201
|
Cuesta-Domínguez Á, León-Rico D, Álvarez L, Díez B, Bodega-Mayor I, Baños R, Martín-Rey MÁ, Santos-Roncero M, Gaspar ML, Martín-Acosta P, Almarza E, Bueren JA, Río P, Fernández-Ruiz E. BCR-JAK2 drives a myeloproliferative neoplasm in transplanted mice. J Pathol 2015; 236:219-28. [DOI: 10.1002/path.4513] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2014] [Revised: 01/28/2015] [Accepted: 02/05/2015] [Indexed: 01/13/2023]
Affiliation(s)
- Álvaro Cuesta-Domínguez
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - Diego León-Rico
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Lara Álvarez
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Begoña Díez
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Irene Bodega-Mayor
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - Rocío Baños
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Miguel Ángel Martín-Rey
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Matilde Santos-Roncero
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| | - María Luisa Gaspar
- Centro Nacional de Microbiología; Instituto de Salud Carlos III (ISCIII); Majadahonda Spain
| | - Paloma Martín-Acosta
- Servicio de Anatomía Patológica; Hospital Universitario Puerta de Hierro; Majadahonda Spain
| | - Elena Almarza
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Juan A. Bueren
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Paula Río
- Division of Haematopoietic Innovative Therapies; CIEMAT/CIBERER; Madrid Spain
- Instituto de Investigaciones Sanitarias Fundación Jiménez Díaz (IIS-FJD, UAM); Madrid Spain
| | - Elena Fernández-Ruiz
- Molecular Biology Unit; Instituto de Investigación Sanitaria Princesa (IIS-P, UAM), Hospital Universitario de La Princesa; Madrid Spain
| |
Collapse
|
202
|
Transgenic mouse model expressing P53(R172H), luciferase, EGFP, and KRAS(G12D) in a single open reading frame for live imaging of tumor. Sci Rep 2015; 5:8053. [PMID: 25623590 PMCID: PMC4306974 DOI: 10.1038/srep08053] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2014] [Accepted: 01/02/2015] [Indexed: 01/21/2023] Open
Abstract
Genetically engineered mouse cancer models allow tumors to be imaged in vivo via co-expression of a reporter gene with a tumor-initiating gene. However, differential transcriptional and translational regulation between the tumor-initiating gene and the reporter gene can result in inconsistency between the actual tumor size and the size indicated by the imaging assay. To overcome this limitation, we developed a transgenic mouse in which two oncogenes, encoding P53R172H and KRASG12D, are expressed together with two reporter genes, encoding enhanced green fluorescent protein (EGFP) and firefly luciferase, in a single open reading frame following Cre-mediated DNA excision. Systemic administration of adenovirus encoding Cre to these mice induced specific transgene expression in the liver. Repeated bioluminescence imaging of the mice revealed a continuous increase in the bioluminescent signal over time. A strong correlation was found between the bioluminescent signal and actual tumor size. Interestingly, all liver tumors induced by P53R172H and KRASG12D in the model were hepatocellular adenomas. The mouse model was also used to trace cell proliferation in the epidermis via live fluorescence imaging. We anticipate that the transgenic mouse model will be useful for imaging tumor development in vivo and for investigating the oncogenic collaboration between P53R172H and KRASG12D.
Collapse
|
203
|
Construction and characterization of VL–VH tail-parallel genetically engineered antibodies against staphylococcal enterotoxins. Immunol Res 2015; 61:281-93. [DOI: 10.1007/s12026-015-8623-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
|
204
|
Osakada F, Takahashi M. Challenges in retinal circuit regeneration: linking neuronal connectivity to circuit function. Biol Pharm Bull 2015; 38:341-57. [PMID: 25757915 DOI: 10.1248/bpb.b14-00771] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tremendous progress has been made in retinal regeneration, as exemplified by successful transplantation of retinal pigment epithelia and photoreceptor cells in the adult retina, as well as by generation of retinal tissue from embryonic stem cells and induced pluripotent cells. However, it remains unknown how new photoreceptors integrate within retinal circuits and contribute to vision restoration. There is a large gap in our understanding, at both the cellular and behavioral levels, of the functional roles of new neurons in the adult retina. This gap largely arises from the lack of appropriate methods for analyzing the organization and function of new neurons at the circuit level. To bridge this gap and understand the functional roles of new neurons in living animals, it will be necessary to identify newly formed connections, correlate them with function, manipulate their activity, and assess the behavioral outcome of these manipulations. Recombinant viral vectors are powerful tools not only for controlling gene expression and reprogramming cells, but also for tracing cell fates and neuronal connectivity, monitoring biological functions, and manipulating the physiological state of a specific cell population. These virus-based approaches, combined with electrophysiology and optical imaging, will provide circuit-level insight into neural regeneration and facilitate new strategies for achieving vision restoration in the adult retina. Herein, we discuss challenges and future directions in retinal regeneration research.
Collapse
Affiliation(s)
- Fumitaka Osakada
- Laboratory of Cellular Pharmacology, Graduate School of Pharmaceutical Sciences, Nagoya University; Systems Neurobiology Laboratory, The Salk Institute for Biological Studies, California 92037, USA; PRESTO, Japan Science and Technology Agency, Saitama 332-0012, Japan.
| | | |
Collapse
|
205
|
Roth JC, Alberti MO, Ismail M, Lingas KT, Reese JS, Gerson SL. MGMT enrichment and second gene co-expression in hematopoietic progenitor cells using separate or dual-gene lentiviral vectors. Virus Res 2014; 196:170-80. [PMID: 25479595 DOI: 10.1016/j.virusres.2014.11.027] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2014] [Revised: 11/20/2014] [Accepted: 11/25/2014] [Indexed: 12/21/2022]
Abstract
The DNA repair gene O(6)-methylguanine-DNA methyltransferase (MGMT) allows efficient in vivo enrichment of transduced hematopoietic stem cells (HSC). Thus, linking this selection strategy to therapeutic gene expression offers the potential to reconstitute diseased hematopoietic tissue with gene-corrected cells. However, different dual-gene expression vector strategies are limited by poor expression of one or both transgenes. To evaluate different co-expression strategies in the context of MGMT-mediated HSC enrichment, we compared selection and expression efficacies in cells cotransduced with separate single-gene MGMT and GFP lentivectors to those obtained with dual-gene vectors employing either encephalomyocarditis virus (EMCV) internal ribosome entry site (IRES) or foot and mouth disease virus (FMDV) 2A elements for co-expression strategies. Each strategy was evaluated in vitro and in vivo using equivalent multiplicities of infection (MOI) to transduce 5-fluorouracil (5-FU) or Lin(-)Sca-1(+)c-kit(+) (LSK)-enriched murine bone marrow cells (BMCs). The highest dual-gene expression (MGMT(+)GFP(+)) percentages were obtained with the FMDV-2A dual-gene vector, but half of the resulting gene products existed as fusion proteins. Following selection, dual-gene expression percentages in single-gene vector cotransduced and dual-gene vector transduced populations were similar. Equivalent MGMT expression levels were obtained with each strategy, but GFP expression levels derived from the IRES dual-gene vector were significantly lower. In mice, vector-insertion averages were similar among cells enriched after dual-gene vectors and those cotransduced with single-gene vectors. These data demonstrate the limitations and advantages of each strategy in the context of MGMT-mediated selection, and may provide insights into vector design with respect to a particular therapeutic gene or hematologic defect.
Collapse
Affiliation(s)
- Justin C Roth
- Molecular Virology Training Program, Case Western Reserve University, Cleveland, OH, USA; Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Michael O Alberti
- Division of Pediatric Infectious Diseases, Department of Pediatrics, University of Alabama at Birmingham, Birmingham, AL, USA.
| | - Mourad Ismail
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Karen T Lingas
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA.
| | - Jane S Reese
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, OH, USA.
| | - Stanton L Gerson
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; Seidman Cancer Center, University Hospitals Case Medical Center, Cleveland, OH, USA; National Center for Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, OH, USA; The Center for Stem Cell and Regenerative Medicine, Cleveland, OH, USA.
| |
Collapse
|
206
|
Kang W, Marasco WA, Tong HI, Byron MM, Wu C, Shi Y, Sun S, Sun Y, Lu Y. Anti-tat Hutat2:Fc mediated protection against tat-induced neurotoxicity and HIV-1 replication in human monocyte-derived macrophages. J Neuroinflammation 2014; 11:195. [PMID: 25416164 PMCID: PMC4256057 DOI: 10.1186/s12974-014-0195-2] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Accepted: 11/05/2014] [Indexed: 01/01/2023] Open
Abstract
BACKGROUND HIV-1 Tat is essential for HIV replication and is also a well-known neurotoxic factor causing HIV-associated neurocognitive disorder (HAND). Currently, combined antiretroviral therapy targeting HIV reverse transcriptase or protease cannot prevent the production of early viral proteins, especially Tat, once HIV infection has been established. HIV-infected macrophages and glial cells in the brain still release Tat into the extracellular space where it can exert direct and indirect neurotoxicity. Therefore, stable production of anti-Tat antibodies in the brain would neutralize HIV-1 Tat and thus provide an effective approach to protect neurons. METHODS We constructed a humanized anti-Tat Hutat2:Fc fusion protein with the goal of antagonizing HIV-1 Tat and delivered the gene into cell lines and primary human monocyte-derived macrophages (hMDM) by an HIV-based lentiviral vector. The function of the anti-Tat Hutat2:Fc fusion protein and the potential side effects of lentiviral vector-mediated gene transfer were evaluated in vitro. RESULTS Our study demonstrated that HIV-1-based lentiviral vector-mediated gene transduction resulted in a high-level, stable expression of anti-HIV-1 Tat Hutat2:Fc in human neuronal and monocytic cell lines, as well as in primary hMDM. Hutat2:Fc was detectable in both cells and supernatants and continued to accumulate to high levels within the supernatant. Hutat2:Fc protected mouse cortical neurons against HIV-1 Tat86-induced neurotoxicity. In addition, both secreted Hutat2:Fc and transduced hMDM led to reducing HIV-1BaL viral replication in human macrophages. Moreover, lentiviral vector-based gene introduction did not result in any significant changes in cytomorphology and cell viability. Although the expression of IL8, STAT1, and IDO1 genes was up-regulated in transduced hMDM, such alternation in gene expression did not affect the neuroprotective effect of Hutat2:Fc. CONCLUSIONS Our study demonstrated that lentivirus-mediated gene transfer could efficiently deliver the Hutat2:Fc gene into primary hMDM and does not lead to any significant changes in hMDM immune-activation. The neuroprotective and HIV-1 suppressive effects produced by Hutat2:Fc were comparable to that of a full-length anti-Tat antibody. This study provides the foundation and insights for future research on the potential use of Hutat2:Fc as a novel gene therapy approach for HAND through utilizing monocytes/macrophages, which naturally cross the blood-brain barrier, for gene delivery.
