1
|
Wang Y, Wang M, Bao R, Wang L, Du X, Qiu S, Yang C, Song H. A novel humanized tri-receptor transgenic mouse model of HAdV infection and pathogenesis. J Med Virol 2023; 95:e29026. [PMID: 37578851 DOI: 10.1002/jmv.29026] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2022] [Revised: 07/17/2023] [Accepted: 07/27/2023] [Indexed: 08/16/2023]
Abstract
Human adenovirus (HAdV) is a highly virulent respiratory pathogen that poses clinical challenges in terms of diagnostics and treatment. Currently, no effective therapeutic drugs or prophylactic vaccines are available for HAdV infections. One factor contributing to this deficiency is that existing animal models, including wild-type and single-receptor transgenic mice, are unsuitable for HAdV proliferation and pathology testing. In this study, a tri-receptor transgenic mouse model expressing the three best-characterized human cellular receptors for HAdV (hCAR, hCD46, and hDSG2) was generated and validated via analysis of transgene insertion, receptor mRNA expression, and protein abundance distribution. Following HAdV-7 infection, the tri-receptor mice exhibited high transcription levels at the early and late stages of the HAdV gene, as well as viral protein expression. Furthermore, the tri-receptor mice infected with HAdV exhibited dysregulated cytokine responses and multiple tissue lesions. This transgenic mouse model represents human HAdV infection and pathogenesis with more accuracy than any other reported animal model. As such, this model facilitates the comprehensive investigation of HAdV pathogenesis as well as the evaluation of potential vaccines and therapeutic modalities for HAdV.
Collapse
Affiliation(s)
- Yawei Wang
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Min Wang
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
- College of Public Heaith, China Medical University, Shenyang, China
| | - Renlong Bao
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Ligui Wang
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Xinying Du
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Shaofu Qiu
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Chaojie Yang
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| | - Hongbin Song
- College of Public Health, Zhengzhou University, Zhengzhou, China
- Infectious Disease Control and Prevention Department, Chinese PLA Center for Disease Control and Prevention, Beijing, China
| |
Collapse
|
2
|
Massi L, Hagihara KM, Courtin J, Hinz J, Müller C, Fustiñana MS, Xu C, Karalis N, Lüthi A. Disynaptic specificity of serial information flow for conditioned fear. SCIENCE ADVANCES 2023; 9:eabq1637. [PMID: 36652513 PMCID: PMC10957099 DOI: 10.1126/sciadv.abq1637] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/22/2022] [Accepted: 12/21/2022] [Indexed: 06/17/2023]
Abstract
Memory encoding and retrieval rely on specific interactions across multiple brain areas. Although connections between individual brain areas have been extensively studied, the anatomical and functional specificity of neuronal circuit organization underlying information transfer across multiple brain areas remains unclear. Here, we combine transsynaptic viral tracing, optogenetic manipulations, and calcium dynamics recordings to dissect the multisynaptic functional connectivity of the amygdala. We identify a distinct basolateral amygdala (BLA) subpopulation that connects disynaptically to the periaqueductal gray (PAG) via the central amygdala (CeA). This disynaptic pathway serves as a core circuit element necessary for the learning and expression of conditioned fear and exhibits learning-related plasticity. Together, our findings demonstrate the utility of multisynaptic approaches for functional circuit analysis and indicate that disynaptic specificity may be a general feature of neuronal circuit organization.
Collapse
Affiliation(s)
- Léma Massi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Kenta M. Hagihara
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| | - Julien Courtin
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Julian Hinz
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| | - Christian Müller
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Maria Sol Fustiñana
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Chun Xu
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- Institute of Neuroscience, Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| | - Nikolaos Karalis
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, Basel CH-4058, Switzerland
- University of Basel, Basel, Switzerland
| |
Collapse
|
3
|
Loxin Reduced the Inflammatory Response in the Liver and the Aortic Fatty Streak Formation in Mice Fed with a High-Fat Diet. Int J Mol Sci 2022; 23:ijms23137329. [PMID: 35806336 PMCID: PMC9266330 DOI: 10.3390/ijms23137329] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2022] [Revised: 06/07/2022] [Accepted: 06/08/2022] [Indexed: 11/17/2022] Open
Abstract
Oxidized low-density lipoprotein (ox-LDL) is the most harmful form of cholesterol associated with vascular atherosclerosis and hepatic injury, mainly due to inflammatory cell infiltration and subsequent severe tissue injury. Lox-1 is the central ox-LDL receptor expressed in endothelial and immune cells, its activation regulating inflammatory cytokines and chemotactic factor secretion. Recently, a Lox-1 truncated protein isoform lacking the ox-LDL binding domain named LOXIN has been described. We have previously shown that LOXIN overexpression blocked Lox-1-mediated ox-LDL internalization in human endothelial progenitor cells in vitro. However, the functional role of LOXIN in targeting inflammation or tissue injury in vivo remains unknown. In this study, we investigate whether LOXIN modulated the expression of Lox-1 and reduced the inflammatory response in a high-fat-diet mice model. Results indicate that human LOXIN blocks Lox-1 mediated uptake of ox-LDL in H4-II-E-C3 cells. Furthermore, in vivo experiments showed that overexpression of LOXIN reduced both fatty streak lesions in the aorta and inflammation and fibrosis in the liver. These findings were associated with the down-regulation of Lox-1 in endothelial cells. Then, LOXIN prevents hepatic and aortic tissue damage in vivo associated with reduced Lox-1 expression in endothelial cells. We encourage future research to understand better the underlying molecular mechanisms and potential therapeutic use of LOXIN.
Collapse
|
4
|
Bertzbach LD, Ip WH, Dobner T. Animal Models in Human Adenovirus Research. BIOLOGY 2021; 10:biology10121253. [PMID: 34943168 PMCID: PMC8698265 DOI: 10.3390/biology10121253] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 11/24/2021] [Accepted: 11/30/2021] [Indexed: 12/31/2022]
Abstract
Simple Summary Animal models are widely used to study various aspects of human diseases and disorders. Likewise, they are indispensable for preclinical testing of medicals and vaccines. Human adenovirus infections are usually self-limiting, and can cause mild respiratory symptoms with fever, eye infection or gastrointestinal symptoms, but occasional local outbreaks with severe disease courses have been reported. In addition, adenovirus infections pose a serious risk for children and patients with a weakened immune system. Human adenovirus research in animal models to study adenovirus-induced disease and tumor development started in the 1950s. Various animal species have been tested for their susceptibility to human adenovirus infection since then, and some have been shown to mimic key characteristics of the infection in humans, including persistent infection. Furthermore, some rodent species have been found to develop tumors upon human adenovirus infection. Our review summarizes the current knowledge on animal models in human adenovirus research, describing the pros and cons along with important findings and future perspectives. Abstract Human adenovirus (HAdV) infections cause a wide variety of clinical symptoms, ranging from mild upper respiratory tract disease to lethal outcomes, particularly in immunocompromised individuals. To date, neither widely available vaccines nor approved antiadenoviral compounds are available to efficiently deal with HAdV infections. Thus, there is a need to thoroughly understand HAdV-induced disease, and for the development and preclinical evaluation of HAdV therapeutics and/or vaccines, and consequently for suitable standardizable in vitro systems and animal models. Current animal models to study HAdV pathogenesis, persistence, and tumorigenesis include rodents such as Syrian hamsters, mice, and cotton rats, as well as rabbits. In addition, a few recent studies on other species, such as pigs and tree shrews, reported promising data. These models mimic (aspects of) HAdV-induced pathological changes in humans and, although they are relevant, an ideal HAdV animal model has yet to be developed. This review summarizes the available animal models of HAdV infection with comprehensive descriptions of virus-induced pathogenesis in different animal species. We also elaborate on rodent HAdV animal models and how they contributed to insights into adenovirus-induced cell transformation and cancer.
Collapse
|
5
|
Tang Y, Song H, Shen Y, Yao Y, Yu Y, Wei G, Long B, Yan W. MiR-155 acts as an inhibitory factor in atherosclerosis-associated arterial pathogenesis by down-regulating NoxA1 related signaling pathway in ApoE -/- mouse. Cardiovasc Diagn Ther 2021; 11:1-13. [PMID: 33708473 DOI: 10.21037/cdt-20-518] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Background To investigate the protective efficacy of miR-155 on down regulating NADPH oxidase isoform subunit A1 (NoxA1) gene expression, resulting in inhibition of VSMC migration and over proliferation and thus ameliorating the progression of arterial atherosclerosis in AS mouse model. Therefore, to further explore the regulatory effect of miR-155 on neointima formation in AS and locate potential anti-atherosclerosis target. Methods The mouse vascular aorta smooth muscle cell (MOVAS) was cultured and transfected with recombinant Pad2YFG adenovirus fluorescent vector with miR-155 fragment into 4 groups. Western blotting and RT-PCR were performed to identify the expression of NoxA1 under different circumstances. Fluorescence microscope was applied to observe the transfection rate of miR-155 into adenovirus. Twelve-week fatty food induced atherosclerotic ApoE-/- mouse model was established as host to accept miR-155 transfected adenovirus transplantation to observe its effect on VSMC in AS progression. Carotid and thoracic artery were extracted at 1 month after dosing. Distribution of miR-155 was quantified via expression levels of protein and RNA to detect NoxA1, Nox1, p47phox and NADPH expression. Immunohistochemistry, fluorescence imaging and other methods were performed in arteries section to compare the thickness of neointima and assess the severity of AS in each group. Results Luciferase reporter gene assay showed significant expression of miR-155 in mimic group indicating that miR-155 had target binding effect with NoxA1 gene. Western blotting and RT-PCR results both showed significantly decreased NoxA1 expression in miR-155 mimic group while increased with its inhibitor. The miR-155 distribution was observed varied at 1 month after in control, miR-155 mimic and inhibitor groups. The NoxA1, NADPH, Nox1 and pp47phox protein expression in VSMC was decreased in mimic group vs control and inhibitor groups (P<0.05); no significant difference of NADPH expression was observed in all groups. The NoxA1, Nox1 and p47phox gene expression in VSMC were both found reduced compared with those of control group at week 4 (P<0.05). Immunohistochemistry staining of artery frozen sections figured out that the thickness of neointima of carotid artery in miR-155 mimic group was significantly lower vs control and inhibitor groups (P<0.01) at week 4. Conclusions miR-155 played an important role in NoxA1-related signaling pathway. miR-155 transfection into VSMC may have anti-inflammatory regulatory effect on NoxA1 expression in vivo and resulting in amelioration of atherosclerotic lesion in AS mouse model. In summary, miR-155 specifically plays in a negative feedback loop and demonstrates a protective role during atherosclerosis-associated VSMC proliferation and neointima formation through the miR-155-NoxA1-p47phox complex signaling pathway.
