels by current blockade during viral vector production. BMC Neurosci 2015; 16:30. [PMID: 25940378 PMCID: PMC4425897 DOI: 10.1186/s12868-015-0159-1] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2015] [Accepted: 04/01/2015] [Indexed: 12/02/2022] Open
Abstract
Background High titers of lentiviral vectors are required for the efficient transduction of a gene of interest. During preparation of lentiviral the vectors, the protein of interest is inevitably expressed in the viral vector-producing cells. This expression may affect the production of the lentiviral vector. Methods We prepared lentiviral vectors expressing inwardly rectifying potassium channel (Lv-Kir2.1), its dominant-negative form (Lv-Kir-DN), and other K+ channels, using the ubiquitously active β-actin and neuron-specific synapsin I promoters. Results The titer of Lv-Kir-DN was higher than that of Lv-Kir2.1, suggesting a negative effect of induced K+ currents on viral titer. We then blocked Kir2.1 currents with the selective blocker Ba2+ during Lv-Kir2.1 production, and obtained about a 5-fold increase in the titer. Higher extracellular K+ concentrations increased the titer of Lv-Kir2.1 about 9-fold. With a synapsin I promoter Ba2+ increased the titer because of the moderate expression of Kir2.1 channel. Channel blockade also increased the titers of the lentivirus expressing Kv1.4 and TREK channels, but not HERG. The increase in titer correlated with the K+ currents generated by the channels expressed. Conclusion In the production of lentivirus expressing K+ channels, titers are increased by blocking K+ currents in the virus-producing cells. This identifies a crucial issue in the production of viruses expressing membrane channels, and should facilitate basic and gene therapeutic research on channelopathies.
Collapse
|
36
|
Gui L, Qian H, Rocco KA, Grecu L, Niklason LE. Efficient intratracheal delivery of airway epithelial cells in mice and pigs. Am J Physiol Lung Cell Mol Physiol 2015; 308:L221-8. [PMID: 25416381 DOI: 10.1152/ajplung.00147.2014] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Cellular therapy via direct intratracheal delivery has gained interest as a novel therapeutic strategy for treating various pulmonary diseases including cystic fibrosis lung disease. However, concerns such as insufficient cell engraftment in lungs and lack of large animal model data remain to be resolved. This study aimed to establish a simple method for evaluating cell retention in lungs and to develop reproducible approaches for efficient cell delivery into mouse and pig lungs. Human lung epithelial cells including normal human bronchial/tracheal epithelial (NHBE) cells and human lung epithelial cell line A549 were infected with pSicoR-green fluorescent protein (GFP) lentivirus. GFP-labeled NHBE cells were delivered via a modified intratracheal cell instillation method into the lungs of C57BL/6J mice. Two days following cell delivery, GFP ELISA-based assay revealed a substantial cell-retention efficiency (10.48 ± 2.86%, n = 7) in mouse lungs preinjured with 2% polidocanol. When GFP-labeled A549 cells were transplanted into Yorkshire pig lungs with a tracheal intubation fiberscope, a robust initial cell attachment (22.32% efficiency) was observed at 24 h. In addition, a lentiviral vector was developed to induce the overexpression and apical localization of cystic fibrosis transmembrane conductance regulator (CFTR)-GFP fusion proteins in NHBE cells as a means of ex vivo CFTR gene transfer in nonprogenitor (relatively differentiated) lung epithelial cells. These results have demonstrated the convenience and efficiency of direct delivery of exogenous epithelial cells to lungs in mouse and pig models and provided important background for future preclinical evaluation of intratracheal cell transplantation to treat lung diseases.
Collapse
|
37
|
Canu N. Design and cloning of short hairpin RNAs (shRNAs) into a lentiviral silencing vector to study the function of selected proteins in neuronal apoptosis. Methods Mol Biol 2015; 1254:115-128. [PMID: 25431061 DOI: 10.1007/978-1-4939-2152-2_9] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 06/04/2023]
Abstract
Double-stranded RNA -mediated interference (RNAi ) is a new simple and fast research tool for shutting down genes and characterizes function of their respective proteins. Many strategies for design and delivery of siRNA to target cells are available. Here, we describe the use of lentiviral short hairpin RNA (shRNA) RNA silencing to identify the involvement of d-serine racemase (SR )- an enzyme that syntheses d-serine to modulate glutamate- N-methyl-D-aspartate receptor- in regulating rat cerebellar granule neurons (CGN ) apoptosis. Apoptosis is induced by serum and KCl withdrawal and is detected with fluorometric caspase 3 assay.