Collapse
Affiliation(s)
- Wen Kang
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China. .,Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Wayne A Marasco
- Department of Cancer Immunology and AIDS, Dana-Farber Cancer Institute, Harvard Medical School, 50 Brookline Avenue, Boston, MA, 02215, USA.
| | - Hsin-I Tong
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Mary Margaret Byron
- Hawaii Center for AIDS, John A. Burns School of Medicine, University of Hawaii, 651 Ilalo St., BSB, Suite 231, Honolulu, HI, 96813, USA.
| | - Chengxiang Wu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yingli Shi
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Si Sun
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| | - Yongtao Sun
- Department of Infectious Diseases, Tangdu Hospital, The Fourth Military Medical University, 569 Xinsi Road, Xi'an, Shaanxi, 710038, China.
| | - Yuanan Lu
- Department of Public Health Sciences, John A. Burns School of Medicine, University of Hawaii, 1960 East-west Road, Honolulu, HI, 96822, USA.
| |
Collapse
|
207
|
Chen H, Gao N, Wu J, Zheng X, Li J, Fan D, An J. Variable effects of the co-administration of a GM-CSF-expressing plasmid on the immune response to flavivirus DNA vaccines in mice. Immunol Lett 2014; 162:140-8. [DOI: 10.1016/j.imlet.2014.08.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2013] [Revised: 08/05/2014] [Accepted: 08/05/2014] [Indexed: 11/26/2022]
|
208
|
Mammalian cell display technology coupling with AID induced SHM in vitro: an ideal approach to the production of therapeutic antibodies. Int Immunopharmacol 2014; 23:380-6. [PMID: 25281392 DOI: 10.1016/j.intimp.2014.09.017] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2014] [Revised: 09/12/2014] [Accepted: 09/12/2014] [Indexed: 11/21/2022]
Abstract
Traditional antibody production technology within non-mammalian cell expression systems has shown many unsatisfactory properties for the development of therapeutic antibodies. Nevertheless, mammalian cell display technology reaps the benefits of producing full-length all human antibodies. Together with the developed cytidine deaminase induced in vitro somatic hypermutation technology, mammalian cell display technology provides the opportunity to produce high affinity antibodies that might be ideal for therapeutic application. This review was concentrated on the development of the mammalian cell display technology as well as the activation-induced cytidine deaminase induced in vitro somatic hypermutation technology and their applications for the production of therapeutic antibodies.
Collapse
|
209
|
Bosch MK, Nerbonne JM, Ornitz DM. Dual transgene expression in murine cerebellar Purkinje neurons by viral transduction in vivo. PLoS One 2014; 9:e104062. [PMID: 25093726 PMCID: PMC4122438 DOI: 10.1371/journal.pone.0104062] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/21/2014] [Accepted: 07/08/2014] [Indexed: 12/19/2022] Open
Abstract
Viral-vector mediated gene transfer to cerebellar Purkinje neurons in vivo is a promising avenue for gene therapy of cerebellar ataxias and for genetic manipulation in functional studies of animal models of cerebellar disease. Here, we report the results of experiments designed to identify efficient methods for viral transduction of adult murine Purkinje neurons in vivo. For these analyses, several lentiviral and an adeno-associated virus (AAV), serotype 1, vector with various promoter combinations were generated and compared for in situ transduction efficiency, assayed by fluorescent reporter protein expression in Purkinje neurons. Additional experiments were also conducted to identify the optimal experimental strategy for co-expression of two proteins in individual Purkinje neurons. Of the viruses tested, AAV1 with a CAG promoter exhibited the highest specificity for Purkinje neurons. To deliver two proteins to the same Purkinje neuron, several methods were tested, including: an internal ribosome entry site (IRES), a 2A sequence, a dual promoter vector, and co-injection of two viruses. Efficient expression of both proteins in the same Purkinje neuron was only achieved by co-injecting two AAV1-CAG viruses. We found that use of an AAV1-CAG virus outperformed similar lentivirus vectors and that co-injection of two AAV1-CAG viruses could be used to efficiently deliver two proteins to the same Purkinje neuron in adult mice. AAV1 with a CAG promoter is highly efficient and selective at transducing adult cerebellar Purkinje neurons and two AAV-CAG viruses can be used to efficiently express two proteins in the same neuron in vivo.
Collapse
Affiliation(s)
- Marie K. Bosch
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - Jeanne M. Nerbonne
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- Internal Medicine, Washington University School of Medicine, Saint Louis, Missouri, United States of America
| | - David M. Ornitz
- Developmental Biology, Washington University School of Medicine, Saint Louis, Missouri, United States of America
- * E-mail:
| |
Collapse
|
210
|
Chuang KH, Hsieh YC, Chiang IS, Chuang CH, Kao CH, Cheng TC, Wang YT, Lin WW, Chen BM, Roffler SR, Huang MY, Cheng TL. High-throughput sorting of the highest producing cell via a transiently protein-anchored system. PLoS One 2014; 9:e102569. [PMID: 25036759 PMCID: PMC4103822 DOI: 10.1371/journal.pone.0102569] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2014] [Accepted: 06/20/2014] [Indexed: 11/19/2022] Open
Abstract
Developing a high-throughput method for the effecient selection of the highest producing cell is very important for the production of recombinant protein drugs. Here, we developed a novel transiently protein-anchored system coupled with fluorescence activated cell sorting (FACS) for the efficient selection of the highest producing cell. A furin cleavage peptide (RAKR) was used to join a human anti-epithelial growth factor antibody (αEGFR Ab) and the extracellular-transmembrane-cytosolic domains of the mouse B7-1 antigen (B7). The furin inhibitor can transiently switch secreted αEGFR Ab into a membrane-anchored form. After cell sorting, the level of membrane αEGFR Ab-RAKR-B7 is proportional to the amount of secreted αEGFR Ab in the medium. We further selected 23 αEGFR Ab expressing cells and demonstrated a high correlation (R2 = 0.9165) between the secretion level and surface expression levels of αEGFR Ab. These results suggested that the novel transiently protein-anchored system can easily and efficiently select the highest producing cells, reducing the cost for the production of biopharmaceuticals.
Collapse
Affiliation(s)
- Kuo-Hsiang Chuang
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
- Ph.D. Program for Clinical Drug Discovery from Botanical Herbs, Taipei Medical University, Taipei, Taiwan
- Master Program for Clinical Pharmacogenomics and Pharmacoproteomics, Taipei Medical University, Taipei, Taiwan
| | - Yuan-Chin Hsieh
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - I-Shiuan Chiang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chih-Hung Chuang
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Chien-Han Kao
- Graduate Institute of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ta-Chun Cheng
- Graduate Institute of Pharmacognosy, Taipei Medical University, Taipei, Taiwan
| | - Yeng-Tseng Wang
- Department of Biochemistry, College of Medicine, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Wen-Wei Lin
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
| | - Bing-Mae Chen
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Steve R. Roffler
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan
| | - Ming-Yii Huang
- Department of Radiation Oncology, Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung Medical University, Kaohsiung, Taiwan
- * E-mail: (MYH); (TLC)
| | - Tian-Lu Cheng
- Department of Biomedical Science and Environmental Biology, Kaohsiung Medical University, Kaohsiung, Taiwan
- Institute of Biomedical Sciences, National Sun Yat-Sen University, Kaohsiung, Taiwan
- Cancer Center, Kaohsiung Medical University Hospital, Kaohsiung, Taiwan
- * E-mail: (MYH); (TLC)
| |
Collapse
|
211
|
Sadikoglou E, Daoutsali E, Petridou E, Grigoriou M, Skavdis G. Comparative analysis of internal ribosomal entry sites as molecular tools for bicistronic expression. J Biotechnol 2014; 181:31-4. [DOI: 10.1016/j.jbiotec.2014.03.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Revised: 03/02/2014] [Accepted: 03/25/2014] [Indexed: 10/25/2022]
|
212
|
Hu K. Vectorology and factor delivery in induced pluripotent stem cell reprogramming. Stem Cells Dev 2014; 23:1301-15. [PMID: 24625220 PMCID: PMC4046209 DOI: 10.1089/scd.2013.0621] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2013] [Accepted: 03/13/2014] [Indexed: 12/26/2022] Open
Abstract
Induced pluripotent stem cell (iPSC) reprogramming requires sustained expression of multiple reprogramming factors for a limited period of time (10-30 days). Conventional iPSC reprogramming was achieved using lentiviral or simple retroviral vectors. Retroviral reprogramming has flaws of insertional mutagenesis, uncontrolled silencing, residual expression and re-activation of transgenes, and immunogenicity. To overcome these issues, various technologies were explored, including adenoviral vectors, protein transduction, RNA transfection, minicircle DNA, excisable PiggyBac (PB) transposon, Cre-lox excision system, negative-sense RNA replicon, positive-sense RNA replicon, Epstein-Barr virus-based episomal plasmids, and repeated transfections of plasmids. This review provides summaries of the main vectorologies and factor delivery systems used in current reprogramming protocols.