Collapse
Affiliation(s)
- Yu Tang
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Haoming Song
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Yuqin Shen
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Yian Yao
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Yunan Yu
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Guolian Wei
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Bangxiang Long
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| | - Wenwen Yan
- Department of Cardiology, Tongji Hospital affiliated to Tongji University, Shanghai, China
| |
Collapse
|
6
|
Hou W, Zhu X, Liu J, Ma J. Inhibition of miR-153 ameliorates ischemia/reperfusion-induced cardiomyocytes apoptosis by regulating Nrf2/HO-1 signaling in rats. Biomed Eng Online 2020; 19:15. [PMID: 32143647 PMCID: PMC7059292 DOI: 10.1186/s12938-020-0759-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2019] [Accepted: 02/23/2020] [Indexed: 01/13/2023] Open
Abstract
Background Previous in vitro studies demonstrated that suppression of microRNAs might protect cardiomyocytes and neurons against oxygen–glucose deprivation and reoxygenation (OGD/R)-induced cell apoptosis. However, whether the protective effect of miR-153-inhibition on cardiomyocytes can be observed in the animal model is unknown. We aimed to address this question using a rat model of ischemia–reperfusion (I/R). Methods Rats were received the intramyocardial injection of saline or adenovirus-carrying target or control gene, and the rats were subjected to ischemia/reperfusion (I/R) treatment. The effects of miR-153 on I/R-induced inflammatory response and oxidative stress in the rat model were assessed using various assays. Results We found that suppression of miR-153 decreased cleaved caspase-3 and Bcl-2-associated X (Bax) expression, and increased B cell lymphoma 2 (Bcl-2) expression. We further confirmed that Nuclear transcription factor erythroid 2-like 2 (Nrf2) is a functional target of miR-153, and Nrf2/Heme oxygenase-1 (HO-1) signaling was involved in miR-153-regulated I/R-induced cardiomyocytes apoptosis. Inhibition of miR-153 reduced I/R-induced inflammatory response and oxidative stress in rat myocardium. Conclusion Suppression of miR-153 exerts a cardioprotective effect against I/R-induced injury through the regulation of Nrf2/HO-1 signaling, suggesting that targeting miR-153, Nrf2, or both may serve as promising therapeutic targets for the alleviation of I/R-induced injury.
Collapse
Affiliation(s)
- Wei Hou
- Department of Emergency, Yidu Central Hospital of Wei Fang, No.4138, South Linglongshan Road, Weifang, 262500, Shandong, China
| | - Xianting Zhu
- Department of Nursing, Yidu Central Hospital of Wei Fang, No.4138, South Linglongshan Road, Weifang, 262500, Shandong, China
| | - Juan Liu
- Department of Pediatrics, Ward 1, Yidu Central Hospital of Wei Fang, No. 4138, South Linglongshan Road, Weifang, 262500, Shandong, China
| | - Jiaguo Ma
- Department of Cardiology, Qing Zhou Traditional Chinese Hospital, No. 2727, Haidai Middle Road, Weifang, 262500, Shandong, China.
| |
Collapse
|
7
|
Enhancement of FAK alleviates ventilator-induced alveolar epithelial cell injury. Sci Rep 2020; 10:419. [PMID: 31942012 PMCID: PMC6962166 DOI: 10.1038/s41598-019-57350-6] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2019] [Accepted: 12/16/2019] [Indexed: 01/19/2023] Open
Abstract
Mechanical ventilation induces lung injury by damaging alveolar epithelial cells (AECs), but the pathogenesis remains unknown. Focal adhesion kinase (FAK) is a cytoplasmic protein tyrosine kinase that is involved in cell growth and intracellular signal transduction pathways. This study explored the potential role of FAK in AECs during lung injury induced by mechanical ventilation. High-volume mechanical ventilation (HMV) was used to create a mouse lung injury model, which was validated by analysis of lung weight, bronchoalveolar lavage fluid and histological investigation. The expression of FAK and Akt in AECs were evaluated. In addition, recombinant FAK was administered to mice via the tail vein, and then the extent of lung injury was assessed. Mouse AECs were cultured in vitro, and FAK expression in cells under stretch was investigated. The effects of FAK on cell proliferation, migration and apoptosis were investigated. The results showed that HMV decreased FAK expression in AECs of mice, while FAK supplementation attenuated lung injury, reduced protein levels/cell counts in the bronchoalveolar lavage fluid and decreased histological lung injury and oedema. The protective effect of FAK promoted AEC proliferation and migration and prevented cells from undergoing apoptosis, which restored the integrity of the alveoli through Akt pathway. Therefore, the decrease in FAK expression by HMV is essential for injury to epithelial cells and the disruption of alveolar integrity. FAK supplementation can reduce AEC injury associated with HMV.
Collapse
|
8
|
Lavi K, Jacobson GA, Rosenblum K, Lüthi A. Encoding of Conditioned Taste Aversion in Cortico-Amygdala Circuits. Cell Rep 2019; 24:278-283. [PMID: 29996089 DOI: 10.1016/j.celrep.2018.06.053] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2017] [Revised: 03/22/2018] [Accepted: 06/11/2018] [Indexed: 12/19/2022] Open
Abstract
Avoidance of potentially toxic food by means of conditioned taste aversion is critical for survival of many animals. However, the underlying neuronal mechanisms are poorly understood. Here, using two-photon calcium imaging of defined gustatory cortex neurons in vivo, we show that conditioned taste aversion dynamically shifts neuronal population coding by stimulus-specific recruitment of neurons that project to the basolateral amygdala.
Collapse
Affiliation(s)
- Karen Lavi
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland; Gonda Multidisciplinary Brain Research Center, Bar Ilan University, Ramat Gan 5290002, Israel; Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel
| | - Gilad A Jacobson
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland
| | - Kobi Rosenblum
- Sagol Department of Neurobiology, University of Haifa, Haifa 3498838, Israel; Center for Gene Manipulation in the Brain, University of Haifa, Haifa 3498838, Israel
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Basel 4058, Switzerland; Biozentrum, University of Basel, Basel 4056, Switzerland.
| |
Collapse
|
9
|
Tippimanchai DD, Nolan K, Poczobutt J, Verzosa G, Li H, Scarborough H, Huang J, Young C, DeGregori J, Nemenoff RA, Malkoski SP. Adenoviral vectors transduce alveolar macrophages in lung cancer models. Oncoimmunology 2018; 7:e1438105. [PMID: 29872579 PMCID: PMC5980415 DOI: 10.1080/2162402x.2018.1438105] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2017] [Revised: 01/29/2018] [Accepted: 02/02/2018] [Indexed: 11/09/2022] Open
Abstract
Adenoviral vectors expressing Cre recombinase are commonly used to initiate tumor formation in murine lung cancer models. While these vectors are designed to target genetic recombination to lung epithelial cells, adenoviruses can infect additional cell types that potentially influence tumor development. Our goal was to explore the consequences of adenoviral-mediated alveolar macrophage (AM) transduction in a Kras-initiated lung tumor model. As expected, treatment of animals harboring the KrasLSL-G12D allele and an inducible green fluorescence protein (GFP) tracking allele with an adenoviral vector expressing Cre recombinase under the control of the cytomegalovirus (CMV) promoter (Ad5-CMV-Cre), caused GFP-positive lung adenocarcinomas. Surprisingly, however, up to 70% of the total GFP+ cells were AM, and GFP+ AM could be detected 6 months after tumor initiation, and transduced AM demonstrated Kras activation and increased proliferation. In contrast, recombination was not detected in other immune cell populations and AM recombination could be eliminated by tumor initiation with an adenovirus expressing Cre recombinase under the control of the surfactant protein C (SPC) promoter. In addition, AM isolated from KrasLSL-G12D animals and transduced by Ad5-CMV-Cre ex vivo displayed prolonged survival in vitro and increased the growth of murine lung adenocarcinoma CMT/167 cells when co-injected in an orthotopic flank model. Given the importance of the immune system in tumor development and progression, inadvertent AM transduction by Ad5-CMV-Cre merits careful consideration during lung cancer model selection particularly if studies evaluating the tumor-immune interactions are planned.
Collapse
Affiliation(s)
- Darinee D Tippimanchai
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Kyle Nolan
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Joanna Poczobutt
- Division of Renal Disease and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Gregory Verzosa
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Howard Li
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Hannah Scarborough
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Jing Huang
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Christian Young
- Department of Pathology, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - James DeGregori
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Raphael A Nemenoff
- Division of Renal Disease and Hypertension, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| | - Stephen P Malkoski
- Division of Pulmonary Sciences and Critical Care Medicine, University of Colorado Denver Anschutz Medical Campus, Aurora, CO
| |
Collapse
|
10
|
Rozeske RR, Jercog D, Karalis N, Chaudun F, Khoder S, Girard D, Winke N, Herry C. Prefrontal-Periaqueductal Gray-Projecting Neurons Mediate Context Fear Discrimination. Neuron 2018; 97:898-910.e6. [DOI: 10.1016/j.neuron.2017.12.044] [Citation(s) in RCA: 90] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 12/07/2017] [Accepted: 12/27/2017] [Indexed: 01/22/2023]
|
11
|
Vogel E, Krabbe S, Gründemann J, Wamsteeker Cusulin JI, Lüthi A. Projection-Specific Dynamic Regulation of Inhibition in Amygdala Micro-Circuits. Neuron 2017; 91:644-51. [PMID: 27497223 DOI: 10.1016/j.neuron.2016.06.036] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2015] [Revised: 05/02/2016] [Accepted: 06/22/2016] [Indexed: 12/15/2022]
Abstract
Cannabinoid receptor type 1 (CB1R)-expressing CCK interneurons are key regulators of cortical circuits. Here we report that retrograde endocannabinoid signaling and CB1R-mediated regulation of inhibitory synaptic transmission onto basal amygdala principal neurons strongly depend on principal neuron projection target. Projection-specific asymmetries in the regulation of local inhibitory micro-circuits may contribute to the selective activation of distinct amygdala output pathways during behavioral changes.
Collapse
Affiliation(s)
- Elisabeth Vogel
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; University of Basel, 4000 Basel, Switzerland
| | - Sabine Krabbe
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | - Jan Gründemann
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland
| | | | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, 4058 Basel, Switzerland; University of Basel, 4000 Basel, Switzerland.
| |
Collapse
|
12
|
Xu C, Krabbe S, Gründemann J, Botta P, Fadok JP, Osakada F, Saur D, Grewe BF, Schnitzer MJ, Callaway EM, Lüthi A. Distinct Hippocampal Pathways Mediate Dissociable Roles of Context in Memory Retrieval. Cell 2016; 167:961-972.e16. [PMID: 27773481 DOI: 10.1016/j.cell.2016.09.051] [Citation(s) in RCA: 182] [Impact Index Per Article: 20.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Revised: 06/29/2016] [Accepted: 09/17/2016] [Indexed: 12/15/2022]
Abstract
Memories about sensory experiences are tightly linked to the context in which they were formed. Memory contextualization is fundamental for the selection of appropriate behavioral reactions needed for survival, yet the underlying neuronal circuits are poorly understood. By combining trans-synaptic viral tracing and optogenetic manipulation, we found that the ventral hippocampus (vHC) and the amygdala, two key brain structures encoding context and emotional experiences, interact via multiple parallel pathways. A projection from the vHC to the basal amygdala mediates fear behavior elicited by a conditioned context, whereas a parallel projection from a distinct subset of vHC neurons onto midbrain-projecting neurons in the central amygdala is necessary for context-dependent retrieval of cued fear memories. Our findings demonstrate that two fundamentally distinct roles of context in fear memory retrieval are processed by distinct vHC output pathways, thereby allowing for the formation of robust contextual fear memories while preserving context-dependent behavioral flexibility.