Collapse
Affiliation(s)
- Nadia Canu
- Department of System Medicine, University of "Tor Vergata" Rome, Via Montpellier 1, Rome, 00137, Italy,
| |
Collapse
|
38
|
Gargiulo G, Serresi M, Cesaroni M, Hulsman D, van Lohuizen M. In vivo shRNA screens in solid tumors. Nat Protoc 2014; 9:2880-902. [PMID: 25411954 DOI: 10.1038/nprot.2014.185] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Loss-of-function (LOF) experiments targeting multiple genes during tumorigenesis can be implemented using pooled shRNA libraries. RNAi screens in animal models rely on the use of multiple shRNAs to simultaneously disrupt gene function, as well as to serve as barcodes for cell fate outcomes during tumorigenesis. Here we provide a protocol for performing RNAi screens in orthotopic mouse tumor models, referring to glioma and lung adenocarcinoma as specific examples. The protocol aims to provide guidelines for applying RNAi to a diverse spectrum of solid tumors and to highlight crucial considerations when designing and performing these studies. It covers shRNA library assembly and packaging into lentiviral particles, and transduction into tumor-initiating cells (TICs), followed by in vivo transplantation, tumor DNA recovery, sequencing and analysis. Depending on the target genes and tumor model, tumor suppressors and oncogenes can be identified or biological pathways can be dissected in 6-9 weeks.
Collapse
Affiliation(s)
- Gaetano Gargiulo
- Division of Molecular Genetics, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Michela Serresi
- Division of Molecular Genetics, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Matteo Cesaroni
- Fels Institute, Temple University School of Medicine, Philadelphia, Pennsylvania, USA
| | - Danielle Hulsman
- Division of Molecular Genetics, the Netherlands Cancer Institute, Amsterdam, the Netherlands
| | - Maarten van Lohuizen
- 1] Division of Molecular Genetics, the Netherlands Cancer Institute, Amsterdam, the Netherlands. [2] Cancer Genomics Centre, the Netherlands
| |
Collapse
|
39
|
Porter SN, Baker LC, Mittelman D, Porteus MH. Lentiviral and targeted cellular barcoding reveals ongoing clonal dynamics of cell lines in vitro and in vivo. Genome Biol 2014; 15:R75. [PMID: 24886633 PMCID: PMC4073073 DOI: 10.1186/gb-2014-15-5-r75] [Citation(s) in RCA: 56] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2013] [Accepted: 05/30/2014] [Indexed: 12/03/2022] Open
Abstract
Background Cell lines are often regarded as clonal, even though this simplifies what is known about mutagenesis, transformation and other processes that destabilize them over time. Monitoring these clonal dynamics is important for multiple areas of biomedical research, including stem cell and cancer biology. Tracking the contributions of individual cells to large populations, however, has been constrained by limitations in sensitivity and complexity. Results We utilize cellular barcoding methods to simultaneously track the clonal contributions of tens of thousands of cells. We demonstrate that even with optimal culturing conditions, common cell lines including HeLa, K562 and HEK-293 T exhibit ongoing clonal dynamics. Starting a population with a single clone diminishes but does not eradicate this phenomenon. Next, we compare lentiviral and zinc-finger nuclease barcode insertion approaches, finding that the zinc-finger nuclease protocol surprisingly results in reduced clonal diversity. We also document the expected reduction in clonal complexity when cells are challenged with genotoxic stress. Finally, we demonstrate that xenografts maintain clonal diversity to a greater extent than in vitro culturing of the human non-small-cell lung cancer cell line HCC827. Conclusions We demonstrate the feasibility of tracking and quantifying the clonal dynamics of entire cell populations within multiple cultured cell lines. Our results suggest that cell heterogeneity should be considered in the design and interpretation of in vitro culture experiments. Aside from clonal cell lines, we propose that cellular barcoding could prove valuable in modeling the clonal behavior of heterogeneous cell populations over time, including tumor populations treated with chemotherapeutic agents.