Collapse
Affiliation(s)
- Kejin Hu
- Department of Biochemistry and Molecular Genetics, UAB Stem Cell Institute, School of Medicine, University of Alabama at Birmingham , Birmingham, Alabama
| |
Collapse
|
213
|
Levin RA, Felsen CN, Yang J, Lin JY, Whitney MA, Nguyen QT, Tsien RY. An optimized triple modality reporter for quantitative in vivo tumor imaging and therapy evaluation. PLoS One 2014; 9:e97415. [PMID: 24816650 PMCID: PMC4016317 DOI: 10.1371/journal.pone.0097415] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 04/17/2014] [Indexed: 11/24/2022] Open
Abstract
We present an optimized triple modality reporter construct combining a far-red fluorescent protein (E2-Crimson), enhanced firefly luciferase enzyme (Luc2), and truncated wild type herpes simplex virus I thymidine kinase (wttk) that allows for sensitive, long-term tracking of tumor growth in vivo by fluorescence, bioluminescence, and positron emission tomography. Two human cancer cell lines (MDA-MB-231 breast cancer and HT-1080 fibrosarcoma cancer) were successfully transduced to express this triple modality reporter. Fluorescence and bioluminescence imaging of the triple modality reporter were used to accurately quantify the therapeutic responses of MDA-MB-231 tumors to the chemotherapeutic agent monomethyl auristatin E in vivo in athymic nude mice. Positive correlation was observed between the fluorescence and bioluminescence signals, and these signals were also positively correlated with the ex vivo tumor weights. This is the first reported use of both fluorescence and bioluminescence signals from a multi-modality reporter construct to measure drug efficacy in vivo.
Collapse
Affiliation(s)
- Rachel A. Levin
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Csilla N. Felsen
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Jin Yang
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - John Y. Lin
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Michael A. Whitney
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Quyen T. Nguyen
- Division of Otolaryngology/Head and Neck Surgery, University of California San Diego, La Jolla, California, United States of America
| | - Roger Y. Tsien
- Department of Pharmacology, UCSD School of Medicine, University of California San Diego, La Jolla, California, United States of America
- Howard Hughes Medical Institute, La Jolla, California, United States of America
| |
Collapse
|
214
|
Kumar P, Ayalew LE, Godson DL, Gaba A, Babiuk LA, Tikoo SK. Mucosal immunization of calves with recombinant bovine adenovirus-3 coexpressing truncated form of bovine herpesvirus-1 gD and bovine IL-6. Vaccine 2014; 32:3300-6. [PMID: 24731813 DOI: 10.1016/j.vaccine.2014.03.073] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2014] [Revised: 03/15/2014] [Accepted: 03/25/2014] [Indexed: 11/30/2022]
Abstract
Previous studies have suggested an important role of the cytokine adjuvant IL-6 in the induction of mucosal immune responses in animals, including mice. Here, we report the in vivo ability of bovine adenovirus (BAdV)-3 expressing bovine (Bo) IL-6, to influence the systemic and mucosal immune responses against bovine herpesvirus (BHV)-1 gDt in calves. To co-express both antigen and cytokine, we first constructed a recombinant BAdV-3 expressing chimeric gDt.BoIL-6 protein (BAV326). Secondly, we constructed another recombinant BAdV-3 simultaneously expressing gDt and BoIL-6 using IRES containing a bicistronic cassette gDt-IRES.IL-6, (BAV327). Recombinant proteins expressed by BAV326 and BAV327 retained antigenicity (gDt) and biological activity (BoIL-6). Intranasal immunization of calves with recombinant BAV326, BAV327 or BAV308 (gDt alone) resulted in demonstrable levels of gDt-specific IgG responses in sera and IgA response in nasal secretions, in all animals. In addition, all calves developed complement-independent neutralizing antibody responses against BHV-1. However, no significant difference could be observed in the induction of systemic or mucosal immune response in animals immunized with recombinant BAV326 or BAV327 co-expressing BoIL-6. Moreover, there was no difference in the protection against BHV-1 challenge particularly in the amount of virus excretion in the nasal cavity in calves immunized with BAV326, BAV327 or BAV308. These data suggest that the BoIL-6 had no modulating effect on the induction of gDt specific mucosal and systemic immune responses in calves.
Collapse
Affiliation(s)
- Pankaj Kumar
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lisanework E Ayalew
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada; Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Dale L Godson
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada
| | - Amit Gaba
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada; Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Lorne A Babiuk
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada; Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada
| | - Suresh K Tikoo
- VIDO-InterVac, University of Saskatchewan, Saskatoon, SK, Canada; Veterinary Microbiology, University of Saskatchewan, Saskatoon, SK, Canada; Vaccinology & Immunotherapeutics Program, School of Public Health, University of Saskatchewan, Saskatoon, SK, Canada.
| |
Collapse
|
215
|
Mufarrege EF, Antuña S, Etcheverrigaray M, Kratje R, Prieto C. Development of lentiviral vectors for transient and stable protein overexpression in mammalian cells. A new strategy for recombinant human FVIII (rhFVIII) production. Protein Expr Purif 2014; 95:50-6. [DOI: 10.1016/j.pep.2013.11.005] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2013] [Revised: 10/17/2013] [Accepted: 11/14/2013] [Indexed: 10/26/2022]
|
216
|
Unprecedented cell-selection using ultra-quick freezing combined with aquaporin expression. PLoS One 2014; 9:e87644. [PMID: 24558371 PMCID: PMC3928110 DOI: 10.1371/journal.pone.0087644] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2013] [Accepted: 12/25/2013] [Indexed: 11/19/2022] Open
Abstract
Freezing is usually used for preservation and storage of biological samples; however, this process may have some adverse effects such as cell membrane damage. Aquaporin (AQP), a water channel protein, has been suggested to play some roles for cryopreservation although its molecular mechanism remains unclear. Here we show that membrane damage caused by ultra-quick freezing is rescued by the expression of AQP4. We next examine if the expression of AQP combined with ultra-quick freezing can be used to select cells efficiently under freezing conditions where most cells are died. CHO cells stably expressing AQP4 were exclusively selected from mixed cell cultures. Having identified the increased expression of AQP4 during ES cell differentiation into neuro-ectoderm using bioinformatics, we confirmed the improved survival of differentiated ES cells with AQP4 expression. Finally we show that CHO cells transiently transfected with Endothelin receptor A and Aqp4 were also selected and concentrated by multiple cycles of freezing/thawing, which was confirmed with calcium imaging in response to endothelin. Furthermore, we found that the expression of AQP enables a reduction in the amount of cryoprotectants for freezing, thereby decreasing osmotic stress and cellular toxicity. Taken together, we propose that this simple but efficient and safe method may be applicable to the selection of mammalian cells for applications in regenerative medicine as well as cell-based functional assays or drug screening protocols.
Collapse
|
217
|
Tong J, Buch S, Yao H, Wu C, Tong HI, Wang Y, Lu Y. Monocytes-derived macrophages mediated stable expression of human brain-derived neurotrophic factor, a novel therapeutic strategy for neuroAIDS. PLoS One 2014; 9:e82030. [PMID: 24505242 PMCID: PMC3914783 DOI: 10.1371/journal.pone.0082030] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Accepted: 10/19/2013] [Indexed: 12/30/2022] Open
Abstract
HIV-1 associated dementia remains a significant public health burden. Clinical and experimental research has shown that reduced levels of brain-derived neurotrophic factor (BDNF) may be a risk factor for neurological complications associated with HIV-1 infection. We are actively testing genetically modified macrophages for their possible use as the cell-based gene delivery vehicle for the central nervous system (CNS). It can be an advantage to use the natural homing/migratory properties of monocyte-derived macrophages to deliver potentially neuroprotective BDNF into the CNS, as a non-invasive manner. Lentiviral-mediated gene transfer of human (h)BDNF plasmid was constructed and characterized. Defective lentiviral stocks were generated by transient transfection of 293T cells with lentiviral transfer plasmid together with packaging and envelope plasmids. High titer lentiviral vector stocks were harvested and used to transduce human neuronal cell lines, primary cultures of human peripheral mononocyte-derived macrophages (hMDM) and murine myeloid monocyte-derived macrophages (mMDM). These transduced cells were tested for hBDNF expression, stability, and neuroprotective activity. The GenomeLab GeXP Genetic Analysis System was used to evaluate transduced cells for any adverse effects by assessing gene profiles of 24 reference genes. High titer vectors were prepared for efficient transduction of neuronal cell lines, hMDM, and mMDM. Stable secretion of high levels of hBDNF was detected in supernatants of transduced cells using western blot and ELISA. The conditioned media containing hBDNF were shown to be protective to neuronal and monocytic cell lines from TNF-α and HIV-1 Tat mediated cytotoxicity. Lentiviral vector-mediated gene transduction of hMDM and mMDM resulted in high-level, stable expression of the neuroprotective factorBDNF in vitro. These findings form the basis for future research on the potential use of BDNF as a novel therapy for neuroAIDS.
Collapse
Affiliation(s)
- Jing Tong
- MOE Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan, People's Republic of China
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Shilpa Buch
- University of Nebraska Medical Center, Pharmacology and Experimental Neuroscience, Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Honghong Yao
- University of Nebraska Medical Center, Pharmacology and Experimental Neuroscience, Nebraska Medical Center, Omaha, Nebraska, United States of America
| | - Chengxiang Wu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Hsin-I Tong
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
| | - Youwei Wang
- MOE Key Laboratory of Combinatorial Biosynthesis and Drug Discovery, Institute of TCM & Natural Products, School of Pharmaceutical Sciences, Wuhan University, Wuhan, People's Republic of China
- * E-mail: (YW); (YL)
| | - Yuanan Lu
- Department of Public Health Sciences, University of Hawaii at Manoa, Honolulu, Hawaii, United States of America
- * E-mail: (YW); (YL)
| |
Collapse
|
218
|
Hutson TH, Kathe C, Menezes SC, Rooney MC, Bueler H, Moon LDF. The use of an adeno-associated viral vector for efficient bicistronic expression of two genes in the central nervous system. Methods Mol Biol 2014; 1162:189-207. [PMID: 24838969 PMCID: PMC5597041 DOI: 10.1007/978-1-4939-0777-9_16] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/16/2023]
Abstract
Recombinant adeno-associated viral (AAV) vectors are one of the most promising therapeutic delivery systems for gene therapy to the central nervous system (CNS). Preclinical testing of novel gene therapies requires the careful design and production of AAV vectors and their successful application in a model of CNS injury. One major limitation of AAV vectors is their limited packaging capacity (<5 kb) making the co-expression of two genes (e.g., from two promoters) difficult. An internal ribosomal entry site has been used to express two genes: However, the second transgene is often expressed at lower levels than the first. In addition to this, achieving high levels of transduction in the CNS can be challenging. In this chapter we describe the cloning of a bicistronic AAV vector that uses the foot-and-mouth disease virus 2A sequence to efficiently express two genes from a single promoter. Bicistronic expression of a therapeutic gene and a reporter gene is desirable so that the axons from transduced neurons can be tracked and, after CNS injury, the amount of axonal sprouting or regeneration quantified. We go on to describe how to perform a pyramidotomy model of CNS injury and the injection of AAV vectors into the sensorimotor cortex to provide efficient transduction and bicistronic gene expression in cortical neurons such that transduced axons are detectable in the dorsal columns of the spinal cord.