Collapse
Affiliation(s)
- Chun Xu
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Sabine Krabbe
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Jan Gründemann
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Paolo Botta
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, 4003 Basel, Switzerland
| | - Jonathan P Fadok
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland
| | - Fumitaka Osakada
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Dieter Saur
- Department of Internal Medicine 2, Technische Universität München, Ismaningerstrasse 22, 81675 Munich, Germany; German Cancer Consortium (DKTK) and German Cancer Research Center (DKFZ), Im Neuenheimer Feld 280, 69120 Heidelberg, Germany
| | - Benjamin F Grewe
- CNC Program, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Mark J Schnitzer
- CNC Program, Howard Hughes Medical Institute, Stanford University, Stanford, CA 94305, USA
| | - Edward M Callaway
- Systems Neurobiology Laboratories, Salk Institute for Biological Studies, La Jolla, CA 92037, USA
| | - Andreas Lüthi
- Friedrich Miescher Institute for Biomedical Research, Maulbeerstrasse 66, 4058 Basel, Switzerland; University of Basel, 4003 Basel, Switzerland.
| |
Collapse
|
13
|
Clausen BE, Brand A, Karram K. Surmounting limited gene delivery into primary immune cell populations: Efficient cell type-specific adenoviral transduction by CAR. Eur J Immunol 2015; 45:1596-9. [PMID: 25903647 DOI: 10.1002/eji.201545685] [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: 04/14/2015] [Revised: 04/14/2015] [Accepted: 04/16/2015] [Indexed: 12/21/2022]
Abstract
Ectopic gene expression studies in primary immune cells have been notoriously difficult to perform due to the limitations in conventional transfection and viral transduction methods. Although replication-defective adenoviruses provide an attractive alternative for gene delivery, their use has been hampered by the limited susceptibility of murine leukocytes to adenoviral infection, due to insufficient expression of the human coxsackie/adenovirus receptor (CAR). In this issue of the European Journal of Immunology, Heger et al. [Eur. J. Immunol. 2015. 45: XXXX-XXXX] report the generation of transgenic mice that enable conditional Cre/loxP-mediated expression of human CAR. The authors demonstrate that this R26/CAG-CAR∆1(StopF) mouse strain facilitates the faithful monitoring of Cre activity in situ as well as the specific and efficient adenoviral transduction of primary immune cell populations in vitro. Further tweaking of the system towards more efficient gene transfer in vivo remains a future challenge.
Collapse
Affiliation(s)
- Björn E Clausen
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Anna Brand
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| | - Khalad Karram
- Institute for Molecular Medicine, University Medical Center of the Johannes Gutenberg-University Mainz, 55131 Mainz, Germany
| |
Collapse
|
14
|
Heger K, Kober M, Rieß D, Drees C, de Vries I, Bertossi A, Roers A, Sixt M, Schmidt-Supprian M. A novel Cre recombinase reporter mouse strain facilitates selective and efficient infection of primary immune cells with adenoviral vectors. Eur J Immunol 2015; 45:1614-20. [DOI: 10.1002/eji.201545457] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2015] [Revised: 02/16/2015] [Accepted: 03/13/2015] [Indexed: 12/21/2022]
Affiliation(s)
- Klaus Heger
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - Maike Kober
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - David Rieß
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| | - Christoph Drees
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
| | - Ingrid de Vries
- Institute of Science and Technology Austria; Klosterneuburg Austria
| | | | - Axel Roers
- Institute for Immunology; Technische Universität Dresden; Dresden Germany
| | - Michael Sixt
- Institute of Science and Technology Austria; Klosterneuburg Austria
| | - Marc Schmidt-Supprian
- Hematology and Oncology; Klinikum rechts der Isar, Technische Universität München; Munich Germany
- Max Planck Institute of Biochemistry; Martinsried Germany
| |
Collapse
|
15
|
Transcriptional targeting of primary and metastatic tumor neovasculature by an adenoviral type 5 roundabout4 vector in mice. PLoS One 2013; 8:e83933. [PMID: 24376772 PMCID: PMC3871592 DOI: 10.1371/journal.pone.0083933] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2013] [Accepted: 11/10/2013] [Indexed: 11/25/2022] Open
Abstract
New approaches targeting metastatic neovasculature are needed. Payload capacity, cellular transduction efficiency, and first-pass cellular uptake following systemic vector administration, motivates persistent interest in tumor vascular endothelial cell (EC) adenoviral (Ad) vector targeting. While EC transductional and transcriptional targeting has been accomplished, vector administration approaches of limited clinical utility, lack of tumor-wide EC expression quantification, and failure to address avid liver sequestration, challenged prior work. Here, we intravenously injected an Ad vector containing 3 kb of the human roundabout4 (ROBO4) enhancer/promoter transcriptionally regulating an enhanced green fluorescent protein (EGFP) reporter into immunodeficient mice bearing 786-O renal cell carcinoma subcutaneous (SC) xenografts and kidney orthotopic (KO) tumors. Initial experiments performed in human coxsackie virus and adenovirus receptor (hCAR) transgenic:Rag2 knockout mice revealed multiple ECs with high-level Ad5ROBO4-EGFP expression throughout KO and SC tumors. In contrast, Ad5CMV-EGFP was sporadically expressed in a few tumor vascular ECs and stromal cells. As the hCAR transgene also facilitated Ad5ROBO4 and control Ad5CMV vector EC expression in multiple host organs, follow-on experiments engaged warfarin-mediated liver vector detargeting in hCAR non-transgenic mice. Ad5ROBO4-mediated EC expression was undetectable in most host organs, while the frequencies of vector expressing intratumoral vessels and whole tumor EGFP protein levels remained elevated. In contrast, AdCMV vector expression was only detectable in one or two stromal cells throughout the whole tumor. The Ad5ROBO4 vector, in conjunction with liver detargeting, provides tractable genetic access for in-vivo EC genetic engineering in malignancies.
Collapse
|
16
|
EGFR-Targeted Adenovirus Dendrimer Coating for Improved Systemic Delivery of the Theranostic NIS Gene. MOLECULAR THERAPY. NUCLEIC ACIDS 2013; 2:e131. [PMID: 24193032 PMCID: PMC3889187 DOI: 10.1038/mtna.2013.58] [Citation(s) in RCA: 48] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/08/2013] [Accepted: 08/10/2013] [Indexed: 12/19/2022]
Abstract
We recently demonstrated tumor-selective iodide uptake and therapeutic efficacy of
combined radiovirotherapy after systemic delivery of the theranostic sodium iodide
symporter (NIS) gene using a dendrimer-coated adenovirus. To further improve shielding and
targeting we physically coated replication-selective adenoviruses carrying the
hNIS gene with a conjugate consisting of cationic poly(amidoamine) (PAMAM)
dendrimer linked to the peptidic, epidermal growth factor receptor (EGFR)-specific ligand
GE11. In vitro experiments demonstrated coxsackie-adenovirus receptor-independent
but EGFR-specific transduction efficiency. Systemic injection of the uncoated adenovirus
in a liver cancer xenograft mouse model led to high levels of NIS expression in the liver
due to hepatic sequestration, which were significantly reduced after coating as
demonstrated by 123I-scintigraphy. Reduction of adenovirus liver pooling
resulted in decreased hepatotoxicity and increased transduction efficiency in peripheral
xenograft tumors. 124I-PET-imaging confirmed EGFR-specificity by significantly
lower tumoral radioiodine accumulation after pretreatment with the EGFR-specific antibody
cetuximab. A significantly enhanced oncolytic effect was observed following systemic
application of dendrimer-coated adenovirus that was further increased by additional
treatment with a therapeutic dose of 131I. These results demonstrate restricted
virus tropism and tumor-selective retargeting after systemic application of coated,
EGFR-targeted adenoviruses therefore representing a promising strategy for improved
systemic adenoviral NIS gene therapy.
Collapse
|
17
|
Kaliberov SA, Kaliberova LN, Hong Lu Z, Preuss MA, Barnes JA, Stockard CR, Grizzle WE, Arbeit JM, Curiel DT. Retargeting of gene expression using endothelium specific hexon modified adenoviral vector. Virology 2013; 447:312-25. [PMID: 24210128 DOI: 10.1016/j.virol.2013.09.020] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 08/27/2013] [Accepted: 09/20/2013] [Indexed: 01/30/2023]
Abstract
Adenovirus serotype 5 (Ad5) vectors are well suited for gene therapy. However, tissue-selective transduction by systemically administered Ad5-based vectors is confounded by viral particle sequestration in the liver. Hexon-modified Ad5 expressing reporter gene under transcriptional control by the immediate/early cytomegalovirus (CMV) or the Roundabout 4 receptor (Robo4) enhancer/promoter was characterized by growth in cell culture, stability in vitro, gene transfer in the presence of human coagulation factor X, and biodistribution in mice. The obtained data demonstrate the utility of the Robo4 promoter in an Ad5 vector context. Substitution of the hypervariable region 7 (HVR7) of the Ad5 hexon with HVR7 from Ad serotype 3 resulted in decreased liver tropism and dramatically altered biodistribution of gene expression. The results of these studies suggest that the combination of liver detargeting using a genetic modification of hexon with an endothelium-specific transcriptional control element produces an additive effect in the improvement of Ad5 biodistribution.
Collapse
Affiliation(s)
- Sergey A Kaliberov
- Department of Radiation Oncology, School of Medicine, Washington University in St. Louis, St. Louis, MO, United States of America.
| | | | | | | | | | | | | | | | | |
Collapse
|
18
|
A myeloid cell-binding adenovirus efficiently targets gene transfer to the lung and escapes liver tropism. Gene Ther 2012; 20:733-41. [PMID: 23171918 DOI: 10.1038/gt.2012.91] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2012] [Revised: 10/15/2012] [Accepted: 10/16/2012] [Indexed: 12/19/2022]
Abstract
Specific and efficient gene delivery to the lung has been hampered by liver sequestration of adenovirus serotype 5 (Ad5) vectors. The complexity of Ad5 liver tropism has largely been unraveled, permitting improved efficacy of Ad5 gene delivery. However, Kupffer cell (KC) scavenging and elimination of Ad5 still represent major obstacles to lung gene delivery strategies. KC uptake substantially reduces bioavailability of Ad5 for target tissues and compensatory dose escalation leads to acute hepatotoxicity and a potent innate immune response. Here, we report a novel lung-targeting strategy through redirection of Ad5 binding to the concentrated leukocyte pool within the pulmonary microvasculature. We demonstrate that this leukocyte-binding approach retargets Ad5 specifically to lung endothelial cells and prevents KC uptake and hepatocyte transduction, resulting in 165,000-fold enhanced lung targeting, compared with Ad5. In addition, myeloid cell-specific binding is preserved in single-cell lung suspensions and only Ad.MBP-coated myeloid cells achieved efficient endothelial cell transduction ex vivo. These findings demonstrate that KC sequestration of Ad5 can be prevented through more efficient uptake of virions in target tissues and suggest that endothelial transduction is achieved by leukocyte-mediated 'hand-off' of Ad.
Collapse
|
19
|
Abstract
Passive transfer of neutralizing antibodies against HIV-1 can prevent infection in macaques and seems to delay HIV-1 rebound in humans. Anti-HIV antibodies are therefore of great interest for vaccine design. However, the basis for their in vivo activity has been difficult to evaluate systematically because of a paucity of small animal models for HIV infection. Here we report a genetically humanized mouse model that incorporates a luciferase reporter for rapid quantitation of HIV entry. An antibody's ability to block viral entry in this in vivo model is a function of its bioavailability, direct neutralizing activity, and effector functions.