Collapse
|
40
|
Yang T, Burrows C, Park JH. Development of a doxycycline-inducible lentiviral plasmid with an instant regulatory feature. Plasmid 2014; 72:29-35. [PMID: 24727543 DOI: 10.1016/j.plasmid.2014.04.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2013] [Revised: 03/23/2014] [Accepted: 04/01/2014] [Indexed: 12/19/2022]
Abstract
Lentiviruses provide highly efficient gene delivery vehicles in both dividing and non-dividing cells. Inducible gene expression systems often employ a specific cell line that constitutively expresses a regulatory protein for transgene expression. As one of such inducible expression systems the Tet-On system uses a cell line expressing reverse tetracycline-responsive transcriptional activator (rtTA). The rtTA protein binds to the tetracycline-responsive element (TRE) in the promoter and activates transcription of a transgene in a doxycycline-dependent manner. To establish a universal and instant regulatory system without generating Tet-On cell lines, the cDNAs of rtTA and a testing target gene (PPM1B) were cloned in the bi-directional TRE-containing promoters. Here, we examined whether a basal leaky expression of rtTA allows instantly inducible expression of both rtTA itself and the target gene, PPM1B in a single plasmid using the two mini-CMV promoters. Transient transfection of the lentiviral plasmids into human embryonic kidney HEK293T cells showed a significant induction of PPM1B expression in response to doxycycline, suggesting that these lentiviral plasmids can be used as an instantly inducible mammalian expression vector. However, the expression of rtTA by lentiviral transduction shows a minimal expression without a consistent response to doxycycline, suggesting that the utility of these lentiviral vectors is limited. A potential solution to overcome lentiviral transgene inactivation is proposed.
Collapse
Affiliation(s)
- Tian Yang
- Institute of Fundamental Sciences, Massey University, Palmerston North, New Zealand; Institute of Somatology, Nanjing Medical University, 140 Hanzhong Road, Nanjing, Jiangsu 210029, PR China
| | - Christopher Burrows
- Institute of Fundamental Sciences, Massey University, Palmerston North, New Zealand
| | - Jeong Hyeon Park
- Institute of Fundamental Sciences, Massey University, Palmerston North, New Zealand.
| |
Collapse
|
41
|
Parker MW, Rossi D, Peterson M, Smith K, Sikström K, White ES, Connett JE, Henke CA, Larsson O, Bitterman PB. Fibrotic extracellular matrix activates a profibrotic positive feedback loop. J Clin Invest 2014; 124:1622-35. [PMID: 24590289 DOI: 10.1172/jci71386] [Citation(s) in RCA: 414] [Impact Index Per Article: 37.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2013] [Accepted: 12/27/2013] [Indexed: 12/13/2022] Open
Abstract
Pathological remodeling of the extracellular matrix (ECM) by fibroblasts leads to organ failure. Development of idiopathic pulmonary fibrosis (IPF) is characterized by a progressive fibrotic scarring in the lung that ultimately leads to asphyxiation; however, the cascade of events that promote IPF are not well defined. Here, we examined how the interplay between the ECM and fibroblasts affects both the transcriptome and translatome by culturing primary fibroblasts generated from IPF patient lung tissue or nonfibrotic lung tissue on decellularized lung ECM from either IPF or control patients. Surprisingly, the origin of the ECM had a greater impact on gene expression than did cell origin, and differences in translational control were more prominent than alterations in transcriptional regulation. Strikingly, genes that were translationally activated by IPF-derived ECM were enriched for those encoding ECM proteins detected in IPF tissue. We determined that genes encoding IPF-associated ECM proteins are targets for miR-29, which was downregulated in fibroblasts grown on IPF-derived ECM, and baseline expression of ECM targets could be restored by overexpression of miR-29. Our data support a model in which fibroblasts are activated to pathologically remodel the ECM in IPF via a positive feedback loop between fibroblasts and aberrant ECM. Interrupting this loop may be a strategy for IPF treatment.
Collapse
|
42
|
Chahal PS, Schulze E, Tran R, Montes J, Kamen AA. Production of adeno-associated virus (AAV) serotypes by transient transfection of HEK293 cell suspension cultures for gene delivery. J Virol Methods 2013; 196:163-73. [PMID: 24239634 PMCID: PMC7113661 DOI: 10.1016/j.jviromet.2013.10.038] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2013] [Revised: 10/11/2013] [Accepted: 10/29/2013] [Indexed: 01/29/2023]
Abstract
Transient transfection of HEK293 suspension cells efficiently produce AAV vectors. Nine different AAV serotypes were produced with yields of 1E+13 Vg/L. AAV2 and AAV6 produced in 3-L bioreactors gave yields comparable to shake-flasks. The process is cGMP compatible using serum-free media and HEK293 master cell bank. Industrialization of the process is possible for manufacturing AAV serotypes.