Collapse
Affiliation(s)
- Thomas Haynes Hutson
- Neurorestoration Group, Wolfson Centre for Age-Related Diseases, King's College London, Guy's Campus, London, SE1 1UL, UK,
| | | | | | | | | | | |
Collapse
|
219
|
Guo P, Dai W, Liu S, Yang P, Cheng J, Liang L, Chen Z, Gao H. Construction and expression of bivalent membrane-anchored DNA vaccine encoding Sjl4FABP and Sj26GST genes. ACTA ACUST UNITED AC 2013; 26:493-6. [PMID: 17219949 DOI: 10.1007/s11596-006-0501-z] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
In order to construct a eukaryotic co-expression plasmid containing membrane-anchored Sjcl4FABP and Sjc26GST genes and identify their expression in vitro, Sj14 and Sj26 genes were obtained by RT-PCR with total RNA of Schistosoma japonicum adult worms as the template and cloned into eukaryotic expression plasmid pVAC to construct recombinant plasmids pVAC-Sj14 and pVAC-Sj26. Then a 23 amino-acid signal peptide of human interleukin-2 (IL-2) upstream Sj14 or Sj26 gene and a membrane-anchored sequence containing 32 amino-acids of carboxyl-terminal of human placental alkaline phosphatase (PLAP) downstream were amplified by PCR as the template of plasmid pVAC-Sj14 or pVAC-Sj26 only to get two gene fragments including Sj14 gene and Sj26 gene. The two modified genes were altogether cloned into a eukaryotic co-expression plasmid pIRES, resulting in another new recombinant plasmid pIRES-Sj26-Sj14. The expression of Sj14 and Sj26 genes was detected by RT-PCR and indirect immunofluorescent assays (IFA) when the plasmid pIRES-Sj26-Sj14 was transfected into eukaryotic Hela cells. Restriction enzyme analysis, PCR and sequencing results revealed that the recombinant plasmids pVAC-Sj14, pVAC-Sj26 and plRES-Sj26-Sj14 were successfully constructed and the expression of modified Sj14 and Sj26 genes could be detected by RT-PCR and IFA. A bivalent membrane-anchored DNA vaccine encoding Sj14 and Sj26 genes was acquired and expressed proteins were proved to be mostly anchored in cellular membranes.
Collapse
Affiliation(s)
- Ping Guo
- Department of Biochemistry and Molecular Biology, School of Basic Medical Science, Tongii Medical College, Huazhong University of Science and Technology, Wuhan 430030, China
| | | | | | | | | | | | | | | |
Collapse
|
220
|
Fagoe ND, Eggers R, Verhaagen J, Mason MRJ. A compact dual promoter adeno-associated viral vector for efficient delivery of two genes to dorsal root ganglion neurons. Gene Ther 2013; 21:242-52. [PMID: 24285216 DOI: 10.1038/gt.2013.71] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2013] [Revised: 10/23/2013] [Accepted: 10/24/2013] [Indexed: 12/19/2022]
Abstract
Adeno-associated viral (AAV) vectors based on serotype 5 are an efficient means to target dorsal root ganglia (DRG) to study gene function in the primary sensory neurons of the peripheral nervous system. In this study, we have developed a compact AAV dual promoter vector composed of the cytomegalovirus (CMV) and chicken beta-actin (CAG) promoters in a back-to-back configuration with a shared enhancer, and show efficient expression of two proteins simultaneously in DRG neurons. We demonstrate how this is useful for experiments on axonal regeneration, by co-expressing a gene of interest and an axonal marker. Using a farnesylated form of eGFP, which is actively transported along axons, we show superior long-distance labelling of axons of DRG neurons compared with normal eGFP. Additionally, we have efficiently transduced lumbar DRG neurons by injecting the AAV dual promoter vector into the dorsal intrathecal space, which is a less invasive delivery method. In summary, we have developed an AAV dual promoter vector designed for simultaneous expression of a gene of interest and a fluorescent protein to label long-distance axonal projections, which allows specific quantification of axons from transduced neurons after injury.
Collapse
Affiliation(s)
- N D Fagoe
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - R Eggers
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| | - J Verhaagen
- 1] Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands [2] Center for Neurogenomics and Cognition Research, Neuroscience Campus Amsterdam, Vrije Universtiteit Amsterdam, Amsterdam, The Netherlands
| | - M R J Mason
- Laboratory for Neuroregeneration, Netherlands Institute for Neuroscience, an Institute of the Royal Academy of Arts and Sciences, Amsterdam, The Netherlands
| |
Collapse
|
221
|
Tang JCY, Szikra T, Kozorovitskiy Y, Teixiera M, Sabatini BL, Roska B, Cepko CL. A nanobody-based system using fluorescent proteins as scaffolds for cell-specific gene manipulation. Cell 2013; 154:928-39. [PMID: 23953120 PMCID: PMC4096992 DOI: 10.1016/j.cell.2013.07.021] [Citation(s) in RCA: 91] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2012] [Revised: 05/31/2013] [Accepted: 07/15/2013] [Indexed: 01/22/2023]
Abstract
Fluorescent proteins are commonly used to label cells across organisms, but the unmodified forms cannot control biological activities. Using GFP-binding proteins derived from Camelid antibodies, we co-opted GFP as a scaffold for inducing formation of biologically active complexes, developing a library of hybrid transcription factors that control gene expression only in the presence of GFP or its derivatives. The modular design allows for variation in key properties such as DNA specificity, transcriptional potency, and drug dependency. Production of GFP controlled cell-specific gene expression and facilitated functional perturbations in the mouse retina and brain. Further, retrofitting existing transgenic GFP mouse and zebrafish lines for GFP-dependent transcription enabled applications such as optogenetic probing of neural circuits. This work establishes GFP as a multifunctional scaffold and opens the door to selective manipulation of diverse GFP-labeled cells across transgenic lines. This approach may also be extended to exploit other intracellular products as cell-specific scaffolds in multicellular organisms.
Collapse
Affiliation(s)
- Jonathan C Y Tang
- Howard Hughes Medical Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | |
Collapse
|
222
|
Akbor MM, Tomobe K, Yamada T, Kim J, Mano H, Kurosawa N, Sasaki K, Nomura Y, Isobe M. Possible involvement of Hcn1 ion channel in learning and memory dysfunction in SAMP8 mice. Biochem Biophys Res Commun 2013; 441:25-30. [DOI: 10.1016/j.bbrc.2013.09.145] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2013] [Accepted: 09/27/2013] [Indexed: 10/26/2022]
|
223
|
Han L, Ravoori M, Wu G, Sakai R, Yan S, Singh S, Xu K, Roth JA, Ji L, Kundra V. Somatostatin Receptor Type 2–Based Reporter Expression after Plasmid-Based in Vivo Gene Delivery to Non–Small Cell Lung Cancer. Mol Imaging 2013; 12:7290.2013.00060. [DOI: 10.2310/7290.2013.00060] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 08/30/2023] Open
Affiliation(s)
- Lin Han
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Murali Ravoori
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Guanglin Wu
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Ryo Sakai
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Shaoyu Yan
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Sheela Singh
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Kai Xu
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Jack A. Roth
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Lin Ji
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| | - Vikas Kundra
- From the Departments of Experimental Diagnostic Imaging, Thoracic and Cardiovascular Surgery, and Diagnostic Radiology, The University of Texas MD Anderson Cancer Center, Houston, TX
| |
Collapse
|
224
|
Zhang XB. Cellular reprogramming of human peripheral blood cells. GENOMICS PROTEOMICS & BIOINFORMATICS 2013; 11:264-74. [PMID: 24060839 PMCID: PMC4357833 DOI: 10.1016/j.gpb.2013.09.001] [Citation(s) in RCA: 40] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 06/25/2013] [Revised: 08/29/2013] [Accepted: 09/03/2013] [Indexed: 12/22/2022]
Abstract
Breakthroughs in cell fate conversion have made it possible to generate large quantities of patient-specific cells for regenerative medicine. Due to multiple advantages of peripheral blood cells over fibroblasts from skin biopsy, the use of blood mononuclear cells (MNCs) instead of skin fibroblasts will expedite reprogramming research and broaden the application of reprogramming technology. This review discusses current progress and challenges of generating induced pluripotent stem cells (iPSCs) from peripheral blood MNCs and of in vitro and in vivo conversion of blood cells into cells of therapeutic value, such as mesenchymal stem cells, neural cells and hepatocytes. An optimized design of lentiviral vectors is necessary to achieve high reprogramming efficiency of peripheral blood cells. More recently, non-integrating vectors such as Sendai virus and episomal vectors have been successfully employed in generating integration-free iPSCs and somatic stem cells.
Collapse
Affiliation(s)
- Xiao-Bing Zhang
- Department of Medicine, Loma Linda University, Loma Linda, CA 92354, USA.
| |
Collapse
|
225
|
Liao X, Cui H, Wang F. Establishment of a transgenic mouse model of corneal dystrophy overexpressing human BIGH3. Int J Mol Med 2013; 32:1110-4. [PMID: 24009044 DOI: 10.3892/ijmm.2013.1480] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2013] [Accepted: 08/22/2013] [Indexed: 11/06/2022] Open
Abstract
This study aimed to establish a transgenic mouse model of corneal dystrophy (CD) overexpressing the human transforming growth factor, β-induced, 68 kDa (TGFBI, also known as BIGH3) gene. A purified and linearized recombinant plasmid carrying the expression cassette BIGH3‑IRES‑EGFP was microinjected into the pronuclei of C57BL/6J mouse fertilized eggs under the control of the phosphoglycerate kinase (PGK) promoter. The expression of human BIGH3 in the transgenic mice was confirmed by PCR using DNA extracted from tail tissue. Four founder transgenic mice were identified by PCR and the increased expression of BIGH3 was observed in the corneas of the transgenic mice by RT-PCR and western blot analysis. The abnormal corneas with central opacity were observed in the transgenic mice by corneal photography. We concluded that the exogenous gene, BIGH3, was integrated successfully into the mouse genome through microinjection. In addition, the phenotype observed in this BIGH3 transgenic mouse model was similar to CD. Therefore, this transgenic model may prove useful in the investigation of the pathogenesis of CD.