Collapse
|
20
|
Alberti MO, Roth JC, Ismail M, Tsuruta Y, Abraham E, Pereboeva L, Gerson SL, Curiel DT. Derivation of a myeloid cell-binding adenovirus for gene therapy of inflammation. PLoS One 2012; 7:e37812. [PMID: 22624065 PMCID: PMC3356302 DOI: 10.1371/journal.pone.0037812] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2012] [Accepted: 04/24/2012] [Indexed: 11/21/2022] Open
Abstract
The gene therapy field is currently limited by the lack of vehicles that permit efficient gene delivery to specific cell or tissue subsets. Native viral vector tropisms offer a powerful platform for transgene delivery but remain nonspecific, requiring elevated viral doses to achieve efficacy. In order to improve upon these strategies, our group has focused on genetically engineering targeting domains into viral capsid proteins, particularly those based on adenovirus serotype 5 (Ad5). Our primary strategy is based on deletion of the fiber knob domain, to eliminate broad tissue specificity through the human coxsackie-and-adenovirus receptor (hCAR), with seamless incorporation of ligands to re-direct Ad tropism to cell types that express the cognate receptors. Previously, our group and others have demonstrated successful implementation of this strategy in order to specifically target Ad to a number of surface molecules expressed on immortalized cell lines. Here, we utilized phage biopanning to identify a myeloid cell-binding peptide (MBP), with the sequence WTLDRGY, and demonstrated that MBP can be successfully incorporated into a knob-deleted Ad5. The resulting virus, Ad.MBP, results in specific binding to primary myeloid cell types, as well as significantly higher transduction of these target populations ex vivo, compared to unmodified Ad5. These data are the first step in demonstrating Ad targeting to cell types associated with inflammatory disease.
Collapse
Affiliation(s)
- Michael O. Alberti
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Justin C. Roth
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (JCR); (DTC)
| | - Mourad Ismail
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- National Center for Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - Yuko Tsuruta
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Edward Abraham
- Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Larisa Pereboeva
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Stanton L. Gerson
- Division of Hematology and Oncology, Department of Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
- National Center for Regenerative Medicine, Case Comprehensive Cancer Center, Case Western Reserve University, Cleveland, Ohio, United States of America
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, Surgery, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail: (JCR); (DTC)
| |
Collapse
|
21
|
Abstract
Progress in vector design and an increased knowledge of mechanisms underlying tumor-induced immune suppression have led to a new and promising generation of Adenovirus (Ad)-based immunotherapies, which are discussed in this review. As vaccine vehicles Ad vectors (AdVs) have been clinically evaluated and proven safe, but a major limitation of the commonly used Ad5 serotype is neutralization by preexistent or rapidly induced immune responses. Genetic modifications in the Ad capsid can reduce intrinsic immunogenicity and facilitate escape from antibody-mediated neutralization. Further modification of the Ad hexon and fiber allows for liver and scavenger detargeting and selective targeting of, for example, dendritic cells. These next-generation Ad vaccines with enhanced efficacy are now becoming available for testing as tumor vaccines. In addition, AdVs encoding immune-modulating products may be used to convert the tumor microenvironment from immune-suppressive and proinvasive to proinflammatory, thus facilitating cell-mediated effector functions that can keep tumor growth and invasion in check. Oncolytic AdVs, that selectively replicate in tumor cells and induce an immunogenic form of cell death, can also be armed with immune-activating transgenes to amplify primed antitumor immune responses. These novel immunotherapy strategies, employing highly efficacious AdVs in optimized configurations, show great promise and warrant clinical exploration.
Collapse
|
22
|
Unity and diversity in the human adenoviruses: exploiting alternative entry pathways for gene therapy. Biochem J 2010; 431:321-36. [DOI: 10.1042/bj20100766] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Human Ads (adenoviruses) have been extensively utilized for the development of vectors for gene transfer, as they infect many cell types and do not integrate their genome into host-cell chromosomes. In addition, they have been widely studied as cytolytic viruses, termed oncolytic adenoviruses in cancer therapy. Ads are non-enveloped viruses with a linear double-stranded DNA genome of 30–38 kb which encodes 30–40 genes. At least 52 human Ad serotypes have been identified and classified into seven species, A–G. The Ad capsid has icosahedral symmetry and is composed of 252 capsomers, of which 240 are located on the facets of the capsid and consist of a trimeric hexon protein and the remaining 12 capsomers, the pentons, are at the vertices and comprise the penton base and projecting fibre protein. The entry of Ads into human cells is a two-step process. In the first step, the fibre protein mediates a primary interaction with the cell, effectively tethering the virus particle to the cell surface via a cellular attachment protein. The penton base then interacts with cell-surface integrins, leading to virus internalization. This interaction of the fibre protein with a number of cell-surface molecules appears to be important in determining the tropism of adenoviruses. Ads from all species, except species B and certain serotypes of species D, utilize CAR (coxsackie and adenovirus receptor) as their primary cellular-attachment protein, whereas most species B Ads use CD46, a complement regulatory protein. Such species-specific differences, as well as adaptations or modifications of Ads required for applications in gene therapy, form the major focus of the present review.
Collapse
|
23
|
Lewis TB, Glasgow JN, Glandon AM, Curiel DT, Standaert DG. Transduction of brain dopamine neurons by adenoviral vectors is modulated by CAR expression: rationale for tropism modified vectors in PD gene therapy. PLoS One 2010; 5. [PMID: 20862245 PMCID: PMC2941453 DOI: 10.1371/journal.pone.0012672] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2010] [Accepted: 08/18/2010] [Indexed: 01/01/2023] Open
Abstract
Background Gene-based therapy is a new paradigm for the treatment of Parkinson disease (PD) and offers considerable promise for precise targeting and flexibility to impact multiple pathobiological processes for which small molecule agents are not available. Some success has been achieved utilizing adeno-associated virus for this approach, but it is likely that the characteristics of this vector system will ultimately create barriers to progress in clinical therapy. Adenovirus (Ad) vector overcomes limitations in payload size and targeting. The cellular tropism of Ad serotype 5 (Ad5)–based vectors is regulated by the Ad attachment protein binding to its primary cellular receptor, the coxsackie and adenovirus receptor (CAR). Many clinically relevant tissues are refractory to Ad5 infection due to negligible CAR levels but can be targeted by tropism-modified, CAR-independent forms of Ad. Our objective was to evaluate the role of CAR protein in transduction of dopamine (DA) neurons in vivo. Methodology/Principal Findings Ad5 was delivered to the substantia nigra (SN) in wild type (wt) and CAR transgenic animals. Cellular tropism was assessed by immunohistochemistry (IHC) in the SN and striatal terminals. CAR expression was assessed by western blot and IHC. We found in wt animals, Ad5 results in robust transgene expression in astrocytes and other non-neuronal cells but poor infection of DA neurons. In contrast, in transgenic animals, Ad5 infects SNc neurons resulting in expression of transduced protein in their striatal terminals. Western blot showed low CAR expression in the ventral midbrain of wt animals compared to transgenic animals. Interestingly, hCAR protein localizes with markers of post-synaptic structures, suggesting synapses are the point of entry into dopaminergic neurons in transgenic animals. Conclusions/Significance These findings demonstrate that CAR deficiency limits infection of wild type DA neurons by Ad5 and provide a rationale for the development of tropism-modified, CAR-independent Ad-vectors for use in gene therapy of human PD.
Collapse
Affiliation(s)
- Travis B. Lewis
- Department of Cell Biology, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Joel N. Glasgow
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Division of Cardiovascular Disease, Department of Medicine, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - Anya M. Glandon
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David T. Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetrics and Gynecology, Pathology, and Surgery, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- Gene Therapy Center, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
| | - David G. Standaert
- Center for Neurodegeneration and Experimental Therapeutics, The University of Alabama at Birmingham, Birmingham, Alabama, United States of America
- * E-mail:
| |
Collapse
|
24
|
Komarova S, Roth J, Alvarez R, Curiel DT, Pereboeva L. Targeting of mesenchymal stem cells to ovarian tumors via an artificial receptor. J Ovarian Res 2010; 3:12. [PMID: 20500878 PMCID: PMC2883983 DOI: 10.1186/1757-2215-3-12] [Citation(s) in RCA: 46] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2010] [Accepted: 05/25/2010] [Indexed: 02/07/2023] Open
Abstract
Background Mesenchymal Progenitor/Stem Cells (MSC) respond to homing cues providing an important mechanism to deliver therapeutics to sites of injury and tumors. This property has been confirmed by many investigators, however, the efficiency of tumor homing needs to be improved for effective therapeutic delivery. We investigated the feasibility of enhancing MSC tumor targeting by expressing an artificial tumor-binding receptor on the MSC surface. Methods Human MSC expressing an artificial receptor that binds to erbB2, a tumor cell marker, were obtained by transduction with genetically modified adenoviral vectors encoding an artificial receptor (MSC-AR). MSC-AR properties were tested in vitro in cell binding assays and in vivo using two model systems: transient transgenic mice that express human erbB2 in the lungs and ovarian xenograft tumor model. The levels of luciferase-labeled MSCs in erbB2-expressing targeted sites were evaluated by measuring luciferase activity using luciferase assay and imaging. Results The expression of AR enhanced binding of MSC-AR to erbB2-expressing cells in vitro, compared to unmodified MSCs. Furthermore, we have tested the properties of erbB2-targeted MSCs in vivo and demonstrated an increased retention of MSC-AR in lungs expressing erbB2. We have also confirmed increased numbers of erbB2-targeted MSCs in ovarian tumors, compared to unmodified MSC. The kinetic of tumor targeting by ip injected MSC was also investigated. Conclusion These data demonstrate that targeting abilities of MSCs can be enhanced via introduction of artificial receptors. The application of this strategy for tumor cell-based delivery could increase a number of cell carriers in tumors and enhance efficacy of cell-based therapy.
Collapse
Affiliation(s)
- Svetlana Komarova
- Division of Human Gene Therapy, Department of Medicine, University of Alabama at Birmingham, Birmingham, Alabama 35294- 2172, USA.
| | | | | | | | | |
Collapse
|
25
|
Tarallo V, Vesci L, Capasso O, Esposito MT, Riccioni T, Pastore L, Orlandi A, Pisano C, De Falco S. A placental growth factor variant unable to recognize vascular endothelial growth factor (VEGF) receptor-1 inhibits VEGF-dependent tumor angiogenesis via heterodimerization. Cancer Res 2010; 70:1804-13. [PMID: 20145150 DOI: 10.1158/0008-5472.can-09-2609] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Angiogenesis is one of the crucial events for cancer development and growth. Two members of the vascular endothelial growth factor (VEGF) family, VEGF-A and placental growth factor (PlGF), which are able to heterodimerize if coexpressed in the same cell, are both required for pathologic angiogenesis. We have generated a PlGF1 variant, named PlGF1-DE in which the residues Asp72 and Glu73 were substituted with Ala, which is unable to bind and activate VEGF receptor-1 but is still able to heterodimerize with VEGF. Here, we show that overexpression in tumor cells by adenoviral delivery or stable transfection of PlGF1-DE variant significantly reduces the production of VEGF homodimer via heterodimerization, determining a strong inhibition of xenograft tumor growth and neoangiogenesis, as well as significant reduction of vessel lumen and stabilization, and monocyte-macrophage infiltration. Conversely, the overexpression of PlGF1wt, also reducing the VEGF homodimer production comparably with PlGF1-DE variant through the generation of VEGF/PlGF heterodimer, does not inhibit tumor growth and vessel density compared with controls but induces increase of vessel lumen, vessel stabilization, and monocyte-macrophage infiltration. The property of PlGF and VEGF-A to generate heterodimer represents a successful strategy to inhibit VEGF-dependent angiogenesis. The PlGF1-DE variant, and not PlGF1wt as previously reported, acts as a "dominant negative" of VEGF and is a new candidate for antiangiogenic gene therapy in cancer treatment.