Adeno-associated virus (AAV) is being used successfully in gene therapy. Different serotypes of AAV target specific organs and tissues with high efficiency. There exists an increasing demand to manufacture various AAV serotypes in large quantities for pre-clinical and clinical trials. A generic and scalable method has been described in this study to efficiently produce AAV serotypes (AAV1-9) by transfection of a fully characterized cGMP HEK293SF cell line grown in suspension and serum-free medium. First, the production parameters were evaluated using AAV2 as a model serotype. Second, all nine AAV serotypes were produced successfully with yields of 1013 Vg/L cell culture. Subsequently, AAV2 and AAV6 serotypes were produced in 3-L controlled bioreactors where productions yielded up to 1013 Vg/L similar to the yields obtained in shake-flasks. For example, for AAV2 1013 Vg/L cell culture (6.8 × 1011 IVP/L) were measured between 48 and 64 h post transfection (hpt). During this period, the average cell specific AAV2 yields of 6800 Vg per cell and 460 IVP per cell were obtained with a Vg to IVP ratio of less than 20. Successful operations in bioreactors demonstrated the potential for scale-up and industrialization of this generic process for manufacturing AAV serotypes efficiently.
Collapse
Affiliation(s)
- Parminder Singh Chahal
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Erica Schulze
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Rosa Tran
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Johnny Montes
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2
| | - Amine A Kamen
- Vaccine Program, Human Health Therapeutics Portfolio, National Research Council of Canada, 6100 Royalmount Avenue, Montreal, QC, Canada H4P2R2.
| |
Collapse
|
43
|
Ramsey JE, Fontes JD. The zinc finger transcription factor ZXDC activates CCL2 gene expression by opposing BCL6-mediated repression. Mol Immunol 2013; 56:768-80. [PMID: 23954399 DOI: 10.1016/j.molimm.2013.07.001] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2013] [Revised: 06/28/2013] [Accepted: 07/04/2013] [Indexed: 12/12/2022]
Abstract
The zinc finger X-linked duplicated (ZXD) family of transcription factors has been implicated in regulating transcription of major histocompatibility complex class II genes in antigen presenting cells; roles beyond this function are not yet known. The expression of one gene in this family, ZXD family zinc finger C (ZXDC), is enriched in myeloid lineages and therefore we hypothesized that ZXDC may regulate myeloid-specific gene expression. Here we demonstrate that ZXDC regulates genes involved in myeloid cell differentiation and inflammation. Overexpression of the larger isoform of ZXDC, ZXDC1, activates expression of monocyte-specific markers of differentiation and synergizes with phorbol 12-myristate 13-acetate (which causes differentiation) in the human leukemic monoblast cell line U937. To identify additional gene targets of ZXDC1, we performed gene expression profiling which revealed multiple inflammatory gene clusters regulated by ZXDC1. Using a combination of approaches we show that ZXDC1 activates transcription of a gene within one of the regulated clusters, chemokine (C-C motif) ligand 2 (CCL2; monocyte chemoattractant protein 1; MCP1) via a previously defined distal regulatory element. Further, ZXDC1-dependent up-regulation of the gene involves eviction of the transcriptional repressor B-cell CLL/lymphoma 6 (BCL6), a factor known to be important in resolving inflammatory responses, from this region of the promoter. Collectively, our data show that ZXDC1 is a regulator in the process of myeloid function and that ZXDC1 is responsible for Ccl2 gene de-repression by BCL6.
Collapse
Affiliation(s)
- Jon E Ramsey
- Department of Biochemistry and Molecular Biology, University of Kansas School of Medicine, 3901 Rainbow Boulevard, MS3030, Kansas City, KS 66160, USA.
| | | |
Collapse
|
44
|
Segura MM, Mangion M, Gaillet B, Garnier A. New developments in lentiviral vector design, production and purification. Expert Opin Biol Ther 2013; 13:987-1011. [PMID: 23590247 DOI: 10.1517/14712598.2013.779249] [Citation(s) in RCA: 86] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
INTRODUCTION Lentiviruses are a very potent class of viral vectors for which there is presently a rapidly growing interest for a number of gene therapy. However, their construction, production and purification need to be performed according to state-of-the-art techniques in order to obtain sufficient quantities of high purity material of any usefulness and safety. AREAS COVERED The recent advances in the field of recombinant lentivirus vector design, production and purification will be reviewed with an eye toward its utilization for gene therapy. Such a review should be helpful for the potential user of this technology. EXPERT OPINION The principal hurdles toward the use of recombinant lentivirus as a gene therapy vector are the low titer at which it is produced as well as the difficulty to purify it at an acceptable level without degrading it. The recent advances in the bioproduction of this vector suggest these issues are about to be resolved, making the retrovirus gene therapy a mature technology.