Collapse
Affiliation(s)
- Xin Liao
- Department of Ophthalmology, Shanghai Tenth People's Hospital Affiliated to Tongji University, Shanghai 200072, P.R. China
| | | | | |
Collapse
|
226
|
Li H, Li X, Liu Q, Shi Z, Shi D. Expression of Biologically Active Human Recombinant Interferon Alpha 2b in Human Breast Cancer Cell Line Bcap-37. Appl Biochem Biotechnol 2013; 171:1535-44. [DOI: 10.1007/s12010-013-0420-y] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2013] [Accepted: 07/30/2013] [Indexed: 10/26/2022]
|
227
|
Santana VC, Diniz MO, Cariri FAMO, Ventura AM, Cunha-Neto E, Almeida RR, Campos MA, Lima GK, Ferreira LCS. Bicistronic DNA vaccines simultaneously encoding HIV, HSV and HPV antigens promote CD8⁺ T cell responses and protective immunity. PLoS One 2013; 8:e71322. [PMID: 23951135 PMCID: PMC3738591 DOI: 10.1371/journal.pone.0071322] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Accepted: 07/04/2013] [Indexed: 12/31/2022] Open
Abstract
Millions of people worldwide are currently infected with human papillomavirus (HPV), herpes simplex virus (HSV) or human immunodeficiency virus (HIV). For this enormous contingent of people, the search for preventive and therapeutic immunological approaches represents a hope for the eradication of latent infection and/or virus-associated cancer. To date, attempts to develop vaccines against these viruses have been mainly based on a monovalent concept, in which one or more antigens of a virus are incorporated into a vaccine formulation. In the present report, we designed and tested an immunization strategy based on DNA vaccines that simultaneously encode antigens for HIV, HSV and HPV. With this purpose in mind, we tested two bicistronic DNA vaccines (pIRES I and pIRES II) that encode the HPV-16 oncoprotein E7 and the HIV protein p24 both genetically fused to the HSV-1 gD envelope protein. Mice i.m. immunized with the DNA vaccines mounted antigen-specific CD8+ T cell responses, including in vivo cytotoxic responses, against the three antigens. Under experimental conditions, the vaccines conferred protective immunity against challenges with a vaccinia virus expressing the HIV-derived protein Gag, an HSV-1 virus strain and implantation of tumor cells expressing the HPV-16 oncoproteins. Altogether, our results show that the concept of a trivalent HIV, HSV, and HPV vaccine capable to induce CD8+ T cell-dependent responses is feasible and may aid in the development of preventive and/or therapeutic approaches for the control of diseases associated with these viruses.
Collapse
Affiliation(s)
- Vinicius C. Santana
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Mariana O. Diniz
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Francisco A. M. O. Cariri
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Armando M. Ventura
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
| | - Edécio Cunha-Neto
- Laboratory of Clinical Immunology and Allergy-LIM60, Division of Clinical Immunology and Allergy, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Rafael R. Almeida
- Laboratory of Clinical Immunology and Allergy-LIM60, Division of Clinical Immunology and Allergy, Department of Medicine, University of São Paulo School of Medicine, São Paulo, Brazil
| | - Marco A. Campos
- René Rachou Research Center, Fiocruz, Belo Horizonte, Brazil
| | | | - Luís C. S. Ferreira
- Department of Microbiology, Biomedical Sciences Institute, University of São Paulo, São Paulo, Brazil
- * E-mail:
| |
Collapse
|
228
|
Lyons SK, Patrick PS, Brindle KM. Imaging mouse cancer models in vivo using reporter transgenes. Cold Spring Harb Protoc 2013; 2013:685-99. [PMID: 23906907 DOI: 10.1101/pdb.top069864] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Imaging mouse models of cancer with reporter transgenes has become a relatively common experimental approach in the laboratory, which allows noninvasive and longitudinal investigation of diverse aspects of tumor biology in vivo. Our goal here is to outline briefly the principles of the relevant imaging modalities, emphasizing particularly their strengths and weaknesses and what the researcher can expect in a practical sense from each of these techniques. Furthermore, we discuss how relatively subtle modifications in the way reporter transgene expression is regulated in the cell underpin the ability of reporter transgenes as a whole to provide readouts on such varied aspects of tumor biology in vivo.
Collapse
Affiliation(s)
- Scott K Lyons
- Department of Molecular Imaging, CRUK Cambridge Research Institute, Li Ka Shing Centre, Cambridge CB2 0RE, United Kingdom
| | | | | |
Collapse
|
229
|
Expression analysis of combinatorial genes using a bi-cistronic T2A expression system in porcine fibroblasts. PLoS One 2013; 8:e70486. [PMID: 23922997 PMCID: PMC3726604 DOI: 10.1371/journal.pone.0070486] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2013] [Accepted: 06/20/2013] [Indexed: 01/16/2023] Open
Abstract
In pig-to-primate xenotransplantation, multiple transgenic pigs are required to overcome a series of transplant rejections. The generation of multiple transgenic pigs either by breeding or the introduction of several mono-cistronic vectors has been hampered by the differential expression patterns of the target genes. To achieve simultaneous expression of multiple genes, a poly-cistronic expression system using the 2A peptide derived from the Thosea asigna virus (T2A) can be considered an alternative choice. Before applying T2A expression system to pig generation, the expression patterns of multiple genes in this system should be precisely evaluated. In this study, we constructed several bi-cistronic T2A expression vectors, which combine target genes that are frequently used in the xenotransplantation field, and introduced them into porcine fibroblasts. The proteins targeted to the same or different subcellular regions were efficiently expressed without affecting the localization or expression levels of the other protein. However, when a gene with low expression efficiency was inserted into the upstream region of the T2A sequences, the expression level of the downstream gene was significantly decreased compared with the expression efficiency without the insertion. A small interfering RNA targeting one gene in this system resulted in the significant downregulation of both the target gene and the other gene, indicating that multiple genes combined into a T2A expression vector can be considered as a single gene in terms of transcription and translation. In summary, the efficient expression of a downstream gene can be achieved if the expression of the upstream gene is efficient.
Collapse
|
230
|
Abstract
Rabies viruses, negative-strand RNA viruses, infect neurons through axon terminals and spread trans-synaptically in a retrograde direction between neurons. Rabies viruses whose glycoprotein (G) gene is deleted from the genome cannot spread across synapses. Complementation of G in trans, however, enables trans-synaptic spreading of G-deleted rabies viruses to directly connected, presynaptic neurons. Recombinant rabies viruses can encode genes of interest for labeling cells, controlling gene expression and monitoring or manipulating neural activity. Cre-dependent or bridge protein-mediated transduction and single-cell electroporation via the EnvA-TVA or EnvB-TVB (envelope glycoprotein and its specific receptor for avian sarcoma leukosis virus subgroup A or B) system allow cell type-specific or single cell-specific targeting. These rabies virus-based approaches permit the linking of connectivity to cell morphology and circuit function for particular cell types or single cells. Here we describe methods for construction of rabies viral vectors, recovery of G-deleted rabies viruses from cDNA, amplification of the viruses, pseudotyping them with EnvA or EnvB and concentration and titration of the viruses. The entire protocol takes 6-8 weeks.
Collapse
|
231
|
Kuzmich AI, Vvedenskii AV, Kopantzev EP, Vinogradova TV. Quantitative comparison of gene co-expression in a bicistronic vector harboring IRES or coding sequence of porcine teschovirus 2A peptide. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2013; 39:454-65. [DOI: 10.1134/s1068162013040122] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
|
232
|
Semple-Rowland SL, Berry J. Use of lentiviral vectors to deliver and express bicistronic transgenes in developing chicken embryos. Methods 2013; 66:466-73. [PMID: 23816789 DOI: 10.1016/j.ymeth.2013.06.026] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/11/2013] [Revised: 05/16/2013] [Accepted: 06/21/2013] [Indexed: 12/16/2022] Open
Abstract
The abilities of lentiviral vectors to carry large transgenes (∼8kb) and to efficiently infect and integrate these genes into the genomes of both dividing and non-dividing cells make them ideal candidates for transport of genetic material into cells and tissues. Given the properties of these vectors, it is somewhat surprising that they have seen only limited use in studies of developing tissues and in particular of the developing nervous system. Over the past several years, we have taken advantage of the large capacity of these vectors to explore the expression characteristics of several dual promoter and 2A peptide bicistronic transgenes in developing chick neural retina, with the goal of identifying transgene designs that reliably express multiple proteins in infected cells. Here we summarize the activities of several of these transgenes in neural retina and provide detailed methodologies for packaging lentivirus and delivering the virus into the developing neural tubes of chicken embryos in ovo, procedures that have been optimized over the course of several years of use in our laboratory. Conditions to hatch injected embryos are also discussed. The chicken-specific techniques will be of highest interest to investigators using avian embryos, development and packaging of lentiviral vectors that reliably express multiple proteins in infected cells should be of interest to all investigators whose experiments demand manipulation and expression of multiple proteins in developing cells and tissues.
Collapse
Affiliation(s)
- Susan L Semple-Rowland
- Department of Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL 32610 0244, United States.
| | - Jonathan Berry
- Department of Neuroscience, University of Florida, McKnight Brain Institute, Gainesville, FL 32610 0244, United States.
| |
Collapse
|
233
|
Abstract
Internal Ribosome Entry Site (IRES) sequences have been widely used to link the expression of two independent proteins on the same mRNA transcript. Genes encoding fluorescent proteins or drug-resistance enzymes are usually placed downstream of IRES, serving as expression indicators or selection markers. In biological applications where the upstream gene-of-interest is to be expressed at extremely high levels, it is often desirable to purposely reduce IRES downstream gene expression to economize the cellular resources and/or to generate more stringent selection pressure. Here we describe a miniature IRES mutant sequence (IRESmut3) with dramatically diminished co-translational efficiency to fulfill these purposes.