Collapse
Affiliation(s)
- Valeria Tarallo
- Angiogenesis Lab and Stem Cell Fate Lab, Institute of Genetics and Biophysics Adriano Buzzati-Traverso, Consiglio Nazionale delle Ricerche, Naples, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Chen SY, Shiau AL, Shieh GS, Su CH, Lee CH, Lee HL, Wang CR, Wu CL. Amelioration of experimental arthritis by a telomerase-dependent conditionally replicating adenovirus that targets synovial fibroblasts. ACTA ACUST UNITED AC 2010; 60:3290-302. [PMID: 19877026 DOI: 10.1002/art.24940] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
OBJECTIVE Synovial fibroblasts (SFs) play a pivotal role in the pathogenesis of rheumatoid arthritis (RA). It has been documented that the phenotype of rheumatoid synovium is similar, in many respects, to that of an aggressive tumor. In this study, a novel, genetically engineered adenovirus was designed to lyse SFs that exhibit high telomerase activity and p53 mutations, and its effects as a novel therapeutic strategy were assessed in an experimental arthritis model. METHODS An E1B-55-kd-deleted adenovirus driven by the human telomerase reverse transcriptase promoter was constructed (designated Ad.GS1). Cytolysis of SFs and productive replication of Ad.GS1 in the SFs of rats with collagen-induced arthritis (CIA), as well as the SFs of patients with RA (RASFs), were assessed in vitro and in vivo. Treatment responses, as well as the presence of disease-related cytokines and enzymes in the ankle joints, were determined in the murine model. RESULTS Ad.GS1 replicated in and induced cytolysis of human RASFs and SFs from arthritic rats, but spared normal fibroblasts. Bioluminescence imaging in vivo also demonstrated replication of Ad.GS1 in arthritic rat joints, but not in normal rat joints. Intraarticular administration of Ad.GS1 significantly reduced the ankle circumference, articular index scores, radiographic scores, and histologic scores and decreased the production of interleukin-1beta, matrix metalloproteinase 9, and prolyl 4-hydroxylase in rats with CIA compared with their control counterparts. CONCLUSION This study is the first to demonstrate the amelioration of arthritic symptoms by a novel, telomerase-dependent adenovirus in the rat CIA model, an experimental model that resembles human RA. In addition, the results suggest that because of its ability to induce cytolysis of SFs, this virus may be further explored as a therapeutic agent in patients with RA.
Collapse
Affiliation(s)
- Shih-Yao Chen
- National Cheng Kung University Medical College, Tainan, Taiwan
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Shimony N, Elkin G, Kolodkin-Gal D, Krasny L, Urieli-Shoval S, Haviv YS. Analysis of adenoviral attachment to human platelets. Virol J 2009; 6:25. [PMID: 19222836 PMCID: PMC2649059 DOI: 10.1186/1743-422x-6-25] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2009] [Accepted: 02/17/2009] [Indexed: 12/01/2022] Open
Abstract
Background Systemic adenoviral (Ad) vector administration is associated with thrombocytopenia. Recently, Ad interaction with mouse platelets emerged as a key player determining liver uptake and platelet clearance. However, whether Ad can activate platelets is controversial. Thus, in vitro analysis of Ad attachment to platelets is of interest. Methods We developed a direct flow cytometry assay to specifically detect Ad particles adherent to human platelets. The method was pre-validated in nucleated cells. Blocking assays were employed to specifically inhibit Ad attachment to platelets. Platelet activation was analyzed using annexin v flow cytometry. Results We found in vitro that Ad binding to human platelets is synergistically enhanced by the combination of platelet activation by thrombin and MnCl2 supplementation. Of note, Ad binding could activate human platelets. Platelets bound Ad displaying an RGD ligand in the fiber knob more efficiently than unmodified Ad. In contrast to a previous report, CAR expression was not detected on human platelets. Integrins appear to mediate Ad binding to platelets, at least partially. Finally, αIIbβ3-deficient platelets from a patient with Glanzmann thrombasthenia could bind Ad 5-fold more efficiently than normal platelets. Conclusion The flow cytometry methodology developed herein allows the quantitative measurement of Ad attachment to platelets and may provide a useful in vitro approach to investigate Ad interaction with platelets.
Collapse
Affiliation(s)
- Nilly Shimony
- Department of Medicine, Hadassah-Hebrew University Medical Center, Jerusalem 91120, Israel.
| | | | | | | | | | | |
Collapse
|
28
|
Mirza M, Petersen C, Nordqvist K, Sollerbrant K. Coxsackievirus and adenovirus receptor is up-regulated in migratory germ cells during passage of the blood-testis barrier. Endocrinology 2007; 148:5459-69. [PMID: 17690169 DOI: 10.1210/en.2007-0359] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The coxsackievirus and adenovirus receptor (CAR) is a cell adhesion molecule expressed in epithelial tight junctions and other cell-cell contacts. Using indirect immunofluorescence, quantitative RT-PCR, and Western blots, the expression and distribution of CAR in developing and adult testis are examined. CAR is highly expressed in both Sertoli and germ cells during perinatal and postnatal development, followed by a rapid down-regulation of both mRNA and protein levels. Interestingly, we find that CAR is a previously unknown downstream target for FSH because CAR mRNA levels were induced in primary cultures of FSH-stimulated Sertoli cells. In contrast to other epithelia, CAR is not a general component of tight junctions in the seminiferous epithelium, and Sertoli cells in the adult testis do not express CAR. Instead, CAR expression is stage dependent and specifically found in migratory germ cells. RT-PCR also demonstrated the presence of junctional adhesion molecule-like (JAML) in the testis. JAML was previously reported by others to form a functional complex with CAR regulating transepithelial migration of leukocytes. The expression of JAML in the testis suggests that a similar functional complex might be present during germ cell migration across the blood-testis barrier. Finally, an intermediate compartment occupied by CAR-positive, migrating germ cells and flanked by two occludin-containing junctions is identified. Together, these results implicate a function for CAR in testis morphogenesis and in migration of germ cells across the blood-testis barrier during spermatogenesis.
Collapse
Affiliation(s)
- Momina Mirza
- Ludwig Institute for Cancer Research, Stockholm Branch, Karolinska Institutet, Box 240, SE-17177, Stockholm, Sweden
| | | | | | | |
Collapse
|
29
|
Abstract
Translating knowledge of genetic disease mechanisms into gene therapies has been slow with limited clinical success. One major reason is that the transfer vectors, which are most often of viral origin, are not targeted sufficiently towards the cells of interest. To achieve successful delivery of genetic material, transductional targeting is often essential to enter the target cell and to avoid side effects from the transduction of non-target cells. Many techniques to target viral vectors to specific cells have been developed. They can be divided into three types: systems that use adaptor proteins from other viruses (pseudotyping); systems that use adaptors to couple the targeting ligand to the vector; and systems that genetically incorporate the targeting moiety into the viral genome. Whereas systems involving adaptor proteins are highly useful in preclinical evaluations, systems that make use of genetically incorporated targeting ligands are advantageous for clinical applications. Combinations of several targeting principles (including ablation of natural tropism, pseudotyping and adaptors) and novel combinations (such as the adeno-associated virus (AAV) genome in a phage vector) allow systemic vector application. An initial clinical study with a targeted retrovirus showed feasibility to transfer laboratory success to patient application, underlining that there are no principal regulatory barriers for targeted vectors. Systemic vector applications will be facilitated by enabling the vector to move beyond the vascular endothelium at specific sites, using transcytosis or cellular vehicles. The application of existing targeting techniques to new viral vector serotypes and new vector classes is extending the therapeutic capabilities further. Obstacles to systemic application of vectors are found in the blood as immune reactions against the vector and as binding of blood proteins to the vector. Some targeting approaches might have the potential to circumvent these obstacles. To preclinically evaluate new targeting strategies, several models that reflect the human situation to varying degrees are available. The use of primary cells, tissue-slice systems and transgenic animals seems to be especially promising. Imaging technologies provide the ability to monitor the vector in vivo in real time without sacrificing the animal model. These techniques facilitate vector targeting and biodistribution studies.
A key challenge in gene therapy is vector targeting to specific cells, while avoiding effects on other tissues. Several strategies have been developed recently to enable targeting of the main viral vectors, moving them a step closer to clinical use. To achieve therapeutic success, transfer vehicles for gene therapy must be capable of transducing target cells while avoiding impact on non-target cells. Despite the high transduction efficiency of viral vectors, their tropism frequently does not match the therapeutic need. In the past, this lack of appropriate targeting allowed only partial exploitation of the great potential of gene therapy. Substantial progress in modifying viral vectors using diverse techniques now allows targeting to many cell types in vitro. Although important challenges remain for in vivo applications, the first clinical trials with targeted vectors have already begun to take place.
Collapse
Affiliation(s)
- Reinhard Waehler
- Division of Human Gene Therapy, 502 Biomedical Research Building II, 901 19th Street, South Birmingham, 35294-2172 Alabama USA
| | - Stephen J. Russell
- Molecular Medicine Program, Mayo Clinic College of Medicine, 200 First Street SW, Rochester, 55905 Minnesota USA
| | - David T. Curiel
- Division of Human Gene Therapy, 502 Biomedical Research Building II, 901 19th Street, South Birmingham, 35294-2172 Alabama USA
| |
Collapse
|
30
|
Pereboeva L, Komarova S, Roth J, Ponnazhagan S, Curiel DT. Targeting EGFR with metabolically biotinylated fiber-mosaic adenovirus. Gene Ther 2007; 14:627-37. [PMID: 17251987 PMCID: PMC2203207 DOI: 10.1038/sj.gt.3302916] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Adenovirus (Ad)-based vectors are useful gene delivery vehicles for a variety of applications. Despite their attractive properties, many in vivo applications require modulation of the viral tropism. Targeting approaches applied to adenoviral vectors included genetic modification of the viral capsid, controlled expression of the transgene and combinatorial approaches that combine two or more targeting elements in single vectors. Most of these studies confirmed successful retargeting in cell cultures, however, in vivo gains of targeted adenoviral vectors have not been widely demonstrated. We have developed a combinatorial retargeting approach utilizing metabolically biotinylated Ad, where the biotin acceptor peptide was incorporated in one of the fibers in a dual fiber viral particle resulting in metabolically biotinylated fiber-mosaic Ad (mBfMAd). We have utilized this vector in complex with epidermal growth factor (EGF)-Streptavidin to retarget fiber-mosaic virus to EGF receptor (EGFR) expressing cells in vitro and confirmed an increased infectivity of the retargeting complex. Most importantly, the utility of this strategy was demonstrated in vivo in two distinct animal models. In both models tested, retargeted mBfMAd demonstrated an increased ratio of gene expression in target tissues compared to the liver expression profile. Thus, metabolically biotinylated fiber-mosaic virus in combination with appropriate adapters can be successfully exploited for adenoviral retargeting strategies.