Collapse
Affiliation(s)
- Maria Mercedes Segura
- Chemical Engineering Department, Universitat Autònoma de Barcelona, Campus Bellaterra, Cerdanyola del Vallès (08193), Barcelona, Spain
| | | | | | | |
Collapse
|
45
|
Abstract
The continuous production of spermatazoa throughout the reproductive lifetime of a male depends on the maintenance of a pool of progenitor cells called spermatogonial stem cells (SSCs). SSCs represent a very small fraction of the cellular population in the testes and lack definitive molecular markers for their identification. The discovery of conditions that allow one to propagate mouse SSCs in vitro essentially indefinitely has truly facilitated studies of the molecular mechanisms regulating SSC function. While multiple conditions for culturing SSCs have now been described, here we detail a method for culturing SSCs that uses a simpler medium than the original formulation. As with numerous other primary and stem cell cultures, it is difficult to introduce DNA into cultured SSCs using standard transfection approaches. However, VSV-G pseudotyped lentivirus efficiently infects cultured SSCs with minimal toxicity. Here we present protocols for producing lentivirus and stably modifying the genome of cultured SSCs using lentiviral vectors.
Collapse
|
46
|
Danos AM, Liao Y, Li X, Du W. Functional inactivation of Rb sensitizes cancer cells to TSC2 inactivation induced cell death. Cancer Lett 2012; 328:36-43. [PMID: 23022476 DOI: 10.1016/j.canlet.2012.09.016] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2012] [Revised: 09/18/2012] [Accepted: 09/19/2012] [Indexed: 11/16/2022]
Abstract
We showed previously that inactivation of TSC2 induces death in cancer cells lacking the Retinoblastoma (Rb) tumor suppressor under stress conditions, suggesting that inactivation of TSC2 can potentially be used as an approach to specifically kill cancers that have lost WT Rb. As Rb is often inactivated in cancers by overexpression of cyclin D1, loss of p16(ink4a) cdk inhibitor, or expression of viral oncoproteins, it will be interesting to determine if such functional inactivation of Rb would similarly sensitize cancer cells to TSC2 inactivation induced cell death. In addition, many cancers lack functional Pten, resulting in increased PI3K/Akt signaling that has been shown to modulate E2F-induced cell death. Therefore it will be interesting to test whether loss of Pten will affect TSC2 inactivation induced killing of Rb mutant cancer cells. Here, we show that overexpression of Cyclin D1 or the viral oncogene E1a sensitizes cancer cells to TSC2 knockdown induced cell death and growth inhibition. On the other hand, knockdown of p16(ink4a) sensitizes cancer cells to TSC2 knockdown induced cell death in a manner that is likely dependant on serum induction of Cyclin D1 to inactivate the Rb function. Additionally, we demonstrate that loss of Pten does not interfere with TSC2 knockdown induced cell death in Rb mutant cancer cells. Together, these results suggest that TSC2 is potentially a useful target for a large spectrum of cancer types with an inactivated Rb pathway.
Collapse
Affiliation(s)
- Arpad M Danos
- Ben May Department for Cancer Research, The University of Chicago, Chicago, IL 60637, United States
| | | | | | | |
Collapse
|
47
|
Garcia JM, Lai JCC. Production of influenza pseudotyped lentiviral particles and their use in influenza research and diagnosis: an update. Expert Rev Anti Infect Ther 2011; 9:443-55. [PMID: 21504401 DOI: 10.1586/eri.11.25] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Pseudotyped viral particles are being used as safe surrogates to mimic the structure and surface of many viruses, including highly pathogenic viruses such as avian influenza H5N1, to investigate biological functions mediated by the envelope proteins derived from these viruses. The first part of this article evaluates and discusses the differences in the production and characterization of influenza pseudoparticles. The second part focuses on the applications that such a flexible tool can provide in modern influenza research, in particular in the fields of drug discovery, molecular biology and diagnosis.
Collapse
Affiliation(s)
- Jean-Michel Garcia
- HKU-Pasteur Research Centre, Dexter HC Man Building, 8 Sassoon Road, Pokfulam, Hong Kong.
| | | |
Collapse
|