Collapse
Affiliation(s)
- Jun Yan
- Institute of Surgery Research, State Key Laboratory of Trauma, Burns and Combined Injury, Research Institute for Traffic Medicine, Daping Hospital, Third Military Medical University, Chongqing 400042, China
| | | | | | | |
Collapse
|
234
|
Tone Y, Kawahara M, Kawaguchi D, Ueda H, Nagamune T. Death signalobody: inducing conditional cell death in response to a specific antigen. Hum Gene Ther Methods 2013; 24:141-50. [PMID: 23470213 DOI: 10.1089/hgtb.2012.147] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
As the possibility of tumorigenesis and undesirable immune responses in patients cannot be completely excluded in gene and cell therapies, a conditional death switch to eliminate the therapeutic cells would be a valuable tool to enhance the safety of these therapies. A few ligand-receptor conditional death switches have already been developed; however, they cannot be used if patients exhibit side effects upon administration of the ligand. Here we demonstrate a death-inducing chimeric antibody named "death signalobody," in which the antigen-antibody system, having virtually infinite ligand-receptor combinations, is utilized for the activation of death signaling. We designed a death signalobody named "SFas," which has an antifluorescein single-chain variable fragment and the cytoplasmic domain of Fas. SFas efficiently induced conditional apoptosis in murine pro-B Ba/F3 cells in response to fluorescein-conjugated bovine serum albumin. Moreover, SFas was also able to induce antigen-dependent conditional apoptosis in human cancer cell lines. The death signalobody technique will be a valuable tool for the conditional elimination of cells of interest in multiple therapeutic applications.
Collapse
Affiliation(s)
- Yuichiro Tone
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, Tokyo 113-8656, Japan
| | | | | | | | | |
Collapse
|
235
|
Cloning and characterization of a hybridoma secreting a 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK)-specific monoclonal antibody and recombinant F(ab). Toxins (Basel) 2013; 5:568-89. [PMID: 23518474 PMCID: PMC3705279 DOI: 10.3390/toxins5030568] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2013] [Revised: 02/28/2013] [Accepted: 03/05/2013] [Indexed: 02/07/2023] Open
Abstract
Smokeless tobacco products have been associated with increased risks of oro-pharyngeal cancers, due in part to the presence of tobacco-specific nitrosamines (TSNAs) such as 4-(methylnitrosamino)-1-(3-pyridyl)-1-butanone (NNK). These potent carcinogens are formed during tobacco curing and as a result of direct nitrosation reactions that occur in the oral cavity. In the current work we describe the isolation and characterization of a hybridoma secreting a high-affinity, NNK-specific monoclonal antibody. A structurally-related benzoyl derivative was synthesized to facilitate coupling to NNK-carrier proteins, which were characterized for the presence of the N-nitroso group using the Griess reaction, and used to immunize BALB/c mice. Splenocytes from mice bearing NNK-specific antibodies were used to create hybridomas. Out of four, one was selected for subcloning and characterization. Approximately 99% of the monoclonal antibodies from this clone were competitively displaced from plate-bound NNKB conjugates in the presence of free NNK. The affinity of the monoclonal antibody to the NNKB conjugates was Kd = 2.93 nM as determined by surface plasmon resonance. Free nicotine was a poor competitor for the NNKB binding site. The heavy and light chain antibody F(ab) fragments were cloned, sequenced and inserted in tandem into an expression vector, with an FMDV Furin 2A cleavage site between them. Expression in HEK 293 cells revealed a functional F(ab) with similar binding features to that of the parent hybridoma. This study lays the groundwork for synthesizing transgenic tobacco that expresses carcinogen-sequestration properties, thereby rendering it less harmful to consumers.
Collapse
|
236
|
Zhang W, Wang F, Yan J, Zhang X, Wang Y, Jiang Y, Wang L, Xu Y, Yu J. Design, expression and characterization of a novel coexpression system of two antiarthritic molecules. Appl Microbiol Biotechnol 2013; 97:6301-14. [PMID: 23463251 DOI: 10.1007/s00253-013-4787-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2012] [Revised: 02/14/2013] [Accepted: 02/17/2013] [Indexed: 11/30/2022]
Abstract
The complexity of rheumatoid arthritis (RA) pathogenesis makes combined blockade of multiple targets an attractive therapeutic strategy. The combination therapy with anti-TNF plus anti-T-cell has been mostly reported to provide greater efficacy than anti-TNF alone. TNFR (p75)-Fc fusion protein, which has been proven effective in clinics, is chosen as the TNF antagonist in this study. CTLA4-FasL fusion molecule, which has been well characterized in our previous studies for its suppressive effect in rat arthritis model, is chosen as the T-cell antagonist. In this study, furin cleavage site and 2A self-processing sequence were introduced to link upstream TNFR-Fc and downstream CTLA4-FasL and mediate separate coexpression of the two fusion proteins in a single recombinant adeno-associated virus (rAAV) vector. Using this expression system, we generated two fusion proteins with same size as their individual counterparts in vitro and in vivo, and the proteins desirably retained their parent biological activities. In vivo results demonstrated that furin-2A technology is able to regulate separate coexpression of these proteins under arthritic inflammatory conditions. This study describes a single rAAV vector for production of two antiarthritic molecules antagonizing both TNF and T cells, which may serve as an attractive expression system for RA gene therapy.
Collapse
Affiliation(s)
- Wei Zhang
- Department of Translational Medicine, Beijing Institute of Basic Medical Sciences, Beijing 100850, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
237
|
Brim BL, Haskell R, Awedikian R, Ellinwood N, Jin L, Kumar A, Foster T, Magnusson K. Memory in aged mice is rescued by enhanced expression of the GluN2B subunit of the NMDA receptor. Behav Brain Res 2013; 238:211-26. [PMID: 23103326 PMCID: PMC3540206 DOI: 10.1016/j.bbr.2012.10.026] [Citation(s) in RCA: 55] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2012] [Revised: 10/09/2012] [Accepted: 10/13/2012] [Indexed: 11/26/2022]
Abstract
The GluN2B subunit of the N-methyl-d-aspartate (NMDA) receptor shows age-related declines in expression across the frontal cortex and hippocampus. This decline is strongly correlated to age-related memory declines. This study was designed to determine if increasing GluN2B subunit expression in the frontal lobe or hippocampus would improve memory in aged mice. Mice were injected bilaterally with either the GluN2B vector, containing cDNA specific for the GluN2B subunit and enhanced green fluorescent protein (eGFP); a control vector or vehicle. Spatial memory, cognitive flexibility, and associative memory were assessed using the Morris water maze. Aged mice, with increased GluN2B subunit expression, exhibited improved long-term spatial memory, comparable to young mice. However, memory was rescued on different days in the Morris water maze; early for hippocampal GluN2B subunit enrichment and later for the frontal lobe. A higher concentration of the GluN2B antagonist, Ro 25-6981, was required to impair long-term spatial memory in aged mice with enhanced GluN2B expression, as compared to aged controls, suggesting there was an increase in the number of GluN2B-containing NMDA receptors. In addition, hippocampal slices from aged mice with increased GluN2B subunit expression exhibited enhanced NMDA receptor-mediated excitatory post-synaptic potentials (EPSP). Treatment with Ro 25-6981 showed that a greater proportion of the NMDA receptor-mediated EPSP was due to the GluN2B subunit in these animals, as compared to aged controls. These results suggest that increasing the production of the GluN2B subunit in aged animals enhances memory and synaptic transmission. Therapies that enhance GluN2B subunit expression within the aged brain may be useful for ameliorating age-related memory declines.
Collapse
Affiliation(s)
- B. L. Brim
- Molecular and Cellular Biosciences Program, Oregon State University, Corvallis, OR, 97331, U.S.A
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, U.S.A
- Healthy Aging Program, Linus Pauling Institute, Oregon State University, Corvallis, OR; 97331, U.S.A
| | - R. Haskell
- ViraQuest, Inc., North Liberty, IA; 52317, U.S.A
| | - R. Awedikian
- Department of Animal Sciences, Iowa State University, Ames, IA, 50011, U.S.A
| | - N.M. Ellinwood
- Department of Animal Sciences, Iowa State University, Ames, IA, 50011, U.S.A
| | - L. Jin
- Molecular and Cellular Biosciences Program, Oregon State University, Corvallis, OR, 97331, U.S.A
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, U.S.A
| | - A. Kumar
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32611, U.S.A
| | - T.C. Foster
- Department of Neuroscience, McKnight Brain Institute, University of Florida, Gainesville, FL, 32611, U.S.A
| | - K. Magnusson
- Molecular and Cellular Biosciences Program, Oregon State University, Corvallis, OR, 97331, U.S.A
- Department of Biomedical Sciences, College of Veterinary Medicine, Oregon State University, Corvallis, OR, 97331, U.S.A
- Healthy Aging Program, Linus Pauling Institute, Oregon State University, Corvallis, OR; 97331, U.S.A
| |
Collapse
|
238
|
Fiumelli H, Briner A, Puskarjov M, Blaesse P, Belem BJ, Dayer AG, Kaila K, Martin JL, Vutskits L. An ion transport-independent role for the cation-chloride cotransporter KCC2 in dendritic spinogenesis in vivo. Cereb Cortex 2013; 23:378-88. [PMID: 22345354 DOI: 10.1093/cercor/bhs027] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
The neuron-specific K-Cl cotransporter, KCC2, is highly expressed in the vicinity of excitatory synapses in pyramidal neurons, and recent in vitro data suggest that this protein plays a role in the development of dendritic spines. The in vivo relevance of these observations is, however, unknown. Using in utero electroporation combined with post hoc iontophoretic injection of Lucifer Yellow, we show that premature expression of KCC2 induces a highly significant and permanent increase in dendritic spine density of layer 2/3 pyramidal neurons in the somatosensory cortex. Whole-cell recordings revealed that this increased spine density is correlated with an enhanced spontaneous excitatory activity in KCC2-transfected neurons. Precocious expression of the N-terminal deleted form of KCC2, which lacks the chloride transporter function, also increased spine density. In contrast, no effect on spine density was observed following in utero electroporation of a point mutant of KCC2 (KCC2-C568A) where both the cotransporter function and the interaction with the cytoskeleton are disrupted. Transfection of the C-terminal domain of KCC2, a region involved in the interaction with the dendritic cytoskeleton, also increased spine density. Collectively, these results demonstrate a role for KCC2 in excitatory synaptogenesis in vivo through a mechanism that is independent of its ion transport function.