Collapse
Affiliation(s)
- L Pereboeva
- Division of Human Gene Therapy, Departments of Medicine, Obstetric and Gynecology, Pathology and Surgery, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Komarova
- Division of Human Gene Therapy, Departments of Medicine, Obstetric and Gynecology, Pathology and Surgery, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - J Roth
- Division of Human Gene Therapy, Departments of Medicine, Obstetric and Gynecology, Pathology and Surgery, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - S Ponnazhagan
- Department of Pathology, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| | - DT Curiel
- Division of Human Gene Therapy, Departments of Medicine, Obstetric and Gynecology, Pathology and Surgery, Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
31
|
Shaw CA, Larochelle N, Dudley RWR, Lochmuller H, Danialou G, Petrof BJ, Karpati G, Holland PC, Nalbantoglu J. Simultaneous dystrophin and dysferlin deficiencies associated with high-level expression of the coxsackie and adenovirus receptor in transgenic mice. THE AMERICAN JOURNAL OF PATHOLOGY 2007; 169:2148-60. [PMID: 17148677 PMCID: PMC1762479 DOI: 10.2353/ajpath.2006.060570] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
The Coxsackie and adenovirus receptor (CAR), a cell adhesion molecule of the immunoglobulin superfamily, is usually confined to the sarcolemma at the neuromuscular junction in mature skeletal muscle fibers. Previously, we reported that adenovirus-mediated gene transfer is greatly facilitated in hemizygous transgenic mice with extrasynaptic CAR expression driven by a muscle-specific promoter. However, in the present study, when these mice were bred to homozygosity, they developed a severe myopathic phenotype and died prematurely. Large numbers of necrotic and regenerating fibers were present in the skeletal muscle of the homozygous CAR transgenics. The myopathy was further characterized by increased levels of caveolin-3 and beta-dystroglycan and decreased levels of dystrophin, dysferlin, and neuronal nitric-oxide synthase. Even the hemizygotes manifested a subtle phenotype, displaying deficits in isometric force generation and perturbed mitogen-activated protein kinase (MAPK-erk1/2) activation during contraction. There are few naturally occurring or engineered mouse lines showing as severe a skeletal myopathy as observed with ectopic expression of CAR in the homozygotes. Taken together, these findings suggest that substantial overexpression of CAR may lead to physiological dysfunction by disturbing sarcolemmal integrity (through dystrophin deficiency), impairing sarcolemmal repair (through dysferlin deficiency), and interfering with normal signaling (through alterations in caveolin-3 and neuronal nitric-oxide synthase levels).
Collapse
Affiliation(s)
- Christian A Shaw
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University Health Center and Meakins-Christie Laboratories, 3801 University St., Montreal, Quebec, Canada H3A 2B4
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Izumi M, Kawakami Y, Glasgow JN, Belousova N, Everts M, Kim-Park S, Yamamoto S, Wang M, Le LP, Reynolds PN, Curiel DT. In vivo analysis of a genetically modified adenoviral vector targeted to human CD40 using a novel transient transgenic model. J Gene Med 2006; 7:1517-25. [PMID: 16170831 DOI: 10.1002/jgm.806] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
BACKGROUND Retargeting is necessary to overcome the limitations of adenovirus (Ad)-based gene therapy vectors. To this end, we previously constructed an adenovirus with the fiber knob domain replaced by a fibritin trimerization motif fused to the CD40 ligand (Ad5Luc.FF/CD40L). We demonstrated the utility of this fiber replacement strategy for targeting CD40 (hCD40) on human dendritic cells in vitro. The in vivo targeting capacity of this virus, however, is unknown, and there is a limited repertoire of animal models that present hCD40 at an accessible site. Therefore, a new animal model for evaluating CD40-targeted vectors is required. METHODS We constructed a recombinant adenovirus that expresses hCD40 under transcriptional control of the flt-1 promoter (AdflthCD40). Expression of hCD40 was validated both in vitro and in transgenic mice expressing the human coxsackie adenovirus receptor (hCAR mice). We then evaluated the targeting efficiency of Ad5Luc.FF/CD40L to hCD40 expressed in the pulmonary vasculature of the hCAR mice. RESULTS Infection of flt-1-positive cells with AdflthCD40 resulted in abundant hCD40 expression in vitro, which could subsequently be targeted by Ad5Luc.FF/CD40L. In vivo administration of AdflthCD40 to hCAR mice resulted in hCD40 expression in the pulmonary vasculature, which was successfully targeted with systemically administered Ad5Luc.FF/CD40L. CONCLUSIONS This is the first data showing that genetically modified Ad5Luc.FF/CD40L can successfully target hCD40 in vivo. Our data also establishes the utility of transcriptionally targeted, Ad-mediated transient expression of human target molecules in the pulmonary vasculature of hCAR mice as models for in vivo analysis of targeted gene therapy vectors.
Collapse
Affiliation(s)
- Miiru Izumi
- Division of Human Gene Therapy, Department of Medicine, Gene Therapy Center, University of Alabama at Birmingham, AL 35294-2172, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
33
|
Allen AM, Dosanjh J, Dassanayake S, Tan G, Thomas WG. Baroreceptor reflex stimulation does not induce cytomegalovirus promoter-driven transgene expression in the ventrolateral medulla in vivo. Auton Neurosci 2006; 126-127:150-5. [PMID: 16624635 DOI: 10.1016/j.autneu.2006.02.024] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2005] [Revised: 02/20/2006] [Accepted: 02/27/2006] [Indexed: 10/24/2022]
Abstract
Adenoviruses are being employed to induce transgene expression in the central nervous system in vivo. In these studies, the cytomegalovirus (CMV) promoter is commonly employed to drive expression of the transgene because of its strong, constitutive activity in a wide range of cell types. However, using this promoter, expression in neurons is variable, with strongest expression being observed in non-neuronal cells. Indeed, even in vitro, CMV driven expression in neurons is variable. In cultured sympathetic ganglion cells it has been demonstrated that CMV-driven expression requires activation of cAMP-response element-binding protein (CREB) and that this can be induced by depolarization. In this study we tested whether depolarization might induce CMV-driven transgene expression, delivered by microinjection of an adenovirus, in the rostral ventrolateral medulla (RVLM) of rats. Prior to stimulation, transgene expression occurs in non-neuronal cells in the RVLM. Some neuronal expression was observed in neighbouring regions, in the nucleus ambiguus and in facial motor neurons. Within the RVLM, depolarization, induced by intraperitoneal administration of the ganglion blocking drug, pentolinium, did not lead to induction of transgene expression. This stimulus is known to induce expression of the immediate early gene c-fos. We conclude that either this experimental paradigm was not sufficient for activation of the CREB pathway or that possibly the virus does not gain access to the neurons of the RVLM. The adoption of specific promoters or viruses with higher neuronal transduction efficiency appears to be essential for the genetic modification of RVLM presympathetic neurons in vivo.
Collapse
Affiliation(s)
- Andrew M Allen
- Department of Physiology, and Howard Florey Institute, University of Melbourne, Victoria, 3010, Australia.
| | | | | | | | | |
Collapse
|
34
|
Wang H, Xie H, Zhang H, Das SK, Dey SK. Conditional gene recombination by adenovirus-driven Cre in the mouse uterus. Genesis 2006; 44:51-6. [PMID: 16416422 PMCID: PMC4267753 DOI: 10.1002/gene.20185] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Cre-mediated conditional gene targeting has been shown to be successful in many cell and tissue types. However, gene recombination in the uterus with heterogeneous cell types by Cre activation is not yet well established. Using recombinant adenoviruses expressing a functional Cre (ADV-Cre) and ROSA26 reporter mice, we show here that ADV-Cre infused intraluminally in a small volume (10 microl) conditionally excises the loxP site, resulting in lacZ expression in uterine luminal epithelial cells without significantly affecting pregnancy. In contrast, a similar intraluminal infusion of ADV-Cre in a larger volume (50 microl) damages the normal architecture and integrity of the luminal epithelium, inducing gene recombination in the underneath stromal cells, with disruption of pregnancy. Further, decidualizing stromal cells at the implantation sites can be targeted by ADV-Cre after intravenous administration on days 5-6. This route of administration also elicits Cre activity in other tissues, including the liver, spleen, ovary, and, more remarkably, in the adrenal cortex. These findings demonstrate the feasibility of achieving conditional expression or deletion of specific genes in uterine cells at desired times and physiological states.
Collapse
Affiliation(s)
- Haibin Wang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Huirong Xie
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Hao Zhang
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sanjoy K. Das
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee
| | - Sudhansu K. Dey
- Department of Pediatrics, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Cell & Developmental Biology, Vanderbilt University Medical Center, Nashville, Tennessee
- Department of Pharmacology, Vanderbilt University Medical Center, Nashville, Tennessee
- Correspondence to: Sudhansu K. Dey, Department of Pediatrics, Division of Reproductive and Developmental Biology, Vanderbilt University Medical Center, MCN-D4100, Nashville, TN 37232-2678,
| |
Collapse
|
35
|
Raschperger E, Thyberg J, Pettersson S, Philipson L, Fuxe J, Pettersson RF. The coxsackie- and adenovirus receptor (CAR) is an in vivo marker for epithelial tight junctions, with a potential role in regulating permeability and tissue homeostasis. Exp Cell Res 2006; 312:1566-80. [PMID: 16542650 DOI: 10.1016/j.yexcr.2006.01.025] [Citation(s) in RCA: 104] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2005] [Revised: 01/16/2006] [Accepted: 01/18/2006] [Indexed: 10/24/2022]
Abstract
The coxsackie- and adenovirus receptor (CAR) is a transmembrane protein belonging to the immunoglobulin superfamily. The function of CAR as a virus receptor has been extensively analyzed, while its physiological role and expression pattern in adult tissues have remained less clear. CAR associates with epithelial tight junctions in vitro and mediates cell-cell adhesion. Using a set of affinity-purified antibodies, we show that CAR is predominantly expressed in epithelial cells lining the body cavities in adult mice, where it specifically co-localizes with the tight junction components ZO-1 and occludin. Notably, CAR could not be detected in endothelial cells of the vasculature, including brain capillaries. CAR expression correlated positively with the maturity of tight junctions and inversely with permeability. With a few exceptions, the two known CAR isoforms were co-expressed in most epithelial cells analyzed. A CAR mutant lacking the intracellular tail over-expressed in transgenic mice was diffusely localized over the plasma membrane, showing the importance of this domain for correct subcellular localization in vivo. We conclude that CAR is localized to epithelial tight junctions in vivo where it may play a role in the regulation of epithelial permeability and tissue homeostasis.
Collapse
|
36
|
Korn WM, Macal M, Christian C, Lacher MD, McMillan A, Rauen KA, Warren RS, Ferrell L. Expression of the coxsackievirus- and adenovirus receptor in gastrointestinal cancer correlates with tumor differentiation. Cancer Gene Ther 2006; 13:792-7. [PMID: 16628228 DOI: 10.1038/sj.cgt.7700947] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Modified adenoviruses represent a new approach to treatment of gastrointestinal cancer. However, their uptake by cells in many cases requires the major receptor for adenoviruses, the coxsackievirus and adenovirus receptor (CAR). Thus, lack of CAR expression is a potential cause of intrinsic resistance of tumor cells to this type of treatment. To evaluate this, we studied the localization of CAR protein in normal and malignant gastrointestinal tissues. In normal tissues, CAR was concentrated at sites of cell-cell interaction, in particular at the apico-lateral cellular surface. Expression was particularly strong around bile and pancreatic ducts, which is in agreement with CAR's physiological function as a tight-junction protein. In GI malignancies (esophageal, pancreatic, colorectal and liver cancer), expression of the receptor varied substantially. Loss of CAR expression at cell-cell junction was evident in many samples. A significant correlation between CAR expression and histological grade was found, with moderately to poorly differentiated tumors most frequently demonstrating loss or reduction of CAR expression. These data indicate that CAR expression is frequently altered in gastrointestinal malignancy, potentially reducing the efficacy of adenovirus-based therapies.
Collapse
Affiliation(s)
- W M Korn
- Division of Gastroenterology, Department of Medicine, University of California San Francisco, San Francisco, CA 94143, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Verhaagh S, de Jong E, Goudsmit J, Lecollinet S, Gillissen G, de Vries M, van Leuven K, Que I, Ouwehand K, Mintardjo R, Weverling GJ, Radošević K, Richardson J, Eloit M, Lowik C, Quax P, Havenga M. Human CD46-transgenic mice in studies involving replication-incompetent adenoviral type 35 vectors. J Gen Virol 2006; 87:255-265. [PMID: 16432010 DOI: 10.1099/vir.0.81293-0] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Wild-type strains of mice do not express CD46, a high-affinity receptor for human group B adenoviruses including type 35. Therefore, studies performed to date in mice using replication-incompetent Ad35 (rAd35) vaccine carriers may underestimate potency or result in altered vector distribution. Here, it is reported that CD46 transgenic mice (MYII-strain) express CD46 in all major organs and that it functions as a receptor for rAd35 vectors. Similar to monkeys and humans, MYII mice highly express CD46 in their lungs and kidneys and demonstrate low expression in muscle. Upon intravenous administration, rAd35 vector genomes as well as expression are detected in lungs of MYII mice, in contrast to wild-type littermates. Expression was predominantly detected in lung epithelial cells. Upon intramuscular administration, the initial level of luciferase expression is higher in MYII mice as compared with wild-type littermates, in spite of the fact that CD46 expression is low in muscle of MYII mice. The higher level of expression in muscle of MYII mice results in prolonged gene expression as assessed by CCD camera imaging for luciferase activity. Finally, a significant dose-sparing effect in MYII mice as compared with wild-type littermates on anti-SIVgag CD8+ T-cell induction following intramuscular vaccination with an rA35.SIVgag vaccine was observed. This dose-sparing effect was also observed when reinfusing dendritic cells derived from MYII mice after exposure to rAd35.SIVgag vaccine as compared with rAd35.SIVgag exposed dendritic cells from wild-type littermates. It was concluded that MYII mice represent an interesting preclinical model to evaluate potency and safety of rAd35 vectors.
Collapse
Affiliation(s)
- Sandra Verhaagh
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | - Esmeralda de Jong
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | - Jaap Goudsmit
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | | | - Gert Gillissen
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | | | | | - Ivo Que
- Department of Endocrinology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Krista Ouwehand
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | - Ratna Mintardjo
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| | | | | | | | - Marc Eloit
- Ecole National Veterinaire d'Alfort, Alfort, France
| | - Clemens Lowik
- Department of Endocrinology, Leiden University Medical Centre, Leiden, The Netherlands
| | - Paul Quax
- Gaubius Laboratory, TNO Leiden, The Netherlands
| | - Menzo Havenga
- Crucell Holland BV, Archimedesweg 4, 2333 CN Leiden, The Netherlands
| |
Collapse
|
38
|
Lacher MD, Tiirikainen MI, Saunier EF, Christian C, Anders M, Oft M, Balmain A, Akhurst RJ, Korn WM. Transforming growth factor-beta receptor inhibition enhances adenoviral infectability of carcinoma cells via up-regulation of Coxsackie and Adenovirus Receptor in conjunction with reversal of epithelial-mesenchymal transition. Cancer Res 2006; 66:1648-57. [PMID: 16452224 DOI: 10.1158/0008-5472.can-05-2328] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Expression of the Coxsackie and Adenovirus Receptor (CAR) is frequently reduced in carcinomas, resulting in decreased susceptibility of such tumors to infection with therapeutic adenoviruses. Because CAR participates physiologically in the formation of tight-junction protein complexes, we examined whether molecular mechanisms known to down-regulate cell-cell adhesions cause loss of CAR expression. Transforming growth factor-beta (TGF-beta)-mediated epithelial-mesenchymal transition (EMT) is a phenomenon associated with tumor progression that is characterized by loss of epithelial-type cell-cell adhesion molecules (including E-cadherin and the tight junction protein ZO-1), gain of mesenchymal biochemical markers, such as fibronectin, and acquisition of a spindle cell phenotype. CAR expression is reduced in tumor cells that have undergone EMT in response to TGF-beta. This down-regulation results from repression of CAR gene transcription, whereas altered RNA stability and increased proteasomal protein degradation play no role. Loss of CAR expression in response to TGF-beta is accompanied by reduced susceptibility to adenovirus infection. Indeed, treatment of carcinoma cells with LY2109761, a specific pharmacologic inhibitor of TGF-beta receptor types I and II kinases, resulted in increased CAR RNA and protein levels as well as improved infectability with adenovirus. This was observed in cells induced to undergo EMT by addition of exogenous TGF-beta and in those that were transformed by endogenous autocrine/paracrine TGF-beta. These findings show down-regulation of CAR in the context of EMT and suggest that combination of therapeutic adenoviruses and TGF-beta receptor inhibitors could be an efficient anticancer strategy.
Collapse
Affiliation(s)
- Markus D Lacher
- Division of Gastroenterology and Hematology/Oncology, University of California-San Francisco Comprehensive Cancer Center, San Francisco, CA 94143, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Everts M, Kim-Park SA, Preuss MA, Passineau MJ, Glasgow JN, Pereboev AV, Mahasreshti PJ, Grizzle WE, Reynolds PN, Curiel DT. Selective induction of tumor-associated antigens in murine pulmonary vasculature using double-targeted adenoviral vectors. Gene Ther 2005; 12:1042-8. [PMID: 15789059 DOI: 10.1038/sj.gt.3302491] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Targeted therapies directed to tumor-associated antigens are being investigated for the treatment of cancer. However, there are few suitable animal models for testing the ability to target these tumor markers. Therefore, we have exploited mice transgenic for the human coxsackie and adenovirus receptor (hCAR) to establish a new model for transient expression of human tumor-associated antigens in the pulmonary vasculature. Systemic administration of Ad in hCAR mice resulted in an increase in transgene expression in the lungs compared to wild-type mice, as determined using a luciferase reporter gene. To reduce transgene expression in the liver, the predominant organ of ectopic Ad localization and transgene expression following systemic administration, we utilized the endothelial-specific flt-1 promoter, which resulted in a further increased lung-to-liver ratio of luciferase expression. Administration of an adenoviral vector encoding the tumor-associated antigen carcinoembryonic antigen (CEA) under transcriptional control of the flt-1 promoter resulted in selective expression of this antigen in the pulmonary vasculature of hCAR mice. Feasibility of targeting to expressed CEA was subsequently demonstrated using adenoviral vectors preincubated with a bifunctional adapter molecule recognizing this tumor-associated antigen, thus demonstrating utility of this transient transgenic animal model.
Collapse
Affiliation(s)
- M Everts
- Division of Human Gene Therapy, Department of Medicine, and the Gene Therapy Center, University of Alabama at Birmingham, Birmingham, AL 35294-2172, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Affiliation(s)
- Yuanming Zhang
- Division of Infectious Diseases, Children's Hospital of Philadelphia, PA, USA
| | | |
Collapse
|
41
|
Jerebtsova M, Liu XH, Ye X, Ray PE. Adenovirus-mediated gene transfer to glomerular cells in newborn mice. Pediatr Nephrol 2005; 20:1395-400. [PMID: 16133067 DOI: 10.1007/s00467-005-1882-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/13/2004] [Revised: 12/29/2004] [Accepted: 12/29/2004] [Indexed: 10/25/2022]
Abstract
The systemic delivery of recombinant adenoviral (rAd) vectors to renal glomeruli has been problematic due to the rapid clearance of the circulating virus by the liver. We have previously shown that prolonged retention of rAd vectors in the circulation by liver bypass improves the transduction of renal glomerular cells in adult mice and rats. This study was done to determine whether newborn mice have a delayed clearance of rAd vectors from the circulation and a more efficient transduction of glomerular cells after a systemic injection of rAd vectors. Newborn (1 day old) and adult (6 months old) C57Bl6/J mice ( n = 20 in each group) were injected with rAd vectors carrying the lacZ gene (rAd.lacZ) through the retro-orbital venous plexus (2 x 10(9 )particles/g body weight). The renal expression of Coxsackie and Adenoviral Receptors (CAR) and lacZ gene were evaluated at different time points by Western blots, immunohistochemistry, beta-galactosidase staining, enzyme assay activity, and RT-PCR studies in newborn and adult mice. The clearance rate of rAd.lacZ was significantly delayed in newborn mice, and the concentration of circulating virus in these mice was almost ten times higher than that in adult mice. Newborn kidneys showed increased expression of CAR, predominately localized in glomerular cells. These findings were associated with an efficient gene transfer of the lacZ gene into glomeruli and tubules of newborn mice. This study demonstrates for the first time the feasibility of using systemic intravenous injections of rAd vectors to express foreign genes in developing glomeruli of young mice.
Collapse
Affiliation(s)
- Marina Jerebtsova
- Center for Genetic Medicine Research, Children's Research Institute, Children's National Medical Center, Washington, DC 20010, USA
| | | | | | | |
Collapse
|
42
|
Bao Y, Peng W, Verbitsky A, Chen J, Wu L, Rauen KA, Sawicki JA. Human coxsackie adenovirus receptor (CAR) expression in transgenic mouse prostate tumors enhances adenoviral delivery of genes. Prostate 2005; 64:401-7. [PMID: 15761871 DOI: 10.1002/pros.20265] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
BACKGROUND Transgenic mice that recapitulate the progression of human diseases are potentially useful models for testing the effectiveness of new therapeutic strategies. Their use in pre-clinical testing of adenovirally-delivered gene therapies, however, is limited because of restricted cell surface expression of Coxsackie adenovirus receptor (CAR) in mice. METHODS To develop a more suitable transgenic mouse model for testing adenoviral-based gene therapies for prostate cancer, we generated prostate specific antigen/human CAR (PSA/hCAR) transgenic mice in which a chimeric enhancer/promoter sequence of the human PSA gene drives expression of a functional hCAR coding sequence. RESULTS Expression of an adenovirally-delivered luciferase reporter gene in prostate tumor cells in bigenic mice (PSA/hCAR + TRAMP) was enhanced compared to the level in tumor cells lacking the PSA/hCAR transgene. CONCLUSIONS Breeding PSA/hCAR mice to existing transgenic mouse models for prostate cancer (e.g., TRAMP) results in improved mouse models for testing adenovirally-delivered therapeutic genes.