Collapse
Affiliation(s)
- Hubert Fiumelli
- Center for Psychiatric Neurosciences, Department of Psychiatry, Lausanne University Hospital, 1008 Prilly-Lausanne, Switzerland
| | | | | | | | | | | | | | | | | |
Collapse
|
239
|
Neurodevelopmental role for VGLUT2 in pyramidal neuron plasticity, dendritic refinement, and in spatial learning. J Neurosci 2013; 32:15886-901. [PMID: 23136427 DOI: 10.1523/jneurosci.4505-11.2012] [Citation(s) in RCA: 54] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
The level and integrity of glutamate transmission during critical periods of postnatal development plays an important role in the refinement of pyramidal neuron dendritic arbor, synaptic plasticity, and cognition. Presently, it is not clear how excitatory transmission via the two predominant isoforms of the vesicular glutamate transporter (VGLUT1 and VGLUT2) participate in this process. To assess a neurodevelopmental role for VGLUT2 in pyramidal neuron maturation, we generated recombinant VGLUT2 knock-out mice and inactivated VGLUT2 throughout development using Emx1-Cre(+/+) knock-in mice. We show that VGLUT2 deficiency in corticolimbic circuits results in reduced evoked glutamate transmission, release probability, and LTD at hippocampal CA3-CA1 synapses during a formative developmental period (postnatal days 11-14). In adults, we find a marked reduction in the amount of dendritic arbor across the span of the dendritic tree of CA1 pyramidal neurons and reduced long-term potentiation and levels of synaptic markers spinophilin and VGLUT1. Loss of dendritic arbor is accompanied by corresponding reductions in the number of dendritic spines, suggesting widespread alterations in synaptic connectivity. Conditional VGLUT2 knock-out mice exhibit increased open-field exploratory activity yet impaired spatial learning and memory, endophenotypes similar to those of NMDA receptor knock-down mice. Remarkably, the impairment in learning can be partially restored by selectively increasing NMDA receptor-mediated glutamate transmission in adult mice by prolonged treatment with d-serine and a d-amino acid oxidase inhibitor. Our data indicate that VGLUT2 expression is pivotal to the proper development of mature pyramidal neuronal architecture and plasticity, and that such glutamatergic deficiency leads to cognitive malfunction as observed in several neurodevelopmental psychiatric disorders.
Collapse
|
240
|
Touihri L, Ahmed SB, Chtourou Y, Daoud R, Bahloul C. Design of different strategies of multivalent DNA-based vaccination against rabies and canine distemper in mice and dogs. Virol J 2012; 9:319. [PMID: 23270301 PMCID: PMC3547725 DOI: 10.1186/1743-422x-9-319] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 12/12/2012] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND During the vaccination campaigns, puppies younger than 3 months old are not targeted and remain unvaccinated for at least the first year of their lives. Almost half of the reported rabid dogs are 6 months or younger. Hence, we should recommend the vaccination against rabies of young puppies. Unfortunately, owing to the exposure of puppies to infections with either canine parvovirus (CPV) or distemper virus (CDV) after the intervention of the vaccinators, owners are reluctant to vaccinate puppies against rabies. Therefore, it is necessary to include the CPV and CDV valences in the vaccine against rabies. Multivalent DNA-based vaccination in dogs, including rabies and distemper valences, could help in raising vaccine coverage. METHODS We have designed monovalent and multivalent DNA-based vaccine candidates for in vitro and in vivo assays. These plasmids encode to the rabies virus glycoprotein and/or the canine distemper virus hemagglutinin. The first strategy of multivalent DNA-based vaccination is by mixing plasmids encoding to a single antigen each. The second is by simply fusing the genes of the antigens together. The third is by adding the foot and mouth disease virus (FMDV) 2A oligopeptide gene into the antigen genes. The last strategy is by the design and use of a bicistronic plasmid with an "Internal Ribosome Entry Site" (IRES) domain. RESULTS The monovalent construct against canine distemper was efficiently validated by inducing higher humoral immune responses compared to cell-culture-derived vaccine both in mice and dogs. All multivalent plasmids efficiently expressed both valences after in vitro transfection of BHK-21 cells. In BALB/c mice, the bicistronic IRES-dependant construct was the most efficient inducer of virus-neutralizing antibodies against both valences. It was able to induce better humoral immune responses compared to the administration of either cell-culture-derived vaccines or monovalent plasmids. The FMDV 2A was also efficient in the design of multivalent plasmids. CONCLUSIONS In a single shot, the design of efficient multivalent plasmids will be very beneficial for DNA-based vaccination against numerous diseases.
Collapse
Affiliation(s)
- Leila Touihri
- Institut Pasteur de Tunis, MMVDB, 13, Place Pasteur BP-74, Tunis-Belvedere, 1002, Tunisia
| | | | | | | | | |
Collapse
|
241
|
Generation of a genome scale lentiviral vector library for EF1α promoter-driven expression of human ORFs and identification of human genes affecting viral titer. PLoS One 2012; 7:e51733. [PMID: 23251614 PMCID: PMC3520899 DOI: 10.1371/journal.pone.0051733] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2012] [Accepted: 11/06/2012] [Indexed: 01/12/2023] Open
Abstract
The bottleneck in elucidating gene function through high-throughput gain-of-function genome screening is the limited availability of comprehensive libraries for gene overexpression. Lentiviral vectors are the most versatile and widely used vehicles for gene expression in mammalian cells. Lentiviral supernatant libraries for genome screening are commonly generated in the HEK293T cell line, yet very little is known about the effect of introduced sequences on the produced viral titer, which we have shown to be gene dependent. We have generated an arrayed lentiviral vector library for the expression of 17,030 human proteins by using the GATEWAY® cloning system to transfer ORFs from the Mammalian Gene Collection into an EF1alpha promoter-dependent lentiviral expression vector. This promoter was chosen instead of the more potent and widely used CMV promoter, because it is less prone to silencing and provides more stable long term expression. The arrayed lentiviral clones were used to generate viral supernatant by packaging in the HEK293T cell line. The efficiency of transfection and virus production was estimated by measuring the fluorescence of IRES driven GFP, co-expressed with the ORFs. More than 90% of cloned ORFs produced sufficient virus for downstream screening applications. We identified genes which consistently produced very high or very low viral titer. Supernatants from select clones that were either high or low virus producers were tested on a range of cell lines. Some of the low virus producers, including two previously uncharacterized proteins were cytotoxic to HEK293T cells. The library we have constructed presents a powerful resource for high-throughput gain-of-function screening of the human genome and drug-target discovery. Identification of human genes that affect lentivirus production may lead to improved technology for gene expression using lentiviral vectors.
Collapse
|
242
|
Liu K, Wang H, Long Y, Ye J, Yuan L. Coordinate lentiviral expression of Cre recombinase and RFP/EGFP mediated by FMDV 2A and analysis of Cre activity. J Cell Biochem 2012; 113:2909-19. [PMID: 22532014 DOI: 10.1002/jcb.24168] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The site-specific recombination mediated by Cre recombinase has been utilized extensively in genetic engineering and gene function studies. Efficient delivery of a Cre enzyme with enzymatic activity and the ability to monitor the enzyme expression are required in applications, and lentiviral constructs with a fluorescent protein (FP) to report the Cre expression are suitable for most studies. However, the current lentiviral vector systems have some deficiencies in precise reporting the Cre expression through fluorescence. To solve the problem, we generated a lentiviral system with Cre and RFP or EGFP bridged by an FMDV 2A sequence in an open reading frame expressed by a CMV promoter. We then examined the capabilities of the constructs to package with VSVG into infectious virus and to mediate expression of the Cre enzyme and fluorescent reporter. Furthermore, we monitored the bioactivities of the expressed products. We demonstrated the coordinate expression of the enzyme and the reporter. The expressed Cre was efficient at removing LoxP-flanked fragments in cells and did not show obvious cellular toxicity, and the expressed FPs allowed direct observation under fluorescent microscope. Therefore, the conjugation of CMV-Cre-2A-FP represents a significant improvement to the current lentiviral Cre delivery systems for obtaining a required Cre activity while accurately monitoring its presence. Our study also provides information concerning application of the established vector system.
Collapse
Affiliation(s)
- Kuancan Liu
- State Key Laboratory of Cellular Stress Biology, School of Life Sciences, Xiamen University, Xiamen 361005, China
| | | | | | | | | |
Collapse
|
243
|
Maier P, Heckmann D, Spier I, Laufs S, Zucknick M, Allgayer H, Fruehauf S, Zeller WJ, Wenz F. F2A sequence linking MGMT(P140K) and MDR1 in a bicistronic lentiviral vector enables efficient chemoprotection of haematopoietic stem cells. Cancer Gene Ther 2012; 19:802-10. [PMID: 23037811 DOI: 10.1038/cgt.2012.67] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
Chemoprotection of haematopoietic stem cells (HSCs) by gene therapeutic transfer of drug-resistance genes represents the encouraging approach to prevent myelosuppression, which is one of the most severe side effects in tumor therapy. Thus, we cloned and evaluated six different bicistronic lentiviral SIN vectors encoding two transgenes, MGMT(P140K) (an O(6)-benzylguanine-resistant mutant of methylguanine-DNA methyltransferase) and MDR1 (multidrug resistance 1), using various linker sequences (IRESEMCV, IRESFMDV and 2A-element of FMDV (F2A)). Expression of both transgenes in HL-60 and in K562 cells was assayed by quantitative real-time PCR. Combination therapy with ACNU plus paclitaxel in HL-60 cells and with carmustin (BCNU) plus doxorubicin in K562 cells resulted in the most significant survival advantage of cells transduced with the lentiviral vector HR'SIN-MGMT(P140K)-F2A-MDR1 compared with untransduced cells. In human HSCs, overexpression of both transgenes by this vector also caused significantly increased survival and enrichment of transduced cells after treatment with BCNU plus doxorubicin or temozolomide plus paclitaxel. In summary, we could show significant chemoprotection by overexpression of MDR1 and MGMT(P140K) with a lentiviral vector using the F2A linker element in two different haematopoietic cell lines and in human primary HSCs with various combination regimens. Consequently, we are convinced that these in vitro investigations will help to improve combination chemotherapy regimens by reducing myelotoxic side effects and increasing the therapeutic efficiency.