Collapse
Affiliation(s)
- Yunhua Bao
- Lankenau Institute for Medical Research, Wynnewood, Pennsylvania 19096, USA
| | | | | | | | | | | | | |
Collapse
|
43
|
Isoform-specific expression of the Coxsackie and adenovirus receptor (CAR) in neuromuscular junction and cardiac intercalated discs. BMC Cell Biol 2004; 5:42. [PMID: 15533241 PMCID: PMC533869 DOI: 10.1186/1471-2121-5-42] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2004] [Accepted: 11/08/2004] [Indexed: 12/22/2022] Open
Abstract
Background The Coxsackie and adenovirus receptor (CAR) has a restricted expression pattern in the adult. In skeletal muscle, although CAR is expressed in immature fibers, its transcript levels are barely detectable in mature muscle. This is in contrast to the robust expression observed in the heart. However, both heart and skeletal muscle are susceptible to infection with the Coxsackie B virus which utilizes primarily CAR for cellular internalization. The specific point of viral entry in skeletal and heart muscle remains unknown. Results Using antibodies directed against the extracellular and the cytoplasmic domains of CAR, we show CAR in normal human and mouse skeletal muscle to be a novel component of the neuromuscular junction. In cardiac muscle, CAR immunoreactivity is observed at the level of intercalated discs. We demonstrate a single isoform of CAR to be expressed exclusively at the human neuromuscular junction whereas both predominant CAR isoforms are expressed at the intercalated discs of non-diseased human heart. Conclusion The localization of CAR to these important junctional complexes suggests that CAR may play both a structural and a regulatory role in skeletal and cardiac muscle, and that these complexes may serve as a point of entry for Coxsackie B virus.
Collapse
|
44
|
Le LP, Everts M, Dmitriev IP, Davydova JG, Yamamoto M, Curiel DT. Fluorescently Labeled Adenovirus with pIX-EGFP for Vector Detection. Mol Imaging 2004; 3:105-16. [PMID: 15296675 DOI: 10.1162/15353500200404100] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Abstract
Adenoviruses are extensively studied in terms of their use as gene therapy vectors and pathogenesis. These vectors have been targeted on both transcriptional and transductional levels to achieve cell-specific gene delivery. Current detection strategies, including reporter gene expression, viral component detection, and vector labeling with fluorophores, have been applied to analyze adenoviral vectors; however, these methods are inadequate for assessing transductional targeting. As an alternative to conventional vector detection techniques, we developed a specific genetic labeling system whereby an adenoviral vector incorporates a fusion between capsid protein IX and EGFP. DNA packaging and thermostability were marginally hampered by the modification while DNA replication, cytopathic effect, and CAR-dependent binding were not affected. The fluorescent label was associated with the virus capsid and conferred a fluorescent property useful in detecting adenoviral particles in flow cytometry, tracking, and tissue sections. We believe our genetic adenovirus labeling system has important implications for vector development, detecting adenovirus vectors in targeting schemes, and studying adenovirus biology. In addition, this technique has potential utility for dynamic monitoring of adenovirus replication and spread.
Collapse
Affiliation(s)
- Long P Le
- University of Alabama, Birmingham, USA
| | | | | | | | | | | |
Collapse
|
45
|
Philipson L, Pettersson RF. The Coxsackie-Adenovirus Receptor—A New Receptor in the Immunoglobulin Family Involved in Cell Adhesion. Curr Top Microbiol Immunol 2004; 273:87-111. [PMID: 14674599 DOI: 10.1007/978-3-662-05599-1_3] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The physiological and cell biological aspects of the Coxsackie-Adenovirus Receptor (CAR) is discussed in this review. The receptor obviously recognizes the group C adenoviruses in vivo, but also fibers from other groups except group B in vitro. The latter viruses seem to utilize a different receptor. The receptor accumulates at, or close to, the tight junction in polarized epithelial cells and probably functions as a cell-cell adhesion molecule. The cytoplasmic tail of the receptor is not required for virus attachment and uptake. Although there is a correlation between CAR and uptake of adenoviruses in several human tumor cells, evidence of an absolute requirement for integrins has not been forthcoming. The implication of these findings for adenovirus gene therapy is discussed.
Collapse
Affiliation(s)
- L Philipson
- Department of Cell and Molecular Biology, Karolinska Institute, Box 285, 17177 Stockholm, Sweden.
| | | |
Collapse
|
46
|
Huang KC, Altinoz M, Wosik K, Larochelle N, Koty Z, Zhu L, Holland PC, Nalbantoglu J. Impact of the coxsackie and adenovirus receptor (CAR) on glioma cell growth and invasion: Requirement for the C-terminal domain. Int J Cancer 2004; 113:738-45. [PMID: 15499626 DOI: 10.1002/ijc.20623] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Expression of the coxsackie and adenovirus receptor (CAR) is downregulated in malignant glioma cell lines and is barely detectable in high-grade primary astrocytoma (glioblastoma multiforme). We determined the effect of forced CAR expression on the invasion and growth of the human glioma cell line U87-MG, which does not express any CAR. Although retrovirally mediated expression of full-length CAR in U87-MG cells did not affect monolayer growth in vitro, it did reduce glioma cell invasion in a 3-dimensional spheroid model. Furthermore, in xenograft experiments, intracerebral implantation of glioma cells expressing full-length CAR resulted in tumors with a significantly reduced volume compared to tumors generated by control vector-transduced U87-MG cells. In contrast, U87-MG cells expressing transmembrane CAR with a deletion of the entire cytoplasmic domain (except for the first 2 intracellular juxtamembrane cysteine amino acids) had rates of invasion and tumor growth that were similar to those of the control cells. This difference in behavior between the 2 forms of CAR was not due to improper cell surface localization of the cytoplasmically deleted CAR as determined by comparable immunostaining of unpermeabilized cells, equivalent adenoviral transduction of the cells and similar extent of fractionation into lipid-rich domains. Taken together, these results suggest that the decrease or loss of CAR expression in malignant glioma may confer a selective advantage in growth and invasion to these tumors.
Collapse
Affiliation(s)
- Kuo-Cheng Huang
- Department of Neurology and Neurosurgery, McGill University and the Montreal Neurological Institute, Montreal, Quebec, Canada
| | | | | | | | | | | | | | | |
Collapse
|
47
|
Hurez V, Hautton RD, Oliver J, Matthews RJ, Weaver CK. Gene delivery into primary T cells: overview and characterization of a transgenic model for efficient adenoviral transduction. Immunol Res 2003; 26:131-41. [PMID: 12403352 DOI: 10.1385/ir:26:1-3:131] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Technologies for transfer of exogenous genes into primary T cells have been limited until recently. The introduction of new approaches for gene transfer via different viral vectors has expanded the options for genetic manipulation of primary T cells and has provided powerful tools for studies of T cell activation and differentiation. We provide a brief overview of the systems currently available and contrast the advantages and disadvantages of each. We also describe a new transgenic model that enables highly efficient gene delivery into primary T cells by nonreplicating adenoviral vectors.
Collapse
Affiliation(s)
- Vincent Hurez
- Department of Pathology, University of Alabama at Birmingham, 35233-2170, USA
| | | | | | | | | |
Collapse
|
48
|
Turturro F. Recombinant adenovirus-mediated cytotoxic gene therapy of lymphoproliferative disorders: is CAR important for the vector to ride? Gene Ther 2003; 10:100-4. [PMID: 12571638 DOI: 10.1038/sj.gt.3301842] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The literature has seen an incredible booming of publications related to the use of recombinant adenoviruses as therapeutic tools for lymphoproliferative disorders over the last decade. Several approaches of adenovirus-mediated gene expression have been used to transfect cell lines that are derived from lymphoid tumors and would have otherwise been refractory to other transfection methods. The identification of high-affinity receptor for human adenoviruses serotype 2 and 5, the coxsackie-adenovirus receptor (CAR), has raised the question about its relevance for the efficacy of recombinant adenovirus-mediated gene therapy. We review published studies that have analyzed the use of recombinant adenovirus vectors expressing cytotoxic genes for gene therapy in lymphomas, chronic lymphocytic leukemia and multiple myeloma. For simplicity, we group all these diseases under the term lymphoproliferative disorders. We analyze the use of recombinant adenovirus-mediated cytotoxicity by assessing the importance of the biochemical and intrinsic signaling pathways interacting with the products of the exogenous viral-mediated expression. Ultimately, we discuss studies that have been finalized to by-pass the limitations of the biodistribution of CAR by modifying or targeting adenovirus to other membrane proteins in cells derived from lymphoproliferative disorders.
Collapse
Affiliation(s)
- F Turturro
- Department of Medicine, Feist-Weiller Cancer Center, Louisiana State University Health Sciences Center, 1501 Kings Highway, Shreveport, LA 71130-3932, USA
| |
Collapse
|
49
|
Yant SR, Ehrhardt A, Mikkelsen JG, Meuse L, Pham T, Kay MA. Transposition from a gutless adeno-transposon vector stabilizes transgene expression in vivo. Nat Biotechnol 2002; 20:999-1005. [PMID: 12244327 DOI: 10.1038/nbt738] [Citation(s) in RCA: 150] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2002] [Accepted: 07/16/2002] [Indexed: 11/08/2022]
Abstract
A major limitation of adenovirus-mediated gene therapy for inherited diseases is the instability of transgene expression in vivo, which originates at least in part from the loss of the linear, extrachromosomal vector genomes. Herein we describe the production of a gene-deleted adenovirus-transposon vector that stably maintains virus-encoded transgenes in vivo through integration into host cell chromosomes. This system utilizes a donor transposon vector that undergoes Flp-mediated recombination and excision of its therapeutic payload in the presence of the Flp and Sleeping Beauty recombinases. Systemic in vivo delivery of this system resulted in efficient generation of transposon circles and stable transposase-mediated integration in mouse liver. Somatic integration was sufficient to maintain therapeutic levels of human coagulation Factor IX for more than six months in mice undergoing extensive liver proliferation. These vectors combine the versatility of adenoviral vectors with the integration capabilities of a eukaryotic DNA transposon and should prove useful in the treatment of genetic diseases.
Collapse
Affiliation(s)
- Stephen R Yant
- Department of Pediatrics and Genetics, Stanford University School of Medicine, Stanford, CA 94305-5208, USA
| | | | | | | | | | | |
Collapse
|
50
|
Bhattacharya D, Logue EC, Bakkour S, DeGregori J, Sha WC. Identification of gene function by cyclical packaging rescue of retroviral cDNA libraries. Proc Natl Acad Sci U S A 2002; 99:8838-43. [PMID: 12084928 PMCID: PMC124385 DOI: 10.1073/pnas.132274799] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023] Open
Abstract
Genes regulating responses in mammalian cells are often difficult to identify by functional cloning strategies limited to a single round of selection. Here we describe a strategy, cyclical packaging rescue (CPR), which allows rapid recovery and retransmission of retroviral cDNA libraries. CPR can be used not only with immortalized cell lines such as fibroblasts and Jurkat T cells, but also with primary B lymphocytes, which can be maintained only in short-term cultures. CPR allows for multiple rounds of selection and enrichment to identify cDNAs regulating responses in mammalian cells. Using CPR, five cDNAs were functionally cloned, which conferred protection against tumor necrosis factor alpha (TNFalpha)-induced apoptosis in RelA(-/-) fibroblasts. Three of the genes, RelA, cellular FLICE-like inhibitory protein (c-FLIP), and a dominant-negative mutant of TNF receptor 1 arising through CPR afforded strong protection against apoptosis. Two of the genes identified, Dbs and Fas-associated death domain protein (FADD), previously identified as a proapoptotic molecule, afforded partial protection against TNFalpha-induced apoptosis. These results suggest that CPR is a versatile method that permits functional identification of both wild-type and dominant-negative gene products that regulate cellular responses.
Collapse
Affiliation(s)
- Deepta Bhattacharya
- Department of Molecular and Cell Biology, University of California, Berkeley, CA 94720, USA
| | | | | | | | | |
Collapse
|