Collapse
Affiliation(s)
- P Maier
- Department of Radiation Oncology, University Medical Center Mannheim, University of Heidelberg, Mannheim, Germany.
| | | | | | | | | | | | | | | | | |
Collapse
|
244
|
Liu M, Yu H. Cocktail production of an endo-β-xylanase and a β-glucosidase from Trichoderma reesei QM 9414 in Escherichia coli. Biochem Eng J 2012. [DOI: 10.1016/j.bej.2012.07.003] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
|
245
|
Gheryani N, Coffelt SB, Gartland A, Rumney RMH, Kiss-Toth E, Lewis CE, Tozer GM, Greaves DR, Dear TN, Miller G. Generation of a novel mouse model for the inducible depletion of macrophages in vivo. Genesis 2012; 51:41-9. [PMID: 22927121 DOI: 10.1002/dvg.22343] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2010] [Revised: 08/22/2012] [Accepted: 08/22/2012] [Indexed: 11/08/2022]
Abstract
Macrophages play an essential role in tissue homeostasis, innate immunity, inflammation, and wound repair. Macrophages are also essential during development, severely limiting the use of mouse models in which these cells have been constitutively deleted. Consequently, we have developed a transgenic model of inducible macrophage depletion in which macrophage-specific induction of the cytotoxic diphtheria toxin A chain (DTA) is achieved by administration of doxycycline. Induction of the DTA protein in transgenic animals resulted in a significant 50% reduction in CD68+ macrophages of the liver, spleen, and bone over a period of 6 weeks. Pertinently, the macrophages remaining after doxycycline treatment were substantially smaller and are functionally impaired as shown by reduced inflammatory cytokine production in response to lipopolysaccharide. This inducible model of macrophage depletion can now be utilized to determine the role of macrophages in both development and animal models of chronic inflammatory diseases.
Collapse
Affiliation(s)
- Nabeia Gheryani
- Department of Cardiovascular Science, University of Sheffield, Sheffield, United Kingdom
| | | | | | | | | | | | | | | | | | | |
Collapse
|
246
|
Wan CK, Shaikh SB, Green CR, Nicholson LFB. Comparison of bidirectional and bicistronic inducible systems for coexpression of connexin genes and fluorescent reporters. Anal Biochem 2012; 431:90-5. [PMID: 22929700 DOI: 10.1016/j.ab.2012.08.016] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2012] [Revised: 08/17/2012] [Accepted: 08/18/2012] [Indexed: 11/18/2022]
Abstract
Gene expression studies often require inducible coexpression of both a gene of interest and a reporter gene. Fusion of fluorescent reporters can, however, modify protein structure and function. We have generated inducible expression systems for two connexin genes: Cx30 and Cx43. It has been reported recently that reporter fusion to connexins can modify their function. Therefore, we compared two methods of independent reporter coexpression and examined colocalization with induced connexin expression. Identical levels of connexin expression were observed for both the bidirectional and bicistronic expression systems. In contrast, however, reporter gene expression by the bidirectional promoter provided brighter average fluorescent pixel intensity than expression of a reporter gene in a bicistronic transcript. Moreover, as a result of this difference in reporter expression, bidirectional expression systems provided equal or better colocalization between the connexins and reporter gene fluorescence. The results of our study indicate that bidirectional reporter expression provides a robust indicator of transfection and gene expression and, therefore, may favor the use of bidirectional over bicistronic reporters in the design of expression systems where the gene of interest, such as a connexin gene, contains translational motifs or long intronic regions.
Collapse
Affiliation(s)
- Carthur K Wan
- Department of Anatomy with Radiology and Centre for Brain Research, Faculty of Medical and Health Sciences, University of Auckland, Auckland 92-019, New Zealand.
| | | | | | | |
Collapse
|
247
|
Isoform-specific dominant-negative effects associated with hERG1 G628S mutation in long QT syndrome. PLoS One 2012; 7:e42552. [PMID: 22876326 PMCID: PMC3411645 DOI: 10.1371/journal.pone.0042552] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2012] [Accepted: 07/09/2012] [Indexed: 11/25/2022] Open
Abstract
Background Mutations in the human ether-a-go-go-related gene 1 (hERG1) cause type 2 long QT syndrome (LQT2). The hERG1 gene encodes a K+ channel with properties similar to the rapidly activating delayed rectifying K+ current in the heart. Several hERG1 isoforms with unique structural and functional properties have been identified. To date, the pathogenic mechanisms of LQT2 mutations have been predominantly described in the context of the hERG1a isoform. In the present study, we investigated the functional consequences of the LQT2 mutation G628S in the hERG1b and hERG1aUSO isoforms. Methods A double-stable, mammalian expression system was developed to characterize isoform-specific dominant-negative effects of G628S-containing channels when co-expressed at equivalent levels with wild-type hERG1a. Western blot and co-immunoprecipitation studies were performed to study the trafficking and co-assembly of wild-type and mutant hERG1 isoforms. Patch-clamp electrophysiology was performed to characterize hERG1 channel function and the isoform-specific dominant-negative effects associated with the G628S mutation. Conclusions The non-functional hERG1a-G628S and hERG1b-G628S channels co-assembled with wild-type hERG1a and dominantly suppressed hERG1 current. In contrast, G628S-induced dominant-negative effects were absent in the context of the hERG1aUSO isoform. hERG1aUSO-G628S channels did not appreciably associate with hERG1a and did not significantly suppress hERG1 current when co-expressed at equivalent ratios or at ratios that approximate those found in cardiac tissue. These results suggest that the dominant-negative effects of LQT2 mutations may primarily occur in the context of the hERG1a and hERG1b isoforms.
Collapse
|
248
|
Wang SB, Tan Y, Lei W, Wang YG, Zhou XM, Jia XY, Zhang KJ, Chu L, Liu XY, Qian WB. Complete eradication of xenograft hepatoma by oncolytic adenovirus ZD55 harboring TRAIL-IETD-Smac gene with broad antitumor effect. Hum Gene Ther 2012; 23:992-1002. [PMID: 22530834 DOI: 10.1089/hum.2011.159] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
Cancer-targeting dual-gene virotherapy (CTGVT-DG) is an important modification of CTGVT, in which two suitable genes are used to obtain an excellent antitumor effect. A key problem is to join the two genes to form one fused gene, and then to clone it into the oncolytic viral vector so that only one investigational new drug application, instead of two, is required for clinical use. Many linkers (e.g., internal ribosome entry site) are used to join two genes together, but they are not all equally efficacious. Here, we describe finding the best linker, that is, sequence encoding the four amino acids IETD, to join the tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) gene and the second mitochondria-derived activator of caspase (Smac) gene to form TRAIL-IETD-Smac and inserting it into oncolytic viral vector ZD55 to construct ZD55-TRAIL-IETD-Smac, which matched ZD55-TRAIL plus ZD55-Smac in completely eliminating xenograft hepatoma. ZD55-TRAIL-IETD-Smac works by quantitative cleavage at IETD↓by inducing caspase-8; activation or inhibition of caspase-8 could up- or downregulate cleavage, respectively. The cleaved product, TRAIL-IETD, does not affect the function of TRAIL. Numerous experiments have shown that the combined use of ZD55-TRAIL plus ZD55-X could completely eradicate many xenograft tumors, and therefore the IETD is potentially a useful linker to construct many antitumor drugs, for example, ZD55-TRAIL-IETD-X, where X has a compensative or synergetic effect on TRAIL. We found that the antitumor effect of ZD55-IL-24-IETD-TRAIL also has an equivalent antitumor effect compared with the combined use of ZD55-IL-24 plus ZD55-TRAIL, because ZD55-IL-24 could also induce caspase-8. This means that IETD, as a two-gene linker, may have broad use.
Collapse
Affiliation(s)
- Shi-Bing Wang
- Xinyuan Institute of Medicine and Biotechnology, School of Life Science, Zhejiang Sci-Tech University, Hangzhou, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
249
|
Abstract
IL-10 is one of the key cytokines preventing inflammation-mediated tissue damage. In an attempt to identify IL-10-producing cells in vivo, several groups have recently developed IL-10 reporter mouse strains. Up until now, in total, eight IL-10 reporter strains have been published. This incomparable interest in IL-10 reporter mice emphasizes the importance and difficulties in tracking and subsequently investigating the role of IL-10-producing cells in infectious, inflammatory, autoimmune and cancer diseases. In this review, I summarize and compare the properties of those published IL-10 reporter mouse models. I also discuss the necessity to develop new strategies to generate 'multi-cytokine' reporter mouse models enabling highly sensitive in/ex vivo detection of many cytokines in the same single cell. Such 'multi-cytokine' reporter mice will enable to reconsider the dichotomy 'T-effector versus T-regulatory' paradigm and to provide an accurate revised model for cellular sources of cytokines. Finally, I propose to launch cooperative, international initiatives to promote and coordinate the generation of accurate, combinatorial, reporter mice for every individual murine cytokine.
Collapse
Affiliation(s)
- H Bouabe
- Department of Bacteriology, Max von Pettenkofer Institute, Munich, Germany.
| |
Collapse
|
250
|
Daniel-Meshulam I, Ya'akobi S, Ankri C, Cohen CJ. How (specific) would like your T-cells today? Generating T-cell therapeutic function through TCR-gene transfer. Front Immunol 2012; 3:186. [PMID: 22783259 PMCID: PMC3390604 DOI: 10.3389/fimmu.2012.00186] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/15/2012] [Indexed: 01/02/2023] Open
Abstract
T-cells are central players in the immune response against both pathogens and cancer. Their specificity is solely dictated by the T-cell receptor (TCR) they clonally express. As such, the genetic modification of T lymphocytes using pathogen- or cancer-specific TCRs represents an appealing strategy to generate a desired immune response from peripheral blood lymphocytes. Moreover, notable objective clinical responses were observed in terminally ill cancer patients treated with TCR-gene modified cells in several clinical trials conducted recently. Nevertheless, several key aspects of this approach are the object of intensive research aimed at improving the reliability and efficacy of this strategy. Herein, we will survey recent studies in the field of TCR-gene transfer dealing with the improvement of this approach and its application for the treatment of malignant, autoimmune, and infectious diseases.
Collapse
Affiliation(s)
- Inbal Daniel-Meshulam
- Laboratory of Tumor Immunology and Immunotherapy, The Mina and Everard Goodman Faculty of Life Sciences , Bar-Ilan University, Ramat Gan, Israel
| | | | | | | |
Collapse
|